Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (71)

Search Parameters:
Keywords = suicide gene therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 4580 KB  
Review
The Rise of Fine-Tuned CAR-Based Therapies Against Acute Myeloid Leukemia
by Alejandro Segura Tudela, Ron Geller, Bruno Paiva, Sara Carmen Torres Sánchez, Elisa González Romero, Pilar Lloret Madrid, Pedro Chorão, Javier de la Rubia, Pau Montesinos and Manuel Guerreiro
Cancers 2025, 17(24), 3892; https://doi.org/10.3390/cancers17243892 - 5 Dec 2025
Viewed by 961
Abstract
Acute myeloid leukemia (AML) is a heterogeneous and aggressive hematologic malignancy with poor prognosis despite multiple available therapies. While chimeric antigen receptor (CAR) T-cell therapy has transformed the treatment of B-cell malignancies, its application in AML has been limited by early relapses and [...] Read more.
Acute myeloid leukemia (AML) is a heterogeneous and aggressive hematologic malignancy with poor prognosis despite multiple available therapies. While chimeric antigen receptor (CAR) T-cell therapy has transformed the treatment of B-cell malignancies, its application in AML has been limited by early relapses and severe toxicities. Unlike B-cell antigens, most AML-associated surface antigens are also expressed on healthy hematopoietic stem and progenitor cells, creating significant risks of on-target/off-tumor toxicity and prolonged myeloablation. To address the scarcity of AML-specific targets, several innovative CAR strategies have been developed to enhance precision, safety, and efficacy. Logic-gated CARs improve selectivity through dual-antigen recognition or conditional activation. Drug-inducible and transient expression systems, as well as pharmacologic or suicide switches, enable controlled modulation or elimination of CAR cells to reduce toxicity. Adapter CAR platforms allow real-time, flexible targeting, while engineered modulation of gene expression or cytokine secretion enhances persistence and antitumor activity. Finally, alternative immune cells, including natural killer (NK) cells and macrophages, provide versatile platforms that may overcome limitations of conventional T-cell therapies, such as fratricide or challenges in allogeneic use. This review provides a comprehensive overview of these emerging CAR approaches, highlighting their advantages, limitations, and potential to expand immunotherapeutic strategies for AML. Full article
(This article belongs to the Special Issue Advances in T-Cell Immunotherapy for Acute Myeloid Leukemia)
Show Figures

Figure 1

27 pages, 4850 KB  
Review
Unlocking the Role of OCT4 in Cancer Lineage Plasticity: A Cross-Cancer Perspective with an Emphasis on Prostate Cancer
by Mohammad Esfini Farahani, Yanquan Zhang, Amos Olalekan Akinyemi, Fatemeh Seilani, Md Rakibul Alam and Xiaoqi Liu
Biomedicines 2025, 13(7), 1642; https://doi.org/10.3390/biomedicines13071642 - 4 Jul 2025
Viewed by 2327
Abstract
Prostate cancer (PCa) is a highly heterogeneous disease, with castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) representing its most aggressive and therapy-resistant forms. Emerging evidence indicates that lineage plasticity—driven by key transcription factors such as Octamer Binding Factor 4 (OCT4)—plays a [...] Read more.
Prostate cancer (PCa) is a highly heterogeneous disease, with castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) representing its most aggressive and therapy-resistant forms. Emerging evidence indicates that lineage plasticity—driven by key transcription factors such as Octamer Binding Factor 4 (OCT4)—plays a crucial role in therapeutic resistance and disease progression. OCT4, in coordination with SOX2 and NANOG, acts as a master regulator of stemness and is frequently upregulated in prostate cancer stem cells (PCSCs). This upregulation contributes to tumor initiation, metastasis, and resistance to both androgen deprivation therapy (ADT) and chemotherapy. In this review, we explore the role of OCT4 in mediating lineage plasticity in prostate cancer, with particular emphasis on its involvement in treatment resistance and neuroendocrine differentiation. We also examine therapeutic strategies aimed at targeting OCT4 directly, such as microRNA-mediated suppression, small-molecule inhibitors, and suicide gene therapy, as well as indirect approaches that modulate OCT4 expression via FGFR and NF-κB signaling pathways. While these strategies offer promising avenues, challenges such as adaptive resistance and the intricate signaling networks within PCSCs remain significant hurdles. A deeper understanding of the molecular mechanisms underlying OCT4-driven plasticity may pave the way for novel therapeutic approaches and improved outcomes in advanced prostate cancer. Full article
(This article belongs to the Special Issue Molecular Biomarkers of Tumors: Advancing Genetic Studies)
Show Figures

