Recent Advances in Gene Editing Cargo Delivery for Hematopoietic Stem and Progenitor Cells

Research Article
Open access

Recent Advances in Gene Editing Cargo Delivery for Hematopoietic Stem and Progenitor Cells

Yuxiang Tang 1*
  • 1 South China University of Technology, Guangzhou, China, 511442    
  • *corresponding author yxiang_tang@163.com
TNS Vol.103
ISSN (Print): 2753-8826
ISSN (Online): 2753-8818
ISBN (Print): 978-1-80590-073-3
ISBN (Online): 978-1-80590-074-0

Abstract

As the cornerstone of human hematopoiesis, hematopoietic stem and progenitor cells (HSPCs) sustain immune homeostasis by continuously generating blood cellular components. Their proliferative capacity and multilineage differentiation potential make them prime targets for gene therapy. The clinical application of HSPC genome editing relies on two key advancements: the development of highly specific and biocompatible genome-editing reagents and the establishment of efficient delivery systems ensuring targeted cellular uptake. Recent innovations in gene editing tools and cargo delivery methods, such as RNA- and protein-based editors, have enabled novel therapeutic strategies for hematological disorders. Current gene delivery platforms for HSPCs include electroporation, synthetic nanoscale carriers (e.g., polymeric and lipid nanoparticles), and engineered viral vectors such as integration-defective lentiviral vectors (IDLVs), adeno-associated viral vectors (AADVs), virus-like particles, and adenovirus vectors (AdVs). While ex vivo gene therapy remain predominant, it requires complex and costly patient conditioning regimens. in vivo approaches, primarily utilizing AdVs and LNPs, offer an alternative but lack sufficient targeting precision and transfection efficiency. By critically analyzing these advancements, this review aims to identify pathways for optimizing genome editing in HSPCs and enhancing therapeutic precision in hematological disorder management.

Keywords:

Gene therapy, gene editing, gene delivery, hematopoietic stem and progenitor cells, blood disorders

Tang,Y. (2025). Recent Advances in Gene Editing Cargo Delivery for Hematopoietic Stem and Progenitor Cells. Theoretical and Natural Science,103,35-41.
Export citation

1. Introduction

A variety of tissues in the adult human body maintain a limited population of adult stem cells, which play essential cytoprotective roles in preserving and replenishing specific cell types, thereby ensuring tissue homeostasis [1]. Among them, blood cells perform vital functions, including immune defense and injury repair. However, significant loss of blood cells can occur due to disease or trauma. Hematopoietic stem cells (HSCs) serve as the fundamental units of hematopoiesis, possessing lifelong capacity for blood system reconstitution. They exhibit two key functional attributes: clonal expansion potential and multilineage differentiation commitment. As adult stem cells, HSCs can replenish all types of blood cells, maintaining hematopoietic homeostasis [2]. Hematopoietic stem cell transplantation (HSCT) is the standard treatment for malignant hematological diseases and congenital disorders. In the 1950s, Rekers et al. carried out the technique of allogeneic HSCT by intravenously delivering bone marrow cells to irradiated mice, thereby preventing their demise and reconstituting hematopoiesis [3]. As of 2022, the global number of HSCT procedures has surpassed 1.5 million [4]. However, allogeneic HSCT is not always the optimal treatment option due to its reliance on a highly matched human leukocyte antigen (HLA) profile. The probability of identifying a suitably matched sibling donor is less than 30%. Furthermore, allogeneic transplantation carries the risk of inducing immune rejection, which can result in severe complications or mortality [5,6]. To overcome these challenges, genetic modification of autologous HSCs has emerged as a promising strategy for treating hematological diseases. This approach involves either ex vivo gene editing followed by transplantation or direct in vivo gene therapy, aiming to correct disease-causing mutations while minimizing immune complications. This review focuses on recent advancements in gene editing cargo delivery for HSPCs, discussing efficient and safe delivery strategies, associated challenges, and existing solutions.

