Abstract

The hope for precision medicine has long been on the drug discovery horizon, well before the Human Genome Project gave it promise at the turn of the 21st century. In oncology, the concept has finally been realized and is now firmly embedded in ongoing drug discovery programs, and with many recent therapies involving some level of patient/disease stratification, including some highly personalized treatments. In addition, several drugs for rare diseases have been recently approved or are in late-stage clinical development, and new delivery modalities in cell and gene therapy and oligonucleotide approaches are yielding exciting new medicines for rare diseases of unmet need. For common complex diseases, however, the GWAS-driven advances in annotation of the genetic architecture over the past decade have not led to a concomitant shift in refined treatments. Similarly, attempts to disentangle treatment responders from non-responders via genetic predictors in pharmacogenetics studies have not met their anticipated success. It is possible that common diseases are simply lagging behind due to the inherent time lag with drug discovery, but it is also possible that their inherent multifactorial nature and their etiological and clinical heterogeneity will prove more resistant to refined treatment paradigms. The emergence of population-based resources in electronic health records, coupled with the rapid expansion of mobile devices and digital health may help to refine the measurement of phenotypic outcomes to match the exquisite detail emerging at the molecular level.

Introduction

The term “precision medicine” and related antecedents “personalized medicine”, “stratified medicine” and “targeted therapies” convey so many different meanings that their usage can sometimes transmit little information of practical utility. Here we aim to describe the impact of genetics, genomics and related technologies on novel drug discovery for specific patient groups. There are a number of exciting developments in this domain, spanning a diverse array of applications that range from the highly personalized advances in ex vivo autologous gene therapy ( 1 ) to the group-level progress in immuno-oncology therapies for patients not defined by a particular somatic mutation or traditional tumor histology ( 2–4 ). We have adopted a definition of precision medicine which subsumes the others: “prevention and treatment that takes individual variability into account” ( 5 ).

Recent therapeutic advances in oncology demonstrate that precision medicine has moved beyond the early phase of hope and hyperbole to a practical reality ( 6 , 7 ). Most ongoing research and clinical programs in cancer treatment have precision medicine at the core, spanning targeted mutations, immunological approaches and combination therapies. Such advances are not exclusive to oncology, as identification of the genes causing rare diseases has been fueled by advances in DNA sequencing and the emergence of social media and patient- (or parent-) driven crowd-sourcing in undiagnosed conditions ( 8 ). Translation of such rare disease findings into new therapies are being enabled by long-awaited progress in alternative drug delivery modalities, with oligonucleotide approaches ( 9–11 ) and cell and gene therapies ( 12–14 ) maturing after decades of challenging development.

Chronic conditions such as Type 2 diabetes, cardiovascular disease and common respiratory conditions have all had multiple major discoveries over the past decade, some via creative studies in highly selected samples, families and/or extreme clinical phenotypes ( 15–17 ) and others via Genome Wide Association Studies (GWAS; 18 ). There is also renewed excitement and promise in challenging areas such as schizophrenia and autism spectrum disorders ( 19 , 20 ). However, with the exception of a few well-publicized examples (particularly PCSK9/LDL cholesterol and SOST/bone mineral density) ( 21 , 22 ) and some general advances in network biology, most of a large volume of findings have not translated into novel or precision directed drug discovery programs, despite considerable investment ( 23 ). Nor has pharmacogenetics—the impact of genetic variation on drug response—shown the early promise of precision medicine in the form of disease/responder refinement, though there have been notable successes in predictors of drug safety ( 24 ).

The differences amongst disease areas in progress toward more precise treatments is likely due to a host of factors including biological architecture, phenotypic complexity, stochastic influences and practical effects of research attention/funding and simply the time it takes to translate discoveries into therapies. Here we consider oncology and rare diseases as examples of success and scope for further promise, and discuss the opportunities and challenges for common chronic conditions and neurodegeneration in particular given the relevance for unmet patient need, opportunities for scientific advance and societal importance ( 25 ).

Oncology

It could be argued that precision medicine in cancer treatment started with the discovery and development of Imatinib for Chronic Myeloid Leukemia (CML), targeting the product of the common CML chromosome 9,22 translocation ( 26 ). Imatinib has revolutionized the treatment of CML and coupled with the treatment of the resistance mutations that arise with other inhibitors, CML has largely become a managed disease. A similar situation prevails with the treatment of acute promyelocytic leukemia (APL) with all trans-retinoic acid as the proliferation of these tumor cells is primarily driven by a translocation of the retinoic acid receptor gene (105).

The routine analysis of somatic mutations in biopsies from most tumors, using gene panels or exome sequencing and the tailoring of the treatment to the mutations that are found, has become virtually routine in oncology clinics ( 27 ). One of the best examples of this kind is in the treatment of activating mutations in the EGF receptor often found in (female) non-smokers with lung cancer. These patients are treated with gefitinib which inhibits the EGFR kinase and provides some short-term mediation of their disease until resistance emerges. New drugs (e.g. osimertinib) directed to the T790M mutation—a commonly arising gefitinib resistance mutation—are now available ( 28 ). This is an example of the most advanced paradigm for precision treatment of tumors: treating the primary proliferative drive in the cells first (e.g. from activation of the EGFR) and following up with a drug directed to the resistance mutations.

The treatment of melanoma with activating mutations in BRAF (V600E) with vemurafinib or dabrafanib provides another example of the precision approach. Again resistance is inevitable, usually mediated by feedback activation of the EGFR and can be treated with trametinib, a MEK inhibitor, in combination with dabrafenib ( 29 ). It may be possible to combine this with the use of checkpoint inhibitors such as anti-CTLA4 if the side effect profiles can be managed ( 30 ). The kinase inhibitors crizotinib and ceretinib, both of which inhibit ALK, an oncogenic kinase involved in anaplastic lymphoma, neuroblastoma and non-small cell lung cancer (as a translocated gene—EML4-ALK) progressed from discovery to the clinic in <5 years, shaving years off traditional drug discovery timelines. One of the reasons for this speed of clinical development was because the drugs were suitable for only the patients with overexpression of the enzyme via gene amplification, activation or translocation. This leads to an important conclusion for mechanistic translation to precision medicine: access to defined patient populations allows rapid proof of concept (ADPP-RPC)

Variations on this theme are also apparent when considering PARP inhibitors which have been shown to work more effectively in breast cancer in patients with BRCA1 mutations ( 31 ). This has been extended to evaluation of efficacy in all cancers where there are mutations in genes involved in homologous DNA repair and gene panels are being developed to identify mutations in these genes ( 32 ).

