You are on page 1of 8

Available online at www.sciencedirect.

com

ScienceDirect

Computational approaches in design of nucleic


acid-based therapeutics
Mark Sherman1 and Lydia Contreras2

Recent advances in computational and experimental methods libraries for target protein inhibition that lead to thera-
have led to novel avenues for therapeutic development. peutic effects. While these efforts have resulted in hun-
Utilization of nucleic acids as therapeutic agents and/or targets dreds of new drug entities, the journey to approval for the
has been recently gaining attention due to their potential as average drug is increasingly costly [1]. The rising cost of
high-affinity, selective molecular building blocks for various drug development can, in part, be attributed to increased
therapies. Notably, development of computational algorithms screening efforts due to low hit rates and to misdirected
for predicting accessible RNA binding sites, identifying investigations based on proteins that show no interactions
therapeutic target sequences, modeling delivery into tissues, with current candidate drug molecule libraries; other
and designing binding aptamers have enhanced therapeutic reasons include the high probability of failure during
potential for this new drug category. Here, we review trends in the scale-up process and the escalating demand by regu-
drug development within the pharmaceutical industry and ways latory bodies for more safety and efficacy data prior to
by which nucleic acid-based drugs have arisen as effective approval [1,2]. Furthermore, many diseases simply can-
therapeutic candidates. In particular, we focus on not be influenced by small molecules due to the inability
computational and experimental approaches to nucleic acid- to identify a binding pocket in key proteins underlying
based drug design, commenting on challenges and outlooks disease pathways to serve as a suitable drug target. In
for future applications. addition, challenges to small molecules arise from the
possibility that disease phenotypes are caused by mecha-
Addresses nisms upstream of protein production, such as improper
1
Cell and Molecular Biology Graduate Program, University of Texas at gene splicing, without a suitable small molecule target.
Austin, 100 E. 24th Street, A6500, Austin, TX 78712, USA
2
McKetta Department of Chemical Engineering, University of Texas at
Austin, 200 E. Dean Keeton St., Stop C0400, Austin, TX 78712, USA
In recent years, drug development efforts have become
more sophisticated, relying less on large-scale screening
Corresponding author: Contreras, Lydia (lcontrer@utexas.edu) of chemical compounds and more on targeted develop-
ment of drugs toward specific molecular entities. Modern
drug development pipelines attempt to target a wider
Current Opinion in Biotechnology 2018, 53:232–239
spectrum of molecules beyond traditional enzyme targets,
This review comes from a themed issue on Pharmaceutical
including metabolites [3], protein–protein interactions
biotechnology
[4], and nucleic acids [5]. These advances have enabled
Edited by Amanda Lewis and Nripen Singh
treatment of various diseases through the development
and engineering of new vaccines (with over 30 active
vaccine-associated clinical trials currently underway (clin-
https://doi.org/10.1016/j.copbio.2017.12.001
icaltrials.gov)), peptide therapeutics such as Parsabiv [6],
nucleotide prodrugs such as Vosevi [7], as well as a variety
0958-1669/ã 2018 Published by Elsevier Ltd.
of antibody-based technologies including antibody–drug
conjugates (used in Mylotarg) [8,9], phage display (used
in development of Humira) [10,11,12], and radiolabelled
diagnostic antibodies as used in Netspot [13–15].

Introduction Incentives and challenges to nucleic-acid


Over almost 80 years of FDA regulation, the field of drug therapeutic development
discovery and development has transformed its focus from While protein-based therapeutics (antibodies, recombi-
pain management of disease symptoms to systematic nant proteins, and peptides) have made up over a quarter
identification of causative factors and targeted mediation of FDA approved drugs since 2015 (FDA.gov), they have
(Figure 1). Early pharmaceutical development stemmed relatively a short shelf life, require refrigerated transport,
from the isolation and concentration of active agents from and rely on live organisms for production, making
natural products by chance discovery. Although the ther- manufacturing them expensive, contamination prone,
apeutic mechanisms of these substances were unclear at and variable across batches [16]. In response to these
the time, most contemporary drug development efforts shortcomings, recent efforts have been made toward
aim to target specific disease-associated proteins. The development of nucleic acid-based drugs, offering the
majority of approaches involve screening small molecule key advantage of chemical manufacturing. As such, they

Current Opinion in Biotechnology 2018, 53:232–239 www.sciencedirect.com


Computational approaches in nucleic acid therapeutics Sherman and Contreras 233

Figure 1

1939-1959 1960-1999 1990-2017


N= 27 N=111 N=357
Current Opinion in Biotechnology

An analysis of indications and usage descriptions from FDA approved drug labels across three time periods from 1939 to 2017. Between
1939 and 1959, approved drugs appear to be associated with pain management and symptomatic relief. Between 1960 and 1989, small molecule
inhibitors appear to become mainstream and birth control is developed. Between 1990 and 2017, inhibition continued to be a major theme for
pharmaceutical development and drugs became more specific toward particular diseases, including viruses such as HIV. During the last 30 years,
the emergence of nucleic acids as targets (nucleosides) also represents a newer phenomenon. Data generated by an analysis on a collection of
drug labels through U.S. National Library of Medicine’s DailyMed website.

maintain a relatively long half life, are easily transport- infusion of chimeric antigen receptor (CAR) T cells
able, are consistent from batch to batch, and have the while others are siRNA (small interfering RNA), shRNA
potential to introduce new therapeutic chemistries [17] to (small hairpin RNA), and mRNA (messenger RNA)
the current, inhibitor-dominated field (Figure 1). For based. CAR T cells are derived from the patients own
instance, DNA has been identified as a target for drug T cells which are genetically modified ex vivo and intro-
development [18] for its gene therapy ability via manip- duced back into the body with enhanced therapeutic
ulation of gene expression or editing of defective genes functions. Currently approved RNA-based drugs are
[19]. Likewise, RNA variants have been implicated in a administered as local injections or systemic infusions
variety of disease phenotypes such as Spinal Muscular (Table 1). As research in nucleic acid-based therapeutics
Atrophy (SMA) [20], Duchenne Muscular Dystrophy progresses, it holds promise to develop drugs that can not
(DMD) [21,22], Alzheimer’s Disease, and cancer only inhibit action of a protein, but could alter gene
[23,24]. Therefore, development of nucleic acid-based expression and metabolic flux through development of
drugs hold promise to provide high affinity and high aptamers (RNA analogs of antibodies), prodrugs,
specificity entities for therapeutic applications; however, siRNAs, miRNAs, and new vaccines with more efficient
progress in this field has been limited by challenges in modes of delivery.
delivery to specific tissues, a very active area of research
(discussed later) [25–31]. Current strategies toward aptamer-based
therapeutic development
In 2016, three nucleic acid-based therapies gained FDA Due to their ability to bind to diverse targets such as ions,
approval: Exondys-51 [22] (for treatment of DMD), dyes, amino acids, RNAs, oligosaccharides, antibodies,
Spinraza [20] (for treatment of SMA), and Defitelio [32] and cells, aptamers have become the main focus of
(for treatment of renal or pulmonary dysfunction). Both nucleic acid-based therapeutic development [33]. Apta-
Spinraza and Exondys-51 are antisense oligonucleotides mers are short, single-stranded DNA or RNA oligonu-
that therapeutically influence splicing patterns of dis- cleotides that bind to a variety of target molecules with
eased proteins. The mechanism for Defitelio has not high affinity and specificity, dependent on their tertiary
been elucidated [32]. Several other RNA-based or structure [34]. The majority of DNA and RNA aptamers
RNA-targeting drugs are currently in clinical trials are experimentally developed via Systematic Evolution
(Table 1). The majority of these candidate drugs involve of Ligands by Exponential Enrichment (SELEX)

www.sciencedirect.com Current Opinion in Biotechnology 2018, 53:232–239


234 Pharmaceutical biotechnology

Table 1

Nucleic acid-based drugs recently approved or currently in clinical trials

Current Active ingredient/indicated study Indicated condition Mode of delivery Intervention


phase
Early Non-Viral, RNA-Redirected Hodgkin Lymphoma Intravenous (IV) infusion CD19 RNA redirected
Phase 1 Autologous T Cells autologous T-cells (RNA
CART19 cells)
Early Autologous T Cells Expressing MET Malignant Melanoma Breast Intravenous (IV) infusion T cells modified with RNA
Phase 1 scFv CAR (RNA CART-cMET) Cancer anti-cMET CAR
Phase 1 RNActive1 Rabies Vaccine (CV7201) Rabies Intravenous (IV) injection mRNA
Phase 1 Hypoxia-inducible Factor 1a (HIF1A) Carcinoma, Hepatocellular Intravenous (IV) infusion mRNA Antagonist
Phase 1 Pbi-shRNATM EWS/FLI1 Type 1 LPX Ewing’s Sarcoma Intravenous (IV) infusion pbi-shRNATM EWS/FLI1
Type 1 LPX
Phase 1 Neutral Liposomal Small Interfering Advanced Cancers Intravenous (IV) infusion siRNA-EphA2-DOPC
RNA Delivery of EphA2
Phase 1 cMet CAR RNA T Cells Metastatic Breast Cancer Intratumor administration cMet RNA CAR T cells
Triple Negative Breast
Cancer
Phase 1 Basiliximab in Treating Patients With Malignant Neoplasms Brain Intravenous (IV) infusion RNA-loaded dendritic cell
Newly Diagnosed Glioblastoma vaccine; Basiliximab
Multiforme Undergoing Targeted
Immunotherapy and
Temozolomide-Caused
Lymphopenia
Phase 1 Gene Transfer Sickle Cell Disease Intravenous (IV) infusion CD34+ HSC cells
transduced with lentiviral
vector containing shRNA
targeting BCL11a
Phase 1 CD19+ CAR T Cells Leukemia Lymphoma Intravenous (IV) infusion Fludarabine
monophosphate;
Cyclophosphamide
Phase 3 Dendritic Cells loaded with Uveal Melanoma Intravenous (IV) infusion Autologous Dendritic Cells
Autologous Tumor RNA loaded with autologous
Tumor RNA
Approved Nusinersen (Spinraza) Spinal Muscular Atrophy Intrathecal injection Antisense Oligonucleotide
(SMA)
Approved Eteplirsen (Exondys 51) Duchenne muscular Intravenous (IV) infusion Antisense Oligonucleotide
dystrophy (DMD)
Approved Defibrotid (Defiteli) Severe hepatic veno- Intravenous (IV) infusion Oligonucleotide
occlusive disease (sVOD)

Data collected from FDA Novel Drug Approval summaries (FDA.gov) and ClinicalTrials.gov.