2. Gene Editing Cargo for HSPCs

A cell’s physiological state is primarily determined by its genetic makeup. Therefore, a key goal in medical research is to precisely modify genetic loci and repair damaged genes. Transient genome-editing activation minimizes off-target effects and reduce editor immunogenicity [7]. For efficient "hit-and-run" genetic modifications, gene-editing cargos are best delivered as proteins or RNA. Jennifer Doudna et al. demonstrated that Cas9 recognizes its target sequence through the seed region within crRNA and requires a downstream protospacer-adjacent motif (PAM) for binding. Engineered RNAs can be custom-designed to direct Cas9 endonuclease activity toward predetermined loci, triggering programmable double-stranded DNA breaks through site-specific cleavage [8]. Expanding on this fundamental work, Feng's laboratory successfully adapted the CRISPR-Cas9 system for efficient genome editing in mammalian cells [9]. The gene-editing payload of CRISPR systems, including Cas9 nucleases delivered in mRNA form, has gained significant traction owing to recent advancements in mRNA technology and the advent of more complex gene editors such as base editors (approximately 4.8 kb) and prime editors (approximately 6.7 kb). These editors incorporate effectors fused with Cas9 variants [10,11]. Base editor mRNA achieves editing efficiencies exceeding 80% across various targets in HSPCs. Additionally, chemical modifications to the gene-edited mRNA further enhance the overall gene editing efficiency [10]. Nucleases can also be delivered directly in their protein form, which facilitates immediate action and rapid cellular turnover, thereby minimizing off-target effects. Currently, one of the most efficient and safe CRISPR-Cas9 gene editing strategies for human stem and progenitor cells involves the simultaneous delivery of pre-assembled ribonucleo-protein complexes (RNP), guided by RNA (gRNA) and Cas9 nuclease, via nucleofection. Moreover, RNP-based approaches do not compromise the survival capacity, clonal expansion potential, and post-transplantation functionality of HSPCs. Additionally, transient gene editing using RNPs minimizes off-target effects, insertion mutagenesis, and immune activation while achieving high editing efficiency.

3. Cargo Delivery Systems to HSPCs

AT primary challenge in delivering gene editors and DNA templates to HSCs, both ex vivo and in vivo, is ensuring efficient and safe genome editing. This is critical to maintaining accurate genetic information in the progeny of these cells. The selection of a delivery system depends on several factors, including the cellular repair pathway involved in editing, the required editing efficiency, the cytotoxicity tolerance of target cells, and the size of the donor DNA template. Current gene-editing delivery strategies can be broadly categorized into viral and non-viral methods. Among non-viral approaches, researchers are actively exploring physical and chemical methods for delivering biomolecular proteins and RNA.

3.1. Electroporation

Electroporation is one of the most effective techniques for delivering biomolecule into cells. It enables efficient intracellular transport of mRNA and protein complexes, making it a powerful tool for gene editing. Additionally, it facilitates multiple gene editing across various target loci by delivering mRNA or RNPs [12-14]. The mechanism of electroporation involves transient disruption of membrane semi-permeability upon exposure to an electrical impulse, allowing RNA or RNP to enter the cell. Efficient homology-directed repair (HDR) relies on the concurrent delivery of donor DNA, nuclease, and sgRNA. Electroporation has been demonstrated to efficiently introduce these three components, facilitating HDR in stem cells. A key advantage of electroporation is its ability to deliver large molecular constructs, including Cas9 fusion proteins, base editors, and prime editors. While electroporation can induce transcriptional changes in human stem cells, their core pluripotency markers and trilineage differentiation potential remain unaffected. Early clinical studies indicate that patients with thalassemia and sickle cell disease, who previously required regular blood transfusions, have achieved transfusion independence [15].

3.2. Nanoparticles

Cationic/ionizable polymer nanoparticles (PNPs) and lipid nanoparticles (LNPs) can encapsulate negatively charged nucleic acids, facilitating intracellular delivery through various endocytosis mechanisms. These nanoparticles are easy to construct, exhibit low immunogenicity and non-invasiveness, enable rapid gene expression, and can be customized for cell- or tissue-specific delivery. For instance, studies have demonstrated effective silencing of CD44 protein expression in CD34+ acute myeloid leukemia (AML) cells using CD44 siRNA encapsulated in PNPs. This suggests that PNPs hold potential for targeting HSPCs [16]. Additionally, Cas9 RNPs encapsulated in poly β-amino esters (PBAE-PNPs) have achieved gene-editing efficiencies of up to 70% in HSPCs when targeting the CD33 and HBG promoters [17]. Notably, PNPs achieved equivalent gene-editing efficiency with only one-third of the Cas9 RNP dose required for electroporation, indicating their potential as a superior alternative [17].

3.3. Adeno-associated Viral Vectors

Adeno-associated virus vectors (AAVs) are among the most clinically validated gene therapy vehicles, with several FDA-approved gene therapy products. Their ability to transduce both dividing and non-dividing cells, coupled with stable transgene expression and a favorable safety profile, positions AAVs as leading vectors for in vivo gene therapy. AAV-based approaches have been utilized to treat various genetic disorders, including hereditary retinal dystrophy and hemophilia [18]. At least ten AAV serotypes have been identified, each exhibiting distinct cell-type specificity. Among these, AAV6 demonstrates a high affinity for HSCs. Modifications to AAV6 vectors have been shown to enhance transduction efficiency [19]. AAV6 has emerged as a leading viral vector for site-specific gene integration in ex vivo HSC therapies, and has been successfully utilized to correct a range of genetic diseases [20-23]. Triple mutation of the AAV6 capsid, combined with high-cell-confluence transduction conditions, has achieved over 90% transduction efficiency in HSPCs [24]. Despite these advantages, AAV-mediated gene therapy faces limitations due to its relatively small cargo capacity (approximately 4.7 kb). To address this limitation, researchers have explored dual-vector strategies and oversized AAV constructs to accommodate larger transgenes [25,26]. However, immune responses targeting AAV-transfected cells poses extra challenges that may constrain therapeutic efficacy [27].

3.4. Integration Defective Lentiviral Vectors

Lentiviral vectors (LVs) represent another class of viral vectors widely utilized for gene editing. These vectors, originally derived from the HIV-1 envelope, have undergone modifications to enhance biosafety, including the removal of replication and integrase-related genes. LVs are predominantly employed for ex vivo delivery and offer a substantial cargo capacity (up to approximately 10kb), making them suitable for delivering Cas proteins and multiple sgRNAs, thereby enabling multi-gene editing.

However, conventional lentiviral vectors pose a risk of genomic integration, leading to sustained expression of gene-editing components and increased off-target indel rates [28-30]. To mitigate this risk, researchers developed integration-defective lentiviral vectors (IDLVs), which incorporate a trans-complementing packaging system and a point mutation in the integrase domain. The IDLV integrase protein interacts with the host cell protein LEDGF/p75 to facilitate homology-directed repair (HDR) in human embryonic stem cells (hESCs) [31]. Additionally, IDLV systems enable efficient HDR in HSPCs with reducing DNA damage signaling and minimizing the integration of viral sequences at target sites compared to AAV6-mediated delivery [32]. IDLVs have been successfully utilized for delivering zinc finger nuclease (ZFNs) and for introducing corrective DNA templates into HSPCs, especially in the context of Primary Immunodeficiency (PIDs) [33].

3.5. Virus-like Particles

While lentiviral vectors enable robust delivery of CRISPR components in HSPCs, the sustained nuclease activity resulting from persistent Cas9-gRNA expression elevates the risk of off-target mutagenesis. Delivering gene-editing nucleases as RNA-protein complexes offers a more favorable alternative by allowing transient expression and reducing unintended genome modifications. Engineered virus-like particles (eVLPs) have emerged as a promising appraoch, potentially replacing viral vectors as delivery vehicles under specific conditions.

VLPs, derived from various viral origins, can encapsulate gene editing nucleases by fusing with viral proteins. During the self-assembly of viral proteins, gene-editing nucleases can be incorporated into VLPs in the form of either RNA or proteins Researchers successfully installed a base editor within VLPs and delivered it in vivo to the liver of a mouse, achieving an editing efficiency of approximately 63% [34]. However, the efficacy of clinical-grade VLP protocols in HSPCs remains to be elucidated.

3.6. in vivo Targeting of HSPCs

HSPC gene editing is generally implemented through two primary approaches: ex vivo manipulation followed by transplantation or direct in vivo administration of editing vectors. In ex vivo autologous gene therapy, a patient's own HSPCs are mobilized and collected via apheresis, followed by gene editing in a controlled laboratory setting. The edited HSPCs are then reinfused into the patient. Following transplantation, patients undergo conditioning regimens involving chemotherapeutic or radiotherapy to eliminate existing cells and create space within the hematopoietic niche. However, in diseases requiring intensive conditioning, these regimens often cause severe adverse effects. Moreover, the patient-specific nature of autologous gene therapy result in high manufacturing costs, making ex vivo gene therapy financially inaccessible for many patients. To expand access to HSC gene therapy, an alternative approach involves non-invasively delivery of gene-editing tools directly to target cells or tissues in vivo. Current in vivo HSPCs delivery approaches predominantly utilize helper-dependent adenoviral vectors, necessitating prior HSPC mobilization to optimize transduction efficiency. Subsequently, low-dose chemotherapy is used to selectively edit these cells.

Another in vivo gene delivery system involves cationic polymers, synthetic macromolecules with a positive charge that facilitate nucleic acid delivery. These polymers deliver large nucleic acids (e.g., plasmids, siRNA, mRNA) by forming nanosized polyplexes through electrostatic interactions, enabling transfection via endosomal escape [35]. Examples include DEAE-dextran and polyamino acids, which offer cargo size-independent complexation, simplified production, and optimized retention duration compared to viral vectors [36, 37]. However, their electrostatic assembly makes them highly sensitive to chemical environments, leading to chemical environments, leading to variable endocytosis and inconsistent endosomal escape efficiency [36]. Furthermore, the positive charge of cationic polymers can induce cytotoxicity by compromising membrane integrity. This inherent cytotoxicity can be mitigated through hydrophilic monomer incorporation (e.g., TEGMA), molecular weight optimization, and pKa modulation [38].