Figure 1

19 pages, 277 KB  
Review
Clinical Trials of Cancer Immunogene Therapies in Companion Animals: An Update (2017–2024)
by Gerardo C. Glikin and Liliana M. E. Finocchiaro
Vet. Sci. 2025, 12(4), 329; https://doi.org/10.3390/vetsci12040329 - 3 Apr 2025
Cited by 2 | Viewed by 4421
Abstract
This review summarizes the findings of veterinary clinical trials on immunogene therapy published between 2017 and 2024. Various tumor types, including melanoma (canine and feline), mastocytoma (canine), mammary adenocarcinoma (canine), osteosarcoma (canine), and sarcoid (equine), were treated using diverse strategies. Non-viral vectors were [...] Read more.
This review summarizes the findings of veterinary clinical trials on immunogene therapy published between 2017 and 2024. Various tumor types, including melanoma (canine and feline), mastocytoma (canine), mammary adenocarcinoma (canine), osteosarcoma (canine), and sarcoid (equine), were treated using diverse strategies. Non-viral vectors were predominantly used to deliver genes encoding tumor-associated antigens, cytokines, or suicide enzymes. Among these non-viral methods, electrotransfer was the most commonly employed technique for introducing therapeutic genes into cells. Generally, these procedures resulted in minimal or no adverse side effects, and treated animals often showed significant improvements, such as enhanced quality of life, delayed or suppressed tumor recurrence or metastasis, and increased survival times. Some of these innovative approaches hold great potential as adjunct therapies to standard treatments. The promising outcomes from immunogene therapy studies in companion animals strongly support their application in veterinary oncology and provide valuable preclinical data (including safety assessments and proof-of-concept studies) for analogous human clinical trials. Full article
(This article belongs to the Special Issue Advanced Therapy in Companion Animals)
26 pages, 2544 KB  
Review
Immortalization of Mesenchymal Stem Cells for Application in Regenerative Medicine and Their Potential Risks of Tumorigenesis
by Natsuki Yamaguchi, Eri Horio, Jukito Sonoda, Miu Yamagishi, Satomi Miyakawa, Fumihiro Murakami, Hideaki Hasegawa, Yasuhiro Katahira, Izuru Mizoguchi, Yasuyuki Fujii, Daichi Chikazu and Takayuki Yoshimoto
Int. J. Mol. Sci. 2024, 25(24), 13562; https://doi.org/10.3390/ijms252413562 - 18 Dec 2024
Cited by 21 | Viewed by 4650
Abstract
Regenerative medicine utilizes stem cells to repair damaged tissues by replacing them with their differentiated cells and activating the body’s inherent regenerative abilities. Mesenchymal stem cells (MSCs) are adult stem cells that possess tissue repair and regenerative capabilities and immunomodulatory properties with a [...] Read more.
Regenerative medicine utilizes stem cells to repair damaged tissues by replacing them with their differentiated cells and activating the body’s inherent regenerative abilities. Mesenchymal stem cells (MSCs) are adult stem cells that possess tissue repair and regenerative capabilities and immunomodulatory properties with a much lower risk of tumorigenicity, making them a focus of numerous clinical trials worldwide. MSCs primarily exert their therapeutic effects through paracrine effects via secreted factors, such as cytokines and exosomes. This has led to increasing interest in cell-free therapy, where only the conditioned medium (also called secretome) from MSC cultures is used for regenerative applications. However, MSCs face certain limitations, including cellular senescence, scarcity, donor heterogeneity, complexity, short survival post-implantation, and regulatory and ethics hurdles. To address these challenges, various types of immortalized MSCs (ImMSCs) capable of indefinite expansion have been developed. These cells offer significant promise and essential tools as a reliable source for both cell-based and cell-free therapies with the aim of translating them into practical medicine. However, the process of immortalization, often involving the transduction of immortalizing genes, poses potential risks of genetic instability and resultant malignant transformation. Cell-free therapy is particularly attractive, as it circumvents the risks of tumorigenicity and ethical concerns associated with live cell therapies. Rigorous safety tests, such as monitoring chromosomal abnormalities, are critical to ensure safety. Technologies like inducible or suicide genes may allow for the controlled proliferation of MSCs and induce apoptosis after their therapeutic task is completed. This review highlights recent advancements in the immortalization of MSCs and the associated risks of tumorigenesis. Full article
(This article belongs to the Special Issue State-of-the-Art Cancer Immunotherapies—2nd Edition)
Show Figures