“Precision” is also being applied in monoclonal antibody (Mab) therapy for breast and other cancers. The seminal discovery that anti-EGFR antibodies (e.g. panitumumab) do not work in colon tumors that harbor KRAS mutations leads to an efficient precision medicine Mab therapy, as a relatively inexpensive diagnostic test can reveal KRAS wild-type or mutant status ( 33 ). Only those with wild-type KRAS are eligible for treatment. Similarly, only breast tumors with Her2 amplification shown by FISH or shown to have increased Her2 protein are treated with an anti-HER2 antibody trastuzumab, sometimes in combination in metastatic disease, with a Mab directed to a different epitope on Her2 (pertuzumab) ( 34 ).

Most recently, immunotherapeutic checkpoint inhibitors have spawned a new era of cancer treatment due to dramatic improvements over existing treatments in some patients. The field of immunotherapy via checkpoint inhibitors was founded on the observation that CTLA4 (CD 152) when blocked, could unlock tumor infiltrating T cells to attack antigens expressed by the tumor ( 35 ). The therapeutic antibody ipilimumab was the first antibody to be developed against CTLA4. This discovery was followed soon after by Mabs recognizing PD1 (nivolumab, pembrolizumab) and more recently PDL1 ( 36 , 37 ). The biggest issue with the new inhibitors concerns the identification of responders and non-responders, especially as some of the latest successes invoke combination therapy with these agents ( 38 ). Extensive research is being undertaken to predict responders and non-responders. Exome sequencing of tumors to assess mutation load and the expression of “neo antigens” is being coupled with HLA typing to determine whether a particular peptide epitope can be presented by the relevant HLA alleles ( 39–41 ). Transcriptional analysis of the tumor microenvironment and even microbiome sequencing is being used to find other genomic correlates ( 42 ). The use and uptake of appropriate biomarkers to guide treatment choices is variable at present. For example, the prescription of pembroluzimab (Keytruda) requires an immunohistochemical test to measure PDL1 levels in tumor biopsies whereas the prescription of nivolumab (Opdivo) does not.

The use of engineered T cells (CAR-T cells), in which T cells are taken from individual patients and engineered to express chimeric antigen receptors with or without the expression of co-stimulatory proteins, is also a more precise way to treat tumors, with particular success in acute lymphoblastic leukemia ( 43 ). This is indeed “personalized” medicine because the patient’s own cells are manipulated and it is precise because the antigens targeted are found on particular tumor cells ( 44 ).

Rare Diseases

Rare genetic diseases offer one of the clearest depictions of precision medicine because the fundamental mutation(s) are increasingly known and offer natural biomarkers for patient selection as well as possible clues to steer medicinal chemistry and dosage away from adverse events. Although it has taken 25 years since the first monogenic disease genes discoveries, targeted cystic fibrosis treatment recently received regulatory approval ( 45 ), and therapies for Duchenne’s Muscular Dystrophy therapies have reached late stage clinical development, albeit not without challenges ( 46–48 ).

Decades of studying fundamental biological mechanisms are enabling drug discovery in rare diseases, underscoring the importance of deep functional biology in an era of broad high-throughput genomics. But the recent progress in translation of rare disease treatment has also benefited greatly from technology advances. In particular, approaches harnessing RNA interference discovered nearly 20 years ago are showing signs of maturity ( 49 ), with medicines reaching regulatory approval or late stage clinical development across a range of disorders including familial hypercholesterolemia, TTR-mediated amyloidosis, hemophilia and spinal muscular atrophy ( 50–52 ). Other approaches are exquisitely tailored, targeting specific mutations or components of pathways derived from those mutations (e.g. BCL11A with beta thalassemia; 53 , 54 ), skipping specific mutations in DMD and working directly on the mechanism of action, such as allele-specific enhancement for dominant negative activity ( 55 ).

Gene therapy, once an area of great promise for Mendelian diseases, is also maturing after decades of research and some early tragic events ( 56 ). Despite estimates of over 1800 clinical trials having been conducted ( 57 ), the first regulatory approval for gene therapy was granted in 2012 (European Medicines Agency, EMA) for Glybera ( 58 ), targeting the LPL gene in individuals with familial lipoprotein lipase deficiency. EMA approval was recently granted for the first retroviral-mediated gene transfer in children with ADA-SCID ( 14 ). The US FDA has not approved any gene therapy products for sale, though a number of ongoing clinical programs are planning for FDA consideration. At present, generalized applicability of gene therapy remains limited due to practical considerations including ‘supply chain’ access and viral production for autologous therapies, potential for adverse immune responses and ongoing concerns about gene insertion sites using viral vectors.