An alternative strategy for in vivo HSPC gene editing involves LNPs, lipid-based nanocarriers composed of four key components: ionizable cationic lipids (for efficient nucleic acid encapsulation and endosomal escape), cholesterol (for membrane stability), helper phospholipids (for structural stabilization and fluidity), and PEGylated lipids (to prevent aggregation) [39]. LNPs have several key advantages that enhance their suitability for in vivo gene-editing: Firstly, they protect nucleic acids from nuclease degradation, improving stability, shelf life, and bioavailability [40]. Secondly, advances in scalable microfluidic hybrid methods have simplified LNPs manufacturing, enabling large-scale production [41]. Thirdly, LNPs allow for repeated administration, making them particularly valuable for immunization and short-term treatment. Fourth, LNP-mediated gene editing achieves comparable levels of efficiency with one-third the required doses to other methods [42]. Lastly, RNP-encapsulated LNPs can be produced and transported in a lyophilized form, enhancing accessibility to treatment. Nevertheless, LNP-based delivery faces certain challenges. Traditional LNPs exhibit low tissue specificity outside the liver when administered intravenously, potentially limiting their therapeutic scope. However, direct injection of mRNA-LNPs into target organs has shown promise in addressing this limitation, with clinical trials already underway for direct mRNA-LNPs tumor injections. Additionally, transient mRNA-LNP systems are preferable to viral vectors with prolonged expression in slow-turnover tissues, as the "hit-and-run" mechanism ensures permanent genomic edits while reducing off-target mutations and immune responses.

A recent breakthrough demonstrated in vivo base editing of HSPCs via LNP-mediated delivery. Researchers successfully encapsulated mRNA encoding a base editor within LNPs and targeted the stem cell factor receptor CD117 in bone marrow HSCs. This lipid nanoparticle-based editing system enabled direct reprogramming of bone marrow stem cells in vivo without requiring donor cells or toxic conditioning regimens like chemotherapy or radiotherapy. The study achieved efficient HSCs editing under non-genotoxic conditions, offering a promising therapeutic approach for genetic disorders [43]. Another study highlighted an innovative LNP formulation with extended PEG-lipid alkyl chains (C18), which significantly improved nanoparticle delivery efficiency to the bone marrow following systemic administration. The refined formulation successfully delivered both short RNA (siCD45) and larger mRNA (Cre mRNA) payloads in vivo [44]. Furthermore, CD117 targeting has been demonstrated using a variety of ionizable lipid formulations.

Overall, in vivo gene editing targeting HSCs holds transformative potential for HSC gene therapy. However, significant advancements are required to achieve clinically viable efficacy.

4. Conclusion

HSPC gene editing holds transformative potential for treating genetic disorders, infectious diseases, and hematological conditions by enabling permanent correction of cellular functions. However, its clinical translation faces significant hurdles. At the laboratory stage, ex vivo approaches—reliant on costly and complex procedures like HSPC isolation, cytokine stimulation, and electroporation—limit accessibility and risk compromising cell viability. in vivo strategies, though promising for bypassing these barriers, struggle with inefficient targeting of HSPCs and risks such as off-target edits, p53-mediated DNA damage responses, and immunogenicity. Furthermore, safety concerns around double-strand breaks and the high costs of specialized manufacturing facilities underscore the need for safer, more scalable delivery systems.

To address these challenges, future efforts must prioritize the development of precision-targeted in vivo delivery platforms, such as lipid nanoparticles (LNPs), which offer advantages like repeatable dosing, reduced toxicity, and simplified production. Innovations in gene-editing tools (e.g., base editors to minimize DNA damage) and delivery technologies (e.g., engineered vectors for HSPC-specific homing) are critical to enhance specificity and safety. Simultaneously, streamlining manufacturing processes and reducing reliance on ex vivo infrastructure will improve cost-effectiveness and global accessibility. By integrating advances in biomaterials, computational biology, and immunology, the field can transition toward therapies that are not only curative but also equitable, ultimately revolutionizing the treatment landscape for diverse diseases.


References

[1]. Post Y, Clevers H (2019). Defining adult stem cell functionat its simplest: the ability to replace lost cells through mitosis. Cell Stem Cell 25:174-183.

[2]. Seita, J., & Weissman, I. L. (2010). Hematopoietic stem cell: self‐renewal versus differentiation. Wiley Interdisciplinary Reviews: Systems Biology and Medicine, 2(6), 640-653.

[3]. Rekers, P. E., Coulter, M. P., & WARREN, S. L. (1950). Effect of transplantation of bone marrow into irradiated animals. Archives of Surgery, 60(4), 635-667.

