Abstract
In Part I of this Review, we introduced recent advances in gene delivery technologies and explained how they have powered some of the current human gene therapy applications. In Part II, we expand the discussion on gene therapy applications, focusing on some of the most exciting clinical uses. To help readers to grasp the essence and to better organize the diverse applications, we categorize them under four gene therapy strategies: (1) gene replacement therapy for monogenic diseases, (2) gene addition for complex disorders and infectious diseases, (3) gene expression alteration targeting RNA, and (4) gene editing to introduce targeted changes in host genome. Human gene therapy started with the simple idea that replacing a faulty gene with a functional copy can cure a disease. It has been a long and bumpy road to finally translate this seemingly straightforward concept into reality. As many disease mechanisms unraveled, gene therapists have employed a gene addition strategy backed by a deep knowledge of what goes wrong in diseases and how to harness host cellular machinery to battle against diseases. Breakthroughs in other biotechnologies, such as RNA interference and genome editing by chimeric nucleases, have the potential to be integrated into gene therapy. Although clinical trials utilizing these new technologies are currently sparse, these innovations are expected to greatly broaden the scope of gene therapy in the near future.
1. Introduction
Human genetic disorders arise from mutations in the DNA genome, which in many cases abrogate the normal function of genes. To tackle these “loss-of-function” diseases, recombinant DNA technology fostered the hope that delivering the normal copy of a mutated gene to the patient will cure the disease. This “gene replacement” approach to treating single gene disorders represents the prototype of gene therapy. In addition to gene replacement, other gene manipulation strategies have been developed to meet the demand of treating mechanistically different diseases (Figure 1). For example, gene addition supplements therapeutic factors that are either protective or curative. Gene knockdown aims at diminishing gain-of-toxicity. Gene editing suits the need of introducing specific changes in the genome. Nowadays, gene therapy utilizes diverse gene delivery vehicles, comprises various gene manipulation strategies, and targets a broader range of human diseases such as infectious diseases. In Part I of this Review, we discussed the recent advances in gene delivery technologies, including using non-viral and viral vectors, in vivo and ex vivo routes of gene transfer. Here, we introduce various gene manipulation strategies employed in current human gene therapy applications, aiming to provide readers with a framework of comprehending the rapidly evolving field of translational gene therapy. For more comprehensive discussion focusing on specific diseases or approaches, readers are encouraged to explore the excellent review articles in References and research articles therein.
Figure 1.

Depiction of four gene therapy strategies. (A) A gene mutation (red dot) abrogates protein synthesis (red cross) and leads to disease. Gene replacement corrects the disease by providing a functional copy of the gene (green helix) and normal proteins (green circles). (B) Multiple genetic factors and/or environmental factors can result in a disease though a complex disease cascade (dashed arrow). Gene addition alleviates the disease by supplementing therapeutic genes that target a specific aspect of the disease mechanism. (C) A gene mutation leads to the production of toxic protein aggregate (clustered red circles) that causes a disease. Gene knockdown utilizes small RNAs (green combs) to inhibit the aberrant mRNA (black comb), thus preventing the synthesis of toxic protein aggregate and correcting the disease. (D) Gene editing by chimeric nucleases (circle and pie) is a versatile approach to make a targeted change from a disease-promoting sequence (red dot) to a disease-preventing sequence (green dot). Also see Figure 4.
2. Gene replacement therapy for monogenic diseases
Many human genetic diseases are clearly defined by a single gene defect, and gene replacement is a straightforward approach to treating these diseases. Not surprisingly, the majority of the most advanced clinical gene therapy development falls into the category of gene replacement, because the conceptually simple design provokes extensive investigation. In addition, animal models that mimic monogenic human diseases are more easily obtained by naturally occurring mutation or genetic manipulation, which provides a valuable platform for pre-clinical testing. Gene replacement can take place either directly in vivo or through ex vivo cell therapy.
Lipoprotein lipase deficiency (LPLD)
The first and only gene therapy product that has obtained marketing authorization in the Western world is Glybera, a recombinant AAV (rAAV) vector for treating a rare disease called LPLD by replacing with a functional LPL gene (Yla-Herttuala, 2012). LPL is an enzyme that breaks down fats, and patients suffering from LPLD are restricted to a low-fat diet and are prone to recurring, life threatening inflammation in pancreas (Gaudet et al., 2012). Glybera consists of a hyperactive copy of human LPL gene packaged in rAAV1 vector. Following one-time intramuscular injections at multiple sites, transduced muscle cells serve as a biofactory to continuously produce and secrete LPL into blood. The secreted LPL docks on the luminal surface of endothelial cells lining blood vessels, and helps to break down fats stored in circulating vesicles and to alleviate disease severity (Gaudet et al., 2013; Stroes et al., 2008). To protect the viral vector from host immune attack and clearance, immune suppression is recommended between three days prior to and twelve weeks after treatment (Ferreira et al., 2014a; Ferreira et al., 2014b). The success of Glybera is partially attributable to targeting an easily accessible and permissive organ, skeletal muscle, which has drawn considerable interest as a platform for gene therapy (Wang et al., 2014a).