[35,36], a process that utilizes multiple rounds of ligand Role of computational approaches in
selection and amplification of RNA or DNA library var- aptamer-based therapeutic design
iants to raise aptamers with high affinity and selectivity to The structural makeup of aptamers are key for their use as
a target ligand. Aptamers can bind in the pM range of effective therapeutics [33,34]; therefore, development of
affinity and exhibit higher specificity and lower off-target structure prediction algorithms represents a significant
effects than their protein counterparts [37]. The utiliza- undertaking in the rational design of aptamer therapeutics.
tion of SELEX has contributed greatly in our understand- Structural predictions from nucleic acid sequences are
ing of RNA biology and development of nucleic acid- challenging due to structural differences observed in vivo
based therapeutics; however, its successful application to versus in vitro [42], a high number of transitional states
aptamers with desirable properties requires 10–20 rounds [43], and the influence of metal ions, other RNAs, small
of selection, representing weeks to months of develop- organic molecules, and proteins on 3D RNA structure
ment effort [16]. While the coupling of SELEX with [33,34,44]. To address these challenges, bioinformatics
other molecular technologies such as high throughput and computational modeling approaches have been con-
sequencing, microarrays and bioinformatics have shown structed to statistically analyze existing datasets to increase
a reduction in selection efforts [38–41], computational the likelihood of successful aptamer design. In particular,
modeling approaches for the rational design of aptamers, databases of RNA-RNA interactions [45], RNA-DNA
particularly RNA aptamers, show promise to greatly interactions [46], and nucleic acid-protein interactions
reduce development time and associated costs [16]. [47] have been compiled to assist in these efforts [16].

Current Opinion in Biotechnology 2018, 53:232–239 www.sciencedirect.com


Computational approaches in nucleic acid therapeutics Sherman and Contreras 235

In this scheme, once a target for the aptamer has been Once tertiary structure is established, models can be
identified, structural data of both the target and the ideal utilized to predict docking location and strength of bind-
aptamer is gathered or generated. This is generally done ing of nucleic acids and their targets based on atomistic
through crystallography-based approaches [48], homology simulations. These simulations calculate minimum free
modeling using modeling software like ModeRNA [49– energy based on parameters such as electrostatic inter-
51], or through the use of a thermodynamics-based model actions, Van der Waals forces, and hydrophobic interac-
such as RNAup [52] (Table 2). Furthermore, several tions. Atomistic modeling tools such as CHARMM [60]
models have been introduced in recent years to derive and AMBER [61] have been used in this fashion to
secondary structure from primary RNA sequence such as simulate RNA–metal interactions and several examples
mFold [53] and ViennaRNA [54] (Table 2). In general, of this approach have recently been reviewed in Sun et al.
these models take into account a series of user defined [50]. For instance, molecular simulations and Markov
constraints to predict RNA secondary structure through state modeling have been applied to predict the kinetics
energy minimization algorithms. Models have also been of RNA conformational state changes, supporting experi-
developed to predict 3D tertiary structure from secondary mental observations that the binding of specific RNAs can
structure (MC-FOLDjMC-Sym [55]) and even directly be relatively slow and involves large conformational rear-
from primary sequence using coarse grained molecular rangements after initial binding [62]. Understanding
dynamics approaches (NAST [56], ERNWIN [57]) these dynamics can allow for efficient use of informatics
(Table 2). A graphical summary of these models based tools toward rational design of therapeutic aptamers, a
on complexity is shown in Figure 2. For modeling RNA– process analogous to approaches used in protein engi-
RNA interactions of uncharacterized RNAs, a desirable neering for therapeutics [63].
metric is the identification of accessible regions of target
RNAs in vivo [50]. To address this challenge, models have Rational design of nucleic acid therapeutic
been developed and benchmarked to investigate the delivery systems
likelihood for RNA–RNA binding. These include While the molecular toolkit of nucleic acid-based drugs is
IntaRNA [58], an RNA–RNA prediction algorithm based being diversified and potential therapeutic mechanisms
on seed constrains, and InTherAcc [43], an informed are being elucidated, the challenge to effectively deliver
biophysical model based on biological data collected in these therapeutic agents to the site of treatment remains a
vivo capturing RNA accessibility for binding using an in major hurdle. That is, for a therapeutic molecule to reach
vivo RNA Structural Sensing System [42]. To help its target site, several aspects need to be addressed; a few
inform crystal structure determination and provide experi- key ones include prevention of immunogenicity, escape
mental validation for models, several biochemical techni- from degradation enzymes, penetration through cellular
ques have been useful; these include hydroxyl radical barriers that inhibit uptake, release from liposomes before
footprinting (probing for protected nucleotides), selective degradation, and (depending on its mechanism) transport
20 OH acylation by primer extension (SHAPE) (chemical to the nucleus to perform intended functions [64,65].
probing for backbone flexibility), and dimethysulfate While nucleic acid-based drugs have been experimentally
sequencing (probing for H bonding) [59]. optimized for individual elements [64], there is not yet an

Table 2

A collection of modeling programs referenced in this article, their URL address, and a short description of their function

Model Website Description


ModeRNA http://genesilico.pl/moderna/ Comparative modeling of RNA 3D structures
MC-FOLDjMC-Sym http://www.major.iric.ca/MC-Fold/ Predicts secondary structures from sequence
NAST https://simtk.org/projects/nast Uses coarse grained molecular dynamics and a knowledge-based
force field to generate RNA structures
ERNWIN http://rna.tbi.univie.ac.at/ernwin Predicts RNA 3D structure using a coarse-grain helix-centered model
IntaRNA http://rna.informatik.uni-freiburg.de/IntaRNA/ Prediction of interactions between two RNA molecules
Input.jsp
RNAup http://rna.tbi.univie.ac.at/cgi-bin/ Predicts RNA-RNA interactions
RNAWebSuite/RNAup.cgi
mFold http://unafold.rna.albany.edu/?q=mfold Predicts nucleic acid folding and hybridization
ViennaRNA https://www.tbi.univie.ac.at/RNA/ A C code library and stand-alone programs for prediction and
comparison of RNA secondary structures
AutoDock Vina http://vina.scripps.edu/ Molecular docking program to calculate free energy scores
HADDOCK http://www.bonvinlab.org/software/haddock2. High Ambiguity Driven Biomolecular Docking based on Biochemical
2/haddock-start/ and/or biophysical information
PatchDock https://bioinfo3d.cs.tau.ac.il/PatchDock/ Molecular Docking Algorithm Based on Shape Complementarity
Principles

Data collected from model-associated publications and their web portals.

www.sciencedirect.com Current Opinion in Biotechnology 2018, 53:232–239


236 Pharmaceutical biotechnology

Figure 2

Current Opinion in Biotechnology

Graphical depiction of nucleic acid-based models and their uses across three levels of complexity: 2D RNA structure prediction from primary
sequence, prediction of RNA–RNA interactions, and prediction of RNA–protein interactions. Data collected from model-associated publications
and their web portals.

effective vehicle for targeted drug delivery. Current distant future, it is possible that genetically engineered
therapeutic delivery relies on transport of naked particles replacement organs could be delivered overnight for
or suboptimal modes of delivery, such as PEGylation and transplantation the following day [71]. The societal
liposomal based delivery [27,66]. Multi-scale computa- outcome of these revolutions may transition the role of
tional approaches to these issues could complement health care from one of fixing health issues to one of
experimental methods. Indeed, these approaches have surveillance followed by immediate treatment in
been implicated both as tools to gain mechanistic insights response to detection of early disease indicators. Nucleic
on biological properties and as inspirations to develop acid-based technologies, such as viral gene therapy and
novel systems for nanoparticle delivery [27]. Specifically, genome editing, enable the potential for permanent
the coarse grained molecular dynamics simulation model, removal of diseases without sustained medication nor
MARTINI [67], has been used to model RNA interac- drug development for treatment of disease phenotypes.
tions with other biomolecules and has been implicated for This type of therapy could reduce health care costs and
use in nanoparticle-mediated delivery modeling could curtail the impact of complex diseases worldwide.
[27,68,69].
Conclusions
Future directions of nucleic acid-based Nucleic acid-based therapeutics are quickly becoming
therapeutics relevant within the pharmaceutical industry. With three
Developments in DNA and RNA sequencing technolo- oligonucleotide based drugs approved in 2016 and over
gies, genome editing capabilities, and bioinformatics 40 privately in development [72] (ClinicalTrials.gov), the
approaches hold promise to replace dependence of a nucleic acid-based therapeutic industry appears to be
comfortable lifestyle on pharmaceuticals with advanced gathering momentum. Additionally, increasing attention
control of gene expression, particularly for complex dis- by the academic research community toward new tools
eases caused by genetic anomalies [70]. As computational and delivery mechanisms for DNA and RNA oligonucleo-
technologies become inextricably incorporated into soci- tides suggests that we are not far off from advances in
ety, their role in biotechnology and therapeutics is likely delivery to specific cell types and tissues, providing a new
to expand. In the near future, these new technologies suite of therapeutic chemistries with fewer side effects
raise the possibility that a drop of blood placed on a device and more efficient modes of action, rivaling antibody-
similar to a glucose monitor could screen the genome, based therapeutics. The development of computational
transcriptome, proteome and metabolome for abnormali- tools to identify target molecules (DNA, RNA, protein,
ties and those anomalies could be medicated appropri- small molecules) and modeling approaches to effectively
ately before the onset of disease. In a perhaps not so predict 3D structure from sequence will likely assist in