[4]. Niederwieser, D., Baldomero, H., Bazuaye, N., Bupp, C., Chaudhri, N., Corbacioglu, S., et al. (2022). One and a half million hematopoietic stem cell transplants: continuous and differential improvement in worldwide access with the use of non-identical family donors. Haematologica, 107(5), 1045-1053.

[5]. Copelan, E. A. (2006). Hematopoietic stem-cell transplantation. New England Journal of Medicine, 354(17), 1813-1826.

[6]. Angelucci E, Matthes-Martin, S., Baronciani, D., et al. (2014). Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: indications and management recommendations from an international expert panel. Haematologica 99:811820.

[7]. Kim, S., Kim, D., Cho, S. et al. (2014). Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome research, 24(6), 1012-1019.

[8]. Jinek, M., Chylinski, K., Fonfara, I., et al. (2012). A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science, 337(6096), 816-821.

[9]. Cong, L., Ran, F. A., Cox, D., et al. (2013). Multiplex genome engineering using CRISPR/Cas systems. Science, 339(6121), 819-823.

[10]. Jiang, T., Henderson, J. M., Coote, K., et al. (2020). Chemical modifications of adenine base editor mRNA and guide RNA expand its application scope. Nature communications, 11(1), 1979.

[11]. Liu, P., Liang, S. Q., Zheng, C., et al. (2021). Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nat. Commun. 12, 2121.

[12]. Bak, R. O., Dever, D. P., Reinisch, A., et al. (2017). Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6. Elife, 6, e27873.

[13]. Zeng, J., Wu, Y., Ren, C., et al. (2020). Therapeutic base editing of human hematopoietic stem cells. Nature Medicine, 26(4), 535-541.

[14]. Venkatesan, V., Christopher, A. C., Rhiel, M., et al. (2023). Editing the core region in HPFH deletions alters fetal and adult globin expression for treatment of β-hemoglobinopathies. Molecular Therapy-Nucleic Acids, 32, 671-688.

[15]. Frangoul, H., Altshuler, D., Cappellini, M. D., et al. (2021). CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. New England Journal of Medicine, 384(3), 252-260.

[16]. Gul-Uludağ, H., Valencia-Serna, J., Kucharski, C., et al. (2014). Polymeric nanoparticle-mediated silencing of CD44 receptor in CD34+ acute myeloid leukemia cells. Leukemia research, 38(11), 1299-1308.

[17]. El-Kharrag, R., Berckmueller, K. E., Madhu, R., et al. (2022). Efficient polymer nanoparticle-mediated delivery of gene editing reagents into human hematopoietic stem and progenitor cells. Molecular Therapy, 30(6), 2186-2198.

[18]. Li, C., & Samulski, R. J. (2020). Engineering adeno-associated virus vectors for gene therapy. Nature Reviews Genetics, 21(4), 255-272.

[19]. Büning, H., & Srivastava, A. (2019). Capsid modifications for targeting and improving the efficacy of AAV vectors. Molecular therapy Methods & clinical development, 12, 248-265.

[20]. Pavel-Dinu, M., Wiebking, V., Dejene, B. T., et al. (2019). Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nature communications, 10(1), 1634.

[21]. Rai, R., Romito, M., Rivers, E., et al. (2020). Targeted gene correction of human hematopoietic stem cells for the treatment of Wiskott-Aldrich Syndrome. Nature communications, 11(1), 4034.

[22]. Scharenberg, S. G., Poletto, E., Lucot, K. L., et al. (2020). Engineering monocyte/macrophage− specific glucocerebrosidase expression in human hematopoietic stem cells using genome editing. Nature Communications, 11(1), 3327.

[23]. Wilkinson, A. C., Dever, D. P., Baik, R., et al. (2021). Cas9-AAV6 gene correction of beta-globin in autologous HSCs improves sickle cell disease erythropoiesis in mice. Nature Communications, 12(1), 686.

[24]. Ling, C., Bhukhai, K., Yin, Z., et al. (2016). High-efficiency transduction of primary human hematopoietic stem/progenitor cells by AAV6 vectors: strategies for overcoming donor-variation and implications in genome editing. Scientific reports, 6(1), 35495.

[25]. Bak, R. O., & Porteus, M. H. (2017). CRISPR-mediated integration of large gene cassettes using AAV donor vectors. Cell reports, 20(3), 750-756.

[26]. Zhi, S., Chen, Y., Wu, G., et al. (2022). Dual-AAV delivering split prime editor system for in vivo genome editing. Molecular Therapy, 30(1), 283-294.

[27]. Mingozzi, F., & A High, K. (2011). Immune responses to AAV in clinical trials. Current gene therapy, 11(4), 321-330.