The technological and delivery breakthroughs and the rapid pace of identifying the causes of rare diseases lie in contrast to the long paths to CF and DMD treatments, highlighting the perseverance required to translate biology to treatment, even when the mechanisms are known. A truism learned from rare diseases, though not restricted to them, is that “not every disease gene forms a good drug target and few good targets lead to new drugs”

ALS as illustration of promise and challenge

Amyotrophic lateral sclerosis (ALS, also known as Motor Neuron disease) is a relatively rare but devastating neurodegenerative disease that exemplifies ongoing progress and opportunity but also the challenge in therapeutic translation. The molecular pathology of this disease is beginning to be understood comprehensively from studying the genetics of both familial and sporadic forms. The first gene to be implicated in ALS was superoxide dismutase (SOD) where potential gain of function mutations were shown to be causative. Since that time several additional genes have been found ( 59 ), one of the most common of which is an expanded repeat in the non-coding region of a gene with unknown function called C9orf72. This gene is also involved in frontal temporal lobe dementia (FTD), providing additional evidence that these diseases are related in molecular pathology ( 60 , 61 ). Research is underway to understand how this dominant gene causes ALS, yielding evidence for both gain of function and haplo-insufficiency. For example, there is evidence that the expanded repeat RNA forms distinct secondary structures called HRE G quadraduplexes ( 62 ) that can be translated by repeat associated non-ATG (RAN) translation to form RAN peptides from both the sense and the antisense transcript ( 63 , 64 ) can form toxic RNA complexes with RNA binding proteins ( 10 ) and disrupt nucleocytoplasmic transport ( 65 , 66 ). Most recently, mice have been made expressing the expanded repeat from BAC transgenic mice with some limited recapitulation of the pathology seen in humans ( 67 , 68 ). Transgenic knockout mice lacking the C9orf72 gene also show evidence of neurodegeneration suggesting that both loss of normal function and gain of toxic function ( 69 ). In view of the frequency of the SOD and C9orf72 mutations in ALS it is reasonable to think that precise drugs will be developed to treat these patients ( 70 ). For example, antisense oligonucleotides and small molecules are being developed against both SOD and C9orf72 mRNAs ( 71–74 ). For other genes such as TDP43 where rarer mutations cause both FTD and ALS, it is less likely that specific medicines will be developed: instead it is that the genetic “signposts” point to drug strategies that may treat all ALS patients based on shared molecular pathology.

Whole exome sequencing of ALS patients has shown that loss of function mutations in TANK binding kinase 1 (TBK1) are involved in the molecular pathology of the disease ( 75 , 76 ). TBK1 has multiple substrates including Optineurin and NFkB. The former is particularly interesting since this gene product is also involved in a familial form of ALS and the protein is intimately involved in mitophagy in association with PINK and Parkin, two genes causing dopaminergic neuron loss in Parkinson’s Disease ( 77–79 ). This provides an illuminating connection between dopaminergic and motor neuron degeneration mediated by mitophagy—suggesting that the homeostasis of mitochondrial function is crucial to neuronal health. It also suggests future points of intervention for both AS and PD targeting this physiology ( 80 ).

Common Complex Diseases

In contrast to the ongoing transformation of oncology drug discovery and a number of approaches in rare diseases, common complex conditions such as cardiovascular disease, diabetes, immunological and inflammatory disorders have not experienced the same level of success. For example, excluding known drug-metabolism loci, only about one-third of all FDA-approved pharmacogenetic biomarkers are for diseases other than cancer ( http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083378.htm ). Furthermore, the majority of these biomarkers are related to drug safety rather than efficacy, where pharmacogenetics has revealed far fewer loci ( 24 ). This differs substantially from progress in human disease genetics, where the number of loci associated with common complex diseases has moved from 10s to 10 000s ( 18 ). GWAS findings are providing a number of biological insights ( 81 , 82 ), e.g. drawing out the fundamental and widespread relevance of autophagy for Crohn’s disease ( 83 ) and implicating the complement pathway in schizophrenia ( 19 ) but the biological and environmental complexity has not yet led broadly to new treatments targeting molecularly refined clinical endpoints. GWAS loci comprise targets of many approved medicines but those treatments were developed prior to the genetic association results ( 84 ).

Table 1.

 Drugs suitable for subsets of patients suffering from particular cancers

IndicationTargetDrugsComment
CMLBcr-AblImatinibFirst targeted therapeutic kinase inhibitor
PMLRARAll trans retinoic acidRAR translocations
NSCLCEGFR activating mutations (ErbB1)Gefitinib, erlotinib, lapatinibCommon in lung cancer in some female non-smokers
GISTActivating cKIt mutations
NSCLCEGFR T790M resistance mutationOsimertinibCovalent inhibitor
MelanomaBRAF V600E activating mutationsVemurafinib, dabrafinibBRAF activating mutations occur in a sub set of melanoma patients
NSCLCAlk translocationsCrizotinib, ceretinibBrigantinib and lorlatinib are third generation inhibitors targeting resistance mutations
Pediatric neuroblastomaAlk amplification and mutationCrizotinib, ceretinib
Breast cancerPARP inhibitorOlaparibSynthetic lethality of PARP inhibition in DNA repair compromised cells
Colon cancerMutant KRASPanitumumabMutant and wild-type KRAS status determine efficacy of antiEGFR antibodies in therapy
Breast cancerHer 2 (ErbB2)Trastuzumab, pertuzumabAntibodies to Her 2 positive tumors work when the receptor is over-expressed
ALL, CLLEngineered T cellsCAR-T“Personalised medicine” as T cells from individual patients that are engineered and re-infused
IndicationTargetDrugsComment
CMLBcr-AblImatinibFirst targeted therapeutic kinase inhibitor
PMLRARAll trans retinoic acidRAR translocations
NSCLCEGFR activating mutations (ErbB1)Gefitinib, erlotinib, lapatinibCommon in lung cancer in some female non-smokers
GISTActivating cKIt mutations
NSCLCEGFR T790M resistance mutationOsimertinibCovalent inhibitor
MelanomaBRAF V600E activating mutationsVemurafinib, dabrafinibBRAF activating mutations occur in a sub set of melanoma patients
NSCLCAlk translocationsCrizotinib, ceretinibBrigantinib and lorlatinib are third generation inhibitors targeting resistance mutations
Pediatric neuroblastomaAlk amplification and mutationCrizotinib, ceretinib
Breast cancerPARP inhibitorOlaparibSynthetic lethality of PARP inhibition in DNA repair compromised cells
Colon cancerMutant KRASPanitumumabMutant and wild-type KRAS status determine efficacy of antiEGFR antibodies in therapy
Breast cancerHer 2 (ErbB2)Trastuzumab, pertuzumabAntibodies to Her 2 positive tumors work when the receptor is over-expressed
ALL, CLLEngineered T cellsCAR-T“Personalised medicine” as T cells from individual patients that are engineered and re-infused
Table 1.

