Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Elias Quijano is active.

Publication


Featured researches published by Elias Quijano.


Journal of Controlled Release | 2012

Surface modified poly(β amino ester)-containing nanoparticles for plasmid DNA delivery.

Rachel J. Fields; Christopher J. Cheng; Elias Quijano; Caroline E. Weller; Nina Kristofik; Nha Duong; Christopher J. Hoimes; Marie E. Egan; W. Mark Saltzman

The use of biodegradable polymers provides a potentially safe and effective alternative to viral and liposomal vectors for the delivery of plasmid DNA to cells for gene therapy applications. In this work we describe the formulation of a novel nanoparticle (NP) system containing a blend of poly(lactic-co-glycolic acid) and a representative poly(beta-amino) ester (PLGA and PBAE respectively) for use as gene delivery vehicles. Particles of different weight/weight (wt/wt) ratios of the two polymers were characterized for size, morphology, plasmid DNA (pDNA) loading and surface charge. NPs containing PBAE were more effective at cellular internalization and transfection (COS-7 and CFBE41o-) than NPs lacking the PBAE polymer. However, along with these delivery benefits, PBAE exhibited cytotoxic effects that presented an engineering challenge. Surface coating of these blended particles with the cell-penetrating peptides (CPPs) mTAT, bPrPp and MPG via a PEGylated phospholipid linker (DSPE-PEG2000) resulted in NPs that reduced surface charge and cellular toxicity to levels comparable with NPs formulated with only PLGA. Additionally, these coated nanoparticles showed an improvement in pDNA loading, intracellular uptake and transfection efficiency, when compared to NPs lacking the surface coating. Although all particles with a CPP coating outperformed unmodified NPs, respectively, bPrPp and MPG coating resulted in 3 and 4.5× more pDNA loading than unmodified particles and approximately an order of magnitude improvement on transfection efficiency in CFBE41o- cells. These results demonstrate that surface-modified PBAE containing NPs are a highly effective and minimally toxic platform for pDNA delivery.


Nature Communications | 2015

Nanoparticles that deliver triplex-forming peptide nucleic acid molecules correct F508del CFTR in airway epithelium

Nicole Ali McNeer; Kavitha Anandalingam; Rachel J. Fields; Christina Caputo; Sascha Kopic; Anisha Gupta; Elias Quijano; Lee A. Polikoff; Yong Kong; Raman Bahal; John P. Geibel; Peter M. Glazer; W. Mark Saltzman; Marie E. Egan

Cystic fibrosis (CF) is a lethal genetic disorder most commonly caused by the F508del mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. It is not readily amenable to gene therapy because of its systemic nature and challenges including in vivo gene delivery and transient gene expression. Here, we use triplex-forming PNA molecules and donor DNA in biodegradable polymer nanoparticles to correct F508del. We confirm modification with sequencing and a functional chloride efflux assay. In vitro correction of chloride efflux occurs in up to 25% of human cells. Deep sequencing reveals negligible off-target effects in partially homologous sites. Intranasal application of nanoparticles in CF mice produces changes in nasal epithelium potential differences consistent with corrected CFTR, with gene correction also detected in lung tissue. This work represents facile genome engineering in vivo with oligonucleotides using a nanoparticle system to achieve clinically relevant levels of gene editing without off-target effects.Cystic fibrosis (CF) is a lethal genetic disorder most commonly caused by the F508del mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. It is not readily amenable to gene therapy because of its systemic nature and challenges including in vivo gene delivery and transient gene expression. Here we use triplex-forming peptide nucleic acids and donor DNA in biodegradable polymer nanoparticles to correct F508del. We confirm modification with sequencing and a functional chloride efflux assay. In vitro correction of chloride efflux occurs in up to 25% of human cells. Deep-sequencing reveals negligible off-target effects in partially homologous sites. Intranasal delivery of nanoparticles in CF mice produces changes in the nasal epithelium potential difference assay, consistent with corrected CFTR function. Also, gene correction is detected in the nasal and lung tissue. This work represents facile genome engineering in vivo with oligonucleotides using a nanoparticle system to achieve clinically relevant levels of gene editing without off-target effects.