[28]. Kotterman, M. A., Chalberg, T. W., & Schaffer, D. V. (2015). Viral vectors for gene therapy: translational and clinical outlook. Annual review of biomedical engineering, 17(1), 63-89.

[29]. Rothe, M., Modlich, U., & Schambach, A. (2013). Biosafety challenges for use of lentiviral vectors in gene therapy. Current gene therapy, 13(6), 453-468.

[30]. Liu, K. C., Lin, B. S., Gao, A. D., et al. (2014). Integrase-deficient lentivirus: opportunities and challenges for human gene therapy. Current gene therapy, 14(5), 352-364.

[31]. Wang, Y., Wang, Y., Chang, T., et al. (2017). Integration-defective lentiviral vector mediates efficient gene editing through homology-directed repair in human embryonic stem cells. Nucleic acids research, 45(5), e29.

[32]. Ferrari, S., Jacob, A., Cesana, D., et al. (2022). Choice of template delivery mitigates the genotoxic risk and adverse impact of editing in human hematopoietic stem cells. Cell Stem Cell, 29(10), 1428-1444.

[33]. Abdul-Razak, H. H., Rocca, C. J., Howe, S. J., et al. (2018). Molecular evidence of genome editing in a mouse model of immunodeficiency. Scientific reports, 8(1), 8214.

[34]. Banskota, S., Raguram, A., Suh, S., Du, S. W., Davis, J. R., Choi, E. H., ... & Liu, D. R. (2022). Engineered virus-like particles for efficient in vivo delivery of therapeutic proteins. Cell, 185(2), 250-265.

[35]. Cai, X., Dou, R., Guo, C., et al. (2023). Cationic polymers as transfection reagents for nucleic acid delivery. Pharmaceutics, 15(5), 1502.

[36]. Rai, R., Alwani, S., & Badea, I. (2019). Polymeric nanoparticles in gene therapy: New avenues of design and optimization for delivery applications. Polymers, 11(4), 745.

[37]. Canine, B. F., & Hatefi, A. (2010). Development of recombinant cationic polymers for gene therapy research. Advanced drug delivery reviews, 62(15), 1524-1529.

[38]. Weiss, A. M., Lopez, M. A., Rawe, B. W., et al. (2023). Understanding How Cationic Polymers’ Properties Inform Toxic or Immunogenic Responses via Parametric Analysis. Macromolecules, 56(18), 7286-7299.

[39]. Kulkarni, J. A., Witzigmann, D., Thomson, S. B., et al. (2021). The current landscape of nucleic acid therapeutics. Nature nanotechnology, 16(6), 630-643.

[40]. Cheng, Q., Wei, T., Farbiak, L., et al. (2020). Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nature nanotechnology, 15(4), 313-320.

[41]. Shepherd, S. J., Han, X., Mukalel, A. J., et al. (2023). Throughput-scalable manufacturing of SARS-CoV-2 mRNA lipid nanoparticle vaccines. Proceedings of the National Academy of Sciences, 120(33), e2303567120.

[42]. El-Kharrag, R., Berckmueller, K. E., Madhu, R., et al. (2022). Efficient polymer nanoparticle-mediated delivery of gene editing reagents into human hematopoietic stem and progenitor cells. Molecular Therapy, 30(6), 2186-2198.

[43]. Breda, L., Papp, T. E., Triebwasser, M. P., et al. (2023). in vivo hematopoietic stem cell modification by mRNA delivery. Science, 381(6656), 436-443.

[44]. Shi, D., Toyonaga, S., & Anderson, D. G. (2023). in vivo RNA delivery to hematopoietic stem and progenitor cells via targeted lipid nanoparticles. Nano Letters, 23(7), 2938-2944.


Cite this article

Tang,Y. (2025). Recent Advances in Gene Editing Cargo Delivery for Hematopoietic Stem and Progenitor Cells. Theoretical and Natural Science,103,35-41.

Data availability

The datasets used and/or analyzed during the current study will be available from the authors upon reasonable request.

Disclaimer/Publisher's Note

The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of EWA Publishing and/or the editor(s). EWA Publishing and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

About volume

Volume title: Proceedings of the 5th International Conference on Biological Engineering and Medical Science

ISBN:978-1-80590-073-3(Print) / 978-1-80590-074-0(Online)
Editor:Alan Wang
Conference website: https://2025.icbiomed.org/
Conference date: 24 October 2025
Series: Theoretical and Natural Science
Volume number: Vol.103
ISSN:2753-8818(Print) / 2753-8826(Online)

© 2024 by the author(s). Licensee EWA Publishing, Oxford, UK. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license. Authors who publish this series agree to the following terms:
1. Authors retain copyright and grant the series right of first publication with the work simultaneously licensed under a Creative Commons Attribution License that allows others to share the work with an acknowledgment of the work's authorship and initial publication in this series.
2. Authors are able to enter into separate, additional contractual arrangements for the non-exclusive distribution of the series's published version of the work (e.g., post it to an institutional repository or publish it in a book), with an acknowledgment of its initial publication in this series.
3. Authors are permitted and encouraged to post their work online (e.g., in institutional repositories or on their website) prior to and during the submission process, as it can lead to productive exchanges, as well as earlier and greater citation of published work (See Open access policy for details).