 Drugs suitable for subsets of patients suffering from particular cancers

IndicationTargetDrugsComment
CMLBcr-AblImatinibFirst targeted therapeutic kinase inhibitor
PMLRARAll trans retinoic acidRAR translocations
NSCLCEGFR activating mutations (ErbB1)Gefitinib, erlotinib, lapatinibCommon in lung cancer in some female non-smokers
GISTActivating cKIt mutations
NSCLCEGFR T790M resistance mutationOsimertinibCovalent inhibitor
MelanomaBRAF V600E activating mutationsVemurafinib, dabrafinibBRAF activating mutations occur in a sub set of melanoma patients
NSCLCAlk translocationsCrizotinib, ceretinibBrigantinib and lorlatinib are third generation inhibitors targeting resistance mutations
Pediatric neuroblastomaAlk amplification and mutationCrizotinib, ceretinib
Breast cancerPARP inhibitorOlaparibSynthetic lethality of PARP inhibition in DNA repair compromised cells
Colon cancerMutant KRASPanitumumabMutant and wild-type KRAS status determine efficacy of antiEGFR antibodies in therapy
Breast cancerHer 2 (ErbB2)Trastuzumab, pertuzumabAntibodies to Her 2 positive tumors work when the receptor is over-expressed
ALL, CLLEngineered T cellsCAR-T“Personalised medicine” as T cells from individual patients that are engineered and re-infused
IndicationTargetDrugsComment
CMLBcr-AblImatinibFirst targeted therapeutic kinase inhibitor
PMLRARAll trans retinoic acidRAR translocations
NSCLCEGFR activating mutations (ErbB1)Gefitinib, erlotinib, lapatinibCommon in lung cancer in some female non-smokers
GISTActivating cKIt mutations
NSCLCEGFR T790M resistance mutationOsimertinibCovalent inhibitor
MelanomaBRAF V600E activating mutationsVemurafinib, dabrafinibBRAF activating mutations occur in a sub set of melanoma patients
NSCLCAlk translocationsCrizotinib, ceretinibBrigantinib and lorlatinib are third generation inhibitors targeting resistance mutations
Pediatric neuroblastomaAlk amplification and mutationCrizotinib, ceretinib
Breast cancerPARP inhibitorOlaparibSynthetic lethality of PARP inhibition in DNA repair compromised cells
Colon cancerMutant KRASPanitumumabMutant and wild-type KRAS status determine efficacy of antiEGFR antibodies in therapy
Breast cancerHer 2 (ErbB2)Trastuzumab, pertuzumabAntibodies to Her 2 positive tumors work when the receptor is over-expressed
ALL, CLLEngineered T cellsCAR-T“Personalised medicine” as T cells from individual patients that are engineered and re-infused

Genetically validated loci form a plausible cornerstone of precision medicine for common diseases, as arguably the most efficient approach to novel drug discovery is to identify a drug target that is known to influence the clinical measure(s) of the patient sub-group of interest and then to develop a new medicine to modulate its product, e.g. as in BRAF for melanoma ( 32 ) and PCSK9 for hypercholesterolemia ( 85 , 86 ). Development of new drugs in the context of rare genetic diseases and then extending the clinical spectrum to include more common conditions has also been cited as a promising approach ( 87 ). More specific therapies could also eventually emerge from the other end of the drug discovery cycle; i.e. in the form of identification of subgroups of responders for existing treatments rather than de novo drug discovery. It is as yet unknown whether genetic or other biomarkers influence drug response at a sufficient level to support such deductive approaches generally but the emerging collections of millions of participants in population-based studies of electronic health records (EHR) will provide the basic data required to address the question, in the form of measures of treatment, plus clinical phenotypes, plus molecular predictors ( 88 ).

Technology advances such as next-generation sequencing, genome editing, and measurement of chromatin accessibility are greatly enhancing the biological resolution of common diseases by enabling increasingly detailed (and dynamic) assessment of DNA, RNA and proteins in humans. Part of the broad uptake and success of these technologies is the exquisite attention paid to quality and accuracy of measurement, which is crucial given that apparently low error rates (in absolute value) can present real problems when considered at the genomic scale. However, the translational utility of these molecular measures for precision medicine ultimately relies on their ability to inform and predict clinical descriptions, which can be highly variable and are not always measured with the same level of reliability. Future success in precision medicine will require at least as much attention to the precision of the phenotypic outcome measures as to that of the molecular and environmental predictors. The movements toward integrating genomic data with clinical information from Electronic Health Records, coupled with advances in mobile health devices and digitization are occurring at an opportune time to facilitate this phenotype refinement ( 89 ).

It is possible that it is yet too early to measure the degree of success in translation of the recent genetics findings and technology advances for common multifactorial diseases, given that many of the discoveries are relatively recent in comparison to the decades-long timescale of drug discovery. However, it is also possible that phenotypic complexity, stochastic influences and the technical difficulty of measuring cellular, sub-cellular and environmental events will not yield a resolution sufficient to clarify the underlying pathways for precision medicine ( 90 ). The extent to which successful new therapies can target specific patient groups with common complex conditions may not be known until the advances in clinical measures are coupled with those ongoing in molecular technologies and ascertained at population-level scales.

Acknowledgements

L.R.C. would like to acknowledge the constructive input of George Davey-Smith, Allen Oliff, Matt Nelson, John Whittaker, Ricardo Macarron and Stephanie Chissoe.

Conflict of Interest statement . LRC is an employee and shareholder of GlaxoSmithKline. TH declares no current conflicts but was until March 2016 an employee of Biogen.

Funding

Authors fully funded by affiliated institutions as declared.

References

1

Booth
C.
Gaspar
H.B.
Thrasher
A.J.
(
2016
)
Treating immunodeficiency through HSC gene therapy
.
Trends Mol. Med
.,
22
,
317
327
.

2

Maus
M.V.
June
C.H.
(
2016
)
Making better chimeric antigen receptors for adoptive T-cell therapy
.
Clin. Cancer Res
.,
22
,
1875
1884
.