Nature Communications | 2016

In vivo correction of anaemia in β-thalassemic mice by γPNA-mediated gene editing with nanoparticle delivery

Raman Bahal; Nicole Ali McNeer; Elias Quijano; Yanfeng Liu; Parker Sulkowski; Audrey Turchick; Yi-Chien Lu; Dinesh C. Bhunia; Arunava Manna; Dale L. Greiner; Michael A. Brehm; Christopher J. Cheng; Francesc López-Giráldez; Adele S. Ricciardi; Diane S. Krause; Priti Kumar; Patrick G. Gallagher; Demetrios T. Braddock; W. Mark Saltzman; Danith H. Ly; Peter M. Glazer

The blood disorder, β-thalassaemia, is considered an attractive target for gene correction. Site-specific triplex formation has been shown to induce DNA repair and thereby catalyse genome editing. Here we report that triplex-forming peptide nucleic acids (PNAs) substituted at the γ position plus stimulation of the stem cell factor (SCF)/c-Kit pathway yielded high levels of gene editing in haematopoietic stem cells (HSCs) in a mouse model of human β-thalassaemia. Injection of thalassemic mice with SCF plus nanoparticles containing γPNAs and donor DNAs ameliorated the disease phenotype, with sustained elevation of blood haemoglobin levels into the normal range, reduced reticulocytosis, reversal of splenomegaly and up to 7% β-globin gene correction in HSCs, with extremely low off-target effects. The combination of nanoparticle delivery, next generation γPNAs and SCF treatment may offer a minimally invasive treatment for genetic disorders of the blood that can be achieved safely and simply by intravenous administration.


Journal of Controlled Release | 2016

Distribution of polymer nanoparticles by convection-enhanced delivery to brain tumors.

Jennifer K. Saucier-Sawyer; Young-Eun Seo; Alice Gaudin; Elias Quijano; Eric Song; Andrew J. Sawyer; Yang Deng; Anita Huttner; W. Mark Saltzman

Glioblastoma multiforme (GBM) is a fatal brain tumor characterized by infiltration beyond the margins of the main tumor mass and local recurrence after surgery. The blood-brain barrier (BBB) poses the most significant hurdle to brain tumor treatment. Convection-enhanced delivery (CED) allows for local administration of agents, overcoming the restrictions of the BBB. Recently, polymer nanoparticles have been demonstrated to penetrate readily through the healthy brain when delivered by CED, and size has been shown to be a critical factor for nanoparticle penetration. Because these brain-penetrating nanoparticles (BPNPs) have high potential for treatment of intracranial tumors since they offer the potential for cell targeting and controlled drug release after administration, here we investigated the intratumoral CED infusions of PLGA BPNPs in animals bearing either U87 or RG2 intracranial tumors. We demonstrate that the overall volume of distribution of these BPNPs was similar to that observed in healthy brains; however, the presence of tumors resulted in asymmetric and heterogeneous distribution patterns, with substantial leakage into the peritumoral tissue. Together, our results suggest that CED of BPNPs should be optimized by accounting for tumor geometry, in terms of location, size and presence of necrotic regions, to determine the ideal infusion site and parameters for individual tumors.


Current Gene Therapy | 2014

Single-stranded γPNAs for in vivo site-specific genome editing via Watson-Crick recognition.