References

[1]. Post Y, Clevers H (2019). Defining adult stem cell functionat its simplest: the ability to replace lost cells through mitosis. Cell Stem Cell 25:174-183.

[2]. Seita, J., & Weissman, I. L. (2010). Hematopoietic stem cell: self‐renewal versus differentiation. Wiley Interdisciplinary Reviews: Systems Biology and Medicine, 2(6), 640-653.

[3]. Rekers, P. E., Coulter, M. P., & WARREN, S. L. (1950). Effect of transplantation of bone marrow into irradiated animals. Archives of Surgery, 60(4), 635-667.

[4]. Niederwieser, D., Baldomero, H., Bazuaye, N., Bupp, C., Chaudhri, N., Corbacioglu, S., et al. (2022). One and a half million hematopoietic stem cell transplants: continuous and differential improvement in worldwide access with the use of non-identical family donors. Haematologica, 107(5), 1045-1053.

[5]. Copelan, E. A. (2006). Hematopoietic stem-cell transplantation. New England Journal of Medicine, 354(17), 1813-1826.

[6]. Angelucci E, Matthes-Martin, S., Baronciani, D., et al. (2014). Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: indications and management recommendations from an international expert panel. Haematologica 99:811820.

[7]. Kim, S., Kim, D., Cho, S. et al. (2014). Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome research, 24(6), 1012-1019.

[8]. Jinek, M., Chylinski, K., Fonfara, I., et al. (2012). A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science, 337(6096), 816-821.

[9]. Cong, L., Ran, F. A., Cox, D., et al. (2013). Multiplex genome engineering using CRISPR/Cas systems. Science, 339(6121), 819-823.

[10]. Jiang, T., Henderson, J. M., Coote, K., et al. (2020). Chemical modifications of adenine base editor mRNA and guide RNA expand its application scope. Nature communications, 11(1), 1979.

[11]. Liu, P., Liang, S. Q., Zheng, C., et al. (2021). Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nat. Commun. 12, 2121.

[12]. Bak, R. O., Dever, D. P., Reinisch, A., et al. (2017). Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6. Elife, 6, e27873.

[13]. Zeng, J., Wu, Y., Ren, C., et al. (2020). Therapeutic base editing of human hematopoietic stem cells. Nature Medicine, 26(4), 535-541.

[14]. Venkatesan, V., Christopher, A. C., Rhiel, M., et al. (2023). Editing the core region in HPFH deletions alters fetal and adult globin expression for treatment of β-hemoglobinopathies. Molecular Therapy-Nucleic Acids, 32, 671-688.

[15]. Frangoul, H., Altshuler, D., Cappellini, M. D., et al. (2021). CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. New England Journal of Medicine, 384(3), 252-260.

[16]. Gul-Uludağ, H., Valencia-Serna, J., Kucharski, C., et al. (2014). Polymeric nanoparticle-mediated silencing of CD44 receptor in CD34+ acute myeloid leukemia cells. Leukemia research, 38(11), 1299-1308.

[17]. El-Kharrag, R., Berckmueller, K. E., Madhu, R., et al. (2022). Efficient polymer nanoparticle-mediated delivery of gene editing reagents into human hematopoietic stem and progenitor cells. Molecular Therapy, 30(6), 2186-2198.

[18]. Li, C., & Samulski, R. J. (2020). Engineering adeno-associated virus vectors for gene therapy. Nature Reviews Genetics, 21(4), 255-272.

[19]. Büning, H., & Srivastava, A. (2019). Capsid modifications for targeting and improving the efficacy of AAV vectors. Molecular therapy Methods & clinical development, 12, 248-265.

[20]. Pavel-Dinu, M., Wiebking, V., Dejene, B. T., et al. (2019). Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nature communications, 10(1), 1634.

[21]. Rai, R., Romito, M., Rivers, E., et al. (2020). Targeted gene correction of human hematopoietic stem cells for the treatment of Wiskott-Aldrich Syndrome. Nature communications, 11(1), 4034.

[22]. Scharenberg, S. G., Poletto, E., Lucot, K. L., et al. (2020). Engineering monocyte/macrophage− specific glucocerebrosidase expression in human hematopoietic stem cells using genome editing. Nature Communications, 11(1), 3327.