Current Opinion in Biotechnology 2018, 53:232–239 www.sciencedirect.com


Computational approaches in nucleic acid therapeutics Sherman and Contreras 237

this effort, potentially identifying targets leading to can- 13. Clark J, O’Hagan D: Strategies for radiolabelling antibody,
antibody fragments and affibodies with fluorine-18 as tracers
cer-specific treatments, orphan disease therapies, and for positron emission tomography (PET). J Fluor Chem 2017,
effective precision medicine. Finally, advancements in 203:31-46.
computational approaches that enable genome-wide 14. Liu JKH: The history of monoclonal antibody development –
identification of disease characteristics could provide progress, remaining challenges and future innovations. Ann
Med Surg 2014, 3:113-116 http://dx.doi.org/10.1016/j.
early detection of diseases to induce lifestyle changes amsu.2014.09.001.
for longer, healthier livelihoods. 15. Netspot [Package Insert]. Advanced Accelerator Applications
USA, Inc., New York. https://www.accessdata.fda.gov/
drugsatfda_docs/label/2016/208547s000lbl.pdf.
Funding
Welch Foundation [F-1756]; Funding for open access 16. Ahirwar R, Nahar S, Aggarwal S, Ramachandran S, Maiti S,
 Nahar P: In silico selection of an aptamer to estrogen receptor
charge: Welch Foundation [F-1756]; National Science alpha using computational docking employing estrogen
Foundation CAREER Program [CBET-1254754]. response elements as aptamer-alike molecules. Sci Rep 2016,
6:1-11 http://dx.doi.org/10.1038/srep21285.
This article uses a bioinformatics and molecular dynamics approaches to
select candidate binding regions for aptamer design. Following the
Acknowledgements analysis, the one variant selected did show binding activity.
We would like to thank Dr. Elebeoba May for careful reading of the
document and providing feedback and Daniel Herrera for assistance with 17. Zumrut HE, Ara MN, Fraile M, Maio G, Mallikaratchy P: Ligand-
data analysis. We would also like to thank K. Alexandra Krippner for guided selection of target-specific aptamers: a screening
assisting with illustrations. technology for identifying specific aptamers against cell-
surface proteins. Nucleic Acid Ther 2016 http://dx.doi.org/
10.1089/nat.2016.0611. nat.2016.0611.
References and recommended reading 18. Rescifina A, Zagni C, Varrica MG, Pistarà V, Corsaro A: Recent
Papers of particular interest, published within the period of review, advances in small organic molecules as DNA intercalating
have been highlighted as: agents: synthesis, activity, and modeling. Eur J Med Chem
2014, 74:95-115 http://dx.doi.org/10.1016/j.ejmech.2013.11.029.
 of special interest
 of outstanding interest 19. Tang L, Zeng Y, Du H et al.: CRISPR/Cas9-mediated gene
 editing in human zygotes using Cas9 protein. Mol Genet
1. Earm K, Earm YE: Integrative approach in the era of failing drug Genomics 2017, 292:525-533 http://dx.doi.org/10.1007/s00438-
 discovery and development. Integr Med Res 2014, 3:211-216. 017-1299-z.
This review discusses challenges that have arisen in the field of drug This article focuses on the use of CRISPR/Cas9 technologies to remedy
discovery and development and ways to circumvent these challenges as disease genotypes in human cell embryos, providing a proof of concept
the search for new therapeutics continues. for gene editing technologies in human cell lines.