Figure 1

20 pages, 798 KB  
Review
Optimizing Pancreatic Cancer Therapy: The Promise of Immune Stimulatory Oncolytic Viruses
by Shivani Thoidingjam, Aseem Rai Bhatnagar, Sushmitha Sriramulu, Farzan Siddiqui and Shyam Nyati
Int. J. Mol. Sci. 2024, 25(18), 9912; https://doi.org/10.3390/ijms25189912 - 13 Sep 2024
Cited by 7 | Viewed by 5180
Abstract
Pancreatic cancer presents formidable challenges due to rapid progression and resistance to conventional treatments. Oncolytic viruses (OVs) selectively infect cancer cells and cause cancer cells to lyse, releasing molecules that can be identified by the host’s immune system. Moreover, OV can carry immune-stimulatory [...] Read more.
Pancreatic cancer presents formidable challenges due to rapid progression and resistance to conventional treatments. Oncolytic viruses (OVs) selectively infect cancer cells and cause cancer cells to lyse, releasing molecules that can be identified by the host’s immune system. Moreover, OV can carry immune-stimulatory payloads such as interleukin-12, which when delivered locally can enhance immune system-mediated tumor killing. OVs are very well tolerated by cancer patients due to their ability to selectively target tumors without affecting surrounding normal tissues. OVs have recently been combined with other therapies, including chemotherapy and immunotherapy, to improve clinical outcomes. Several OVs including adenovirus, herpes simplex viruses (HSVs), vaccinia virus, parvovirus, reovirus, and measles virus have been evaluated in preclinical and clinical settings for the treatment of pancreatic cancer. We evaluated the safety and tolerability of a replication-competent oncolytic adenoviral vector carrying two suicide genes (thymidine kinase, TK; and cytosine deaminase, CD) and human interleukin-12 (hIL12) in metastatic pancreatic cancer patients in a phase 1 trial. This vector was found to be safe and well-tolerated at the highest doses tested without causing any significant adverse events (SAEs). Moreover, long-term follow-up studies indicated an increase in the overall survival (OS) in subjects receiving the highest dose of the OV. Our encouraging long-term survival data provide hope for patients with advanced pancreatic cancer, a disease that has not seen a meaningful increase in OS in the last five decades. In this review article, we highlight several preclinical and clinical studies and discuss future directions for optimizing OV therapy in pancreatic cancer. We envision OV-based gene therapy to be a game changer in the near future with the advent of newer generation OVs that have higher specificity and selectivity combined with personalized treatment plans developed under AI guidance. Full article
(This article belongs to the Special Issue Virus Engineering and Applications: 2nd Edition)
Show Figures