Raman Bahal; Elias Quijano; Nicole Ali McNeer; Yanfeng Liu; Dinesh C. Bhunia; Francesco Lopez-Giraldez; Rachel J. Fields; William Mark Saltzman; Danith H. Ly; Peter M. Glazer

Triplex-forming peptide nucleic acids (PNAs) facilitate gene editing by stimulating recombination of donor DNAs within genomic DNA via site-specific formation of altered helical structures that further stimulate DNA repair. However, PNAs designed for triplex formation are sequence restricted to homopurine sites. Herein we describe a novel strategy where next generation single-stranded gamma PNAs (γPNAs) containing miniPEG substitutions at the gamma position can target genomic DNA in mouse bone marrow at mixed-sequence sites to induce targeted gene editing. In addition to enhanced binding, γPNAs confer increased solubility and improved formulation into poly(lactic-co-glycolic acid) (PLGA) nanoparticles for efficient intracellular delivery. Single-stranded γPNAs induce targeted gene editing at frequencies of 0.8% in mouse bone marrow cells treated ex vivo and 0.1% in vivo via IV injection, without detectable toxicity. These results suggest that γPNAs may provide a new tool for induced gene editing based on Watson-Crick recognition without sequence restriction.


Advanced Healthcare Materials | 2015

Modified Poly(lactic-co-glycolic Acid) Nanoparticles for Enhanced Cellular Uptake and Gene Editing in the Lung

Rachel J. Fields; Elias Quijano; Nicole Ali McNeer; Christina Caputo; Raman Bahal; Kavi Anandalingam; Marie E. Egan; Peter M. Glazer; W. Mark Saltzman

Surface-modified poly(lactic-co-glycolic acid) (PLGA)/poly(β-aminoester)(PBAE)nanoparticles (NPs) have shown great promise in gene delivery. In this work, the pulmonary cellular uptake of these NPs is evaluated and surface-modified PLGA/PBAE NPs are shown to achieve higher cellular association and gene editing than traditional NPs composed of PLGA or PLGA/PBAE blends alone.


Proceedings of the National Academy of Sciences of the United States of America | 2018

From in silico hit to long-acting late-stage preclinical candidate to combat HIV-1 infection

Shalley N. Kudalkar; Elias Quijano; Krasimir A. Spasov; Won-Gil Lee; José A. Cisneros; W. Mark Saltzman; Priti Kumar; William L. Jorgensen; Karen S. Anderson

Significance Nonnucleoside reverse transcriptase inhibitors (NNRTIs) are essential components of highly active antiretroviral therapy; however, concerns about poor pharmacological properties, dose restriction because of toxicity, and drug resistance have limited treatment options. Our computational and structure-guided design studies for lead optimization have transformed a 5 µM virtual screening hit into a class of NNRTIs with remarkable potency, safety, drug resistance profile, and pharmacological properties. We report a representative, compound I, with marked synergy with existing HIV-1 drugs and antiviral efficacy in HIV-1–infected humanized mice. A single dose of long-acting nanoformulation of compound I retains sustained levels and efficacy for ∼3 weeks, confirming potential as a late-stage preclinical candidate. Additionally, these properties of compound I suggest that it may be a promising candidate to evaluate for preexposure prophylaxis. The HIV-1 pandemic affecting over 37 million people worldwide continues, with nearly one-half of the infected population on highly active antiretroviral therapy (HAART). Major therapeutic challenges remain because of the emergence of drug-resistant HIV-1 strains, limitations because of safety and toxicity with current HIV-1 drugs, and patient compliance for lifelong, daily treatment regimens. Nonnucleoside reverse transcriptase inhibitors (NNRTIs) that target the viral polymerase have been a key component of the current HIV-1 combination drug regimens; however, these issues hamper them. Thus, the development of novel more effective NNRTIs as anti–HIV-1 agents with fewer long-term liabilities, efficacy on new drug-resistant HIV-1 strains, and less frequent dosing is crucial. Using a computational and structure-based design strategy to guide lead optimization, a 5 µM virtual screening hit was transformed to a series of very potent nanomolar to picomolar catechol diethers. One representative, compound I, was shown to have nanomolar activity in HIV-1–infected T cells, potency on clinically relevant HIV-1 drug-resistant strains, lack of cytotoxicity and off-target effects, and excellent in vivo pharmacokinetic behavior. In this report, we show the feasibility of compound I as a late-stage preclinical candidate by establishing synergistic antiviral activity with existing HIV-1 drugs and clinical candidates and efficacy in HIV-1–infected humanized [human peripheral blood lymphocyte (Hu-PBL)] mice by completely suppressing viral loads and preventing human CD4+ T-cell loss. Moreover, a long-acting nanoformulation of compound I [compound I nanoparticle (compound I-NP)] in poly(lactide-coglycolide) (PLGA) was developed that shows sustained maintenance of plasma drug concentrations and drug efficacy for almost 3 weeks after a single dose.