Leber congenital amaurosis (LCA)
The eye also is considered as an ideal organ for gene therapy due to its small size, compartmentalized organization, slow cell turnover, and low risk of immunogenicity (Sahel & Roska, 2013). One form of LCA (a group of diseases with severe retinal dystrophy and blindness) is caused by loss-of-function mutations in the retinal pigment epithelium-specific protein 65kDa gene (RPE65). The RPE65 protein is an enzyme located in retinal pigment epithelium, and catalyzes biochemical conversion of molecules that are involved in phototransduction (Kiser & Palczewski, 2010). In a naturally occurring RPE65-deficient dog model, a single subretinal injection of rAAV-RPE65 restored retinal response and vision function, and the restoration was stable for 3 years (Petersen-Jones et al., 2012). The safety of a similar approach in human use was proved in three phase I clinical trials, and stable clinical benefits were documented for more than 30 treated patients (Bainbridge et al., 2008; Hauswirth et al., 2008; Jacobson et al., 2012; Maguire et al., 2008; Stein et al., 2011). Recently, readministration of rAAV-RPE65 to the contralateral eye was shown to be safe and effective in three patients (Bennett et al., 2012). However, the treatment does not prevent retinal degeneration, though it corrects retinal dysfunction, calling for a combination approach for long-term therapeutic efficacy (Cideciyan et al., 2013). Nevertheless, the RPE65 gene replacement trials opened avenues to gene therapy development for other monogenic eye diseases (Sahel & Roska, 2013).
Blood disorders
Hemophilia B is caused by the deficiency in the gene encoding blood coagulation factor IX (FIX). The FIX gene replacement programs that have been under extensive clinical evaluation aim to deliver rAAV-FIX to liver or muscle, so that the FIX synthesized therein can be secreted to bloodstream and function. Clinical efficacy has been encouraging, as treated patients show vector dose-dependent FIX production, and require less frequent or no infusion with recombinant FIX protein (High et al., 2014; Patel et al., 2014). An important finding from FIX replacement clinical trials is AAV capsid-induced cytotoxic T cell immune response that eliminates transduced cells and compromises therapeutic efficacy (Mingozzi et al., 2009). Although a concomitant immune suppression regimen has been shown to partially reduce the immune response and to improve efficacy, efforts are underway to better address this complication (Mingozzi & High, 2013). The clinical development of hemophilia B gene therapy provides valuable insight to future AAV-based gene therapies targeting liver or muscle as a biofactory to produce therapeutic proteins. Primary immunodeficiencies (PIDs) are a group of heterogeneous diseases characterized by an abnormal immune system. Many PIDs are monogenic, loss-of-function disorders that specifically impair hematopoietic stem cell (HSC) lineages that give rise to a variety of immune cells. The most severe form of PID is severe combined immunodeficiencies (SCIDs) that affect both T- and B-cells. Since the deleterious effects of genetic defects in PIDs are within HSC lineages, ex vivo HSC gene replacement is a rational therapeutic approach (Nienhuis, 2013; Zhang et al., 2013). Integrating viral vectors such as gamma-retroviral vectors and lentiviral vectors have been used to deliver genes into isolated HSCs, and to replace the adenosine deaminase (ADA) gene and the interleukin-2 receptor subunit gamma (IL-2RG) gene for ADA-SCID (Aiuti et al., 2009; Gaspar et al., 2011) and X-linked SCID (Hacein-Bey-Abina et al., 2010), respectively. Although the early clinical trials revealed the risk of insertional mutagenesis due to gamma-retroviral vector integration, lentiviral vector proves to be safer. Furthermore, integrating vector design has been refined and improved safety along with promising efficacy was demonstrated in recent preclinical studies (Carbonaro et al., 2014). The current experience with PID gene replacement has laid the foundation for developing other ex vivo gene therapies, which commonly utilize integrated gene transfer and require careful design and monitoring for genotoxicity.
Other promising gene replacement clinical trials involve treating respiratory diseases (such as cystic fibrosis and alpha 1 anti-trypsin deficiency) (Griesenbach & Alton, 2013; Mueller & Flotte, 2013) and leukodystrophies (degeneration of the white matter in the brain) (Biffi et al., 2011). Taken together, these human gene replacement applications have pioneered the gene therapy field. AAV vectors proved to be the most favorable for in vivo gene delivery due to high efficiency and low risks of immunogenicity and genotoxicity, whereas the most recent generations of integrating vectors with improved safety profile have shown encouraging results in ex vivo gene therapy.
3. Gene addition for complex disorders and infectious diseases
In contrast to a single gene defect underlying monogenic diseases, the combination effects of multiple genes and environmental factors cause complex disorders such as cancer and heart diseases, rendering gene replacement not feasible for these disorders. Complex disorders are often common, and represent the most urgent unmet medical needs. In addition to genetic disorders, infectious diseases also debilitate or kill a large population worldwide. Therefore, significant efforts of gene therapy development have been devoted to these diseases (Figure 2), which usually require a deep understanding of the disease mechanisms, and utilize gene addition to supplement a therapeutic agent that targets a common aspect of the disease.
Figure 2.

Breakdown of approved gene therapy clinical trials worldwide since 1989 according to disease applications. Data are from Wiley Gene Therapy Clinical Trials Worldwide (http://www.abedia.com/wiley/).
Heart failure
A common cellular defect in heart failure (poor heart function resulting in reduced blood flow insufficient to meet the body’s needs) is altered calcium transport in cardiac muscle cells, which is associated with reduced expression and activity of the calcium handling protein sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) not necessarily due to any defect in the corresponding genes (Kranias & Hajjar, 2012). Studies have shown that overexpression of SERCA2a in cardiac muscle cells improves cardiac function in various heart failure animal models (Sikkel et al., 2014). The Calcium Up-regulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) gene therapy clinical trials aim to deliver the human SERCA2A gene to heart using a rAAV1 vector as a treatment for heart failure. In the CUPID 1 trial (phase 2a) involving 39 patients with advanced heart failure, a single intracoronary infusion of high dose rAAV1-SERCA2A improved cardiac function and reduced hospitalizations when examined at 12 months after treatment (Jessup et al., 2011; Zsebo et al., 2014). The phase 2b CUPID 2 trial involving 250 patients is underway to further evaluate the efficacy of high dose rAAV1-SERCA2A.