2. Hao G, Jiang W, Ye Y et al.: ACFIS: a web server for fragment- 20. Aartsma-Rus A: FDA approval of Nusinersen for spinal
based drug discovery. Nucleic Acids Res 2016, 44:550-556 muscular atrophy makes 2016 the year of splice modulating
http://dx.doi.org/10.1093/nar/gkw393. oligonucleotides. Nucleic Acid Ther 2017, 27:67-69 http://dx.doi.
org/10.1089/nat.2017.0665.
3. Xuriden [Package Insert]. Gaithersburg, MD: Wellstat Therapeutics
Corporation. https://www.accessdata.fda.gov/drugsatfda_docs/ 21. Aartsma-Rus A, Straub V, Hemmings R et al.: Development of
label/2015/208169s000lbl.pdf [accessed 23.10.17]. exon skipping therapies for Duchenne muscular dystrophy: a
critical review and a perspective on the outstanding issues.
4. AbbVie Inc. VENCLEXTA (venetoclax). FDA Prescr Information; Nucleic Acid Ther 2017, 27:251-259 http://dx.doi.org/10.1089/
2016. Reference ID: 3915259. nat.2017.0682.
5. Spinraza [Package Insert]. Cambridge, MA: Biogen Inc. https:// 22. Aartsma-Rus A, Krieg AM: FDA approves eteplirsen for
www.accessdata.fda.gov/drugsatfda_docs/label/2016/  Duchenne muscular dystrophy: the next chapter in the
209531lbl.pdf [accessed 23.10.17]. eteplirsen saga. Nucleic Acid Ther 2017, 27:1-3 http://dx.doi.org/
10.1089/nat.2016.0657.
6. PARSABIV [Package Insert]. Thousand Oaks, CA: Amgen Inc.; This article describes the pathway to FDA approval for the oligonucleotide
2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/ therapeutics recently approved for use by the FDA.
2017/208325Orig1s000Lbledt.pdf [accessed 23.10.17].
23. Chen X, Yan CC, Zhang X, You Z-H: Long non-coding RNAs and
7. Vosevi [Package Insert]. Foster City, CA: Gilead Sciences, Inc. complex diseases: from experimental results to
https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/ computational models. Brief Bioinform 2016, 18:558-576 http://
209195s000lbl.pdf [accessed 23.10.17]. dx.doi.org/10.1093/bib/bbw060.
8. Junutula JR, Gerber HP: Next-generation antibody-drug
24. Mihailovic MK, Chen A, Gonzalez-rivera JC, Contreras LM:
conjugates (ADCs) for cancer therapy. ACS Med Chem Lett
 Defective ribonucleoproteins, mistakes in RNA processing,
2016, 7:972-973 http://dx.doi.org/10.1021/
and diseases. Biochemistry 2017 http://dx.doi.org/10.1021/acs.
acsmedchemlett.6b00421.
biochem.6b01134.
9. Pfizer: MYLOTARG (gemtuzumab ozogamicin). 2017:1-19. This article reviews the result of ribonucleoprotein defects, what effect
they have on human health, and offers insight on the promise of Nucleic
10. Stan CD, Dra?gan M, Ta?ta?rı̂nga? G, Stan CI, Tuchiluş CG, acid-based therapeutic development.
Mircea C: Monoclonal antibodies – past, present and future.
Pharmacy 2017, 121:444-450. 25. Remaut K, De Clercq E, Andries O et al.: Aerosolized non-viral
nucleic acid delivery in the vaginal tract of pigs. Pharm Res
11. Frenzel A, Schirrmann T, Hust M: Phage display-derived human 2016, 33:384-394 http://dx.doi.org/10.1007/s11095-015-1796-x.
 antibodies in clinical development and therapy. MAbs 2016,
8:1177-1194 http://dx.doi.org/10.1080/19420862.2016.1212149. 26. Miyata K: Smart polymeric nanocarriers for small nucleic acid
This paper provides a review of the human antibodies developed for delivery. Drug Discov Ther 2016, 10:236-247 http://dx.doi.org/
therapeutic use that have been engineered through phage display and 10.5582/ddt.2016.01061.
provides a summary table of their status as of 2016.
27. Bunker A, Magarkar A, Viitala T: Rational design of liposomal
12. Kupper H, Salfeld J, Tracey D, Kalden JR: Adalimumab (Humira) drug delivery systems, a review: combined experimental and
anti-TNF. Handb Ther Antibodies 2008, 3:696-732 http://dx.doi. computational studies of lipid membranes, liposomes and
org/10.1002/9783527619740.ch27. their PEGylation. Biochim Biophys Acta – Biomembr 2016,

www.sciencedirect.com Current Opinion in Biotechnology 2018, 53:232–239


238 Pharmaceutical biotechnology

1858:2334-2352 http://dx.doi.org/10.1016/j. This article seeks to experimentally validate a previously published


bbamem.2016.02.025. method by the group which allows the in vivo binding of RNA substrates
to the protein of interest.
28. Xu X, Wu J, Liu Y et al.: Multifunctional envelope-type siRNA
delivery nanoparticle platform for prostate cancer therapy. 44. Dawson WK, Bujnicki JM: Computational modeling of RNA 3D
ACS Nano 2017, 11:2618-2627 http://dx.doi.org/10.1021/ structures and interactions. Curr Opin Struct Biol 2016, 37:22-28
acsnano.6b07195. http://dx.doi.org/10.1016/j.sbi.2015.11.007.