3

Smethurst
D.
(
2013
)
A pharmacologic perspective on newly emerging T-cell manipulation technologies
.
Br. J. Clin. Pharmacol
.,
76
,
173
187
.

4

Hoos
A.
(
2016
)
Development of immuno-oncology drugs – from CTLA4 to PD1 to the next generations
.
Nat. Rev. Drug Discov
.,
15
,
235
247
.

5

Collins
F.S.
Varmus
H.
(
2015
)
A new initiative on precision medicine
.
N. Engl. J. Med
.,
372
,
793
795
.

6

Salari
K.
Watkins
H.
Ashley
E.A.
(
2012
)
Personalized medicine: hope or hype?
Eur. Heart J
.,
33
,
1564
1570
.

7

Manolio
T.A.
Chisholm
R.L.
Ozenberger
B.
Roden
D.M.
Williams
M.S.
Wilson
R.
Bick
D.
Bottinger
E.P.
Brilliant
M.H.
Eng
C.
et al. . (
2013
)
Implementing genomic medicine in the clinic: the future is here
.
Genet. Med
.,
15
,
258
267
.

8

Boycott
K.M.
Vanstone
M.R.
Bulman
D.E.
MacKenzie
A.E.
(
2013
)
Rare-disease genetics in the era of next-generation sequencing: discovery to translation
.
Nat. Rev
.,
14
,
681
691
.

9

Wittrup
A.
Lieberman
J.
(
2015
)
Knocking down disease: a progress report on siRNA therapeutics
.
Nat. Rev
.,
16
,
543
552
.

10

Lee
R.G.
Crosby
J.
Baker
B.F.
Graham
M.J.
Crooke
R.M.
(
2013
)
Antisense technology: an emerging platform for cardiovascular disease therapeutics
.
J. Cardiovasc. Transl. Res
.,
6
,
969
980
.

11

Juliano
R.L.
(
2016
)
The delivery of therapeutic oligonucleotides
.
Nucleic Acids Res
., in press.

12

Gaudet
D.
Brisson
D.
(
2015
)
Gene-based therapies in lipidology: current status and future challenges
.
Curr. Opin. Lipidol
.,
26
,
553
565
.

13

Kastelein
J.J.
Ross
C.J.
Hayden
M.R.
(
2013
)
From mutation identification to therapy: discovery and origins of the first approved gene therapy in the Western world
.
Hum. Gene Ther
.,
24
,
472
478
.

14

Cicalese
M.P.
Ferrua
F.
Castagnaro
L.
Pajno
R.
Barzaghi
F.
Giannelli
S.
Dionisio
F.
Brigida
I.
Bonopane
M.
Casiraghi
M
. et al. . (
2016
)
Update on the safety and efficacy of retroviral gene therapy for immunodeficiency due to adenosine deaminase deficiency
.
Blood
, in press.

15

Huang-Doran
I.
Sleigh
A.
Rochford
J.J.
O'Rahilly
S.
Savage
D.B.
(
2010
)
Lipodystrophy: metabolic insights from a rare disorder
.
J. Endocrinol
.,
207
,
245
255
.

16

Angulo
I.
Vadas
O.
Garcon
F.
Banham-Hall
E.
Plagnol
V.
Leahy
T.R.
Baxendale
H.
Coulter
T.
Curtis
J.
Wu
C.
et al. . (
2013
)
Phosphoinositide 3-kinase delta gene mutation predisposes to respiratory infection and airway damage
.
Science
,
342
,
866
871
.

17

Cohen
J.C.
Hobbs
H.H.
(
2013
)
Genetics. Simple genetics for a complex disease
.
Science
,
340
,
689
690
.

18

Welter
D.
MacArthur
J.
Morales
J.
Burdett
T.
Hall
P.
Junkins
H.
Klemm
A.
Flicek
P.
Manolio
T.
Hindorff
L.
et al. . (
2014
)
The NHGRI GWAS Catalog, a curated resource of SNP-trait associations
.
Nucleic Acids Res
.,
42
,
D1001
D1006
.

19

Sekar
A.
Bialas
A.R.
de Rivera
H.
Davis
A.
Hammond
T.R.
Kamitaki
N.
Tooley
K.
Presumey
J.
Baum
M.
Van Doren
V.
et al. . (
2016
)
Schizophrenia risk from complex variation of complement component 4
.
Nature
,
530
,
177
183
.

20

de la Torre-Ubieta
L.
Won
H.
Stein
J.L.
Geschwind
D.H.
(
2016
)
Advancing the understanding of autism disease mechanisms through genetics
.
Nat. Med
.,
22
,
345
361
.

21

Plenge
R.M.
Scolnick
E.M.
Altshuler
D.
(
2013
)
Validating therapeutic targets through human genetics
.
Nat. Rev. Drug Discov
.,
12
,
581
594
.

22

McClung
M.R.
Grauer
A.
(
2014
)
Romosozumab in postmenopausal women with osteopenia
.
N. Engl. J. Med
.,
370
,
1664
1665
.

23

Hodes
R.J.
Buckholtz
N.
(
2016
)
Accelerating medicines partnership: Alzheimer's disease (AMP-AD) knowledge portal aids Alzheimer's drug discovery through open data sharing
.
Expert Opin. Ther. Targets
,
20
,
389
391
.

24

Nelson
M.R.
Johnson
T.
Warren
L.
Hughes
A.R.
Chissoe
S.L.
Xu
C.F.
Waterworth
D.M.
(
2016
)
The genetics of drug efficacy: opportunities and challenges
.
Nat. Rev
.,
17
,
197
206
.

25

Hamburg
M.A.
Collins
F.S.
(
2010
)
The path to personalized medicine
.
N. Engl. J. Med
.,
363
,
301
304
.

26

Capdeville
R.
Buchdunger
E.
Zimmermann
J.
Matter
A.
Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug
.
Nat. Rev. Drug Discov
.,
1
,
493
502
.

27

Garraway
L.A.
Lander
E.S.
(
2013
)
Lessons from the cancer genome
.
Cell
,
153
,
17
37
.