Cancer
The heterogeneity and refractoriness of cancer inspired gene therapists to develop innovative therapeutic approaches. Although many gene therapy attempts to treat cancer have been somewhat disappointing (Brenner et al., 2013), we highlight two gene addition strategies that have shown promising results, and are currently under clinical investigation. The first strategy involves a herpex simplex viral (HSV) vector with certain viral gene deletions, so that it only replicates in tumor cells and leads to oncolysis. Importantly, it delivers the gene encoding an immune stimulating cytokine known as granulocyte macrophage colony-stimulating factor (GM-CSF), which eventually helps immune effector cells to attack tumor cells (Goins et al., 2014). The critical role of GM-CSF gene addition in triggering a systemic anti-tumor immune response was demonstrated in a recent Phase III clinical trial treating melanoma, as intratumor injection of HSVGM-CSF reduced the size of not only injected tumors, but also metastasized tumors (Andtbacka et al., 2014). The second strategy is to transfer a gene encoding a chimeric antigen receptor (CAR) recognizing CD19 (a B-cell surface marker) to ex vivo T cells, which are then reinfused to patients to treat B-cell malignancies. The T cells armed with anti-CD19 CAR were shown to reverse advanced-stage lymphoma in one patient (Kochenderfer et al., 2010). Currently, this strategy is being extensively evaluated in several independent clinical trials to treat B-cell malignancies (Kochenderfer & Rosenberg, 2013).
Infectious diseases
Using muscle-directed gene addition to provide antibody (also called vectored antibody) was recently exploited as an innovative approach to treating infectious diseases. In several pre-clinical animal studies, AAV vectors were used to deliver genes encoding antibodies against pathogen-derived antigens to muscle (Wang et al., 2014a). As a result, the secreted pathogen-specific antibodies in bloodstream continuously provide surveillance and clearance of future infectious pathogens such as HIV (Balazs et al., 2012; Johnson et al., 2009) and influenza viruses (Balazs et al., 2013). Clinical trials to treat HIV infection are at preparation phase with high expectation.
In addition, supplementing trophic factors is an actively pursued approach to treating cardiovascular diseases (Bradshaw & Baker, 2013) and neurodegenerative diseases (Simonato et al., 2013). It should be noted that gene addition should not be considered merely as a “Plan B” when gene replacement is not feasible. Rather, if proven to be successful in a certain application, the same gene addition approach is relatively easy and likely to be adopted for another similar disease condition, thus benefiting an even larger population of patients.
4. Gene expression alteration targeting RNA
RNA can be an intermediate (e.g. messenger RNA) or final (e.g. microRNA) gene product, with diverse functions in biology and disease. Due to the versatile roles of RNA molecules in controlling gene expression, gene therapy strategies specifically designed to target RNA or to produce effector RNA molecules deserve close attention. Here we briefly summarize two commonly utilized gene therapy strategies based on RNA biology.
Gene knockdown by RNA interference (RNAi)
In contrast to loss-of-function gene mutations that are often suitable to gene replacement, gain-of-toxicity mutations lead to the production of toxic gene products, and therefore require a gene knockdown strategy to diminish the toxicity (Figure 1C). RNAi is an RNA-mediated gene silencing process widely found in many eukaryotes including mammals (Deng et al., 2014). Since its discovery in 1998, RNAi has been extensively studied, and matured as one of the most powerful tools for gene silencing in both basic research and therapy development (Fellmann & Lowe, 2014) (Figure 1C). The effector small interfering RNA (siRNA, ~22 nt long, single-stranded) can be initially expressed in the form of short hairpin RNA (shRNA), followed by processing into siRNA to repress gene expression by triggering RNA degradation and/or inhibiting protein synthesis (Borel et al., 2014). Although the cellular toxicity associated with overexpressing shRNA is debatable, the design of artificial microRNA shuttle seems to be safer and can achieve acceptable gene knockdown efficacy (Boudreau et al., 2011; Boudreau et al., 2009). RNAi finds its use in treating diseases caused by toxic proteins derived from gain-of-toxicity gene mutations (San Sebastian et al., 2013), such as superoxide dismutase I in amyotrophic lateral sclerosis (Wang et al., 2014b). One challenge is that RNAi usually indiscriminately reduces the toxic mutant protein and the normal counterpart (the latter being expressed from the second gene copy in a diploid human genome), which can impair the gene’s normal function. In this case, allele-specific RNAi that targets only the mutant gene copy has been developed for diminishing the toxic effect of mutant huntingtin protein in Huntingtont’s disease, while preserving the normal gene copy’s function (Pfister & Zamore, 2009). RNAi has also been used to downregulate specific cellular pathways that promote complex diseases such as cancer (Deng et al., 2014). For diseases that are attributable to both loss-of-function and gain-of-toxicity, a dual vector can be designed to simultaneously knockdown toxic mutated gene product and replace with a normal gene, as exemplified in the gene therapy strategy for alpha 1 anti-trypsin deficiency (Mueller et al., 2012).