29. Shabanpoor F, Hammond SM, Abendroth F, Hazell G, Wood MJA, 45. Zhang X, Wu D, Chen L et al.: BIOINFORMATICS RAID: a
Gait MJ: Identification of a peptide for systemic brain delivery comprehensive resource for human RNA-associated (RNA–
of a morpholino oligonucleotide in mouse models of spinal RNA/RNA–protein) interaction. RNA 2014:989-993 http://dx.doi.
muscular atrophy. Nucleic Acid Ther 2017, 27:130-143 http://dx. org/10.1261/rna.044776.114.4.
doi.org/10.1089/nat.2016.0652.
46. Bader GD, Betel D, Tope syreHogue CWV: BIND: the
30. Davis ME, Zuckerman JE, Choi CHJ et al.: Evidence of RNAi in biomolecular interaction network database. Nucleic Acids Res
humans from systemically administered siRNA via targeted 2003, 31:248-250 http://dx.doi.org/10.1093/nar/gkg056.
nanoparticles. Nature 2010, 464:1067-1070 http://dx.doi.org/
47. Kirsanov DD, Zanegina ON, Aksianov EA, Spirin SA, Karyagina AS,
10.1038/nature08956.
Alexeevski AV: NPIDB: nucleic acid–protein interaction
31. Muralidhara BK, Baid R, Bishop SM, Huang M, Wang W, Nema S: database. Nucleic Acids Res 2013, 41(D1):D517-D523 http://dx.
Critical considerations for developing nucleic acid doi.org/10.1093/nar/gks1199.
macromolecule based drug products. Drug Discov Today 2016, 48. Edwards AL, Garst AD, Batey RT: Determining structures of RNA
21:430-444 http://dx.doi.org/10.1016/j.drudis.2015.11.012. aptamers and riboswitches by X-ray crystallography. Methods
32. Stein CA, Castanotto D: FDA-approved oligonucleotide Mol Biol 2009, 535:135-163 http://dx.doi.org/10.1007/978-1-
therapies in 2017. Mol Ther 2017, 25:1069-1075 http://dx.doi.org/ 59745-557-2.
10.1016/j.ymthe.2017.03.023. 49. Howe JA, Wang H, Fischmann TO et al.: Selective small-
molecule inhibition of an RNA structural element. Nature 2015,
33. Liu J, Cao Z, Lu Y: Functional nucleic acid sensors. Chem Rev
526:672-677 http://dx.doi.org/10.1038/nature15542.
2009, 109 http://dx.doi.org/10.1021/cr030183i.
50. Sun L-Z, Zhang D, Chen S-J: Theory and modeling of RNA
34. Nutiu R, Li Y: Structure-switching signaling aptamers. JACS structure and interactions with metal ions and small
2003:4771-4778 http://dx.doi.org/10.1002/anie.200461848. molecules. Annu Rev Biophys 2017:227-246 http://dx.doi.org/
35. Tuerk C, Gold L: Systematic evolution of ligands by exponential 10.1007/s00210-015-1172-8.
 enrichment: RNA ligands to bacteriophage T4 DNA 51. Rother M, Rother K, Puton T, Bujnicki JM: ModeRNA: a tool for
polymerase. Science 1990, 249:505-510 http://dx.doi.org/ comparative modeling of RNA 3D structure. Nucleic Acids Res
10.1126/science.2200121. 2011, 39:4007-4022 http://dx.doi.org/10.1093/nar/gkq1320.
This paper describes the SELEX process, a common protocol that can be
used to develop candidate apatmers raised to a specific antigen. 52. Mückstein U, Tafer H, Hackermüller J, Bernhart SH, Stadler PF,
Hofacker IL: Thermodynamics of RNA–RNA binding.
36. Ellington AD, Szostak JW: In vitro selection of RNA molecules Bioinformatics 2006, 22:1177-1182 http://dx.doi.org/10.1093/
that bind specific ligands. Nature 1990, 346:818-822 http://dx. bioinformatics/btl024.
doi.org/10.1038/346818a0.
53. Zuker M: Mfold web server for nucleic acid folding and
37. Clawson GA, Abraham T, Pan W et al.: A cholecystokinin B hybridization prediction. Nucleic Acids Res 2003, 31:3406-3415
receptor-specific DNA aptamer for targeting pancreatic http://dx.doi.org/10.1093/nar/gkg595.
ductal adenocarcinoma. Nucleic Acid Ther 2017, 27:23-35 http://
dx.doi.org/10.1089/nat.2016.0621. 54. Lorenz R, Bernhart SH, Höner zu Siederdissen C et al.: ViennaRNA
Package 2.0. Algorithms Mol Biol 2011, 6:26 http://dx.doi.org/
38. Hu WP, Kumar JV, Huang CJ, Chen WY: Computational 10.1186/1748-7188-6-26.
selection of RNA aptamer against angiopoietin-2 and
experimental evaluation. Biomed Res Int 2015:2015 http://dx. 55. Parisien M, Major F: The MC-fold and MC-Sym pipeline infers
doi.org/10.1155/2015/658712. RNA structure from sequence data. Nature 2008, 452:51-55
http://dx.doi.org/10.1038/nature06684.
39. Cho M, Xiao Y, Nie J et al.: Quantitative selection of DNA
aptamers through microfluidic selection and high-throughput 56. Jonikas MA, Radmer RJ, Laederach A et al.: Coarse-grained
sequencing. Proc Natl Acad Sci 2010, 107:15373-15378 http:// modeling of large RNA molecules with knowledge-based
dx.doi.org/10.1073/pnas.1009331107. potentials and structural filters. RNA 2009, 15:189-199 http://
dx.doi.org/10.1261/rna.1270809.
40. Caroli J, Taccioli C, De La Fuente A, Serafini P, Bicciato SJ,
Caroli C, Taccioli A, De La Fuente P, Serafini S: Bicciato; APTANI: 57. Kerpedjiev P, Höner C, Siederdissen Z, Hofacker IL: Predicting
a computational tool to select aptamers through sequence- RNA 3D structure using a coarse-grain helix-centered model.
structure motif analysis of HT-SELEX data. Bioinformatics RNA 2015, 21:1110-1121 http://dx.doi.org/10.1261/
2016, 32:161-164 In: https://doi-org.ezproxy.lib.utexas.edu/10. rna.047522.114.
1093/bioinformatics/btv545.
58. Mann M, Wright PR, Backofen R: IntaRNA 2.0: Enhanced and
41. Luo X, McKeague M, Pitre S et al.: Computational approaches customizable prediction of RNA-RNA interactions. Nucleic
toward the design of pools for the in vitro selection of complex Acids Res 2017, 45(W1):W435-W439 http://dx.doi.org/10.1093/
aptamers. RNA 2010, 16:2252-2262 http://dx.doi.org/10.1261/ nar/gkx279.
rna.2102210. 59. Schlick T, Pyle AM: Opportunities and challenges in RNA
42. Sowa SW, Vazquez-Anderson J, Clark CA et al.: Exploiting post- structural modeling and design. Biophys J 2017:1-10 http://dx.
 transcriptional regulation to probe RNA structures in vivo via doi.org/10.1016/j.jmb.2016.02.012.
fluorescence. Nucleic Acids Res 2015, 43:e13 http://dx.doi.org/ 60. Brooks BR, Iii CLB, Mackerell AD et al.: CHARMM: the
10.1093/nar/gku1191. biomolecular simulation program. J Comput Chem 2009,
This article describes the development of a fluorescent based assay that 30:1545-1614 http://dx.doi.org/10.1002/jcc.21287.
can determine accessible binding regions on RNA molecules.
61. Wang J, Wolf RM, Caldwell JW, Kollman PA, Case DA:
43. Vazquez-anderson J, Mihailovic MK, Baldridge KC et al.: Development and testing of a general amber force field. J
 Optimization of a novel biophysical model using large scale in Comput Chem 2004:1-14.
vivo antisense hybridization data displays improved
prediction capabilities of structurally accessible RNA regions. 62. Warfield BM, Anderson PC: Molecular simulations and Markov
Nucleic Acids Res 2017, 45:5523-5538 http://dx.doi.org/10.1093/ state modeling reveal the structural diversity and dynamics of
nar/gkx115. a theophylline-binding RNA aptamer in its unbound state.