Figure 1

22 pages, 7938 KB  
Article
The Histogenetic Origin of Malignant Cells Predicts Their Susceptibility towards Synthetic Lethality Utilizing the TK.007 System
by Fabian Bernhard Pallasch, Vera Freytag, Malte Kriegs, Dennis Gatzemeier, Thomas Mair, Hannah Voss, Kristoffer Riecken, Mona Dawood, Boris Fehse, Thomas Efferth, Hartmut Schlüter and Udo Schumacher
Cancers 2024, 16(12), 2278; https://doi.org/10.3390/cancers16122278 - 19 Jun 2024
Viewed by 1949
Abstract
Background: Remarkable differences exist in the outcome of systemic cancer therapies. Lymphomas and leukemias generally respond well to systemic chemotherapies, while solid cancers often fail. We engineered different human cancer cells lines to uniformly express a modified herpes simplex virus thymidine kinase TK.007 [...] Read more.
Background: Remarkable differences exist in the outcome of systemic cancer therapies. Lymphomas and leukemias generally respond well to systemic chemotherapies, while solid cancers often fail. We engineered different human cancer cells lines to uniformly express a modified herpes simplex virus thymidine kinase TK.007 as a suicide gene when ganciclovir (GCV) is applied, thus in theory achieving a similar response in all cell lines. Methods: Fifteen different cell lines were engineered to express the TK.007 gene. XTT-cell proliferation assays were performed and the IC50-values were calculated. Functional kinome profiling, mRNA sequencing, and bottom-up proteomics analysis with Ingenuity pathway analysis were performed. Results: GCV potency varied among cell lines, with lymphoma and leukemia cells showing higher susceptibility than solid cancer cells. Functional kinome profiling implies a contribution of the SRC family kinases and decreased overall kinase activity. mRNA sequencing highlighted alterations in the MAPK pathways and bottom-up proteomics showed differences in apoptotic and epithelial junction signaling proteins. Conclusions: The histogenetic origin of cells influenced the susceptibility of human malignant cells towards cytotoxic agents with leukemias and lymphomas being more sensitive than solid cancer cells. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

16 pages, 2923 KB  
Article
Cytosine Deaminase-Overexpressing hTERT-Immortalized Human Adipose Stem Cells Enhance the Inhibitory Effects of Fluorocytosine on Tumor Growth in Castration Resistant Prostate Cancer
by Jae Heon Kim, Hee Jo Yang, Sang Hun Lee and Yun Seob Song
Int. J. Mol. Sci. 2024, 25(10), 5519; https://doi.org/10.3390/ijms25105519 - 18 May 2024
Cited by 2 | Viewed by 1898
Abstract
A promising de novo approach for the treatment of Castration-resistant prostate cancer (CRPC) exploits cell-mediated enzyme prodrug therapy comprising cytosine deaminase (CD) and fluorouracil (5-FC). The aim of this study was to determine the potential of bacterial CD-overexpressing hTERT-immortalized human adipose stem cells [...] Read more.
A promising de novo approach for the treatment of Castration-resistant prostate cancer (CRPC) exploits cell-mediated enzyme prodrug therapy comprising cytosine deaminase (CD) and fluorouracil (5-FC). The aim of this study was to determine the potential of bacterial CD-overexpressing hTERT-immortalized human adipose stem cells (hTERT-ADSC.CD) to suppress CRPC. A lentiviral vector encoding a bacterial CD gene was used to transfect and to generate the hTERT-ADSC.CD line. The ability of the cells to migrate selectively towards malignant cells was investigated in vitro. PC3 and hTERT-ADSC.CD cells were co-cultured. hTERT-ADSC.CD and 1 × 106 PC3 cells were administered to nude mice via intracardiac and subcutaneous injections, respectively, and 5-FC was given for 14 days. hTERT-ADSC.CD were successfully engineered. Enhanced in vitro hTERT-ADSC.CD cytotoxicity and suicide effect were evident following administration of 5 μM 5-FC. hTERT-ADSC.CD, together with 5-FC, augmented the numbers of PC3 cells undergoing apoptosis. In comparison to controls administered hTERT-ADSC.CD monotherapy, hTERT-ADSC.CD in combination with 5-FC demonstrated a greater suppressive effect on tumor. In CPRC-bearing mice, tumor suppression was enhanced by the combination of CD-overexpressing ADSC and the prodrug 5-FC. Stem cells exhibiting CD gene expression are a potential novel approach to treatment for CRPC. Full article
(This article belongs to the Special Issue Cancer Suicide Gene Therapy)
Show Figures

