Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 10(21): e2301377, 2023 07.
Article in English | MEDLINE | ID: mdl-37171801

ABSTRACT

Multiple myeloma (MM) is a cancer of differentiated plasma cells that occurs in the bone marrow (BM). Despite the recent advancements in drug development, most patients with MM eventually relapse and the disease remains incurable. RNA therapy delivered via lipid nanoparticles (LNPs) has the potential to be a promising cancer treatment, however, its clinical implementation is limited due to inefficient delivery to non-hepatic tissues. Here, targeted (t)LNPs designed for delivery of RNA payload to MM cells are presented. The tLNPs consist of a novel ionizable lipid and are coated with an anti-CD38 antibody (αCD38-tLNPs). To explore their therapeutic potential, it is demonstrated that LNPs encapsulating small interference RNA (siRNA) against cytoskeleton-associated protein 5 (CKAP5) lead to a ≈90% decrease in cell viability of MM cells in vitro. Next, a new xenograft MM mouse model is employed, which clinically resembles the human disease and demonstrates efficient homing of MM cells to the BM. Specific delivery of αCD38-tLNPs to BM-residing and disseminated MM cells and the improvement in therapeutic outcome of MM-bearing mice treated with αCD38-tLNPs-siRNA-CKAP5 are shown. These results underscore the potential of RNA therapeutics for treatment of MM and the importance of developing effective targeted delivery systems and reliable preclinical models.


Subject(s)
Multiple Myeloma , Humans , Animals , Mice , Multiple Myeloma/drug therapy , Bone Marrow , Neoplasm Recurrence, Local , RNA, Small Interfering/therapeutic use
2.
Adv Sci (Weinh) ; 10(19): e2301929, 2023 07.
Article in English | MEDLINE | ID: mdl-37092557

ABSTRACT

Ionizable lipid-based nanoparticles (LNPs) are the most advanced non-viral drug delivery systems for RNA therapeutics and vaccines. However, cell type-specific, extrahepatic mRNA delivery is still a major hurdle, hampering the development of novel therapeutic modalities. Herein, a novel ionizable lipid library is synthesized by modifying hydrophobic tail chains and linkers. Combined with other helper lipids and utilizing a microfluidic mixing approach, stable LNPs are formed. Using Luciferase-mRNA, mCherry mRNA, and Cre mRNA together with a TdTomato animal model, superior lipids forming LNPs for potent cell-type specific mRNA delivery are identified. In vitro assays concluded that combining branched ester tail chains with hydroxylamine linker negatively affects mRNA delivery efficiency. In vivo studies identify Lipid 23 as a liver-trophic, superior mRNA delivery lipid and Lipid 16 as a potent cell type-specific ionizable lipid for the CD11bhi macrophage population without an additional targeting moiety. Finally, in vivo mRNA delivery efficiency and toxicity of these LNPs are compared with SM-102-based LNP (Moderna's LNP formulation) and are shown to be cell-specific compared to SM-102-based LNPs. Overall, this study suggests that a structural combination of tail and linker can drive a novel functionality of LNPs in vivo.


Subject(s)
Nanoparticles , Animals , RNA, Messenger/genetics , Nanoparticles/chemistry , Lipids/chemistry
3.
Sci Adv ; 9(14): eade4800, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37018392

ABSTRACT

The potential of microtubule-associated protein targets for cancer therapeutics remains largely unexplored due to the lack of target-specific agents. Here, we explored the therapeutic potential of targeting cytoskeleton-associated protein 5 (CKAP5), an important microtubule-associated protein, with CKAP5-targeting siRNAs encapsulated in lipid nanoparticles (LNPs). Our screening of 20 solid cancer cell lines demonstrated selective vulnerability of genetically unstable cancer cell lines in response to CKAP5 silencing. We identified a highly responsive chemo-resistant ovarian cancer cell line, in which CKAP5 silencing led to significant loss in EB1 dynamics during mitosis. Last, we demonstrated the therapeutic potential in an in vivo ovarian cancer model, showing 80% survival rate of siCKAP5 LNPs-treated animals. Together, our results highlight the importance of CKAP5 as a therapeutic target for genetically unstable ovarian cancer and warrants further investigation into its mechanistic aspects.