28

Ayeni
D.
Politi
K.
Goldberg
S.B.
(
2015
)
Emerging agents and new mutations in EGFR-mutant lung cancer
.
Clin. Cancer Res
.,
21
,
3818
3820
.

29

Sun
C.
Wang
L.
Huang
S.
Heynen
G.J.
Prahallad
A.
Robert
C.
Haanen
J.
Blank
C.
Wesseling
J.
Willems
S.M.
et al. . (
2014
)
Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma
.
Nature
,
508
,
118
122
.

30

Ledford
H.
(
2016
)
Cocktails for cancer with a measure of immunotherapy
.
Nature
,
532
,
162
164
.

31

Benafif
S.
Hall
M.
(
2015
)
An update on PARP inhibitors for the treatment of cancer
.
Onco Targets Ther
.,
8
,
519
528
.

32

Mateo
J.
Carreira
S.
Sandhu
S.
Miranda
S.
Mossop
H.
Perez-Lopez
R.
Nava Rodrigues
D.
Robinson
D.
Omlin
A.
Tunariu
N.
et al. . (
2015
)
DNA-repair defects and olaparib in metastatic prostate cancer
.
N. Engl. J. Med
.,
373
,
1697
1708
.

33

Amado
R.G.
Wolf
M.
Peeters
M.
Van Cutsem
E.
Siena
S.
Freeman
D.J.
Juan
T.
Sikorski
R.
Suggs
S.
Radinsky
R.
et al. . (
2008
)
Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer
.
J. Clin. Oncol
.,
26
,
1626
1634
.

34

Wong
D.J.
Hurvitz
S.A.
(
2014
)
Recent advances in the development of anti-HER2 antibodies and antibody-drug conjugates
.
Ann. Transl. Med
.,
2
,
122.

35

Leach
D.R.
Krummel
M.F.
Allison
J.P.
(
1996
)
Enhancement of antitumor immunity by CTLA-4 blockade
.
Science
,
271
,
1734
1736
.

36

Lesokhin
A.M.
Callahan
M.K.
Postow
M.A.
Wolchok
J.D.
(
2015
)
On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation
.
Sci. Transl. Med
.,
7
,
280sr281.

37

Sharma
P.
Allison
J.P.
(
2015
)
The future of immune checkpoint therapy
.
Science
,
348
,
56
61
.

38

Belvin
M.
Mellman
I.
(
2015
)
Is all cancer therapy immunotherapy?
Sci. Transl. Med
.,
7
,
315fs348.

39

Snyder
A.
Makarov
V.
Merghoub
T.
Yuan
J.
Zaretsky
J.M.
Desrichard
A.
Walsh
L.A.
Postow
M.A.
Wong
P.
Ho
T.S.
et al. . (
2014
)
Genetic basis for clinical response to CTLA-4 blockade in melanoma
.
N. Engl. J. Med
.,
371
,
2189
2199
.

40

Rizvi
N.A.
Hellmann
M.D.
Snyder
A.
Kvistborg
P.
Makarov
V.
Havel
J.J.
Lee
W.
Yuan
J.
Wong
P.
Ho
T.S.
et al. . (
2015
)
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
,
348
,
124
128
.

41

Gubin
M.M.
Schreiber
R.D.
(
2015
)
CANCER. The odds of immunotherapy success
.
Science
,
350
,
158
159
.

42

Tirosh
I.
Izar
B.
Prakadan
S.M.
Wadsworth
M.H.
2nd
Treacy
D.
Trombetta
J.J.
Rotem
A.
Rodman
C.
Lian
C.
Murphy
G.
et al. . (
2016
)
Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq
.
Science
,
352
,
189
196
.

43

June
C.H.
Riddell
S.R.
Schumacher
T.N.
(
2015
)
Adoptive cellular therapy: a race to the finish line
.
Sci. Transl. Med
.,
7
,
280ps287.

44

Rosenberg
S.A.
Restifo
N.P.
(
2015
)
Adoptive cell transfer as personalized immunotherapy for human cancer
.
Science
,
348
,
62
68
.

45

Quon
B.S.
Rowe
S.M.
(
2016
)
New and emerging targeted therapies for cystic fibrosis
.
BMJ
,
352
,
i859.

46

Peltz
S.W.
Morsy
M.
Welch
E.M.
Jacobson
A.
(
2013
)
Ataluren as an agent for therapeutic nonsense suppression
.
Annu. Rev. Med
.,
64
,
407
425
.

47

Mendell
J.R.
Rodino-Klapac
L.R.
Sahenk
Z.
Roush
K.
Bird
L.
Lowes
L.P.
Alfano
L.
Gomez
A.M.
Lewis
S.
Kota
J.
et al. . (
2013
)
Eteplirsen for the treatment of Duchenne muscular dystrophy
.
Ann. Neurol
.,
74
,
637
647
.

48

Voit
T.
Topaloglu
H.
Straub
V.
Muntoni
F.
Deconinck
N.
Campion
G.
De Kimpe
S.J.
Eagle
M.
Guglieri
M.
Hood
S.
et al. . (
2014
)
Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory, randomised, placebo-controlled phase 2 study
.
Lancet Neurol
.,
13
,
987
996
.

49

Hayden
E.C.
(
2014
)
RNA interference rebooted
.
Nature
,
508
,
443.

50

Raal
F.J.
Santos
R.D.
Blom
D.J.
Marais
A.D.
Charng
M.J.
Cromwell
W.C.
Lachmann
R.H.
Gaudet
D.
Tan
J.L.
Chasan-Taber
S.
et al. . (
2010
)
Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial
.
Lancet
,
375
,
998
1006
.

51

Ackermann
E.J.
Guo
S.
Booten
S.
Alvarado
L.
Benson
M.
Hughes
S.
Monia
B.P.
(
2012
)
Clinical development of an antisense therapy for the treatment of transthyretin-associated polyneuropathy
.
Amyloid
,
19
,
43
44
.

52

Adams
D.
Coelho
T.
Obici
L.
Merlini
G.
Mincheva
Z.
Suanprasert
N.
Bettencourt
B.R.
Gollob
J.A.
Gandhi
P.J.
Litchy
W.J.
et al. . (
2015
)
Rapid progression of familial amyloidotic polyneuropathy: a multinational natural history study
.
Neurology
,
85
,
675
682
.

53

Bauer
D.E.
Kamran
S.C.
Lessard
S.
Xu
J.
Fujiwara
Y.
Lin
C.
Shao
Z.
Canver
M.C.
Smith
E.C.
Pinello
L.
et al. . (
2013
)
An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level
.
Science
,
342
,
253
257
.

54

Vierstra
J.
Reik
A.
Chang
K.H.
Stehling-Sun
S.
Zhou
Y.
Hinkley
S.J.
Paschon
D.E.
Zhang
L.
Psatha
N.
Bendana
Y.R.
et al. . (
2015
)
Functional footprinting of regulatory DNA
.
Nat. Methods
,
12
,
927
930
.

55

Murray
S.F.
Jazayeri
A.
Matthes
M.T.
Yasumura
D.
Yang
H.
Peralta
R.
Watt
A.
Freier
S.
Hung
G.
Adamson
P.S.
et al. . (
2015
)
Allele-specific inhibition of rhodopsin with an antisense oligonucleotide slows photoreceptor cell degeneration
.
Invest. Ophthalmol. Vis. Sci
.,
56
,
6362
6375
.

56

Naldini
L.
(
2015
)
Gene therapy returns to centre stage
.
Nature
,
526
,
351
360
.

57

Ginn
S.L.
Alexander
I.E.
Edelstein
M.L.
Abedi
M.R.
Wixon
J.
(
2013
)
Gene therapy clinical trials worldwide to 2012 – an update
.
J. Gene Med
.,
15
,
65
77
.

58

Gaudet
D.
Methot
J.
Dery
S.
Brisson
D.
Essiembre
C.
Tremblay
G.
Tremblay
K.
de Wal
J.
Twisk
J.
van den Bulk
N.
et al. . (
2013
)
Efficacy and long-term safety of alipogene tiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open-label trial
.
Gene Ther
.,
20
,
361
369
.

59

Renton
A.E.
Chio
A.
Traynor
B.J.
(
2014
)
State of play in amyotrophic lateral sclerosis genetics
.
Nat. Neurosci
.,
17
,
17
23
.

60

Gendron
T.F.
Belzil
V.V.
Zhang
Y.J.
Petrucelli
L.
Mechanisms of toxicity in C9FTLD/ALS
.
Acta Neuropathol
.,
127
,
359
376
.

61

Budd Haeberlein
S.L.
Harris
T.J.
(
2015
)
Promising targets for the treatment of neurodegenerative diseases
.
Clin. Pharmacol. Ther
.,
98
,
492
501
.

62

Haeusler
A.R.
Donnelly
C.J.
Periz
G.
Simko
E.A.
Shaw
P.G.
Kim
M.S.
Maragakis
N.J.
Troncoso
J.C.
Pandey
A.
Sattler
R.
et al. . (
2014
)
C9orf72 nucleotide repeat structures initiate molecular cascades of disease
.
Nature
,
507
,
195
200
.

63

Ash
P.E.
Bieniek
K.F.
Gendron
T.F.
Caulfield
T.
Lin
W.L.
Dejesus-Hernandez
M.
van Blitterswijk
M.M.
Jansen-West
K.
Paul
J.W.
3rd
Rademakers
R.
et al. . (
2013
)
Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS
.
Neuron
,
77
,
639
646
.

64

Kwon
I.
Xiang
S.
Kato
M.
Wu
L.
Theodoropoulos
P.
Wang
T.
Kim
J.
Yun
J.
Xie
Y.
McKnight
S.L.
(
2014
)
Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells
.
Science
,
345
,
1139
1145
.

65

Freibaum
B.D.
Lu
Y.
Lopez-Gonzalez
R.
Kim
N.C.
Almeida
S.
Lee
K.H.
Badders
N.
Valentine
M.
Miller
B.L.
Wong
P.C.
et al. . (
2015
)
GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport
.
Nature
,
525
,
129
133
.

66

Zhang
K.
Donnelly
C.J.
Haeusler
A.R.
Grima
J.C.
Machamer
J.B.
Steinwald
P.
Daley
E.L.
Miller
S.J.
Cunningham
K.M.
Vidensky
S.
et al. . (
2015
)
The C9orf72 repeat expansion disrupts nucleocytoplasmic transport
.
Nature
,
525
,
56
61
.

67

O'Rourke
J.G.
Bogdanik
L.
Muhammad
A.K.
Gendron
T.F.
Kim
K.J.
Austin
A.
Cady
J.
Liu
E.Y.
Zarrow
J.
Grant
S.
et al. . (
2015
)
C9orf72 BAC transgenic mice display typical pathologic features of ALS/FTD
.
Neuron
,
88
,
892
901
.

68

Peters
O.M.
Cabrera
G.T.
Tran
H.
Gendron
T.F.
McKeon
J.E.
Metterville
J.
Weiss
A.
Wightman
N.
Salameh
J.
Kim
J.
et al. . (
2015
)
Human C9ORF72 hexanucleotide expansion reproduces RNA foci and dipeptide repeat proteins but not neurodegeneration in BAC transgenic mice
.
Neuron
,
88
,
902
909
.

69

O'Rourke
J.G.
Bogdanik
L.
Yanez
A.
Lall
D.
Wolf
A.J.
Muhammad
A.K.
Ho
R.
Carmona
S.
Vit
J.P.
Zarrow
J.
et al. . (
2016
)
C9orf72 is required for proper macrophage and microglial function in mice
.
Science
,
351
,
1324
1329
.

70

Van Damme
P.
Robberecht
W.
(
2013
)
Clinical implications of recent breakthroughs in amyotrophic lateral sclerosis
.
Curr. Opin. Neurol
.,
26
,
466
472
.

71

Smith
R.A.
Miller
T.M.
Yamanaka
K.
Monia
B.P.
Condon
T.P.
Hung
G.
Lobsiger
C.S.
Ward
C.M.
McAlonis-Downes
M.
Wei
H.
et al. . (
2006
)
Antisense oligonucleotide therapy for neurodegenerative disease
.
J. Clin. Investig
.,
116
,
2290
2296
.

72

Donnelly
C.J.
Zhang
P.W.
Pham
J.T.
Haeusler
A.R.
Mistry
N.A.
Vidensky
S.
Daley
E.L.
Poth
E.M.
Hoover
B.
Fines
D.M.
et al. . (
2013
)
RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention
.
Neuron
,
80
,
415
428
.

73

Lagier-Tourenne
C.
Baughn
M.
Rigo
F.
Sun
S.
Liu
P.
Li
H.R.
Jiang
J.
Watt
A.T.
Chun
S.
Katz
M.
et al. . (
2013
)
Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration
.
Proc. Natl. Acad. Sci. U. S. A
.,
110
,
E4530
E4539
.

74

Jiang
J.
Zhu
Q.
Gendron
T.F.
Saberi
S.
McAlonis-Downes
M.
Seelman
A.
Stauffer
J.E.
Jafar-Nejad
P.
Drenner
K.
Schulte
D.
et al. . (
2016
)
Gain of toxicity from ALS/FTD-linked repeat expansions in C9ORF72 is alleviated by antisense oligonucleotides targeting GGGGCC-containing RNAs
.
Neuron
,
90
,
535
550
.

75

Cirulli
E.T.
Lasseigne
B.N.
Petrovski
S.
Sapp
P.C.
Dion
P.A.
Leblond
C.S.
Couthouis
J.
Lu
Y.F.
Wang
Q.
Krueger
B.J.
et al. . (
2015
)
Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways
.
Science
,
347
,
1436
1441
.

76

Freischmidt
A.
Wieland
T.
Richter
B.
Ruf
W.
Schaeffer
V.
Muller
K.
Marroquin
N.
Nordin
F.
Hubers
A.
Weydt
P.
et al. . (
2015
)
Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia
.
Nat. Neurosci
.,
18
,
631
636
.

77

Ordureau
A.
Sarraf
S.A.
Duda
D.M.
Heo
J.M.
Jedrychowski
M.P.
Sviderskiy
V.O.
Olszewski
J.L.
Koerber
J.T.
Xie
T.
Beausoleil
S.A.
et al. . (
2014
)
Quantitative proteomics reveal a feedforward mechanism for mitochondrial PARKIN translocation and ubiquitin chain synthesis
.
Mol. Cell
,
56
,
360
375
.

78

Matsuda
N.
Tanaka
K.
(
2015
)
Cell biology: tagged tags engage disposal
.
Nature
,
524
,
294
295
.

79

Lazarou
M.
Sliter
D.A.
Kane
L.A.
Sarraf
S.A.
Wang
C.
Burman
J.L.
Sideris
D.P.
Fogel
A.I.
Youle
R.J.
(
2015
)
The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy
.
Nature
,
524
,
309
314
.

80

Wong
Y.C.
Holzbaur
E.L.
(
2014
)
Optineurin is an autophagy receptor for damaged mitochondria in parkin-mediated mitophagy that is disrupted by an ALS-linked mutation
.
Proc. Natl. Acad. Sci. U. S. A
.,
111
,
E4439
E4448
.

81

Fugger
L.
McVean
G.
Bell
J.I.
(
2012
)
Genomewide association studies and common disease–realizing clinical utility
.
N. Engl. J. Med
.,
367
,
2370
2371
.

82

Housley
W.J.
Fernandez
S.D.
Vera
K.
Murikinati
S.R.
Grutzendler
J.
Cuerdon
N.
Glick
L.
De Jager
P.L.
Mitrovic
M.
Cotsapas
C.
et al. . (
2015
)
Genetic variants associated with autoimmunity drive NFkappaB signaling and responses to inflammatory stimuli
.
Sci. Transl. Med
.,
7
,
291ra293.

83

McGovern
D.P.
Kugathasan
S.
Cho
J.H.
(
2015
)
Genetics of inflammatory bowel diseases
.
Gastroenterology
,
149
,
1163
1176
e1162.

84

Sanseau
P.
Agarwal
P.
Barnes
M.R.
Pastinen
T.
Richards
J.B.
Cardon
L.R.
Mooser
V.
(
2012
)
Use of genome-wide association studies for drug repositioning
.
Nat. Biotechnol
.,
30
,
317
320
.

85

Robinson
J.G.
Farnier
M.
Krempf
M.
Bergeron
J.
Luc
G.
Averna
M.
Stroes
E.S.
Langslet
G.
Raal
F.J.
El Shahawy
M.
et al. . (
2015
)
Efficacy and safety of alirocumab in reducing lipids and cardiovascular events
.
N. Engl. J. Med
.,
372
,
1489
1499
.

86

Sabatine
M.S.
Giugliano
R.P.
Wiviott
S.D.
Raal
F.J.
Blom
D.J.
Robinson
J.
Ballantyne
C.M.
Somaratne
R.
Legg
J.
Wasserman
S.M.
et al. . (
2015
)
Efficacy and safety of evolocumab in reducing lipids and cardiovascular events
.
N. Engl. J. Med
.,
372
,
1500
1509
.

87

Fishman
M.C.
(
2013
)
Power of rare diseases: found in translation
.
Sci. Transl. Med
.,
5
,
201ps211.

88

Aronson
S.J.
Rehm
H.L.
(
2015
)
Building the foundation for genomics in precision medicine
.
Nature
,
526
,
336
342
.

89

Topol
E.J.
(
2014
)
Individualized medicine from prewomb to tomb
.
Cell
,
157
,
241
253
.

90

Smith
G.D.
(
2011
)
Epidemiology, epigenetics and the 'Gloomy Prospect': embracing randomness in population health research and practice
.
Int. .J Epidemiol
.,
40
,
537
562
.