Figure 1

12 pages, 1241 KB  
Review
Genetic Mechanisms Driving Uterine Leiomyoma Pathobiology, Epidemiology, and Treatment
by Malini S. Ramaiyer, Eslam Saad, Irem Kurt and Mostafa A. Borahay
Genes 2024, 15(5), 558; https://doi.org/10.3390/genes15050558 - 27 Apr 2024
Cited by 13 | Viewed by 6987
Abstract
Uterine leiomyomas (ULs) are the most common benign tumor of the uterus. They can be associated with symptoms including abnormal uterine bleeding, pelvic pain, urinary frequency, and pregnancy complications. Despite the high prevalence of UL, its underlying pathophysiology mechanisms have historically been poorly [...] Read more.
Uterine leiomyomas (ULs) are the most common benign tumor of the uterus. They can be associated with symptoms including abnormal uterine bleeding, pelvic pain, urinary frequency, and pregnancy complications. Despite the high prevalence of UL, its underlying pathophysiology mechanisms have historically been poorly understood. Several mechanisms of pathogenesis have been suggested, implicating various genes, growth factors, cytokines, chemokines, and microRNA aberrations. The purpose of this study is to summarize the current research on the relationship of genetics with UL. Specifically, we performed a literature review of published studies to identify how genetic aberrations drive pathophysiology, epidemiology, and therapeutic approaches of UL. With regards to pathophysiology, research has identified MED12 mutations, HMGA2 overexpression, fumarate hydratase deficiency, and cytogenetic abnormalities as contributors to the development of UL. Additionally, epigenetic modifications, such as histone acetylation and DNA methylation, have been identified as contributing to UL tumorigenesis. Specifically, UL stem cells have been found to contain a unique DNA methylation pattern compared to more differentiated UL cells, suggesting that DNA methylation has a role in tumorigenesis. On a population level, genome-wide association studies (GWASs) and epidemiologic analyses have identified 23 genetic loci associated with younger age at menarche and UL growth. Additionally, various GWASs have investigated genetic loci as potential drivers of racial disparities in UL incidence. For example, decreased expression of Cytohesin 4 in African Americans has been associated with increased UL risk. Recent studies have investigated various therapeutic options, including ten-eleven translocation proteins mediating DNA methylation, adenovirus vectors for drug delivery, and “suicide gene therapy” to induce apoptosis. Overall, improved understanding of the genetic and epigenetic drivers of UL on an individual and population level can propel the discovery of novel therapeutic options. Full article
(This article belongs to the Special Issue Genetics and Genomics of Female Reproduction)
Show Figures

Figure 1

23 pages, 1888 KB  
Review
Immunotherapy of Hematological Malignancies of Human B-Cell Origin with CD19 CAR T Lymphocytes
by Darya Khvorost, Brittany Kendall and Ali R. Jazirehi
Cells 2024, 13(8), 662; https://doi.org/10.3390/cells13080662 - 9 Apr 2024
Cited by 6 | Viewed by 5223
Abstract
Acute lymphoblastic leukemia (ALL) and non-Hodgkin’s lymphoma (NHL) are hematological malignancies with high incidence rates that respond relatively well to conventional therapies. However, a major issue is the clinical emergence of patients with relapsed or refractory (r/r) NHL or ALL. In such circumstances, [...] Read more.
Acute lymphoblastic leukemia (ALL) and non-Hodgkin’s lymphoma (NHL) are hematological malignancies with high incidence rates that respond relatively well to conventional therapies. However, a major issue is the clinical emergence of patients with relapsed or refractory (r/r) NHL or ALL. In such circumstances, opportunities for complete remission significantly decline and mortality rates increase. The recent FDA approval of multiple cell-based therapies, Kymriah (tisagenlecleucel), Yescarta (axicabtagene ciloleucel), Tecartus (Brexucabtagene autoleucel KTE-X19), and Breyanzi (Lisocabtagene Maraleucel), has provided hope for those with r/r NHL and ALL. These new cell-based immunotherapies use genetically engineered chimeric antigen receptor (CAR) T-cells, whose success can be attributed to CAR’s high specificity in recognizing B-cell-specific CD19 surface markers present on various B-cell malignancies and the subsequent initiation of anti-tumor activity. The efficacy of these treatments has led to promising results in many clinical trials, but relapses and adverse reactions such as cytokine release syndrome (CRS) and neurotoxicity (NT) remain pervasive, leaving areas for improvement in current and subsequent trials. In this review, we highlight the current information on traditional treatments of NHL and ALL, the design and manufacturing of various generations of CAR T-cells, the FDA approval of Kymriah, Yescarta Tecartus, and Breyanzi, and a summary of prominent clinical trials and the notable disadvantages of treatments. We further discuss approaches to potentially enhance CAR T-cell therapy for these malignancies, such as the inclusion of a suicide gene and use of FDA-approved drugs. Full article
(This article belongs to the Special Issue Resistance of Hematological Malignancies to CAR T Cell Therapy)
Show Figures

Figure 1

12 pages, 2518 KB  
Article
Targeted Suicide Gene Therapy with Retroviral Replicating Vectors for Experimental Canine Cancers
by Emiko Sonoda-Fukuda, Yuya Takeuchi, Nao Ogawa, Shunsuke Noguchi, Toru Takarada, Noriyuki Kasahara and Shuji Kubo
Int. J. Mol. Sci. 2024, 25(5), 2657; https://doi.org/10.3390/ijms25052657 - 24 Feb 2024
Cited by 3 | Viewed by 2786
Abstract
Cancer in dogs has increased in recent years and is a leading cause of death. We have developed a retroviral replicating vector (RRV) that specifically targets cancer cells for infection and replication. RRV carrying a suicide gene induced synchronized killing of cancer cells [...] Read more.
Cancer in dogs has increased in recent years and is a leading cause of death. We have developed a retroviral replicating vector (RRV) that specifically targets cancer cells for infection and replication. RRV carrying a suicide gene induced synchronized killing of cancer cells when administered with a prodrug after infection. In this study, we evaluated two distinct RRVs derived from amphotropic murine leukemia virus (AMLV) and gibbon ape leukemia virus (GALV) in canine tumor models both in vitro and in vivo. Despite low infection rates in normal canine cells, both RRVs efficiently infected and replicated within all the canine tumor cells tested. The efficient intratumoral spread of the RRVs after their intratumoral injection was also demonstrated in nude mouse models of subcutaneous canine tumor xenografts. When both RRVs encoded a yeast cytosine deaminase suicide gene, which converts the prodrug 5-fluorocytosine (5-FC) to the active drug 5-fluorouracil, they caused tumor-cell-specific 5-FC-induced killing of the canine tumor cells in vitro. Furthermore, in the AZACF- and AZACH-cell subcutaneous tumor xenograft models, both RRVs exerted significant antitumor effects. These results suggest that RRV-mediated suicide gene therapy is a novel therapeutic approach to canine cancers. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

21 pages, 1083 KB  
Review
Cellular Strategies for Separating GvHD from GvL in Haploidentical Transplantation
by Mauro Di Ianni, Carmine Liberatore, Nicole Santoro, Paola Ranalli, Francesco Guardalupi, Giulia Corradi, Ida Villanova, Barbara Di Francesco, Stefano Lattanzio, Cecilia Passeri, Paola Lanuti and Patrizia Accorsi
Cells 2024, 13(2), 134; https://doi.org/10.3390/cells13020134 - 11 Jan 2024
Cited by 2 | Viewed by 3637
Abstract
GvHD still remains, despite the continuous improvement of transplantation platforms, a fearful complication of transplantation from allogeneic donors. Being able to separate GvHD from GvL represents the greatest challenge in the allogeneic transplant setting. This may be possible through continuous improvement of cell [...] Read more.
GvHD still remains, despite the continuous improvement of transplantation platforms, a fearful complication of transplantation from allogeneic donors. Being able to separate GvHD from GvL represents the greatest challenge in the allogeneic transplant setting. This may be possible through continuous improvement of cell therapy techniques. In this review, current cell therapies are taken into consideration, which are based on the use of TCR alpha/beta depletion, CD45RA depletion, T regulatory cell enrichment, NK-cell-based immunotherapies, and suicide gene therapies in order to prevent GvHD and maximally amplify the GvL effect in the setting of haploidentical transplantation. Full article
Show Figures