Reprogramming messenger RNA (mRNA) splicing by antisense oligonucleotides (AONs)
In mammals, most newly transcribed pre-mRNAs undergo splicing to form mature mRNAs composed of exons, which direct protein synthesis. Therefore, mRNA splicing regulates gene expression by inclusion or exclusion of specific pre-mRNA sequences for translation. In Duchene muscular dystrophy (DMD), many mutations in the DMD gene (encoding the dystrophin protein in muscle cells) disrupt reading frame and generate a premature stop codon in mature mRNA. Since the DMD gene is too big to be packaged in the most efficient in vivo gene delivery vector AAV for gene replacement, DNA or vector-derived RNA AONs (the latter being expressed using the U7 small nuclear RNA backbone) are designed to induce exon skipping, and to restore the correct reading frame, rather than replacing the entire DMD gene. (Goyenvalle et al., 2012a; Goyenvalle & Davies, 2011; Goyenvalle et al., 2012b) (Figure 3A). Although the resulting dystrophin protein is shorter than the normal version, it is partially functional and can significantly reduce disease severity. When designed properly, AONs can also induce exon inclusion. For example, spinal muscular atrophy (SMA) is caused by mutations in the survival of motor neuron 1 (SMN1) gene and loss of SMN protein. A similar gene, SMN2, differs from SMN1 by a single nucleotide change that disrupts a splicing enhancing signal and excludes exon 7 in most mature SMN2 mRNA molecules, thus resulting in no functional SMN protein production (Figure 3B). An AON was designed to force the inclusion of exon 7 in SMN2 mRNA, and therefore to restore SMN protein. This was achieved by designing the AON as a bi-functional molecule – binding to the target SMN2 mRNA and recruiting splicing factors that carry out desired splicing events (Hua et al., 2007; Madocsai et al., 2005; Mitrpant et al., 2013; Skordis et al., 2003) (Figure 3B).
Figure 3.

Reprogramming mRNA splicing by AONs. (A) Exon skipping to restore dystrophin in DMD. Normal splicing of DMD pre-mRNA leads to mature mRNA consisting of all exons that direct the synthesis of full-length dystrophin. In DMD, mutations in exon 50 result in abnormal splicing that excludes exon 50 in mature mRNA, which shifts reading frame and abrogates dystrophin production. AON binding blocks splicing signal in exon 51, which excludes exon 51 together with exon 50, thus restoring original reading frame and producing partially functional, albeit shortened, dystrophin. (B) Exon inclusion to restore SMN protein in SMA. Under normal condition, only the SMN1 gene produces SMN protein, whereas SMN2 gene is largely silenced due to a single nucleotide change in exon 7 (yellow bar), which excludes exon 7 in mature mRNA. In SMA, SMN1 is mutated (e.g. gene deletion), resulting in loss of SMN protein. A bi-functional AON binds to exon 7 of SMN2 pre-mRNA and recruits splicing factor (SF) that mediates the splicing pattern as in normal SMN1 pre-mRNA. Therefore, the silenced SMN2 gene is reactivated and provides therapeutic SMN proteins. Boxes: exons. Curved arrows: protein synthesis. Black stop signs: normal stop codons. Red stop sign: premature stop codon caused by frameshifting. Green stem loops: RNA AON embedded in modified U7 small nuclear RNA. DSB: double-strand break. DMD: Duchene muscular dystrophy. AON: antisense oligonucleotide. SMA: spinal muscular atrophy. SMN: survival of motor neuron.
It should be noted that we only focus on RNA effectors that are derived from corresponding gene transfer and transcription. Alternatively, synthetic DNA or RNA AONs can be delivered and serve similar functions in RNAi and modulating mRNA splicing. Such drug-like nucleic acid molecules are currently more widely used in clinical trials than gene transfer vector-derived RNA effectors (Deng et al., 2014). As gene delivery vector and vector design for expressing RNA are improving, we expect vector-derived RNA will play a greater role in human gene therapy.
5. Gene editing to introduce targeted changes in host genome
Target sequence-specific, designer nucleases have become a powerful tool kit for genome engineering. These chimeric nucleases consist of a sequence-specific and customizable DNA recognition module and a DNA cleavage domain (Gaj et al., 2013). To achieve sequence specificity, Zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) utilize DNA-binding proteins (Joung & Sander, 2013; Palpant & Dudzinski, 2013), whereas the clustered regulatory interspaced short palindromic repeat (CRISPR)/Cas system is based on RNA-guided DNA recognition (Mali et al., 2013). Regardless of the detailed mechanisms, these designer nucleases first induce double-strand breaks (DSBs) in targeted DNA sequence, and then realize a range of DNA modifications through two distinct DNA repair mechanisms, namely nonhomologous end joining (NHEJ) and homologous recombination (HR).
Gene disruption by NHEJ
A DSB can trigger NHEJ, a DNA repair mechanism that ligates the broken DNA fragments by introducing small insertions or deletions. This outcome is ideal for disrupting disease-promoting gene function (Figure 4A). For example, HIV infection of human T cells requires the presence of a T cell co-receptor called chemokine receptor 5 (CCR5). Utilizing ZFN to disrupt the CCR5 gene in T cells was shown to render the modified T cell resistant to HIV infection (Cannon & June, 2011). Several early phase clinical trials involving ex vivo CCR5 disruption for treating HIV infection are ongoing or completed (NCT01252641, NCT00842634, NCT01044654), with results being revealed in the near future.
Figure 4.

Gene editing by ZFN to introduce targeted changes in host genome. (A) Gene disruption through NHEJ. ZFN-induced DSB (white gap) in a disease-promoting gene (black box) is repaired through random insertion/deletion of a few nucleotides (red gap), which diminishes the disease-promoting function of the gene (red cross). (B) In the presence of a donor template with homology to the targeted genomic region, ZFN-induced DSB triggers homology-mediated recombination (dashed crosses), so that the therapeutic gene carried in the donor template is added to the host genome. If the DSB is targeted to a mutated gene region, HR can replace the faulty gene with a normal copy in the donor template. Black boxes: genes in host genome. Green boxes: therapeutic genes carried in donor templates. NHEJ: nonhomologous end joining. ZFN: zinc-finger nuclease. DSB: double-strand break. HR: homologous recombination.
Gene replacement and gene addition by HR
In the presence of a donor template consisting of similar sequences as the targeted DNA, a DSB can also trigger homology-mediated recombination, which can lead to either gene addition or gene replacement depending on where the DSB is targeted and the donor template design (Figure 4B). For example, in a hemophilia mouse model carrying a mutant human FIX gene, intraperitoneal injection of two AAV vectors expressing ZFN and a donor template, respectively, yielded addition of a partial FIX gene fragment harboring exons 2–8 to the mutant FIX transgene. The outcome was that the exon 1 of the mutant FIX transgene was fused with exons 2–8 from the donor template to reconstitute a full-length FIX open reading frame (Li et al., 2011).
The potential of genome editing in human gene therapy applications is evident, as a number of successful studies in human cells and model organisms have been reported (Li et al., 2014; Lisa Li et al., 2014). However, the specificity of editing needs to be rigorously tested to address possible off-target effects before widespread clinical use (Guilinger et al., 2014). In addition, editing efficiency needs to be further improved to meet therapeutic needs, particularly in in vivo gene therapy settings that require gene editing in an enormous number of cells throughout an organ or the whole body.
6. Conclusion
Among the current human gene therapy applications, gene replacement for treating monogenic diseases has been the most mature strategy, evidenced by successful marketing of Glybera and other late-stage clinical development. Gene addition benefits from the experience gained through gene replacement trials, and starts to show promising results in some of the most difficult-to-treat diseases after generations of vector refinement. In combination with some of the most exciting advancements in molecular biology, RNA-targeting gene expression modulation and DNA-targeting gene editing have opened new avenues for treating a much broader range of human diseases. We would like to emphasize that the classification of gene therapy strategies in this Review only aims to ease description and to help with understanding. In future gene therapy applications, the boundaries are expected to blur and cross, with continuous merging with novel biological discoveries and vectorology innovation.
ACKNOWLEDGMENTS
The authors thank the grant supports from University of Massachusetts Medical School (an internal grant), National Institutes of Health (R01NS076991-01, 1R21DA031952-01A, 2P01HL059407, 1P01AI100263-01), the Will Foundation, Jacob’s Cure, NTSAD Foundation, Canavan Foundation, and partial support from a grant from the National High Technology Research and Development Program (“863” Program) of China (2012AA020810).
Footnotes
DISCLOSURE
G. Gao is a founder of Voyager Therapeutics and holds equity in the company. G. Gao is an inventor on patents with potential royalties licensed to Voyager Therapeutics.
REFERENCES
- Aiuti A, Cattaneo F, Galimberti S, Benninghoff U, Cassani B, Callegaro L, Scaramuzza S, Andolfi G, Mirolo M, Brigida I, Tabucchi A, Carlucci F, Eibl M, Aker M, Slavin S, Al-Mousa H, Al Ghonaium A, Ferster A, Duppenthaler A, Notarangelo L, et al. Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med. 2009;360(5):447–458. doi: 10.1056/NEJMoa0805817. [DOI] [PubMed] [Google Scholar]
- Andtbacka RH, Ross MI, Delman K, Noyes D, Zager JS, Hsueh E, Ollila DW, Amatruda T, Chen L, Kaufman H. Society of Surgical Oncology Cancer Symposium. Phoenix, Arizona: 2014. Responses of Injected and Uninjected Lesions to Intralesional Talimogene Laherparepvec (T-VEC) in the OPTiM Study and the Contribution of Surgery to Response. [Google Scholar]
- Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, Viswanathan A, Holder GE, Stockman A, Tyler N, Petersen-Jones S, Bhattacharya SS, Thrasher AJ, Fitzke FW, Carter BJ, Rubin GS, Moore AT, Ali RR. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med. 2008;358(21):2231–2239. doi: 10.1056/NEJMoa0802268. [DOI] [PubMed] [Google Scholar]
- Balazs AB, Bloom JD, Hong CM, Rao DS, Baltimore D. Broad protection against influenza infection by vectored immunoprophylaxis in mice. Nat Biotechnol. 2013;31(7):647–652. doi: 10.1038/nbt.2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balazs AB, Chen J, Hong CM, Rao DS, Yang L, Baltimore D. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature. 2012;481(7379):81–84. doi: 10.1038/nature10660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennett J, Ashtari M, Wellman J, Marshall KA, Cyckowski LL, Chung DC, Mccague S, Pierce EA, Chen Y, Bennicelli JL, Zhu X, Ying GS, Sun J, Wright JF, Auricchio A, Simonelli F, Shindler KS, Mingozzi F, High KA, Maguire AM. AAV2 gene therapy readministration in three adults with congenital blindness. Sci Transl Med. 2012;4(120):120ra115. doi: 10.1126/scitranslmed.3002865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biffi A, Aubourg P, Cartier N. Gene therapy for leukodystrophies. Hum Mol Genet. 2011;20(R1):R42–R53. doi: 10.1093/hmg/ddr142. [DOI] [PubMed] [Google Scholar]
- Borel F, Kay MA, Mueller C. Recombinant AAV as a Platform for Translating the Therapeutic Potential of RNA Interference. Mol Ther. 2014;22(4):692–701. doi: 10.1038/mt.2013.285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boudreau RL, Garwick-Coppens SE, Liu J, Wallace LM, Harper SQ. Rapid Cloning and Validation of MicroRNA Shuttle Vectors: A Practical Guide. In: Harper SQ, editor. RNA Interference Techniques. Springer Science+Business Media, LLC; 2011. pp. 19–37. [Google Scholar]
- Boudreau RL, Martins I, Davidson BL. Artificial microRNAs as siRNA shuttles: improved safety as compared to shRNAs in vitro and in vivo. Mol Ther. 2009;17(1):169–175. doi: 10.1038/mt.2008.231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bradshaw AC, Baker AH. Gene therapy for cardiovascular disease: perspectives and potential. Vascul Pharmacol. 2013;58(3):174–181. doi: 10.1016/j.vph.2012.10.008. [DOI] [PubMed] [Google Scholar]
- Brenner MK, Gottschalk S, Leen AM, Vera JF. Is cancer gene therapy an empty suit? Lancet Oncol. 2013;14(11):e447–e456. doi: 10.1016/S1470-2045(13)70173-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cannon P, June C. Chemokine receptor 5 knockout strategies. Curr Opin HIV AIDS. 2011;6(1):74–79. doi: 10.1097/COH.0b013e32834122d7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carbonaro DA, Zhang L, Jin X, Montiel-Equihua C, Geiger S, Carmo M, Cooper A, Fairbanks L, Kaufman ML, Sebire NJ, Hollis RP, Blundell MP, Senadheera S, Fu PY, Sahaghian A, Chan RY, Wang X, Cornetta K, Thrasher AJ, Kohn DB, et al. Preclinical demonstration of lentiviral vector-mediated correction of immunological and metabolic abnormalities in models of adenosine deaminase deficiency. Mol Ther. 2014;22(3):607–622. doi: 10.1038/mt.2013.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Jacobson SG, Beltran WA, Sumaroka A, Swider M, Iwabe S, Roman AJ, Olivares MB, Schwartz SB, Komaromy AM, Hauswirth WW, Aguirre GD. Human retinal gene therapy for Leber congenital amaurosis shows advancing retinal degeneration despite enduring visual improvement. Proc Natl Acad Sci U S A. 2013;110(6):E517–E525. doi: 10.1073/pnas.1218933110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deng Y, Wang CC, Choy KW, Du Q, Chen J, Wang Q, Li L, Chung TK, Tang T. Therapeutic potentials of gene silencing by RNA interference: principles, challenges, and new strategies. Gene. 2014;538(2):217–227. doi: 10.1016/j.gene.2013.12.019. [DOI] [PubMed] [Google Scholar]
- Fellmann C, Lowe SW. Stable RNA interference rules for silencing. Nature cell biology. 2014;16(1):10–18. doi: 10.1038/ncb2895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferreira V, Petry H, Salmon F. Immune Responses to AAV-Vectors, the Glybera Example from Bench to Bedside. Front Immunol. 2014a;5:82. doi: 10.3389/fimmu.2014.00082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferreira V, Twisk J, Kwikkers K, Aronica E, Brisson D, Methot J, Petry H, Gaudet D. Immune responses to intramuscular administration of alipogene tiparvovec (AAV1-LPL(S447X)) in a phase II clinical trial of lipoprotein lipase deficiency gene therapy. Hum Gene Ther. 2014b;25(3):180–188. doi: 10.1089/hum.2013.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaj T, Gersbach CA, Barbas CF., 3rd ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013;31(7):397–405. doi: 10.1016/j.tibtech.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Zhang F, Adams S, Bjorkegren E, Bayford J, Brown L, Davies EG, Veys P, Fairbanks L, Bordon V, Petropoulou T, Kinnon C, Thrasher AJ. Hematopoietic stem cell gene therapy for adenosine deaminase-deficient severe combined immunodeficiency leads to long-term immunological recovery and metabolic correction. Sci Transl Med. 2011;3(97):97ra80. doi: 10.1126/scitranslmed.3002716. [DOI] [PubMed] [Google Scholar]
- Gaudet D, Methot J, Dery S, Brisson D, Essiembre C, Tremblay G, Tremblay K, De Wal J, Twisk J, Van Den Bulk N, Sier-Ferreira V, Van Deventer S. Efficacy and long-term safety of alipogene tiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open-label trial. Gene Ther. 2013;20(4):361–369. doi: 10.1038/gt.2012.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaudet D, Methot J, Kastelein J. Gene therapy for lipoprotein lipase deficiency. Curr Opin Lipidol. 2012;23(4):310–320. doi: 10.1097/MOL.0b013e3283555a7e. [DOI] [PubMed] [Google Scholar]
- Goins WF, Huang S, Cohen JB, Glorioso JC. Engineering HSV-1 Vectors for Gene Therapy. Methods Mol Biol. 2014;1144:63–79. doi: 10.1007/978-1-4939-0428-0_5. [DOI] [PubMed] [Google Scholar]
- Goyenvalle A, Babbs A, Wright J, Wilkins V, Powell D, Garcia L, Davies KE. Rescue of severely affected dystrophin/utrophin-deficient mice through scAAV-U7snRNA-mediated exon skipping. Hum Mol Genet. 2012a;21(11):2559–2571. doi: 10.1093/hmg/dds082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goyenvalle A, Davies KE. Engineering exon-skipping vectors expressing U7 snRNA constructs for Duchenne muscular dystrophy gene therapy. Methods Mol Biol. 2011;709:179–196. doi: 10.1007/978-1-61737-982-6_11. [DOI] [PubMed] [Google Scholar]
- Goyenvalle A, Wright J, Babbs A, Wilkins V, Garcia L, Davies KE. Engineering multiple U7snRNA constructs to induce single and multiexon-skipping for Duchenne muscular dystrophy. Mol Ther. 2012b;20(6):1212–1221. doi: 10.1038/mt.2012.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griesenbach U, Alton EW. Moving forward: cystic fibrosis gene therapy. Hum Mol Genet. 2013;22(R1):R52–R58. doi: 10.1093/hmg/ddt372. [DOI] [PubMed] [Google Scholar]
- Guilinger JP, Pattanayak V, Reyon D, Tsai SQ, Sander JD, Joung JK, Liu DR. Broad specificity profiling of TALENs results in engineered nucleases with improved DNA-cleavage specificity. Nat Methods. 2014;11(4):429–435. doi: 10.1038/nmeth.2845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC, Martinache C, Rieux-Laucat F, Latour S, Belohradsky BH, Leiva L, Sorensen R, Debre M, Casanova JL, Blanche S, Durandy A, Bushman FD, Fischer A, Cavazzana-Calvo M. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2010;363(4):355–364. doi: 10.1056/NEJMoa1000164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauswirth WW, Aleman TS, Kaushal S, Cideciyan AV, Schwartz SB, Wang L, Conlon TJ, Boye SL, Flotte TR, Byrne BJ, Jacobson SG. Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum Gene Ther. 2008;19(10):979–990. doi: 10.1089/hum.2008.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- High KH, Nathwani A, Spencer T, Lillicrap D. Current status of haemophilia gene therapy. Haemophilia. 2014;20(Suppl 4):43–49. doi: 10.1111/hae.12411. [DOI] [PubMed] [Google Scholar]
- Hua Y, Vickers TA, Baker BF, Bennett CF, Krainer AR. Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol. 2007;5(4):e73. doi: 10.1371/journal.pbio.0050073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Ratnakaram R, Heon E, Schwartz SB, Roman AJ, Peden MC, Aleman TS, Boye SL, Sumaroka A, Conlon TJ, Calcedo R, Pang JJ, Erger KE, Olivares MB, Mullins CL, Swider M, Kaushal S, Feuer WJ, Iannaccone A, et al. Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Arch Ophthalmol. 2012;130(1):9–24. doi: 10.1001/archophthalmol.2011.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jessup M, Greenberg B, Mancini D, Cappola T, Pauly DF, Jaski B, Yaroshinsky A, Zsebo KM, Dittrich H, Hajjar RJ Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease I. Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID): a phase 2 trial of intracoronary gene therapy of sarcoplasmic reticulum Ca2+-ATPase in patients with advanced heart failure. Circulation. 2011;124(3):304–313. doi: 10.1161/CIRCULATIONAHA.111.022889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson PR, Schnepp BC, Zhang J, Connell MJ, Greene SM, Yuste E, Desrosiers RC, Clark KR. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat Med. 2009;15(8):901–906. doi: 10.1038/nm.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. 2013;14(1):49–55. doi: 10.1038/nrm3486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiser PD, Palczewski K. Membrane-binding and enzymatic properties of RPE65. Prog Retin Eye Res. 2010;29(5):428–442. doi: 10.1016/j.preteyeres.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kochenderfer JN, Rosenberg SA. Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat Rev Clin Oncol. 2013;10(5):267–276. doi: 10.1038/nrclinonc.2013.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I, Raffeld M, Nathan DA, Lanier BJ, Morgan RA, Rosenberg SA. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116(20):4099–4102. doi: 10.1182/blood-2010-04-281931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kranias EG, Hajjar RJ. Modulation of cardiac contractility by the phospholamban/SERCA2a regulatome. Circulation research. 2012;110(12):1646–1660. doi: 10.1161/CIRCRESAHA.111.259754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H, Haurigot V, Doyon Y, Li T, Wong SY, Bhagwat AS, Malani N, Anguela XM, Sharma R, Ivanciu L, Murphy SL, Finn JD, Khazi FR, Zhou S, Paschon DE, Rebar EJ, Bushman FD, Gregory PD, Holmes MC, High KA. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature. 2011;475(7355):217–221. doi: 10.1038/nature10177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M, Suzuki K, Kim NY, Liu GH, Izpisua Belmonte JC. A cut above the rest: targeted genome editing technologies in human pluripotent stem cells. J Biol Chem. 2014;289(8):4594–4599. doi: 10.1074/jbc.R113.488247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lisa Li H, Nakano T, Hotta A. Genetic correction using engineered nucleases for gene therapy applications. Dev Growth Differ. 2014;56(1):63–77. doi: 10.1111/dgd.12107. [DOI] [PubMed] [Google Scholar]
- Madocsai C, Lim SR, Geib T, Lam BJ, Hertel KJ. Correction of SMN2 Pre-mRNA splicing by antisense U7 small nuclear RNAs. Mol Ther. 2005;12(6):1013–1022. doi: 10.1016/j.ymthe.2005.08.022. [DOI] [PubMed] [Google Scholar]
- Maguire AM, Simonelli F, Pierce EA, Pugh EN, Jr, Mingozzi F, Bennicelli J, Banfi S, Marshall KA, Testa F, Surace EM, Rossi S, Lyubarsky A, Arruda VR, Konkle B, Stone E, Sun J, Jacobs J, Dell'osso L, Hertle R, Ma JX, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358(21):2240–2248. doi: 10.1056/NEJMoa0802315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mali P, Esvelt KM, Church GM. Cas9 as a versatile tool for engineering biology. Nat Methods. 2013;10(10):957–963. doi: 10.1038/nmeth.2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mingozzi F, High KA. Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood. 2013 doi: 10.1182/blood-2013-01-306647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mingozzi F, Meulenberg JJ, Hui DJ, Basner-Tschakarjan E, Hasbrouck NC, Edmonson SA, Hutnick NA, Betts MR, Kastelein JJ, Stroes ES, High KA. AAV-1-mediated gene transfer to skeletal muscle in humans results in dose-dependent activation of capsid-specific T cells. Blood. 2009;114(10):2077–2086. doi: 10.1182/blood-2008-07-167510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitrpant C, Porensky P, Zhou H, Price L, Muntoni F, Fletcher S, Wilton SD, Burghes AH. Improved antisense oligonucleotide design to suppress aberrant SMN2 gene transcript processing: towards a treatment for spinal muscular atrophy. PLoS One. 2013;8(4):e62114. doi: 10.1371/journal.pone.0062114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller C, Flotte TR. Gene-based therapy for alpha-1 antitrypsin deficiency. COPD. 2013;10(Suppl 1):44–49. doi: 10.3109/15412555.2013.764978. [DOI] [PubMed] [Google Scholar]