[23]. Wilkinson, A. C., Dever, D. P., Baik, R., et al. (2021). Cas9-AAV6 gene correction of beta-globin in autologous HSCs improves sickle cell disease erythropoiesis in mice. Nature Communications, 12(1), 686.

[24]. Ling, C., Bhukhai, K., Yin, Z., et al. (2016). High-efficiency transduction of primary human hematopoietic stem/progenitor cells by AAV6 vectors: strategies for overcoming donor-variation and implications in genome editing. Scientific reports, 6(1), 35495.

[25]. Bak, R. O., & Porteus, M. H. (2017). CRISPR-mediated integration of large gene cassettes using AAV donor vectors. Cell reports, 20(3), 750-756.

[26]. Zhi, S., Chen, Y., Wu, G., et al. (2022). Dual-AAV delivering split prime editor system for in vivo genome editing. Molecular Therapy, 30(1), 283-294.

[27]. Mingozzi, F., & A High, K. (2011). Immune responses to AAV in clinical trials. Current gene therapy, 11(4), 321-330.

[28]. Kotterman, M. A., Chalberg, T. W., & Schaffer, D. V. (2015). Viral vectors for gene therapy: translational and clinical outlook. Annual review of biomedical engineering, 17(1), 63-89.

[29]. Rothe, M., Modlich, U., & Schambach, A. (2013). Biosafety challenges for use of lentiviral vectors in gene therapy. Current gene therapy, 13(6), 453-468.

[30]. Liu, K. C., Lin, B. S., Gao, A. D., et al. (2014). Integrase-deficient lentivirus: opportunities and challenges for human gene therapy. Current gene therapy, 14(5), 352-364.

[31]. Wang, Y., Wang, Y., Chang, T., et al. (2017). Integration-defective lentiviral vector mediates efficient gene editing through homology-directed repair in human embryonic stem cells. Nucleic acids research, 45(5), e29.

[32]. Ferrari, S., Jacob, A., Cesana, D., et al. (2022). Choice of template delivery mitigates the genotoxic risk and adverse impact of editing in human hematopoietic stem cells. Cell Stem Cell, 29(10), 1428-1444.

[33]. Abdul-Razak, H. H., Rocca, C. J., Howe, S. J., et al. (2018). Molecular evidence of genome editing in a mouse model of immunodeficiency. Scientific reports, 8(1), 8214.

[34]. Banskota, S., Raguram, A., Suh, S., Du, S. W., Davis, J. R., Choi, E. H., ... & Liu, D. R. (2022). Engineered virus-like particles for efficient in vivo delivery of therapeutic proteins. Cell, 185(2), 250-265.

[35]. Cai, X., Dou, R., Guo, C., et al. (2023). Cationic polymers as transfection reagents for nucleic acid delivery. Pharmaceutics, 15(5), 1502.

[36]. Rai, R., Alwani, S., & Badea, I. (2019). Polymeric nanoparticles in gene therapy: New avenues of design and optimization for delivery applications. Polymers, 11(4), 745.

[37]. Canine, B. F., & Hatefi, A. (2010). Development of recombinant cationic polymers for gene therapy research. Advanced drug delivery reviews, 62(15), 1524-1529.

[38]. Weiss, A. M., Lopez, M. A., Rawe, B. W., et al. (2023). Understanding How Cationic Polymers’ Properties Inform Toxic or Immunogenic Responses via Parametric Analysis. Macromolecules, 56(18), 7286-7299.

[39]. Kulkarni, J. A., Witzigmann, D., Thomson, S. B., et al. (2021). The current landscape of nucleic acid therapeutics. Nature nanotechnology, 16(6), 630-643.

[40]. Cheng, Q., Wei, T., Farbiak, L., et al. (2020). Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nature nanotechnology, 15(4), 313-320.

[41]. Shepherd, S. J., Han, X., Mukalel, A. J., et al. (2023). Throughput-scalable manufacturing of SARS-CoV-2 mRNA lipid nanoparticle vaccines. Proceedings of the National Academy of Sciences, 120(33), e2303567120.

[42]. El-Kharrag, R., Berckmueller, K. E., Madhu, R., et al. (2022). Efficient polymer nanoparticle-mediated delivery of gene editing reagents into human hematopoietic stem and progenitor cells. Molecular Therapy, 30(6), 2186-2198.

[43]. Breda, L., Papp, T. E., Triebwasser, M. P., et al. (2023). in vivo hematopoietic stem cell modification by mRNA delivery. Science, 381(6656), 436-443.

[44]. Shi, D., Toyonaga, S., & Anderson, D. G. (2023). in vivo RNA delivery to hematopoietic stem and progenitor cells via targeted lipid nanoparticles. Nano Letters, 23(7), 2938-2944.