Figure 1

29 pages, 6372 KB  
Article
Serum-Resistant Ternary DNA Polyplexes for Suicide Gene Therapy of Uterine Leiomyoma
by Anna Egorova, Sofia Shtykalova, Marianna Maretina, Svetlana Freund, Alexander Selutin, Natalia Shved, Sergei Selkov and Anton Kiselev
Int. J. Mol. Sci. 2024, 25(1), 34; https://doi.org/10.3390/ijms25010034 - 19 Dec 2023
Cited by 5 | Viewed by 2058
Abstract
Uterine leiomyoma (UL) is a prevalent benign tumor in women that frequently gives rise to a multitude of reproductive complications. The use of suicide gene therapy has been proposed as a highly promising method for treating UL. To achieve successful gene therapy, it [...] Read more.
Uterine leiomyoma (UL) is a prevalent benign tumor in women that frequently gives rise to a multitude of reproductive complications. The use of suicide gene therapy has been proposed as a highly promising method for treating UL. To achieve successful gene therapy, it is essential to develop carriers that can efficiently transport nucleic acids into targeted cells and tissues. The instability of polyplexes in blood and other biological fluids is a crucial factor to consider when using non-viral carriers. In this study, we present serum-resistant and cRGD-modified DNA complexes for targeted delivery genes to UL cells. Ternary polyplexes were formed by incorporating cystine-cross-linked polyglutamic acid modified with histidine residues. We employed two techniques in the production of cross-linked polyanionic coating: matrix polymerization and oxidative polycondensation. In this study, we investigated the physicochemical properties of ternary DNA complexes, including the size and zeta-potential of the nanoparticles. Additionally, we evaluated cellular uptake, toxicity levels, transfection efficiency and specificity in vitro. The study involved introducing the HSV-TK gene into primary UL cells as a form of suicide gene therapy modeling. We have effectively employed ternary peptide-based complexes for gene delivery into the UL organtypic model. By implementing in situ suicide gene therapy, the increase in apoptosis genes expression was detected, providing conclusive evidence of apoptosis occurring in the transfected UL tissues. The results of the study strongly suggest that the developed ternary polyplexes show potential as a valuable tool in the implementation of suicide gene therapy for UL. Full article
(This article belongs to the Special Issue Biopolymers in Drug and Gene Delivery Systems 3.0)
Show Figures

Figure 1

39 pages, 4591 KB  
Review
Hosts and Heterologous Expression Strategies of Recombinant Toxins for Therapeutic Purposes
by Luana di Leandro, Martina Colasante, Giuseppina Pitari and Rodolfo Ippoliti
Toxins 2023, 15(12), 699; https://doi.org/10.3390/toxins15120699 - 13 Dec 2023
Cited by 5 | Viewed by 5601
Abstract
The production of therapeutic recombinant toxins requires careful host cell selection. Bacteria, yeast, and mammalian cells are common choices, but no universal solution exists. Achieving the delicate balance in toxin production is crucial due to potential self-intoxication. Recombinant toxins from various sources find [...] Read more.
The production of therapeutic recombinant toxins requires careful host cell selection. Bacteria, yeast, and mammalian cells are common choices, but no universal solution exists. Achieving the delicate balance in toxin production is crucial due to potential self-intoxication. Recombinant toxins from various sources find applications in antimicrobials, biotechnology, cancer drugs, and vaccines. “Toxin-based therapy” targets diseased cells using three strategies. Targeted cancer therapy, like antibody–toxin conjugates, fusion toxins, or “suicide gene therapy”, can selectively eliminate cancer cells, leaving healthy cells unharmed. Notable toxins from various biological sources may be used as full-length toxins, as plant (saporin) or animal (melittin) toxins, or as isolated domains that are typical of bacterial toxins, including Pseudomonas Exotoxin A (PE) and diphtheria toxin (DT). This paper outlines toxin expression methods and system advantages and disadvantages, emphasizing host cell selection’s critical role. Full article
(This article belongs to the Special Issue Immunotoxins: Current Status and Future Perspectives)
Show Figures