- Mueller C, Tang Q, Gruntman A, Blomenkamp K, Teckman J, Song L, Zamore PD, Flotte TR. Sustained miRNA-mediated knockdown of mutant AAT with simultaneous augmentation of wild-type AAT has minimal effect on global liver miRNA profiles. Mol Ther. 2012;20(3):590–600. doi: 10.1038/mt.2011.292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nienhuis AW. Development of gene therapy for blood disorders: an update. Blood. 2013;122(9):1556–1564. doi: 10.1182/blood-2013-04-453209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palpant NJ, Dudzinski D. Zinc finger nucleases: looking toward translation. Gene Ther. 2013;20(2):121–127. doi: 10.1038/gt.2012.2. [DOI] [PubMed] [Google Scholar]
- Patel N, Reiss U, Davidoff AM, Nathwani AC. Progress towards gene therapy for haemophilia B. Int J Hematol. 2014;99(4):372–376. doi: 10.1007/s12185-014-1523-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petersen-Jones SM, Annear MJ, Bartoe JT, Mowat FM, Barker SE, Smith AJ, Bainbridge JW, Ali RR. Gene augmentation trials using the Rpe65-deficient dog: contributions towards development and refinement of human clinical trials. Adv Exp Med Biol. 2012;723:177–182. doi: 10.1007/978-1-4614-0631-0_24. [DOI] [PubMed] [Google Scholar]
- Pfister EL, Zamore PD. Huntington's disease: silencing a brutal killer. Exp Neurol. 2009;220(2):226–229. doi: 10.1016/j.expneurol.2009.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahel JA, Roska B. Gene therapy for blindness. Annual review of neuroscience. 2013;36:467–488. doi: 10.1146/annurev-neuro-062012-170304. [DOI] [PubMed] [Google Scholar]
- San Sebastian W, Samaranch L, Kells AP, Forsayeth J, Bankiewicz KS. Gene therapy for misfolding protein diseases of the central nervous system. Neurotherapeutics. 2013;10(3):498–510. doi: 10.1007/s13311-013-0191-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sikkel MB, Hayward C, Macleod KT, Harding SE, Lyon AR. SERCA2a gene therapy in heart failure: an anti-arrhythmic positive inotrope. Br J Pharmacol. 2014;171(1):38–54. doi: 10.1111/bph.12472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simonato M, Bennett J, Boulis NM, Castro MG, Fink DJ, Goins WF, Gray SJ, Lowenstein PR, Vandenberghe LH, Wilson TJ, Wolfe JH, Glorioso JC. Progress in gene therapy for neurological disorders. Nature reviews. Neurology. 2013;9(5):277–291. doi: 10.1038/nrneurol.2013.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skordis LA, Dunckley MG, Yue B, Eperon IC, Muntoni F. Bifunctional antisense oligonucleotides provide a trans-acting splicing enhancer that stimulates SMN2 gene expression in patient fibroblasts. Proc Natl Acad Sci U S A. 2003;100(7):4114–4119. doi: 10.1073/pnas.0633863100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stein L, Roy K, Lei L, Kaushal S. Clinical gene therapy for the treatment of RPE65-associated Leber congenital amaurosis. Expert Opin Biol Ther. 2011;11(3):429–439. doi: 10.1517/14712598.2011.557358. [DOI] [PubMed] [Google Scholar]
- Stroes ES, Nierman MC, Meulenberg JJ, Franssen R, Twisk J, Henny CP, Maas MM, Zwinderman AH, Ross C, Aronica E, High KA, Levi MM, Hayden MR, Kastelein JJ, Kuivenhoven JA. Intramuscular administration of AAV1-lipoprotein lipase S447X lowers triglycerides in lipoprotein lipase-deficient patients. Arterioscler Thromb Vasc Biol. 2008;28(12):2303–2304. doi: 10.1161/ATVBAHA.108.175620. [DOI] [PubMed] [Google Scholar]
- Wang D, Zhong L, Nahid MA, Gao G. The potential of adeno-associated viral vectors for gene delivery to muscle tissue. Expert Opin Drug Deliv. 2014a;11(3):345–364. doi: 10.1517/17425247.2014.871258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Yang B, Qiu L, Yang C, Kramer J, Su Q, Guo Y, Brown RH, Jr, Gao G, Xu Z. Widespread spinal cord transduction by intrathecal injection of rAAV delivers efficacious RNAi therapy for amyotrophic lateral sclerosis. Hum Mol Genet. 2014b;23(3):668–681. doi: 10.1093/hmg/ddt454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yla-Herttuala S. Endgame: glybera finally recommended for approval as the first gene therapy drug in the European union. Mol Ther. 2012;20(10):1831–1832. doi: 10.1038/mt.2012.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Thrasher AJ, Gaspar HB. Current progress on gene therapy for primary immunodeficiencies. Gene Ther. 2013;20(10):963–969. doi: 10.1038/gt.2013.21. [DOI] [PubMed] [Google Scholar]
- Zsebo K, Yaroshinsky A, Rudy JJ, Wagner K, Greenberg B, Jessup M, Hajjar RJ. Long-term effects of AAV1/SERCA2a gene transfer in patients with severe heart failure: analysis of recurrent cardiovascular events and mortality. Circulation research. 2014;114(1):101–108. doi: 10.1161/CIRCRESAHA.113.302421. [DOI] [PubMed] [Google Scholar]