Subject(s)
Nanoparticles , Ovarian Neoplasms , Humans , Animals , Female , Gene Silencing , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/genetics , Microtubules/metabolism , Ovarian Neoplasms/genetics
4.
Sci Adv ; 9(10): eadg1036, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36888708

ABSTRACT

Messenger RNA (mRNA) lipid nanoparticle (LNP) vaccines have emerged as an effective vaccination strategy. Although currently applied toward viral pathogens, data concerning the platform's effectiveness against bacterial pathogens are limited. Here, we developed an effective mRNA-LNP vaccine against a lethal bacterial pathogen by optimizing mRNA payload guanine and cytosine content and antigen design. We designed a nucleoside-modified mRNA-LNP vaccine based on the bacterial F1 capsule antigen, a major protective component of Yersinia pestis, the etiological agent of plague. Plague is a rapidly deteriorating contagious disease that has killed millions of people during the history of humankind. Now, the disease is treated effectively with antibiotics; however, in the case of a multiple-antibiotic-resistant strain outbreak, alternative countermeasures are required. Our mRNA-LNP vaccine elicited humoral and cellular immunological responses in C57BL/6 mice and conferred rapid, full protection against lethal Y. pestis infection after a single dose. These data open avenues for urgently needed effective antibacterial vaccines.


Subject(s)
Plague Vaccine , Plague , Yersinia pestis , Mice , Animals , Plague/prevention & control , Plague Vaccine/genetics , Bacterial Proteins/genetics , Mice, Inbred C57BL , Yersinia pestis/genetics , Antigens, Bacterial/genetics
5.
Adv Mater ; 34(13): e2106350, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35044699

ABSTRACT

Chemo-immunotherapy is a combination of "standard-of-care" chemotherapy with immunotherapy and it is considered the most advanced therapeutic modality for various types of cancers. However, many cancer patients still poorly respond to current regimen of chemo-immunotherapy and suggest nanotherapeutics as a boosting agent. Recently, heme oxygenase-1 (HO1) is shown to act as an immunotherapeutic molecule in tumor myeloid cells, in addition to general chemoresistance function in cancer cells suggesting that HO1-targeted therapeutics can become a novel, optimal strategy for boosting chemo-immunotherapy in the clinic. Currently the available HO1-inhibitors demonstrate serious adverse effects in clinical use. Herein, tumor myeloid cell- and cancer cell-dual targeted HO1-inhibiting lipid nanotherapeutic boost (T-iLNTB) is developed using RNAi-loaded lipid nanoparticles. T-iLNTB-mediated HO1-inhibition sensitizes cancer cells to "standard-of-care" chemotherapeutics by increasing immunogenic cell death, and directly reprograms tumor myeloid cells with distinguished phenotype. Furthermore, tumor myeloid cell reprogramming by T-iLNTB induces CD8+ cytotoxic T cell recruitment, which drives "Cold-to-Hot" transition and correlates with improved responsiveness to immune checkpoint inhibitor in combination therapy. Finally, ex vivo study proves that HO1-inhibition directly affects tumor macrophage differentiation. This study demonstrates the potential of T-iLNTB as a novel therapeutic modality for boosting chemo-immunotherapy.


Subject(s)
Nanoparticles , Neoplasms , Humans , Immunotherapy , Lipids , Liposomes , Neoplasms/drug therapy , Tumor Microenvironment
6.
J Control Release ; 337: 378-389, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34303750

ABSTRACT

Currently there are no specific therapies addressing the distinctive biology of human papillomavirus (HPV)-induced cancer approved for clinical use. Short interfering RNA (siRNA) has much potential for therapeutic manipulation of HPV E6/E7 oncoproteins. Lipid-based nanoparticles (LNPs) can be utilized for systemic transportation and delivery of siRNA at target site. We recently developed a recombinant protein linker that enables uniform conjugation of targeting antibodies to the LNPs. Herein, we demonstrate the therapeutic efficacy of anti-E6/E7 siRNA delivered via targeted LNPs (tLNPs) in a xenograft HPV-positive tumor model. We show that anti-epidermal growth factor receptor (EGFR) antibodies, anchored to the LNPs as targeting moieties, facilitate cargo delivery but also mediate anti-tumor activity. Treatment with siE6 via tLNPs resulted in 50% greater reduction of tumor volume compared to treatment with siControl encapsulated in isoLNPs (coated with isotype control antibodies). We demonstrate superior suppression of HPV oncogenes and higher induction of apoptosis by the tLNPs both in vitro and in vivo. Altogether, the coupling of inhibitory siE6 with anti-EGFR antibodies, that further elicited anti-tumor effects, successfully restricted tumor progression. This system that combines potent siRNA and therapeutically functional tLNPs can be modulated against various cancer models.