Figure 1

17 pages, 2876 KB  
Article
Suicide-Gene-Modified Extracellular Vesicles of Human Primary Uveal Melanoma in Future Therapies
by Jana Jakubechova, Bozena Smolkova, Alena Furdova, Lucia Demkova, Ursula Altanerova, Andreas Nicodemou, Tatiana Zeleznikova, Daniela Klimova and Cestmir Altaner
Int. J. Mol. Sci. 2023, 24(16), 12957; https://doi.org/10.3390/ijms241612957 - 19 Aug 2023
Cited by 6 | Viewed by 2146
Abstract
Extracellular vesicles secreted from uveal melanoma (UM) cells are involved in the establishment of the premetastatic niche and display transforming potential for the formation of metastases, preferentially in the liver. In this study, we cultivated human primary UM cells and uveal melanoma-associated fibroblasts [...] Read more.
Extracellular vesicles secreted from uveal melanoma (UM) cells are involved in the establishment of the premetastatic niche and display transforming potential for the formation of metastases, preferentially in the liver. In this study, we cultivated human primary UM cells and uveal melanoma-associated fibroblasts in vitro to be transduced by infection with a retrovirus containing the suicide gene—fused yeast cytosine deaminase::uracil phospho-ribosyl transferase (yCD::UPRT). A homogenous population of yCD::UPRT-UM cells with the integrated provirus expressed the gene, and we found it to continuously secrete small extracellular vesicles (sEVs) possessing mRNA of the suicide gene. The yCD::UPRT-UM-sEVs were internalized by tumor cells to the intracellular conversion of the prodrug 5-fluorocytosine (5-FC) to the cytotoxic drug 5-fluorouracil (5-FU). The host range of the yCD::UPRT-UM-sEVs was not limited to UMs only. The yCD::UPRT-UM-sEVs inhibited the growth of the human cutaneous melanoma cell line A375 and uveal melanoma cell line MP38, as well as other primary UMs, to various extents in vitro. The yCD::UPRT-UM-sEVs hold the therapeutic and prophylactic potential to become a therapeutic drug for UM. However, the use of yCD::UPRT-UM-sEVs must first be tested in animal preclinical studies. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

12 pages, 2779 KB  
Article
CRISISS: A Novel, Transcriptionally and Post-Translationally Inducible CRISPR/Cas9-Based Cellular Suicide Switch
by Maximilian Amberger, Esther Grueso and Zoltán Ivics
Int. J. Mol. Sci. 2023, 24(12), 9799; https://doi.org/10.3390/ijms24129799 - 6 Jun 2023
Cited by 4 | Viewed by 3015
Abstract
With the ever-increasing developing rate of gene and cellular therapy applications and growing accessibility due to products receiving regulatory approval, the need for effective and reliable safety mechanisms to prevent or eliminate potentially fatal side effects is of the utmost importance. In this [...] Read more.
With the ever-increasing developing rate of gene and cellular therapy applications and growing accessibility due to products receiving regulatory approval, the need for effective and reliable safety mechanisms to prevent or eliminate potentially fatal side effects is of the utmost importance. In this study, we present the CRISPR-induced suicide switch (CRISISS) as a tool to eliminate genetically modified cells in an inducible and highly efficient manner by targeting Cas9 to highly repetitive Alu retrotransposons in the human genome, causing irreparable genomic fragmentation by the Cas9 nuclease and resulting cell death. The suicide switch components, including expression cassettes for a transcriptionally and post-translationally inducible Cas9 and an Alu-specific single-guide RNA, were integrated into the genome of target cells via Sleeping-Beauty-mediated transposition. The resulting transgenic cells did not show signs of any impact on overall fitness when uninduced, as unintended background expression, background DNA damage response and background cell killing were not observed. When induced, however, a strong expression of Cas9, a strong DNA damage response and a rapid halt of cell proliferation coupled with near complete cell death within four days post-induction were seen. With this proof-of-concept study, we present a novel and promising approach for a robust suicide switch with potential utility for gene and cell therapy in the future. Full article
(This article belongs to the Special Issue State-of-the-Art Molecular Genetics and Genomics in Germany)
Show Figures

Figure 1

Back to TopTop