Current Opinion in Biotechnology 2018, 53:232–239 www.sciencedirect.com


Computational approaches in nucleic acid therapeutics Sherman and Contreras 239

PLOS ONE 2017, 12 http://dx.doi.org/10.1371/journal. biomolecular simulations. J Phys Chem B 2007, 111:7812-7824
pone.0176229. http://dx.doi.org/10.1021/jp071097f.
63. Shim J, MacKerell AD Jr: Computational ligand-based rational 68. Uusitalo JJ, Ingólfsson HI, Marrink SJ, Faustino I: Martini coarse-
design: role of conformational sampling and force fields in grained force field: extension to RNA. Biophys J 2017, 113:246-
model development. Med Chem Commun 2011, 2:356-370 256 http://dx.doi.org/10.1016/j.bpj.2017.05.043.
http://dx.doi.org/10.1039/c1md00044f.
69. Vácha R, Martinez-Veracoechea FJ, Frenkel D: Intracellular
64. Geinguenaud F, Guenin E, Lalatonne Y, Motte L: Vectorization of release of endocytosed nanoparticles upon a change of
nucleic acids for therapeutic approach: tutorial review. ACS ligand–receptor interaction. ACS Nano 2012, 6:10598-10605
Chem Biol 2016, 11:1180-1191 http://dx.doi.org/10.1021/ http://dx.doi.org/10.1021/nn303508c.
acschembio.5b01053.
70. Ma H, Marti-Gutierrez N, Park S-W et al.: Correction of a
65. Ding HM, Ma YQ: Theoretical and computational investigations pathogenic gene mutation in human embryos. Nature 2017,
of nanoparticle–biomembrane interactions in cellular delivery. 548:413-419 http://dx.doi.org/10.1038/nature23305.
Small 2015, 11:1055-1071 http://dx.doi.org/10.1002/
smll.201401943. 71. Niu D, Wei H, Lin L et al.: Inactivation of porcine endogenous
 retrovirus in pigs using CRISPR-Cas9. Science (80-) 2017,
cek J, Zboril R:
66. Ulbrich K, Holá K, ubr V, Bakandritsos A, Tu9 357:1303-1307.
Targeted drug delivery with polymers and magnetic This paper describes the establishment of a pig line that has all of porcine
nanoparticles: covalent and noncovalent approaches, release endogenous retroviruses (PERVs) removed to enable human xenotrans-
control, and clinical studies. Chem Rev 2016, 116:5338-5431 plantation without immunogenic response.
http://dx.doi.org/10.1021/acs.chemrev.5b00589.
72. Crooke ST: Molecular mechanisms of antisense
67. Marrink SJ, Risselada HJ, Yefimov S, Tieleman DP, De Vries AH: oligonucleotides. Nucleic Acid Ther 2017, 27:70-77 http://dx.doi.
The MARTINI force field: coarse grained model for org/10.1089/nat.2016.0656.

www.sciencedirect.com Current Opinion in Biotechnology 2018, 53:232–239

You might also like