Subject(s)
Head and Neck Neoplasms , Nanoparticles , Oncogene Proteins, Viral , Uterine Cervical Neoplasms , Cell Line, Tumor , Female , Head and Neck Neoplasms/drug therapy , Humans , Lipids , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , RNA, Small Interfering , Repressor Proteins
7.
Nat Nanotechnol ; 16(9): 1030-1038, 2021 09.
Article in English | MEDLINE | ID: mdl-34140675

ABSTRACT

The successful in vivo implementation of gene expression modulation strategies relies on effective, non-immunogenic delivery vehicles. Lipid nanoparticles are one of the most advanced non-viral clinically approved nucleic-acid delivery systems. Yet lipid nanoparticles accumulate naturally in liver cells upon intravenous administration, and hence, there is an urgent need to enhance uptake by other cell types. Here we use a conformation-sensitive targeting strategy to achieve in vivo gene silencing in a selective subset of leukocytes and show potential therapeutic applications in a murine model of colitis. In particular, by targeting the high-affinity conformation of α4ß7 integrin, which is a hallmark of inflammatory gut-homing leukocytes, we silenced interferon-γ in the gut, resulting in an improved therapeutic outcome in experimental colitis. The lipid nanoparticles did not induce adverse immune activation or liver toxicity. These results suggest that our lipid nanoparticle targeting strategy might be applied for selective delivery of payloads to other conformation-sensitive targets.


Subject(s)
Colitis/therapy , Gene Silencing , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Animals , Colitis/genetics , Gene Expression Regulation/drug effects , Humans , Integrin alpha4/chemistry , Integrin alpha4/genetics , Integrin beta Chains/chemistry , Integrin beta Chains/genetics , Lipids/chemistry , Lipids/pharmacology , Liver/drug effects , Mice , Nanoparticles/therapeutic use , RNA, Small Interfering/genetics
8.
Small ; 17(19): e2100287, 2021 05.
Article in English | MEDLINE | ID: mdl-33825318

ABSTRACT

Ovarian cancer is an aggressive tumor owing to its ability to metastasize from stage II onward. Herein, lipid nanoparticles (LNPs) that encapsulate combination of small interfering RNAs (siRNAs), polo-like kinase-1 (PLK1), and eukaryotic translation-initiation factor 3c (eIF3c), to target different cellular pathways essential for ovarian cancer progression are generated. The LNPs are further modified with hyaluronan (tNPs) to target cluster of differentiation 44 (CD44) expressing cells. Interestingly, hyaluronan-coated LNPs (tNPs) prolong functional activity and reduce growth kinetics of spheroids in in vitro assay as compared to uncoated LNPs (uNPs) due to ≈1500-fold higher expression of CD44. Treatment of 2D and 3D cultured ovarian cancer cells with LNPs encapsulating both siRNAs result in 85% cell death and robust target gene silencing. In advanced orthotopic ovarian cancer model, intraperitoneal administration of LNPs demonstrates CD44 specific tumor targeting of tNPs compared to uNPs and robust gene silencing in tissues involved in ovarian cancer pathophysiology. At very low siRNA dose, enhanced overall survival of 60% for tNPs treated mice is observed compared to 10% and 20% for single siRNA-, eIF3c-tNP, and PLK1-tNP treatment groups, respectively. Overall, LNPs represent promising platform in the treatment of advanced ovarian cancer by improving median- and overall-survival.


Subject(s)
Nanoparticles , Ovarian Neoplasms , Animals , Female , Gene Silencing , Humans , Lipids , Mice , RNA, Small Interfering
9.
Mol Ther ; 29(7): 2239-2252, 2021 07 07.
Article in English | MEDLINE | ID: mdl-33744469

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression by post-transcriptional inhibition of target genes. Proangiogenic small extracellular vesicles (sEVs; popularly identified with the name "exosomes") with a composite cargo of miRNAs are secreted by cultured stem cells and present in human biological fluids. Lipid nanoparticles (LNPs) represent an advanced platform for clinically approved delivery of RNA therapeutics. In this study, we aimed to (1) identify the miRNAs responsible for sEV-induced angiogenesis; (2) develop the prototype of bioinspired "artificial exosomes" (AEs) combining LNPs with a proangiogenic miRNA, and (3) validate the angiogenic potential of the bioinspired AEs. We previously reported that human sEVs from bone marrow (BM)-CD34+ cells and pericardial fluid (PF) are proangiogenic. Here, we have shown that sEVs secreted from saphenous vein pericytes and BM mesenchymal stem cells also promote angiogenesis. Analysis of miRNA datasets available in-house or datamined from GEO identified the let-7 family as common miRNA signature of the proangiogenic sEVs. LNPs with either hsa-let-7b-5p or cyanine 5 (Cy5)-conjugated Caenorhabditis elegans miR-39 (Cy5-cel-miR-39; control miRNA) were prepared using microfluidic micromixing. let-7b-5p-AEs did not cause toxicity and transferred functionally active let-7b-5p to recipient endothelial cells (ECs). let-7b-AEs also improved EC survival under hypoxia and angiogenesis in vitro and in vivo. Bioinspired proangiogenic AEs could be further developed into innovative nanomedicine products targeting ischemic diseases.


Subject(s)
Exosomes/metabolism , Extracellular Vesicles/metabolism , Liposomes/chemistry , MicroRNAs/metabolism , Nanoparticles/chemistry , Neovascularization, Physiologic , Pericardial Fluid/physiology , Animals , Exosomes/genetics , Extracellular Vesicles/genetics , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Mice , MicroRNAs/genetics
10.
Nanoscale ; 12(3): 1894-1903, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31904048

ABSTRACT

The failure of cancer therapies in clinical settings is often attributed to the lack of a relevant tumor model and pathological heterogeneity across tumor types in the clinic. The objective of this study was to develop a robust in vivo tumor model that better represents clinical tumors for the evaluation of anti-cancer therapies. We successfully developed a simple mouse tumor model based on 3D cell culture by injecting a single spheroid and compared it to a tumor model routinely used by injecting cell suspension from 2D monolayer cell culture. We further characterized both tumors with cellular markers for the presence of myofibroblasts, pericytes, endothelial cells and extracellular matrix to understand the role of the tumor microenvironment. We further investigated the effect of chemotherapy (doxorubicin), nanomedicine (Doxil®), biological therapy (Avastin®) and their combination. Our results showed that the substantial blood vasculature in the 3D spheroid model enhances the delivery of Doxil® by 2.5-fold as compared to the 2D model. Taken together, our data suggest that the 3D tumors created by simple subcutaneous spheroid injection represents a robust and more vascular murine tumor model which is a clinically relevant platform to test anti-cancer therapy in solid tumors.


Subject(s)
Bevacizumab/pharmacology , Doxorubicin/analogs & derivatives , Neoplasms, Experimental , Neovascularization, Pathologic , Ovarian Neoplasms , Spheroids, Cellular , Animals , Cell Line, Tumor , Doxorubicin/pharmacology , Female , Heterografts , Humans , Mice , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Polyethylene Glycols/pharmacology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
11.
J Control Release ; 313: 33-41, 2019 11 10.
Article in English | MEDLINE | ID: mdl-31634546

ABSTRACT

Interferon regulatory factor 8 (IRF8) protein plays a critical role in the differentiation, polarization, and activation of mononuclear phagocytic cells. In light of previous studies, we explored the therapeutic potential of IRF8 inhibition as immunomodulatory therapy for inflammatory bowel disease (IBD). To this end, we utilized siRNA-loaded lipid-based nanoparticles (siLNPs) and demonstrated a ∼90% reduction of IRF8 mRNA levels in vitro (PV < 0.0001), alongside a notable reduction in IRF8 protein. Moreover, silencing IRF8 ex vivo in splenocytes lead to a profound downregulation of IRF8 protein, followed by an immunomodulatory effect, as represented by a decrease in the secretion of TNFα, IL6 and IL12/IL23 (IL12p40) proinflammatory cytokines (PV = 0.0045, 0.0330, <0.0001, respectively). In order to silence IRF8 in vivo, selectively in inflammatory leukocytes, we used siLNPs that were coated with anti-Ly6C antibodies via our recently published ASSET targeting approach. Through this strategy, we have demonstrated a selective binding of the targeted-LNPs (T-LNPs) to Ly6C + inflammatory leukocytes. Finally, an immunomodulatory effect was demonstrated in vivo in an IBD mouse model with a profound decrease of TNFα, IL6, IL12/IL23, and IL1ß pro-inflammatory cytokines (n = 5, PV < 0.0001, <0.0001, <0.0001, 0.02, respectively) and an improvement of colon-morphology as assessed by colon-length measurements and colonoscopy (PV < 0.0001). Overall, using antibody-targeted siLNPs, we showed a notable reduction of IRF8 mRNA and protein and demonstrated a targeted immunomodulation therapeutic effect ex vivo and in vivo, in the DSS colitis model. We claim that a selective silencing of IRF8 in inflammatory leukocytes (such as Ly6C+) may serve as a therapeutic approach for treating inflammatory disorders.


Subject(s)
Anti-Inflammatory Agents/metabolism , Inflammatory Bowel Diseases/therapy , Interferon Regulatory Factors/genetics , Leukocytes/metabolism , Lipids/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/metabolism , Animals , Antibodies/chemistry , Antibodies/metabolism , Cholesterol/chemistry , Disease Models, Animal , Female , Genetic Therapy , Humans , Immunomodulation , Interferon Regulatory Factors/metabolism , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , RAW 264.7 Cells , Surface Properties , Transfection , Tumor Necrosis Factor-alpha/metabolism
12.
Nat Commun ; 9(1): 4493, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30374059

ABSTRACT

Therapeutic alteration of gene expression in vivo can be achieved by delivering nucleic acids (e.g., mRNA, siRNA) using nanoparticles. Recent progress in modified messenger RNA (mmRNA) synthesis facilitated the development of lipid nanoparticles (LNPs) loaded with mmRNA as a promising tool for in vivo protein expression. Although progress have been made with mmRNA-LNPs based protein expression in hepatocytes, cell specificity is still a major challenge. Moreover, selective protein expression is essential for an improved therapeutic effect, due to the heterogeneous nature of diseases. Here, we present a precision protein expression strategy in Ly6c+ inflammatory leukocytes in inflammatory bowel disease (IBD) induced mice. We demonstrate a therapeutic effect in an IBD model by targeted expression of the interleukin 10 in Ly6c+ inflammatory leukocytes. A selective mmRNA expression strategy has tremendous therapeutic potential in IBD and can ultimately become a novel therapeutic modality in many other diseases.


Subject(s)
Drug Delivery Systems , Interleukin-10/therapeutic use , Leukocytes/metabolism , Animals , Antigens, Ly/metabolism , Colitis/drug therapy , Colitis/pathology , Disease Models, Animal , Female , Lipids/chemistry , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanoparticles/ultrastructure , RAW 264.7 Cells , RNA, Messenger/metabolism
13.
Nat Nanotechnol ; 13(3): 214-219, 2018 03.
Article in English | MEDLINE | ID: mdl-29379205

ABSTRACT

Previous studies have identified relevant genes and signalling pathways that are hampered in human disorders as potential candidates for therapeutics. Developing nucleic acid-based tools to manipulate gene expression, such as short interfering RNAs1-3 (siRNAs), opens up opportunities for personalized medicine. Yet, although major progress has been made in developing siRNA targeted delivery carriers, mainly by utilizing monoclonal antibodies (mAbs) for targeting4-8, their clinical translation has not occurred. This is in part because of the massive development and production requirements and the high batch-to-batch variability of current technologies, which rely on chemical conjugation. Here we present a self-assembled modular platform that enables the construction of a theoretically unlimited repertoire of siRNA targeted carriers. The self-assembly of the platform is based on a membrane-anchored lipoprotein that is incorporated into siRNA-loaded lipid nanoparticles that interact with the antibody crystallizable fragment (Fc) domain. We show that a simple switch of eight different mAbs redirects the specific uptake of siRNAs by diverse leukocyte subsets in vivo. The therapeutic potential of the platform is demonstrated in an inflammatory bowel disease model by targeting colon macrophages to reduce inflammatory symptoms, and in a Mantle Cell Lymphoma xenograft model by targeting cancer cells to induce cell death and improve survival. This modular delivery platform represents a milestone in the development of precision medicine.


Subject(s)
Colitis/therapy , Drug Delivery Systems/methods , Liposomes/chemistry , RNA, Small Interfering/administration & dosage , RNAi Therapeutics/methods , Animals , Antibodies, Monoclonal/chemistry , Colitis/genetics , Female , Lipoproteins/chemistry , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/therapeutic use
14.
Bioconjug Chem ; 28(2): 556-564, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28040889

ABSTRACT

The immunocompability of polyelectrolyte capsules synthesized by layer-by-layer deposition has been investigated. Capsules of different architecture and composed of either non-degradable or biodegradable polymers, with either positively or negatively charged outer surface, and with micrometer size, have been used, and the capsule uptake by different cell lines has been studied and quantified. Immunocompatibility studies were performed with peripheral blood mononuclear cells (PBMCs). Data demonstrate that incubation with capsules, at concentrations relevant for practical applications, did not result in a reduced viability of cells, as it did not show an increased apoptosis. Presence of capsules also did not result in an increased expression of TNF-α, as detected with antibody staining, as well as at mRNA level. It also did not result in increased expression of IL-6, as detected at mRNA level. These results indicate that the polyelectrolyte capsules used in this study are immunocompatible.


Subject(s)
Cell Survival/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Polyelectrolytes/adverse effects , A549 Cells , Apoptosis/drug effects , Capsules , Cell Line , Cells, Cultured , Humans , Leukocytes, Mononuclear/metabolism , Polyelectrolytes/pharmacokinetics , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
15.
Proc Natl Acad Sci U S A ; 113(1): E16-22, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26699502

ABSTRACT

Despite progress in systemic small interfering RNA (siRNA) delivery to the liver and to solid tumors, systemic siRNA delivery to leukocytes remains challenging. The ability to silence gene expression in leukocytes has great potential for identifying drug targets and for RNAi-based therapy for leukocyte diseases. However, both normal and malignant leukocytes are among the most difficult targets for siRNA delivery as they are resistant to conventional transfection reagents and are dispersed in the body. We used mantle cell lymphoma (MCL) as a prototypic blood cancer for validating a novel siRNA delivery strategy. MCL is an aggressive B-cell lymphoma that overexpresses cyclin D1 with relatively poor prognosis. Down-regulation of cyclin D1 using RNA interference (RNAi) is a potential therapeutic approach to this malignancy. Here, we designed lipid-based nanoparticles (LNPs) coated with anti-CD38 monoclonal antibodies that are specifically taken up by human MCL cells in the bone marrow of xenografted mice. When loaded with siRNAs against cyclin D1, CD38-targeted LNPs induced gene silencing in MCL cells and prolonged survival of tumor-bearing mice with no observed adverse effects. These results highlight the therapeutic potential of cyclin D1 therapy in MCL and present a novel RNAi delivery system that opens new therapeutic opportunities for treating MCL and other B-cell malignancies.


Subject(s)
B-Lymphocytes/immunology , Lymphoma, B-Cell/therapy , Lymphoma, Mantle-Cell/therapy , Nanomedicine/methods , RNA Interference , RNA, Small Interfering/administration & dosage , ADP-ribosyl Cyclase 1/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Cyclin D1/genetics , Down-Regulation , Gene Silencing , Humans , Lipids , Lymphoma, B-Cell/immunology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/immunology , Mice , Nanoparticles , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
16.
ACS Nano ; 9(7): 6706-16, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26042619

ABSTRACT

Modulating T cell function by down-regulating specific genes using RNA interference (RNAi) holds tremendous potential in advancing targeted therapies in many immune-related disorders including cancer, inflammation, autoimmunity, and viral infections. Hematopoietic cells, in general, and primary T lymphocytes, in particular, are notoriously hard to transfect with small interfering RNAs (siRNAs). Herein, we describe a novel strategy to specifically deliver siRNAs to murine CD4(+) T cells using targeted lipid nanoparticles (tLNPs). To increase the efficacy of siRNA delivery, these tLNPs have been formulated with several lipids designed to improve the stability and efficacy of siRNA delivery. The tLNPs were surface-functionalized with anti-CD4 monoclonal antibody to permit delivery of the siRNAs specifically to CD4(+) T lymphocytes. Ex vivo, tLNPs demonstrated specificity by targeting only primary CD4(+) T lymphocytes and no other cell types. Systemic intravenous administration of these particles led to efficient binding and uptake into CD4(+) T lymphocytes in several anatomical sites including the spleen, inguinal lymph nodes, blood, and the bone marrow. Silencing by tLNPs occurs in a subset of circulating and resting CD4(+) T lymphocytes. Interestingly, we show that tLNP internalization and not endosome escape is a fundamental event that takes place as early as 1 h after systemic administration and determines tLNPs' efficacy. Taken together, these results suggest that tLNPs may open new avenues for the manipulation of T cell functionality and may help to establish RNAi as a therapeutic modality in leukocyte-associated diseases.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Gene Silencing , Nanoparticles/metabolism , RNA, Small Interfering/administration & dosage , Animals , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Lipids/chemistry , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , RNAi Therapeutics/methods
17.
ACS Nano ; 9(2): 1581-91, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25558928

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most infiltrating, aggressive, and poorly treated brain tumors. Progress in genomics and proteomics has paved the way for identifying potential therapeutic targets for treating GBM, yet the vast majority of these leading drug candidates for the treatment of GBM are ineffective, mainly due to restricted passages across the blood-brain barrier. Nanoparticles have been emerged as a promising platform to treat different types of tumors due to their ability to transport drugs to target sites while minimizing adverse effects. Herein, we devised a localized strategy to deliver RNA interference (RNAi) directly to the GBM site using hyaluronan (HA)-grafted lipid-based nanoparticles (LNPs). These LNPs having an ionized lipid were previously shown to be highly effective in delivering small interfering RNAs (siRNAs) into various cell types. LNP's surface was functionalized with hyaluronan (HA), a naturally occurring glycosaminoglycan that specifically binds the CD44 receptor expressed on GBM cells. We found that HA-LNPs can successfully bind to GBM cell lines and primary neurosphers of GBM patients. HA-LNPs loaded with Polo-Like Kinase 1 (PLK1) siRNAs (siPLK1) dramatically reduced the expression of PLK1 mRNA and cumulated in cell death even under shear flow that simulate the flow of the cerebrospinal fluid compared with control groups. Next, a human GBM U87MG orthotopic xenograft model was established by intracranial injection of U87MG cells into nude mice. Convection of Cy3-siRNA entrapped in HA-LNPs was performed, and specific Cy3 uptake was observed in U87MG cells. Moreover, convection of siPLK1 entrapped in HA-LNPs reduced mRNA levels by more than 80% and significantly prolonged survival of treated mice in the orthotopic model. Taken together, our results suggest that RNAi therapeutics could effectively be delivered in a localized manner with HA-coated LNPs and ultimately may become a therapeutic modality for GBM.


Subject(s)
Drug Resistance, Neoplasm , Glioblastoma/genetics , Glioblastoma/therapy , Hyaluronic Acid/chemistry , Lipids/chemistry , Nanoparticles/chemistry , RNAi Therapeutics/methods , Animals , Biological Transport , Cell Cycle Proteins/deficiency , Cell Death/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Drug Carriers/chemistry , Drug Carriers/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Grading , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins/deficiency , Polo-Like Kinase 1
18.
ACS Nano ; 8(3): 1958-65, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24660817

ABSTRACT

The treatment of neurodegenerative diseases remains a tremendous challenge due to the limited access of molecules across the blood-brain barrier, especially large molecules such as peptides and proteins. As a result, at most, a small percentage of a drug that is administered systemically will reach the central nervous system in its active form. Currently, research in the field focuses on developing safer and more effective approaches to deliver peptides and proteins into the central nervous system. Multiple strategies have been developed for this purpose. However, noninvasive approaches, such as nanostructured protein delivery carriers and intranasal administration, seem to be the most promising strategies for the treatment of chronic diseases, which require long-term interventions. These approaches are both target-specific and able to rapidly bypass the blood-brain barrier. In this Perspective, we detail some of these strategies and discuss some of the potential pitfalls and opportunities in this field. The next generation strategies will most likely be more cell-type-specific. Devising these strategies to target the brain may ultimately become a novel therapeutic modality to treat neurodegenerative diseases.


Subject(s)
Nanomedicine/methods , Neurodegenerative Diseases/therapy , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Humans
19.
Nanoscale ; 6(7): 3742-52, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24569711

ABSTRACT

Hyaluronan (HA), a naturally occurring high Mw (HMw) glycosaminoglycan, has been shown to play crucial roles in cell growth, embryonic development, healing processes, inflammation, and tumor development and progression. Low Mw (LMw, <10 kDa) HA has been reported to provoke inflammatory responses, such as induction of cytokines, chemokines, reactive nitrogen species and growth factors. Herein, we prepared and characterized two types of HA coated (LMw and HMw) lipid-based targeted and stabilized nanoparticles (tsNPs) and tested their binding to tumor cells expressing the HA receptor (CD44), systemic immunotoxicity, and biodistribution in tumor bearing mice. In vitro, the Mw of the surface anchored HA had a significant influence on the affinity towards CD44 on B16F10 murine melanoma cells. LMw HA-tsNPs exhibited weak binding, while binding of tsNPs coated with HMw HA was characterized by high binding. Both types of tsNPs had no measured effect on cytokine induction in vivo following intravenous administration to healthy C57BL/6 mice suggesting no immune activation. HMw HA-tsNPs showed enhanced circulation time and tumor targeting specificity, mainly by accumulating in the tumor and its vicinity compared with LMw HA-tsNPs. Finally, we show that methotrexate (MTX), a drug commonly used in cancer chemotherapy, entrapped in HMw HA-tsNPs slowly diffused from the particles with a half-life of 13.75 days, and improved the therapeutic outcome in a murine B16F10 melanoma model compared with NPs suggesting an active cellular targeting beyond the Enhanced Permeability and Retention (EPR) effect. Taken together, these findings have major implications for the use of high molecular weight HA in nanomedicine as a selective and safe active cellular targeting moiety.


Subject(s)
Hyaluronic Acid/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Carbocyanines/chemistry , Cell Line, Tumor , Cytokines/metabolism , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Female , Half-Life , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/metabolism , Methotrexate/administration & dosage , Methotrexate/chemistry , Mice , Mice, Inbred C57BL , Molecular Weight , Nanomedicine , Nanoparticles/metabolism , Protein Binding , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tissue Distribution
20.
Cancer Lett ; 334(2): 221-7, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-22935680

ABSTRACT

RNA interference (RNAi), a natural cellular mechanism for RNA-guided regulation of gene expression could in fact become new therapeutic modality if an appropriate efficient delivery strategy that is also reproducible and safe will be developed. Numerous efforts have been made for the past eight years to address this challenge with only mild success. The majority of these strategies are based on cationic formulations that condense the RNAi payload and deliver it into the cell cytoplasm. However, most of these formulations also evoke adverse effects such as mitochondrial damage, interfering with blood coagulation cascade, induce interferon response, promote cytokine induction and activate the complement. Herein, we present a strategy that is devised from neutral phospholipids and cholesterol that self-assembled into lipid-based nanoparticles (LNPs). These LNPs were then coated with the glycosaminoglycan, hyaluronan (HA). HA-LNPs bound and internalized specifically into cancer cells compared with control, non-coated particles. Next, loaded with siRNAs against the multidrug resistance extrusion pump, p-glycoprotein (P-gp), HA-LNPs efficiently and specifically reduced mRNA and P-gp protein levels compared with control particles and with HA-LNPs loaded with control, non-targeted siRNAs. In addition, no cellular toxicity or cytokine induction was observed when these particles were cultured with human Peripheral Blood Mononuclear Cells (PBMCs). The HA-LNPs may offer an alternative approach to cationic lipid-based formulations for RNAi delivery into cancer cells in an efficient and safe manner.


Subject(s)
Hyaluronic Acid/administration & dosage , Lipids/administration & dosage , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Transfection/methods , Humans , Hyaluronic Acid/chemistry , Lipids/chemistry , Nanoparticles , Neoplasms/genetics , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...