Molecules | 2018

Peptide Nucleic Acids as a Tool for Site-Specific Gene Editing

Adele S. Ricciardi; Elias Quijano; Rachael Putman; William Mark Saltzman; Peter M. Glazer

Peptide nucleic acids (PNAs) can bind duplex DNA in a sequence-targeted manner, forming a triplex structure capable of inducing DNA repair and producing specific genome modifications. Since the first description of PNA-mediated gene editing in cell free extracts, PNAs have been used to successfully correct human disease-causing mutations in cell culture and in vivo in preclinical mouse models. Gene correction via PNAs has resulted in clinically-relevant functional protein restoration and disease improvement, with low off-target genome effects, indicating a strong therapeutic potential for PNAs in the treatment or cure of genetic disorders. This review discusses the progress that has been made in developing PNAs as an effective, targeted agent for gene editing, with an emphasis on recent in vivo, nanoparticle-based strategies.


Molecular therapy. Nucleic acids | 2017

Anti-tumor Activity of miniPEG-γ-Modified PNAs to Inhibit MicroRNA-210 for Cancer Therapy

Anisha Gupta; Elias Quijano; Yanfeng Liu; Raman Bahal; Susan E. Scanlon; Eric Song; Wei-Che Hsieh; Demetrios E. Braddock; Danith H. Ly; W. Mark Saltzman; Peter M. Glazer

MicroRNAs (miRs) are frequently overexpressed in human cancers. In particular, miR-210 is induced in hypoxic cells and acts to orchestrate the adaptation of tumor cells to hypoxia. Silencing oncogenic miRs such as miR-210 may therefore offer a promising approach to anticancer therapy. We have developed a miR-210 inhibition strategy based on a new class of conformationally preorganized antisense γ peptide nucleic acids (γPNAs) that possess vastly superior RNA-binding affinity, improved solubility, and favorable biocompatibility. For cellular delivery, we encapsulated the γPNAs in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs). Our results show that γPNAs targeting miR-210 cause significant delay in growth of a human tumor xenograft in mice compared to conventional PNAs. Further, histopathological analyses show considerable necrosis, fibrosis, and reduced cell proliferation in γPNA-treated tumors compared to controls. Overall, our work provides a chemical framework for a novel anti-miR therapeutic approach using γPNAs that should facilitate rational design of agents to potently inhibit oncogenic microRNAs.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Reply to Pandey et al.: Understanding the efficacy of a potential antiretroviral drug candidate in humanized mouse model of HIV infection

Shalley N. Kudalkar; Elias Quijano; Krasimir A. Spasov; Won-Gil Lee; José A. Cisneros; W. Mark Saltzman; Priti Kumar; William L. Jorgensen; Karen S. Anderson

We appreciate Pandey et al.’s (1) interest in further understanding our published work (2). Our responses are given below the excerpts from the Letter.Our first and foremost concern is the data presented in their figure 6C. We fail to understand why there was a fall in the plasma viral loads after day 19 in the control group (i.e., the group not given any intervention). Furthermore, not only was the fall in the plasma viral load significant (to below the limit of detection; <150 copies per milliliter) but also it was more rapid (showing a steeper decline) compared with the treated groups by day 32. If the viral loads are decreasing so rapidly and significantly in the noninterventional (control group), can we draw conclusions about the efficacy of … [↵][1]1To whom correspondence should be addressed. Email: william.jorgensen{at}yale.edu. [1]: #xref-corresp-1-1

Collaboration


Dive into the Elias Quijano's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raman Bahal

Carnegie Mellon University

View shared research outputs
Top Co-Authors

Avatar

Danith H. Ly

Carnegie Mellon University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge