Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HEALTHCARE DIAGNOSTIC
Document Type and Number:
WIPO Patent Application WO/2016/024101
Kind Code:
A1
Abstract:
A health-ageing biomarker is provided which has utility in assessing the biological age of an individual. The biomarker has particular utility in the prediction of the likelihood of an individual developing an ageing-related disease, screening for anti-ageing drugs and to 5 assist with the diagnosis of an ageing-related disease, or assessing the likelihood of an organ being successfully used or matched to a donor patient. Also presented are methods utilising the biomarker and methods of identifying such biomarkers.

Inventors:
TIMMONS JAMES ARCHIBALD (GB)
Application Number:
PCT/GB2015/052314
Publication Date:
February 18, 2016
Filing Date:
August 11, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TIMMONS JAMES ARCHIBALD (GB)
International Classes:
C12Q1/68
Domestic Patent References:
WO2003016573A12003-02-27
WO2003025122A22003-03-27
Foreign References:
FR2900936A12007-11-16
Other References:
CARLUCCI F ET AL: "A 57-gene expression signature in B-cell chronic lymphocytic leukemia", BIOMEDICINE AND PHARMACOTHERAPY, ELSEVIER, FR, vol. 63, no. 9, 1 November 2009 (2009-11-01), pages 663 - 671, XP026719333, ISSN: 0753-3322, [retrieved on 20090225], DOI: 10.1016/J.BIOPHA.2009.02.001
NICOLE NOREN HOOTEN ET AL: "Age-related changes in microRNA levels in serum", AGING, 1 October 2013 (2013-10-01), United States, pages 725 - 740, XP055219930, Retrieved from the Internet
VALERIE VANHOOREN ET AL: "Serum N-glycan profile shift during human ageing", EXPERIMENTAL GERONTOLOGY., vol. 45, no. 10, 1 October 2010 (2010-10-01), GB, pages 738 - 743, XP055219932, ISSN: 0531-5565, DOI: 10.1016/j.exger.2010.08.009
AVIK CHOUDHURI ET AL: "Non-core subunit eIF3h of translation initiation factor eIF3 regulates zebrafish embryonic development", DEVELOPMENTAL DYNAMICS., vol. 239, no. 6, 1 June 2010 (2010-06-01), US, pages 1632 - 1644, XP055219924, ISSN: 1058-8388, DOI: 10.1002/dvdy.22289
Attorney, Agent or Firm:
GLEAVE, Robert (Fugro HouseHithercroft Road, Wallingford Oxfordshire OX10 9RB, GB)
Download PDF:
Claims:
CLAIMS

1 . A method of predicting the likelihood of an individual developing an ageing-related disease, or to assist with the diagnosis of an ageing-related disease, which comprises the steps of:

(a) quantifying, in a biological sample from the individual, the level of expression of each of a panel of genes, the panel of genes comprising at least EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2; and

(b) comparing the level of expression quantified in step (a) with control levels of expression for each of the panel of genes;

such that changes in the levels of expression of the panel of genes are indicative of the individual's risk to developing the ageing-related disease or the presence of the ageing related disease.

2. A method according to claim 1 wherein the panel of genes comprises at least 30, at least 50, at least 70, or at least 120 of the genes listed in Table 2.

3. A method according to claim 1 wherein the panel of genes comprises the 150 genes listed in Table 2. 4. A method according to claim 1 wherein the panel of genes comprises at least 30, at least 50, at least 70, at least 120, or at least 150 of the genes listed in Table 1 .

5. A method according to any preceding claim in which the biological sample is a blood sample, such as whole blood or blood plasma.

6. A method according to any one of claims 1 to 4 in which the biological sample is a tissue sample, such as a tissue sample obtained from the skin, hair, oral mucosa, brain, heart, liver, lungs, stomach, pancreas, kidney, bladder, skeletal muscle, cardiac muscle or smooth muscle.

7. A method according to any preceding claim in which the ageing-related disease is Alzheimer's disease, mild cognitive impairment or dementia.

8. A method according to any preceding claim in which the ageing-related disease is characterised by a deterioration in renal function.

9. A method of predicting the likelihood of an organ from an individual over > 50 years of age being successfully used for transplantation into a donor patient which comprises the steps of:

(a) quantifying, in a biological sample from the individual, the level of expression of each a panel of genes, the panel of genes comprising EIF3H, JMJD8, CDK13, TNK2,

TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2; and

(b) comparing the levels of expression quantified in step (a) with control levels of expression for each of the panel of genes;

such that changes in the levels of expression of the panel of genes is indicative of a successful organ transplantation.

10. A method according to claim 9 wherein the panel of genes comprises at least 30, at least 50, at least 70, or at least 120 of the genes listed in Table 2. 1 1 . A method according to claim 9 wherein the panel of genes comprises the 150 genes listed in Table 2.

12. A method according to claim 9 wherein the panel of genes comprises at least 30, at least 50, at least 70, at least 120, or at least 150 of the genes listed in Table 1 .

13. A method of assessing the ageing effect of a test compound which comprises the steps of :

(a) incubating the test compound with a biological sample;

(b) quantifying the level of expression of each of a panel of genes, the panel of genes comprising EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A,

SIN3A, TFRC, TGFBR3 and U2AF2; and

(c) comparing the levels of expression quantified in step (b), with the levels of expression of each of the panel of genes in the biological sample in the absence of the test compound; such that a changes in the level of expression is indicative of the ageing effect of the test compound.

14. A method according to claim 13 wherein the panel of genes comprises at least 30, at least 50, at least 70, or at least 120 of the genes listed in Table 2 or comprises the 150 genes listed in Table 2.

15. The method according to claim 13 wherein the panel of genes comprises at least 30, at least 50, at least 70, at least 120, or at least 150 of the genes listed in Table 1

16. Use of a panel of genes comprising at least EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2 in a method of predicting the likelihood of an individual developing an ageing-related disease, or in a method to assist with the diagnosis of an ageing-related disease, or in a method of predicting the likelihood of an organ from an individual over > 50 years of age being successfully used for transplantation into a donor patient.

17. The use according to claim 16 wherein the panel of genes comprises at least 30, at least 50, at least 70, or at least 120 of the genes listed in Table 2 or comprises the 150 genes listed in Table 2.

18. The use according to claim 17 wherein the panel of genes comprises at least 30, at least 50, at least 70, at least 120, or at least 150 of the genes listed in Table 1

Description:
HEALTHCARE DIAGNOSTIC

FIELD OF THE INVENTION

This invention relates to the use of genes, and gene expression, as a biomarker in the context of healthcare and medical diagnostics, and related medical tests and methods, in relation to the ageing of an individual and ageing-related diseases.

BACKGROUND OF THE INVENTION

As the number of people routinely living into their eighth decade and beyond rises, the incidence of ageing-related diseases has significantly increased. For example, skeletal muscle atrophy and dysfunction (sarcopenia) has become an increasing age-related health problem, with economic and social consequences (Janssen, I. et al. J. Am. Geriatr. Soc. 52, 80-5 (2004)). This is matched by neuromuscular decline, including an increased prevalence of dementia. To maintain effective performance in any job role attainment of healthy ageing is essential. Furthermore, age is a rough but major parameter in most clinical decision making trees. Identifying the molecular processes governing human ageing and longevity are of great medical importance, but there have been few, human based, discoveries mainly due to the inability to effectively account for influential physiological and environmental factors. There are no diagnostics for healthy ageing in humans.

From epidemiological studies, aerobic fitness (often defined as maximal aerobic capacity) has emerged as one of the most consistent and powerful predictors of long-term health and mortality (Blair et al (1989) Jama 262: 2395-2401 ; Lee et al (201 1 ) Br J Sports Med 45: 504-510) and the present inventor has established that aerobic fitness is substantially determined by genetic factors (Lortie et a/ (1982) Hum Biol 54: 801 -812;

Timmons et a/ (2010) J Appl Physiol 108: 1487-1496). Accurate determination of aerobic fitness in the laboratory, which is time-consuming, costly and unpleasant for the patient, is used to personalize medicinal decision, e.g. determine the appropriateness of cardiac transplantation or some surgical procedures (Myers et al (2013) Circ Heart Fail 6: 21 1 -218; Voduc (2013) Thorac Surg Clin 23: 233-245).

In fact personalized treatment strategies are, slowly, impacting modern medical practice (Vargas et al (2013) PLoS currents, 5; Wiesweg et a/ (2013) Eur J Cancer 49: 3076-3082). Novel, easy to administer diagnostics that accurately and sensitively predict future health risk or help guide preventative measures would enable the evaluation of tailored treatment strategies for the individual. Such a method or diagnostic would ideally be applied to healthy middle-aged subjects that have not yet developed clinical disease to provide the greatest opportunity to enhance healthy ageing. Personalized treatment strategies are slowly impacting on modern medical practice (Wiesweg et a/ (2013)), however none yet offer the possibility to personalize advice to tackle the most frequent causes of morbidity.

In the Uppsala Longitudinal Study of Adult Men (ULSAM) it was found that combining easy to measure risk-factors for cardiovascular disease (e.g. blood pressure) with 4 single protein and biochemical measures in older participants without signs of cardiac disease ('healthy') provided a modest improvement in the C-statistic for diagnostic performance (Zethelius et al (2008) N Engl J Med 358: 2107-21 16). A greater circulating cystatin-C concentration at baseline, a parameter that informs about renal function (Inker et a/ (2012) N Engl J Med 367: 20-29), was related to 10 year mortality in participants with pre-existing disease, but is on its own unable to predict cardiovascular deaths in 'healthy' older subjects. Thus, the use of novel single molecule biomarkers, in younger or healthy population samples typically offer very modest improvements in the C-statistic (Wallentin et al (2013) PLoS One 8: e78797; Daniels et a/ (201 1 ) Circulation 123: 2101-21 10) over pre-existing disease markers or the use of chronological age (Rohatgi et a/ (2014) Clin Chem 58: 172-182). Thus to date we still lack powerful diagnostics of 'healthy ageing', tests which do not rely on biomarkers of emerging disease, and which could be applied to disease-free middle-aged subjects.

There are numerous challenges to both the development of, and the technical implementation of, diagnostics for personalized medicine (Goldberger and Buxton (2013) JAMA 309: 2559-2560), including economic considerations. Further, there are multiple competing technological platforms that yield plentiful data, but so far progress in integrating divergent data formats to yield robust and sensitive diagnostics for clinical decision making remains slow (Goldberger and Buxton (2013), supra). Personalized approaches to cancer diagnosis and treatment have been influenced by DNA sequence analysis (Tokuda et al (2009) Breast Cancer 16: 295-300; Patnaik et a/ (2010) Cancer Res 70: 36-45), and cancer arguably represents where the greatest progress has been made in terms of personalized medicine. Genome-wide association analysis has also identified 281 DNA variants which explained a yet to be verified -17% of exceptional longevity in humans (Sebastiani et al (2012) PLoS One 7: e29848). The utility of information on DNA sequence variation to guide treatment of cardiovascular disease or neurodegeneration is just being explored (Sawhney et al (2012) Curr Genomics 13: 446^462), however this approach will be severely limited by the total contribution that DNA variants make to the heterogeneity of these types of diseases. Global RNA (Passtoors et a/ (2012) PLoS One 7: e27759; Passtoors et a/ (2013) Aging Cell 12: 24-31 ; Gheorghe et al (2014) BMC Genomics 15: 132; Phillips et al (2013) PLoS Genet 9: e1003389; Glass et a/ (2013) Genome Biol 14: R75) and DNA methylation profiling (Christensen et al (2009) PLoS Genet 5: e1000602; Horvath (2013) Genome Biol 14, R1 15; Bell et a/ (2012) PLoS Genet 8: e1002629) have been utilised to search for consistent molecular events correlating with age, where samples come from cross-sectional samples spanning 5-8 decades. Such correlation analyses yield highly significant linear associations, yet by design, such models must be influenced by disease as much as the ageing process per se. For example, Hannum et al built a multi-tissue linear model of DNA methylation age-related changes that correlated with chronological age over seven decades (Hannum et a/ (2013) Mol Cell 49: 359-367). Furthermore, this molecular profile would not, for example, be useful for distinguishing how successful a person was ageing among a group with the same birth-year (Horvath (2013), supra; Hannum et a/ (2013), supra) as chronological age and methylation status co-vary tightly. Further, detectable changes in methylation would need to precede the emergence of disease by decades for it to be of practical use.

In Alzheimer's disease (AD), non-invasive blood-based diagnostics (protein or RNA) are being developed to complement clinical and brain-imaging diagnosis of AD and dramatically expand the screening capacity of the health services (Hodges, J. Alzheimers. Dis. 33, 737-53 (2013)). At best, blood RNA diagnostics are 75% accurate at distinguishing AD patients from controls, and work best in later stages of the disease. Further, while very expensive MRI based technology may be 85% accurate, epidemiological analysis indicates there is neither the equipment nor skilled work-force capacity to cope with the numbers of people at risk.

There is therefore an urgent need for an accurate molecular diagnostic of healthy physiological age and/or a molecular model of ageing that diverges sufficiently enough from chronological age.

SUMMARY OF THE INVENTION

The invention relates to the use of one or more genes as a biomarker for predicting the likelihood of an individual developing an ageing-related disease or to assist with the diagnosis of an ageing-related disease, to a method of predicting the likelihood of an individual developing an ageing-related disease or to assist with the diagnosis of an ageing- related disease, to the use of one or more genes for assessing the ageing effect of a test compound, to a method of assessing the ageing effect of a test compound, to test compounds identified by the invention as having an age-regulating effect and to a kit for assessing the ageing effect of a test compound. Furthermore, use of the biomarker is proposed in a method for identifying drug doses in patients, for rationalization of treatment decisions in a clinical setting or for estimating long-term drug safety. Furthermore, use of the biomarker is proposed as a method for stratifying donor organ status to allow the organ to be matched to the most appropriate recipient for a transplantation procedure. Furthermore, the use of the biomarker is proposed as a method to inform on future sporting performance, industrial performance or to more accurately assess life insurance or health care cost premiums.

According to a first aspect of the invention, there is provided the use of one or more analytes selected from the 670 genes listed in Table 1 as a biomarker for predicting the likelihood of an individual developing an ageing-related disease, or having an age-related clinical adverse event, or to assist with the diagnosis of an ageing-related disease.

Table 1

Gene ID Gene Name Gene ID Gene Name

217700_at CNPY4 230228_at SSC5D

234495_at KLK15 201806_s_at ATXN2L

89476_r_at NPEPL1 215377_at CTBP2

244707_at HCN4 AS 235491 _at ZBTB10

244193_at DNAJC22 206889_at PDIA2

21 1 180_x_at RUNX1 238313_at 238313_at

243906_at 243906_at 218819_at INTS6

214213_x_at LMNA 219835_at PRDM8

217079_at 217079_at 229381 _at C1 orf64

220024_s_at PRX 230561 _s_at KANSL1 L

2401 16_at 2401 16_at 231268_at MYBL1

229047_at PLEKHB1 221758_at ARMC6

241427_x_at FBXW7 238916_at LINC00938

230044_at PCYT2 210499_s_at PQBP1

216327_s_at SIGLEC8 209966_x_at ESRRG

219967_at MRM1 244218_at 244218_at

239125_at SLC25A5 205312_at SPI1

234748_x_at KIF20B 218827_s_at CEP192

206080_at PLCH2 214375_at PPFIBP1

230345_at SEMA7A 227468_at CPT1 C

238046_x_at 238046_x_at 212208_at MED13L

214209_s_at ABCB9 226428_at TNP02 Gene ID Gene Name Gene ID Gene Name

208232_x_at NRG1 230131_x_at ARSD

221309_at RBM17 238263_at EPHA1 -AS1

207883_s_at TFR2 228074_at ITPRIPL2

218762_at ZNF574 237646_x_at PLEKHG5

239523_at TUSC5 202587_s_at AK1

240241_at 240241 _at 222957_at NEU4

227563_at FAM27E3 217040_x_at SOX15

240325_x_at SOX30P1 233938_at C1 1 orf86

228279_s_at TNK2 213177_at MAPK8IP3

205050_s_at MAPK8IP2 227772_at LATS1

217410_at AGRN 21 1901_s_at PDE4A

241563_at RP1 1 -384L8.1 210332_at LOC100134498

231242_at BHLHE41 205390_s_at ANK1

223153_x_at TMUB1 205629_s_at CRH

226871_s_at ATG4D 34408_at RTN2

239837_at ADAM 1 1 206827_s_at TRPV6

214316_x_at CALR 241921_x_at 241921_x_at

209983_s_at NRXN2 239251 _at 239251 _at

222197_s_at LOC100128008 230046_at AC005789.1 1

233894_x_at COL26A1 238849_at ACY1

209097_s_at JAG1 225612_s_at B3GNT5

220849_at EPN2 219893_at CCDC71

230576_at BLOC1 S3 243239_at SAMM50

203842_s_at MAPRE3 232568_at MGC24103

212512_s_at CARM1 204249_s_at LM02

235879_at MBNL1 216647_at TCF3

227287_at CITED2 221493_at TSPYL1

207914_x_at EVX1 237144_at LTBP3

236845_at TRIM62 218834_s_at TMEM132A

238406_x_at SEZ6L2 232012_at CAPN1

213433_at ARL3 215492_x_at PTCRA

240686_x_at TFRC 34031_i_at KRIT1

210364_at SCN2B 226675_s_at MALAT1

231402_at LOC100129105 226907_at PPP1 R14C

226706_at FAM20C 239356_at LOC100129122

234342_at 234342_at 1569006_at CTB-167G5.5

239060_at 239060_at 205075_at SERPINF2

244182_at 244182_at 233073_at 233073_at

219756_s_at POF1 B 238866_at C19orf68

236269_at ZNF628 215058_at DENND5B Gene ID Gene Name Gene ID Gene Name

234400_at 234400_at 230625_s_at TSPAN12

210483_at TNFRSF10C 24121 1_at 24121 1_at

21 1837_s_at PTCRA 239152_at 239152_at

213987_s_at CDK13 217203_at GLUL

202588_at AK1 234021 _at EML2

203876_s_at MMP1 1 230907_at GPRC5C

220529_at FLJ1 1710 212177_at SFRS18

204362_at SKAP2 207468_s_at SFRP5

236278_at HIST1 H3E 231480_at SLC6A19

231520_at SLC35F3 234746_at 234746_at

217046_s_at AGER 206620_at GRAP

230375_at PNISR 229341 _at TFCP2L1

240098_at RIF1 234491_s_at SAV1

239522_at IL12RB1 215979_s_at SLC7A1

225693_s_at CAMTA1 215676_at BRF1

239422_at GPC2 237534_at 237534_at

237046_x_at IL34 53071_s_at OGFOD3

228876_at BAIAP2L2 226359_at GTPBP1

244591_x_at RNF207 240051 _at TPD52L3

22721 1_at PHF19 225571 _at LIFR

221589_s_at ALDH6A1 208661 _s_at TTC3

204974_at RAB3A 213321_at BCKDHB

234003_at ENOX2 1554274_a_at SSH1

214125_s_at NENF 207274_at CHRNE

225072_at ZCCHC3 235432_at NPHP3

234536_at SARDH 227391 _x_at LRRFIP1

215026_x_at SCNN1 A 221 136_at GDF2

217696_at FUT7 203203_s_at KRR1

206906_at I CAM 5 225428_s_at DDX54

230693_at ATP2A1 213956_at CEP350

217074_at SMOX 212845_at SAMD4A

229508_at U2AF2 21 1 1 19_at ESR2

223137_at ZDHHC4 235916_at YPEL4

234694_at CNTROB 205586_x_at VGF

220096_at RNASET2 213939_s_at RUFY3

208129_x_at RUNX1 242503_at CHST13

226141_at CCDC149 202482_x_at RANBP1

222080_s_at SIRT5 219636_s_at ARMC9

241789_at RBMS3 236479_at SCN8A

203055_s_at ARHGEF1 244212_at 244212_at Gene ID Gene Name Gene ID Gene Name

213690_s_at 213690_s_at 231974_at MLL2

215488_at 215488_at 202401 _s_at SRF

239446_x_at DCBLD2 201882_x_at B4GALT1

227781_x_at FAM57B 231 161_x_at 231 161_x_at

231764_at CHRAC1 222560_at LANCL2

219737_s_at PCDH9 221754_s_at COR01 B

229730_at SMTNL2 237463_at ZFPM1

213052_at PRKAR2A 209202_s_at EXTL3

227720_at ANKRD13B 202700_s_at TMEM63A

204731_at TGFBR3 23441 1_x_at CD44

220482_s_at SERGEF 231728_at CAPS

215649_s_at MVK 204104_at SNAPC2

238125_at ADAMTS16 223004_s_at TIMMDC1

244164_at FAM223B 209992_at PFKFB2

219150_s_at ADAP1 214312_at FOXA2

220989_s_at AMN 208607_s_at SAA1

205224_at SURF2 213922_at TTBK2

206416_at ZNF205 239643_at RP13-61613.1

239629_at CFLAR 227520_at CXorf 15

242197_x_at CD36 203437_at TMEM1 1

1556095_at UNC13C 225639_at SKAP2

229343_at GTSE1 212771_at FAM171 A1

216980_s_at SPN 214798_at ATP2C2

236091_at HMGB2 240624_x_at LOC100134685

209280_at MRC2 232534_at LIN37

228684_at ZNF503 201452_at RHEB

229607_at LOC100652912 229714_at HS6ST3

218063_s_at CDC42EP4 232480_at FLJ27365

2121 14_at ATXN7L3B 221333_at FOXP3

240147_at C7ORF50 234714_x_at ATP2B2

223426_s_at EPB41 L4B 209765_at ADAM 19

202312_s_at COL1 A1 229335_at CADM4

235671_at 235671 _at 225290_at ETNK1

226674_at SHISA4 205640_at ALDH3B1

227456_s_at C6orf 136 206646_at GLI1

231 199_at RP1 1 -271 C24.3 226439_s_at NBEA

244504_x_at ARF1 201300_s_at PRNP

236030_at RCOR2 203792_x_at PCGF2

238006_at SIN3A 242744_s_at CASR

212649_at DHX29 239368_at 239368_at Gene ID Gene Name Gene ID Gene Name

228677_s_at RASAL3 214037_s_at CCDC22

201592_at EIF3H 202305_s_at FEZ2

215844_at TNP02 241894_at VM01

240550_at OTUB2 225545_at EEF2K

227738_s_at ARMC5 223464_at OSBPL5

236746_at GALNT1 237334_at SFXN2

224886_at JMJD8 21 1322_s_at SARDH

223415_at RPP25 206820_at AGFG2

222323_at CRYGEP 222346_at LAMA1

244566_at 244566_at 237764_at AC062017.1

241618_at 241618_at 1558747_at SMCHD1

216289_at GPR144 241 125_at 241 125_at

230474_at UBIAD1 206179_s_at TPPP

208102_s_at PSD 239555_at 239555_at

213170_at GPX7 202005_at ST14

224003_at TTTY14 203124_s_at SLC1 1A2

232394_at RP1 1 -517C16.2 1552343_s_at PDE7A

243567_at 243567_at 201921_at GNG10

239508_x_at CCDC108 201750_s_at ECE1

1556096_s_at UNC13C 231030_at LOC100132618

241795_at RHEB 214917_at PRKAA1

228405_at RHPN1 235047_x_at NACC1

236885_at MEX3A 212417_at SCAMP1

232091_s_at ZDHHC24 2291 12_at SIRT5

231224_x_at PRKAG2 238080_at B4GALNT4

204375_at CLSTN3 205212_s_at ACAP1

21 1638_at IGHA1 215695_s_at GYG2

241961_at SRD5A2L2 210613_s_at SYNGR1

225239_at NEAT1 238082_at 238082_at

1568248_x_at SNORA71 B 219694_at FAM105A

234010_at 234010_at 217081_at OR2H2

207005_s_at BCL2 1556136_at MYLK4

230368_at ERF 224431_s_at SUV420H2

214105_at SOCS3 240210_at ATAD3C

222543_at DERL1 244057_s_at VSTM4

214122_at PDLIM7 240875_at CTC1

241629_at 241629_at 224932_at CHCHD10

237370_at 237370_at 227989_at LTBP4

206146_s_at RHAG 229719_s_at DERL3

209266_s_at SLC39A8 213345_at NFATC4 Gene ID Gene Name Gene ID Gene Name

234280_at REG3A 229353_s_at NUCKS1

231561_s_at APOC2 230429_at 230429_at

222066_at EPB41 L1 233128_at 233128_at

231998_at SART1 237013_at 237013_at

1558678_s_at MALAT1 242457_at 242457_at

215661_at MAST2 227991 _x_at ZBTB43

209971_x_at JTV1 207434_s_at FXYD2

243260_x_at C8orf5 207532_at CRYGD

209446_s_at PKM2 218045_x_at PTMS

243029_at KREMEN1 223266_at STRADB

214471_x_at LHB 21 1252_x_at PTCRA

236348_at TMEM176B 213306_at MPDZ

234918_at GLTSCR2 210783_x_at CLEC1 1A

21 1733_x_at SCP2 204837_at MTMR9

RP1 1 -

235929_s_at 209442_x_at ANK3

399K21 .13

238325_s_at ODF3B 243285_at LOC283335

218707_at ZNF444 210126_at PSG9

21 1476_at MYOZ2 228625_at CITED4

234928_x_at RUNX3 206278_at PTAFR

21751 1_at KAZALD1 244104_at MGAT3

230170_at OSM 217898_at EMC7

221557_s_at LEF1 208874_x_at PPP2R4

203986_at STBD1 222040_at HNRNPA1

216256_at GRM8 213971_s_at SUZ12

223147_s_at WDR33 202571 _s_at DLGAP4

228219_s_at UPB1 224996_at ASPH

213700_s_at PK 237075_at AC 104653.1

239933_x_at CCDC176 222667_s_at ASH1 L

241671_x_at CASC15 228319_at FAM84A

208104_s_at TSC22D4 203891_s_at DAPK3

209979_at ADARB1 223554_s_at RANGRF

241670_x_at LOC729177 200686_s_at SFRS1 1

21 1357_s_at ALDOB 237454_at 237454_at

1559641 _at 1559641_at 212487_at G PATCH 8

236303_at ARF3 240280_at UFSP1

21 1576_s_at SLC19A1 208809_s_at C6orf62

229434_at 229434_at 230580_at 230580_at

202138_x_at AIMP2 207643_s_at TNFRSF1A

236317_at 236317_at 224731_at HMGB1

243267_x_at 243267_x_at 227259_at CD47 Gene ID Gene Name Gene ID Gene Name

229758_at TIGD5 204144_s_at PIGQ

227684_at S1 PR2 223970_at RETNLB

236744_at PHPT1 231710_at CAPS

212958_x_at PAM 229483_at 229483_at

216821_at KRT8 239689_at 239689_at

207025_at GJC2 229709_at ATP1 B3

205424_at TBKBP1 229638_at IRX3

206338_at ELAVL3 2151 1 1_s_at TSC22D1

221013_s_at APOL2 225807_at JUB

206763_at FKBP6 214142_at ZG16

236904_x_at TECTA 229693_at TMEM220

216180_s_at SYNJ2 226400_at CDC42

206824_at CES4 228651 _at VWA1

234496_x_at NYX 244279_at SOBP

222154_s_at SPATS2L 1553702_at ZNF697

229519_at FXR1 225874_at FAM100A

243651_at CPEB3 230384_at ANKRD23

221968_s_at ZNF771 227455_at C6orf136

242287_at CLIP1 206349_at LGI1

226846_at PHYHD1 231818_x_at SLC20A2

230466_s_at 230466_s_at 232323_s_at TTC17

231558_at 231558_at 203282_at GBE1

218606_at ZDHHC7 210201_x_at BIN1

213389_at ZNF592 239920_at UBTF

218235_s_at UTP1 1 L 202146_at IFRD1

209359_x_at RUNX1 217858_s_at ARMCX3

241929_at 241929_at 213976_at CIZ1

235817_at TMEM184A 37831_at SIPA1 L3

225709_at ARL6IP6 239613_at 239613_at

213693_s_at MUC1 220641 _at NOX5

231 108_at FUS 236318_x_at FBLL1

201963_at ACSL1 236689_at RNF151

201424_s_at CUL4A 232933_at KIAA1656

209697_at 209697_at 230247_at 230247_at

215256_x_at SNX26 213125_at OLFML2B

223795_at TSPAN10 230374_at PPP1 R14B

222228_S_at ALKBH4 226903_s_at SLC6A10P

234380_x_at LOC728649 216214_at 216214_at

219417_s_at C17orf59 207106_s_at LTK

227362_at SLC2A4RG 223956_at TMPRSS13 Gene ID Gene Name Gene ID Gene Name

21301 1_s_at TPI1 207339_s_at LTB

228105_at 228105_at 201 140_s_at RAB5C

217058_at GNAS 208450_at LGALS2

213156_at 213156_at 236356_at NDUFS1

223151_at DCUN1 D5 21491 1_s_at BRD2

206986_at FGF18 207105_s_at PIK3R2

230035_at BOC 213517_at PCBP2

225480_at C1 orf122 212331_at RBL2

214335_at RPL18 212205_at H2AFV

236737_at ENTHD2 212705_x_at PNPLA2

200608_s_at RAD21 230745_s_at TOX3

209449_at LSM2 233674_at 233674_at

241935_at SHROOM1 201374_x_at PPP2CB

208474_at CLDN6 230453_s_at ATP 2 A3

241799_x_at 241799_x_at 239203_at LSMEM1

242425_at 242425_at 221763_at JMJD1 C

223801_s_at APOL4 235741_at PPIA

227937_at MYPOP 224743_at IMPAD1

208176_at DUX1 201745_at TWF1

208272_at RANBP3 232988_at KIAA0182

228823_at POLR2J2 201557_at VAMP2

236033_at ASB12 230756_at ZNF683

214056_at MCL1 222662_at PPP1 R3B

228798_x_at MAZ 228231 _at SNX8

221256_s_at HDHD3 237018_at 237018_at

216345_at ZSWIM8 200602_at APP

229040_at ITGB2-AS1 239243_at ZNF638

20561 1_at TNFSF12 214024_s_at DGCR6L

235734_at PACSIN3 2191 14_at C3orf18

231782_s_at KLK4 229198_at USP35

204692_at LRCH4 208615_s_at PTP4A2

229717_at AMIG03 214817_at UNC13A

242246_x_at MIR770 217549_at 217549_at

21 1867_s_at PCDHA10 217231_s_at MAST1

205362_s_at PFDN4 210663_s_at KYNU

233679_at MAP3K7IP1 241451_s_at 241451_s_at

RP1 1 -

229617_x_at AP2A1 232732_at

793H13.3

239428_at RAB1A 217062_at DMPK

205387_s_at CGB 243017_at USP27X-AS1

226857_at ARHGEF19 212618_at ZNF609 Gene ID Gene Name Gene ID Gene Name

244580_at 244580_at 215860_at SYT12

201375_s_at PPP2CB 21 1248_s_at CHRD

215454_x_at SFTPC 230531 _at KCNC3

201996_s_at SPEN 219051_x_at METRN

230439_at RBAK-RBAKDN 236439_at 236439_at

235383_at MY07B 1554171_at ZMYM3

236724_at CFC1 234669_x_at C1 1 orf30

208412_s_at RARB 240949_x_at 240949_x_at

227294_at ZNF689 201448_at TIA1

213740_s_at TMEM262 219654_at PTPLA

244656_at RASL10B 228668_x_at FLJ36031

223514_at CARD1 1 227167_s_at RASSF3

207667_s_at MAP2K3 223904_at PRKAG3

210393_at LGR5 205332_at RCE1

214237_x_at PAWR 209262_s_at NR2F6

228648_at LRG1 236978_at 236978_at

230221_at BAT5 225424_at GPAM

218447_at CMC2 226704_at UBE2J2

215367_at KIAA1614 244617_at GPR26

203027_s_at MVD 229852_at NMNAT1

237993_at CHCHD5 237450_at LOC389332

236258_at RBBP8NL 227662_at SYNP02

241669_x_at PRKD2 210561_s_at WSB1

232328_at ZNF552 209850_s_at CDC42EP2

239700_at ZNF710 242467_at 242467_at

215353_at 215353_at 219963_at DUSP13

205665_at TSPAN9 1553749_at FAM76B

227935_s_at PCGF5 208470_s_at HPR

204635_at RPS6KA5 212471_at AVL9

205105_at MAN2A1 207353_s_at HMX1

238345_at SLC38A10 205714_s_at ZMYND10

203996_s_at C21 orf2 234795_at 234795_at

238153_at PDE6B 229670_at 229670_at

Whilst in principle useful information may be obtained from the levels of expression of individual genes, it has been found that more accurate and reliable information can be obtained by combining information about the levels of expression of each of a panel of several genes, in a linear or non-linear manner. In one embodiment, all of the 670 genes listed in Table 1 are used as a specific panel of analyte biomarkers for predicting the likelihood of an individual developing an ageing-related disease or to assist with the diagnosis of an ageing-related disease.

Information obtained regarding the level of expression of each of the panel of biomarkers may be combined in a linear or non-linear manner.

Data is presented herein which demonstrates a number of advantageous properties for the 670 genes listed in Table 1 . For example, the 670 genes were able to distinguish between disease-free old and young brain samples from independent clinical sources and produced under independent laboratory conditions (see Table 7). In addition, the 670 genes demonstrated good classification success in sets of human skin profiles (78%, see Table 7), confirming that the muscle-derived gene-expression signature appears to be a universal diagnostic of human tissue age and able to operate across technology platforms. The panel of genes may comprise or consist all of the genes identified in Table 1 , or at least 30, 50, 70, 100, 120, 130, 140, 150, 200, 300, 500, 600 or 650 of the genes identified in Table 1 .

In one embodiment, the panel of genes selected from Table 1 does not include one or more of SKAP2, CEP192, RBM17, NPEPL1 , PDLIM7, APP or BIN1 . In a further embodiment the panel of genes selected from Table 1 does not include one or more of 1559641_at, 209697_at, 213156_at, 213690_s_at, 215353_at, 215488_at, 216214_at, 217079_at, 217549_at, 228105_at, 229434_at, 229483_at, 229670_at, 230247_at,

230429_at, 230466_s_at, 230580_at, 231 161_x_at, 231558_at, 233073_at, 233128_at, 233674_at, 234010_at, 234342_at, 234400_at, 234746_at, 234795_at, 235671_at,

236317_at, 236439_at, 236978_at, 237013_at, 237018_at, 237370_at, 237454_at,

237534_at, 238046_x_at, 238082_at, 238313_at, 239060_at, 239152_at, 239251_at, 239368_at, 239555_at, 239613_at, 239689_at, 2401 16_at, 240241_at, 240949_x_at, 241 125_at, 24121 1_at, 241451_s_at, 241618_at, 241629_at, 241799_x_at, 241921_x_at, 241929_at, 242425_at, 242457_at, 242467_at, 243267_x_at, 243567_at, 243906_at, 244182_at, 244212_at, 244218_at, 244566_at, or 244580_at.

It has been found that particularly advantageous panels of genes for use in a method of predicting the likelihood of an individual developing an ageing-related disease, or to assist with the diagnosis of an ageing-related disease, comprise at least EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2. Data is presented herein which demonstrates a number of advantageous properties for such panels of genes. For example, the 13 genes were able to distinguish between old and young muscle tissue and are shown to have utility in distinguishing patients with Alzheimer's Disease (AD) or Mild Cognitive Impairment (MCI) from controls using blood samples. In other embodiments, the panel of genes comprises EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2 as members of a panel of genes comprising at least 30, at least 50, at least 70, at least 120, or at least 150 of the genes listed in Table 1 or may consist of EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2 as members of a panel of genes comprising 30, 50, 70, 120, or 150 of the genes listed in Table 1

In a further embodiment, the one or more genes listed in Table 1 are selected from one or more, or each, of ALDH3B1 , CAPN1 , CDC42EP2, COR01 B, LTBP3, NRXN2, PPP1 R14B, RCE1 , RCOR2, SART1 , SYT12, and ZDHHC24. This embodiment of the invention provides the advantage of representing a panel of genes within the same genomic region, i.e. chromosome 1 1 q13. In another embodiment, the one or more genes listed in Table 1 are selected from one or more, or each, of ALDH3B1 , CAPN1 , CD44, CDC42EP2, COR01 B, LM02, LTBP3, NRXN2, PPP1 R14B, RCE1 , RCOR2, SART1 , SYT12, TTC17 and ZDHHC24.

In a further embodiment, the one or more genes listed in Table 1 are selected from one or more, or each, of FXYD2, SCN2B and TMPRSS13. This embodiment of the invention provides the advantage of representing a panel of genes within the same genomic region, i.e. chromosome 1 1 q23.

In one embodiment, the genes are selected from the 150 genes listed in Table 2. Thus, according to a further aspect of the invention, there is provided the use of one or more analytes selected from the 150 genes listed in Table 2 as a biomarker for predicting the likelihood of an individual developing an ageing-related disease or having an age-related clinical adverse event, or to assist with the diagnosis of an ageing-related disease.

Table 2

Gene ID Gene Name Gene ID Gene Name

217700_at CNPY4 239522_at IL12RB1

234495_at KLK15 225693_s_at CAMTA1

89476_r_at NPEPL1 239422_at GPC2

244707_at HCN4 AS 237046_x_at IL34 Gene ID Gene Name Gene ID Gene Name

244193_at DNAJC22 228876_at BAIAP2L2

21 1 180_x_at RUNX1 244591_x_at RNF207

243906_at 243906_at 22721 1_at PHF19

214213_x_at LMNA 221589_s_at ALDH6A1

217079_at 217079_at 204974_at RAB3A

220024_s_at PRX 234003_at ENOX2

2401 16_at 2401 16_at 214125_s_at NENF

229047_at PLEKHB1 225072_at ZCCHC3

241427_x_at FBXW7 234536_at SARDH

230044_at PCYT2 215026_x_at SCNN1 A

216327_s_at SIGLEC8 217696_at FUT7

219967_at MRM1 206906_at ICAM5

239125_at SLC25A5 230693_at ATP2A1

234748_x_at KIF20B 217074_at SMOX

206080_at PLCH2 229508_at U2AF2

230345_at SEMA7A 223137_at ZDHHC4

238046_x_at 238046_x_at 234694_at CNTROB

214209_s_at ABCB9 220096_at RNASET2

208232_x_at NRG1 208129_x_at RUNX1

221309_at RBM17 226141_at CCDC149

207883_s_at TFR2 222080_s_at SIRT5

218762_at ZNF574 241789_at RBMS3

239523_at TUSC5 203055_s_at ARHGEF1

240241_at 240241 _at 213690_s_at 213690_s_at

227563_at FAM27E3 215488_at 215488_at

240325_x_at SOX30P1 239446_x_at DCBLD2

228279_s_at TNK2 227781_x_at FAM57B

205050_s_at MAPK8IP2 231764_at CHRAC1

217410_at AGRN 219737_s_at PCDH9

241563_at RP1 1 -384L8.1 229730_at SMTNL2

231242_at BHLHE41 213052_at PRKAR2A

223153_x_at TMUB1 227720_at ANKRD13B

226871_s_at ATG4D 204731_at TGFBR3

239837_at ADAM 1 1 220482_s_at SERGEF Gene ID Gene Name Gene ID Gene Name

214316_x_at CALR 215649_s_at MVK

209983_s_at NRXN2 238125_at ADAMTS16

222197_s_at LOC100128008 244164_at FAM223B

233894_x_at COL26A1 219150_s_at ADAP1

209097_s_at JAG1 220989_s_at AMN

220849_at EPN2 205224_at SURF2

230576_at BLOC1 S3 206416_at ZNF205

203842_s_at MAPRE3 239629_at CFLAR

212512_s_at CARM1 242197_x_at CD36

235879_at MBNL1 1556095_at UNC13C

227287_at CITED2 229343_at GTSE1

207914_x_at EVX1 216980_s_at SPN

236845_at TRIM62 236091_at HMGB2

238406_x_at SEZ6L2 209280_at MRC2

213433_at ARL3 228684_at ZNF503

240686_x_at TFRC 229607_at LOC100652912

210364_at SCN2B 218063_s_at CDC42EP4

231402_at LOC100129105 2121 14_at ATXN7L3B

226706_at FAM20C 240147_at C7ORF50

234342_at 234342_at 223426_s_at EPB41 L4B

239060_at 239060_at 202312_s_at COL1A1

244182_at 244182_at 235671 _at 235671_at

219756_s_at POF1 B 226674_at SHISA4

236269_at ZNF628 227456_s_at C6orf 136

234400_at 234400_at 231 199_at RP1 1 -271 C24.3

210483_at TNFRSF10C 244504_x_at ARF1

21 1837_s_at PTCRA 236030_at RCOR2

213987_s_at CDK13 238006_at SIN3A

202588_at AK1 212649_at DHX29

203876_s_at MMP1 1 228677_s_at RASAL3

220529_at FLJ1 1710 201592_at EIF3H

204362_at SKAP2 215844_at TNP02

236278_at HIST1 H3E 240550_at OTUB2

231520_at SLC35F3 227738_s_at ARMC5 Gene ID Gene Name Gene ID Gene Name

217046_s_at AGER 236746_at GALNT1

230375_at PNISR 224886_at JMJD8

240098 at RIF1 223415 at RPP25

In one embodiment, all of the 150 genes listed in Table 2 are used as a specific panel of analyte biomarkers for predicting the likelihood of an individual developing an ageing-related disease or to assist with the diagnosis of an ageing-related disease.

Data is presented herein which demonstrates a number of advantageous properties for the 150 genes listed in Table 2. For example, it was found that use of the 150 genes listed in Table 2 enabled the prediction of 20 year survival (p=0.025) in a cox-regression model, with gene score as a continuous variable. It was also found that healthy controls had a significantly higher gene rank score using the 150 genes listed in Table 2 than subjects with cognitive impairment (Figure 6).

Preferably, the panel of genes may comprise all of the genes identified in Table 2, or at least 30, 50, 70, 100, 120, 130, 140, 145 or 149 of the genes identified in Table 2, or consist of 30, 50, 70, 100, 120, 130, 140, 145, 149 or 150 of the genes identified in Table 2. In other embodiments, the panel of genes comprises EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2 as members of a panel of genes comprising at least 30, at least 50, at least 70, or at least 120, of the genes listed in Table 2 or may consist of EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2 as members of a panel of genes comprising 30, 50, 70, or 120 of the genes listed in Table 2.

In one embodiment, the panel of genes selected from Table 2 does not include one or more of SKAP2, RBM17, or NPEPL1 . In a further embodiment the panel of genes selected from Table 2 does not include one or more of 213690_s_at, 215488_at, 217079_at, 234342_at, 234400_at, 235671_at, 238046_x_at, 239060_at, 2401 16_at, 240241_at, 243906_at or 244182_at.

In one embodiment, the analytes are selected from the 30 genes listed in Table 3. The analytes of this embodiment provide the advantage of yielding an optimised n=30 gene diagnostic for gene-score versus renal function at 82 years (see the data provided herein). Thus, according to a further aspect of the invention, there is provided the use of one or more analytes selected from the 30 genes listed in Table 3 as a biomarker for predicting the likelihood of an individual developing an ageing-related disease or having an age-related clinical adverse event, or to assist with the diagnosis of an ageing-related disease, such as a renal related disease or disorder or a disease characterized by a deterioration in renal function.

Table 3

Gene ID Gene Name Affymetrix EXON chip ID

223554_s_at RANGRF 3709590

205640_at ALDH3B1 3379305

229730_at SMTNL2 3742194

201300_s_at PRNP 3874751

234918_at GLTSCR2 3837464

24121 1_at 24121 1_at 3451787

220024_s_at PRX 2722787

206906_at I CAM 5 3850187

236303_at ARF3 3413680

232568_at MGC24103 3163530

231520_at SLC35F3 2461457

216289_at GPR144 3188780

202138_x_at AIMP2 2988882

218045_x_at PTMS 3442306

223147_s_at WDR33 2504766

232732_at RP1 1 -793H13.3 3898694

236278_at HIST1 H3E 2899233

213987_s_at CDK13 3047189

220096_at RNASET2 2984884

224003_at TTTY14 3422257

208661_s_at TTC3 3931320

235383_at MY07B 2574966

215661_at MAST2 2410468

231782_s_at KLK4 3868728

203986_at STBD1 27741 17

225072_at ZCCHC3 3894128

232480_at FLJ27365 3948898

212417_at SCAMP1 2817053

215454_x_at SFTPC 3089192 Gene ID Gene Name Affymetrix EXON chip ID

206646_at GLI1 3418120

In one embodiment, all of the 30 genes listed in Table 3 are used as a specific panel of analyte biomarkers for predicting the likelihood of an individual developing an ageing- related disease, or to assist with the diagnosis of an ageing-related disease.

In one embodiment, the analytes are selected from the 30 genes listed in Table 4. The analytes of this embodiment provide the advantage of yielding a strong diagnostic of mortality as demonstrated by logistic regression analysis of gene-score (continuous variable) versus mortality, where a four-fold range in gene-score alone related to up to a 70% probability of death during the 20 year follow-up period (see data presented herein, in particular Figure 4A). Thus, according to a further aspect of the invention, there is provided the use of one or more analytes selected from the 30 genes listed in Table 4 as a biomarker for predicting the likelihood of an individual developing an ageing-related disease or having an age-related clinical adverse event, such as a disease or disorder likely to result in death of the individual, or to assist with the diagnosis of an ageing-related disease.

Table 4

Gene ID Gene Name Affymetrix EXON chip ID

209765_at ADAM 19 2837413

201921_at GNG10 3362636

203055_s_at ARHGEF1 3626426

230035_at BOC 2689034

220024_s_at PRX 2722787

203027_s_at MVD 3673597

213170_at GPX7 2336439

212649_at DHX29 2857131

205586_x_at VGF 3400621

230576_at BLOC1 S3 3836135

226706_at FAM20C 3034889

234928_x_at RUNX3 2325665

218045_x_at PTMS 3442306

205362_s_at PFDN4 3222991

204104_at SNAPC2 3819312

221493_at TSPYL1 2922624 Gene ID Gene Name Affymetrix EXON chip ID

239920_at UBTF 3758967

212208_at MED13L 3433369

214125_s_at NENF 2454715

230384_at ANKRD23 2565532

213125_at OLFML2B 2364003

242425_at 242425_at 261 1238

22721 1_at PHF19 3187533

209983_s_at NRXN2 3334682

243260_x_at C8orf5 3124227

230375_at PNISR 2918542

201806_s_at ATXN2L 2991090

237534_at 237534_at 3056443

238866_at C19orf68 2976954

209262_s_at NR2F6 3824146

In one embodiment, all of the 30 genes listed in Table 4 are used as a specific panel of analyte biomarkers for predicting the likelihood of an individual developing an ageing- related disease or to assist with the diagnosis of an ageing-related disease.

In one embodiment, the analytes are selected from the 30 genes listed in Table 5. The analytes of this embodiment provide the advantage of having very high specificity and sensitivity. Thus, according to a further aspect of the invention, there is provided the use of one or more analytes selected from the 30 genes listed in Table 5 as a biomarker for predicting the likelihood of an individual developing an ageing-related disease, or having an age-related clinical adverse event, such as a skin related disease (e.g. failed wound healing) or disorder, or to assist with the diagnosis of an ageing-related disease.

Table 5

Gene Name Gene ID lllumina Chip ID

GPATCH8 212487_at ILMN 1764617

MAPK8IP3 213177_at ILMNJ 81 1574

TPPP 206179_s_at ILMNJ 718687

IMPAD1 224743_at ILMN 169631 1

CTBP2 215377_at ILMN 1691294

SIRT5 222080_s_at ILMN 1738983 Gene Name Gene ID lllumina Chip ID

RAB3A 204974_at ILMN 1755369

OLFML2B 213125_at ILMN 1765557

GNG10 201921_at ILMN 1652003

RNF207 244591_x_at ILMN 1802203

PPP2R4 208874_x_at ILMN 1652249

U2AF2 229508_at ILMN 1768930

TTC17 232323_s_at ILMN 1660810

NPEPL1 89476_r_at ILMN 1724194

ASPH 224996_at ILMN 1693771

PTMS 218045_x_at ILMN 1721046

NOX5 220641_at ILMN 1775298

PLEKHG5 237646_x_at ILMN 1765109

AK1 202588_at ILMN 1691736

METRN 219051_x_at ILMNJ 712583

PRKAG3 223904_at ILMNJ 716754

LIFR 225571_at ILMN 1709094

MY07B 235383_at ILMN 1793529

B4GALT1 201882_x_at ILMN 1766221

MAP2K3 207667_s_at ILMN 1680777

ABCB9 214209_s_at ILMN 1788928

SSH1 1554274_a_at ILMN 1727671

NRXN2 209983_s_at ILMN 1738684

SKAP2 225639_at I LMN_2125010

MVD 203027_s_at ILMN 1657550

In one embodiment, all of the 30 genes listed in Table 5 are used as a specific panel of analyte biomarkers for predicting the likelihood of an individual developing an ageing- related disease or to assist with the diagnosis of an ageing-related disease.

Preferably, the panel of genes may comprise all of the genes identified in any one of Table 3, Table 4 or Table 5, or at least 15, 20, 25, or 27 of the genes identified in any one of Table 3, Table 4 or Table 5, or may consist of 15, 20, 25, or 27 of the genes identified in any one of Table 3, Table 4 or Table 5. References herein to "biomarker" refer to a distinctive biological or biologically derived indicator of a process, event, or condition.

A major advantage of the invention is that the identified biomarkers are not affected by various extraneous physiological factors affecting the biological sample in which the level of analyte biomarkers are measured (such as body mass index, aerobic capacity, impaired glucose tolerance and physical fitness). This has the effect that the ageing signature can be used to accurately predict the likelihood of an individual developing an ageing-related disease in a wider range of test subjects.

It will be appreciated that references herein to "likelihood" refer to the probability that a particular event will occur. The biomarkers of the invention provide a novel way to assess whether an individual has a higher or lower probability, or risk, of developing an ageing- related disease, depending on the expression levels of the biomarkers defined herein.

References herein to "ageing-related disease" refer to various diseases that have been associated with the increasing biological age of an individual. Such diseases can also be referred to as "ageing-associated diseases", "degenerative diseases" or "diseases of the elderly". An individual has an increased risk of developing an ageing-related disease as their biological age increases.

Ageing-related diseases include a range of diseases such as, cardiovascular disease, atherosclerosis, coronary heart disease, cardiomyopathy, congestive heart failure, hypertensive heart disease, hypertension, arthritis, osteoarthritis, rheumatoid arthritis, type 2 diabetes, multiple system atrophy, inflammatory bowel disease, Crohn's disease, age- related cancer, shingles, cataracts, glaucoma, age-related macular degeneration, osteoporosis, sarcopenia, fibromyalgia, Parkinson's disease, Alzheimer's disease, dementia, vascular dementia, frontotemporal dementia, progressive dementia, Lewy Body dementia, semantic dementia, mild-cognitive impairment (MCI) and diseases characterised by a deterioration in renal function. Age-related conditions would also include impaired recovery from a surgical intervention, accelerated loss of muscle tissue following a fracture or accident or illness induced bed-rest, susceptibility to impaired wound healing and hence infection, susceptibility for motor-skill impairments and falls. Further, the severity of conditions that present as a type of accelerated ageing, such as multiple sclerosis, ALS (amyotrophic lateral sclerosis, often referred to as Lou Gehrig's Disease) and laminin related diseases would benefit from a more accurate prognosis of the time-course of the disease, using the diagnostic.

As the incidence of ageing-related diseases increases, along with the increasing strain on the healthcare system, it is advantageous to be able to predict an individual's likelihood of developing an ageing-related disease as this permits initiation of appropriate therapy, or preventive measures, e.g. managing risk factors. This information may also be advantageously be used to select patients to participate in clinical trials who have a higher risk of developing an ageing-related disease.

According to a further aspect of the invention there is provided the use of one or more genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or of a panel of genes as defined herein, as a biomarker for assessing the potential duration of a sporting career e.g. Major League Baseball, Grid-Iron or Soccer.

According to a further aspect of the invention there is provided the sum or alternative arithmetic conversion of the levels of expression of 2 or more genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or of the level of expression of each of a panel of genes as defined herein, to create a biological (as opposed to a chronological) ageing index for use individually or as a component of a clinical decision making nomogram or decision tree.

According to a further aspect of the invention there is provided the sum or alternative arithmetic conversion of the levels of expression of 2 or more genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or of the level of expression of each of a panel of genes as defined herein, to create a biological (as opposed to a chronological) ageing index for use individually or as a component of a decision making nomogram for trading or purchasing professional athletes. According to a further aspect of the invention there is provided the sum or alternative arithmetic conversion of the levels of expression of 2 or more genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or of the level of expression of each of a panel of genes as defined herein, to create a biological (as opposed to a chronological) ageing index for use individually or as a component of a decision making nomogram for estimating insurance costs related to health and life-span. It has been found that the 670 genes listed in Table 1 were over represented at certain genomic loci. Thus, according to a further aspect of the invention there is provided a method of predicting the likelihood of an individual developing an ageing-related disease or having an age-related clinical adverse event which comprises the step of detecting the presence of a genetic variation or a significant difference in gene expression compared with a control subject within one or more of the following regions of the human genome: 7q22, 1 1 q13 and 1 1 q23. In one embodiment, the region of the human genome is selected from 1 1 q13 and 1 1 q23. In a further embodiment, the region of the human genome is selected from 1 1 q13 and the method comprises the detection of a genetic variation within one or more, or each, of the following genes: ALDH3B1 , CAPN1 , CDC42EP2, COR01 B, LTBP3, NRXN2, PPP1 R14B, RCE1 , RCOR2, SART1 , SYT12 and ZDHHC24. In a further embodiment, the region of the human genome is selected from 1 1 q23 and the method comprises the detection of a genetic variation within one or more, or each, of the following genes: FXYD2, SCN2B and TMPRSS13.

References herein to "genetic variation" include any variation in the native, non- mutant or wild type genetic code of the gene under analysis. Examples of such genetic variations include: mutations {e.g. point mutations), substitutions, deletions, insertions, single nucleotide polymorphisms (SNPs), haplotypes, chromosome abnormalities, Copy Number Variation (CNV), epigenetics and DNA inversions. According to a further aspect of the invention, there is provided a method of predicting the likelihood of an individual developing an ageing-related disease, or to assist with the diagnosis of an ageing-related disease, or predicting the likelihood of an organ from an individual over > 50 years of age being successfully used for transplantation into a donor patient, which comprises the steps of:

(a) quantifying, in a biological sample from the individual, the level of expression of one or more analyte biomarkers as defined herein; and

(b) comparing the level of expression quantified in step (a), with a control level of

expression of the one or more analyte biomarkers;

such that a change in expression is indicative of the individual's risk to developing an ageing-related disease or death, or the presence of the ageing related disease, or of a successful organ transplantation. Preferably, the level of expression of each of a panel of genes, as defined herein, is quantified in the biological sample from the individual and compared with the control levels of expression for each of the panel of genes. In one embodiment, the panel of genes comprises at least EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2. In another embodiment, the panel of genes comprises at least EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2 as members of a panel of genes comprising at least 30, at least 70, at least 120, or at least 150 of the genes listed in Table 1 , or at least 30, at least 70, or at least 120 of the genes listed in Table 2. In further embodiments, the panel of genes comprises at least 30 of the 670 genes listed in Table 1 , such as at least the 30 genes listed in any one of Table 3, Table 4 and Table 5, or at least 150 of the 670 genes listed in Table 1 , such as at least the 150 genes listed in Table 2.

Information from the method of predicting the likelihood of an individual developing an ageing-related disease as defined herein may be used in a method of selecting individuals to participate in a clinical trial, such as a clinical trial to assess the efficacy of a new method of treatment of the ageing-related disease, for example Alzheimer's disease. The information obtained relating to the likelihood of the development of the ageing-related disease for each individual may be used to stratify the individuals, enabling individuals with a high risk of the disease to be selected to participate in the clinical trial. For example, to screen new Alzheimer's disease drugs in 2015, 1 million older people are required to undergo an initial assessment to find the most suitable 100,000. The present method could reduce the initial numbers 500% and so speed up drug development 5-fold. According to a further aspect of the invention there is provided a method of predicting the likelihood of an individual developing an ageing-related disease, or to assist with the diagnosis of an ageing-related disease, or predicting the likelihood of an organ from an individual over > 50 years of age being successfully used for transplantation into a donor patient, which comprises the steps of (i) quantifying, in a biological sample from the individual, the level of expression of each of a panel of genes; and (ii) comparing the levels of expression quantified in step (i), with control levels of expression for each of the panel of genes; such that changes in the levels of expression are indicative of the individual's risk to developing the ageing-related disease or of a successful organ transplantation; and wherein the panel of genes is selected using a method comprising the steps of: (a) obtaining a biological sample from one or more young human subjects; (b) obtaining a biological sample from one or more older human subjects wherein said older human subjects are disease free; (c) conducting gene expression analysis upon each of the samples obtained in steps (a) and (b) and selecting a panel of genes which show a significant difference in gene expression between the samples obtained in steps (a) and (b).

It will be appreciated that the term "quantifying" refers to calculating the amount of analyte biomarker, such as the amount of each of a panel of genes, in a sample. This may include determining the concentration of the analyte biomarker present in a sample.

Quantification may be performed directly on the sample, indirectly on an extract therefrom, or on a dilution. In one embodiment, the level of gene expression may be quantified using a method comprising the following steps: (i) reverse transcription of RNA to cDNA; (ii) hybridization with at least one oligonucleotide probe; (iii) quantification of gene expression levels. The method may additionally include the step of labeling the cDNA, for example, prior to hybridization. As an alternative, the oligonucleotide probes may be labelled. The quantification of gene expression levels may be carried out, for example, using an analysis of fluorescence or radioisotope levels, depending on the method of labelling utilized.

Quantification may be carried out using at least one DNA microarray, with analysis carried out, for example, utilising a DNA microarray scanner.

Therefore, in a further aspect of the invention there is provided a method of predicting the likelihood of an individual developing an ageing-related disease, or to assist with the diagnosis of an ageing-related disease, or predicting the likelihood of an organ from a person over > 50 years of age being successfully used for transplantation into a donor patient, which comprises the steps of:

(a) contacting, under conditions allowing hybridization between complementary

sequences, the nucleic acids from a biological sample from a test subject and a panel of probes, the panel of probes, for example, comprising at least 30 of the probe sets identified in Table 1 , Table 2, Table 3, Table 4 or Table 5, in order to obtain an expression profile; and

(b) comparing the expression profile generated in step (a), with a control level of

expression;

such that a change in expression is indicative of an individual's risk to developing an ageing- related disease, or the presence of the ageing related disease, or of a successful organ transplantation.

The panel of probes may comprise at least 30, 50, 70, 100, 120, 130, 140, 150, 200, 300, 500, 600 or 650 of the probesets identified in Table 1 (by Gene IDs), or at least 30, 50, 70, 100, 120, 130, 140, 145 or 149 of the probesets identified in Table 2, or at least 15, 20, 25, or 27 of the probesets identified in any one of Table 3, Table 4 or Table 5, or may alternatively comprise probesets with a complementary sequence to the panels of probes defined herein. Preferably, the panel of probes comprises at least the probesets 204974_at, 201592_at, 209983_s_at, 240686_x_at, 238006_at, 229508_at, 214316_x_at, 204731_at, 224886_at, 213987_s_at, 215844_at, 212512_s_at and, 228279_s_at.

The "control level" used in the methods of the invention may be provided as a reference value for the expression level of the chosen analyte, or of each of a panel of analytes, in a test subject of the corresponding age range. A reference value may be devised from a statistical assessment of the expression levels of a particular analyte, or of a panel of analytes, generated from biological samples taken from a plurality or statistically- significant number of test subjects of the corresponding age range. The control level of a particular analyte, or of each of a panel of analytes, may also be derived from externally available gene expression data sets. In one embodiment, the control level value of a particular analyte, such as each of a panel of analytes, may be generated by measuring the expression level of an analyte defined herein, in skeletal muscle biopsies. In a further embodiment the control level values may be generated from samples obtained from at least 10, at least 20, or in particular at least 30 test subjects of a selected age range.

Human skeletal muscle provides the ideal starting tissue from which to generate a 'clean' ageing molecular classifier, as skeletal muscle RNA is easily accessible and its functional status can be studied in great detail prior to tissue sampling in all age groups. This lies in very distinct contrast to using brain, myocardium or any one of a number of other potential human tissue sources because the function of the latter examples can not be measured at the time of tissue sampling.

A change in expression level of the analyte biomarkers defined herein, is indicative of an individual's risk of developing an ageing-related disease. If the ageing signature is opposed or inhibited, i.e. the expression of an analyte which is up-regulated with age is decreased compared to the control value or an analyte which is down-regulated with age is increased compared to the control value, this is indicative of an individual having a greater risk of developing an ageing-related disease, or the presence of the ageing-related disease, or having a higher mortality (Figure 4B). If the ageing signature is activated or induced, i.e. the expression of an analyte which is up-regulated with age is increased compared to the control value or an analyte which is down-regulated with age is decreased compared to the control value, this is indicative of an individual having activated the 'healthy age' programme with the concomitant improved mortality or functional capacity.

The change in expression levels may be assessed, for example, using a gene- ranking approach. Each of the gene expression levels, obtained by quantification of the biological sample from the individual, may be compared with the level of expression of the same gene in each of multiple biological samples taken from multiple different test subjects. The gene expression level may then be ranked in comparison with the levels of expression observed in the samples from test subjects. The order of the ranking takes into account whether the gene is up-regulated or down-regulated during healthy-ageing, such as whether the gene was up-regulated or down-regulated between the young and old samples in the 'Stockholm' data set. The rankings of all of the genes of the panel may then be combined, for example using the sum, median, mean or alternative arithmetic conversion. It is advantageous to be able to assess an individual's biological age accurately, so that if individuals are identified as having a high risk of developing an ageing-related disease they can act accordingly to reduce their risk, such as through lifestyle changes or prophylactic treatment. The analyte biomarkers defined herein have a further advantage because they can provide insight into which physiological traits have potential links to longevity.

In one embodiment the biological sample from the individual and/ or the biological sample from the young and/or older human subjects is a tissue sample. This may be a tissue homogenate, tissue section and biopsy specimens taken from a live subject, or taken postmortem. The samples can be prepared, for example where appropriate diluted or concentrated, and stored in the usual manner.

The analyte biomarkers provided by the invention, have the considerable advantage of accurately predicting the biological ageing in a variety of tissues, and hence the likelihood of an individual developing an ageing-related disease. This allows the method to be carried out on any tissue that is the most cost-effective and readily available.

In a further embodiment the tissue sample is obtained from the skin, hair, oral mucosa, brain, heart, liver, lungs, stomach, pancreas, kidney, bladder, skeletal muscle, cardiac muscle or smooth muscle. In a further embodiment, the tissue sample is obtained from skeletal muscle. In one embodiment, the biological sample is a sample of cells. In one embodiment the biological sample from the individual and/or the biological sample from the young and/or older human subjects is a blood sample, such as whole blood, blood serum or blood plasma. In one embodiment the quantification of analyte biomarkers is performed using a biosensor.

In one embodiment the ageing-related disease is Alzheimer's disease (AD), mild cognitive impairment (MCI) or dementia. In another embodiment, ageing-related disease is AD, MCI, or dementia and the biological sample from the individual is a blood sample, such as whole blood, blood serum or blood plasma. In a further embodiment, the ageing-related disease is AD, MCI, or dementia, the biological sample from the individual is a blood sample, such as whole blood, blood serum or blood plasma, and the biological sample from the young and older human subjects is a tissue sample obtained from skeletal muscle or skin. It will be appreciated that the use of the analyte biomarkers described herein advantageously provides a diagnostic of cognitive impairment utilizing only peripheral samples. The analyte biomarkers may additionally be combined with alternative diagnostic tests utilising other biomarkers of cognitive impairment, or with diagnostics based on clinical parameters, to enhance the performance of such diagnostics.

It will be appreciated that the methodology of identifying the analyte biomarkers of the invention constitutes a novel and inventive aspect of the invention not used in previous studies. For example, it is common practice to identify an age related biomarker by comparing analyte levels (via gene expression levels) in a sample obtained from a young subject with analyte levels in a sample obtained from an elderly subject. By contrast, the present invention obtained samples from young subjects (i.e. subjects under 28 years of age) and older subjects (i.e. subjects over 59 years of age) who were free from clinical metabolic and cardiovascular disease. In addition, the young and older subjects may be selected to have equivalent aerobic fitness levels as determined using gas analysis and a maximal exercise protocol.

The advantage of the method of the invention is that the genes identified should associate with, or reflect, healthy physiological age rather than disease as older subjects were specifically selected to be disease free.

In one embodiment, the young human subjects are under 30 years of age. In a further embodiment, the young human subjects are between 18 and 30 years of age. In a yet further embodiment, the young human subjects are selected from any one of the following ages: 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 , 20, 19 or 18 years of age, such as younger than 28 years of age.

References herein to "disease free" refer to a subject not presenting with any symptoms of a diagnosable disease or disorder. In one embodiment, disease free comprises free from metabolic and cardiovascular disease. In a further embodiment, said older human subjects comprise subjects having a good aerobic fitness and glucose tolerance. Preferably, the young and old subjects are selected to have equivalent aerobic fitness levels as determined using gas analysis and a maximal exercise protocol. In one embodiment, the ageing-related disease is AD or MCI and the older human subjects are free from AD and / or MCI.

In one embodiment, the older human subjects are older than the young human subjects sampled in step (a) of the described aspects of the invention. In a further embodiment, the older human subjects are between 55 and 70 years of age. In a yet further embodiment, the older human subjects are selected from any one of the following ages: 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69 or 70 years of age, such as greater than 59 years of age. In another embodiment the young human subjects are under 30 years of age and the older subjects are greater than 59 years of age or the older subjects were between 55 and 70 years of age. In yet another embodiment the young human subjects are between 18 and 30 years of age and the older subjects are between 55 and 70 years of age.

According to a further aspect of the invention there is provided a method of identifying a biomarker for predicting the likelihood of an individual developing an ageing- related disease, or having an age-related clinical adverse event, or to assist with the diagnosis of an ageing-related disease wherein said method comprises the steps of:

(a) obtaining a biological sample from one or more young human subjects;

(b) obtaining a biological sample from one or more older human subjects wherein said older human subjects are disease free;

(c) conducting gene expression analysis upon each of the samples obtained in steps (a) and (b);

wherein a significant difference in gene expression between the samples obtained in steps (a) and (b) is indicative of a biomarker for predicting the likelihood of an individual developing an ageing-related disease, or having an age-related clinical adverse event, or the presence of the ageing related disease. According to a further aspect of the invention, there is provided a biomarker for predicting the likelihood of an individual developing an ageing-related disease, or having an age-related clinical adverse event, or the presence of the ageing related disease identified by the method defined herein.

In one embodiment, the biomarker is one or more analytes selected from the genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5. Preferably the biomarker is a panel of genes as defined herein. According to a further aspect of the invention, there is provided a biomarker as defined herein for use in predicting the likelihood of an organ from a person over > 50 years of age being successfully used for transplantation into a donor patient. Furthermore, there is provided a biomarker as defined herein for use in a method of stratifying donor organ status to enable matching the organ to the most appropriate recipient for transplantation. In one embodiment, the biomarker is one or more analytes selected from the genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5. Preferably the biomarker is a panel of genes as defined herein.

References herein to "biosensor" refer to anything capable of detecting the presence of the biomarker. For example, the biosensor may comprise a high throughput screening technology, e.g. configured in an array format, such as a chip or as a multi-well array. High- throughput screening technologies are particularly suitable to monitor biomarker signatures for the identification of potentially useful ageing compounds. A biosensor may also comprise a ligand or ligands capable of specific binding to the analyte biomarker, such as an antibody or biomarker-binding fragment thereof, or other oligonucleotide, or ligand, e.g. aptamer, or peptide, capable of specifically binding the biomarker. The ligand may possess a detectable label, such as a luminescent, fluorescent or radioactive label, and/or an affinity tag.

Suitably, biosensors for detection of one or more biomarkers of the invention combine biomolecular recognition with appropriate means to convert detection of the presence, or quantification, of the biomarker in the sample into a signal. According to a further aspect of the invention, there is provided the use of one or more analytes selected from the genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or of a panel of genes as defined herein, as a biomarker for assessing the ageing effect of a test compound. Analyte biomarkers can be used in, for example, clinical screening, drug screening and development. Biomarkers and uses thereof are important in the identification of novel compounds in in vitro and/or in vivo assays.

The biomarkers described herein may also be referred to collectively as an "ageing molecular classifier", "healthy ageing diagnostic" or "longevity diagnostic". They are part of the first accurate multi-tissue molecular classifier of ageing, as supported by the data provided herein.

Therefore, the biomarkers provided by the invention can act as a valuable indicator to establish whether a test compound has an effect on ageing in a variety of tissues. They represent a new resource for developing small-molecule drugs targeted at modifying ageing biology. The biomarkers described herein can also be used as suitable toxicology biomarkers to be used in drug-safety screening. In particular, they can be used to predict whether a compound will have any long-term side-effects on the premature ageing of a tissue.

According to a further aspect of the invention there is therefore provided the use of one or more genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or of a panel of genes as defined herein, as a biomarker for assessing the safety effect of a test compound.

Ageing can have an effect upon the physiological condition of a cell, tissue or organism. References herein to "ageing effect" refer to both a pro- and anti-ageing effect. An "anti healthy ageing" effect results when the ageing signature, as described herein, is opposed, whereas a "pro healthy ageing" effect results when the ageing signature is induced. The invention has the advantage of distinguishing whether a test compound has an anti-health, a pro-health or no effect on healthy ageing at all (for drug safety).

References herein to "test compound" can refer to a chemical or pharmaceutical substance to be tested using the analyte biomarkers described herein. The test compound may be a known substance or a novel synthetic or natural chemical entity, or a combination of two or more of the aforesaid substances.

In one embodiment each of the genes listed in Table 1 or Table 2 or Table 3 or Table 4 or Table 5, or a panel of genes as defined herein, are used as a specific panel of analyte biomarkers for assessing the ageing effect of a test compound. According to a further aspect of the invention, there is provided a method of assessing the ageing effect of a test compound which comprises the steps of:

(a) incubating the test compound with a biological sample;

(b) quantifying the level of expression of one or more of the analyte biomarkers as defined herein; and

(c) comparing the level of expression quantified in step (b), with the level of expression of the one or more analyte biomarkers in said biological sample in the absence of the test compound;

such that a change in expression is indicative of the ageing effect of the test compound.

It will be understood that activation of the health ageing expression pattern is indicative of a test compound having a beneficial effect, whereas inhibition of the health ageing expression pattern is indicative of a test compound having a pro-ageing or unhealthy effect.

The invention described herein, has the advantage of distinguishing whether a compound has a pro healthy ageing or an anti healthy ageing effect in a single procedure, depending on whether the ageing signature is opposed or induced directly in human material. This helps to cut down costs when screening multiple test compounds using accurate, but expensive, microarray technologies.

A further advantage of the invention is that the identified biomarkers are not affected by various extraneous physiological factors affecting the biological sample that the compounds are tested on (such as body mass index, aerobic capacity, impaired glucose tolerance and physical fitness). This indicates that the compounds identified by the analyte biomarkers to have an ageing effect, are more likely to work on a wider range of consumers.

Preferably, the analyte biomarkers are a panel of genes as defined herein. In one embodiment the biological sample is a tissue sample. This may be a tissue homogenate, tissue section and biopsy specimens taken from a live subject, or taken postmortem. The samples can be prepared, for example where appropriate diluted or concentrated, and stored in the usual manner. The analyte biomarkers provided by the invention, have the considerable advantage of accurately predicting the ageing effect of a test compound in a variety of tissues. This allows the method to be carried out on any tissue that is the most cost-effective and readily available.

In a further embodiment the tissue sample is obtained from the skin, hair, oral mucosa, brain, heart, liver, lungs, stomach, pancreas, kidney, bladder, skeletal muscle, cardiac muscle or smooth muscle. In a further embodiment, the tissue sample is obtained from skeletal muscle.

In one embodiment, the biological sample is a sample of cells. In a further embodiment the sample of cells is derived from a cancer cell line. Cancer cell lines can be grown reproducibly and stably in a test tube and therefore provides a suitable biological sample to measure the in vitro effect of a test compound on the healthy ageing signature.

In one example, the ageing signature may be measured in a sample of cancer cells obtained from a patient to provide information on the potential aggression of a tumour, or its ability to survive therapy. If the healthy ageing signature is reduced by a chosen therapeutic, then this is indicative of a pro-survival effect on the cancer cells within the target tumour.

In one embodiment the quantification of analyte biomarkers is performed using a biosensor.

A further aspect of the invention provides a method of treating an ageing-related disease in an individual, which comprises assessing the risk of said individual developing an ageing-related disease according to any of the methods defined herein and if the individual is identified as being at risk of developing an ageing-related disease, treating said individual to prevent or reduce the onset of an ageing-related disease.

A further aspect of the invention provides a compound obtainable by the method as defined herein.

Compounds that activate the ageing signature can be considered "pro healthy ageing" compounds and can be used as effective therapeutics. In particular, pro-ageing compounds can provide a novel anti-cancer therapeutic by enhancing surveillance for cancerous tumor cells. In another example, a pro-ageing compound may be used to activate the healthy ageing signature in skin cells to help accelerate wound healing. Compounds that inhibit the ageing signature can be considered "anti healthy ageing" compounds. Drugs which create this pattern of expression would be important to identify during the drug discovery and development process. In one example an identified anti healthy ageing compound may in the long term damage tissues, such as heart or muscle tissue, and the proposed screen would identify these unwanted and/or negative effect.

In one embodiment, the compound is a nutraceutical compound. References herein to "nutraceutical" refer to any substance that is a food or a part of a food that provides medical or health benefits, including the prevention and treatment of disease. Such products may range from isolated nutrients, dietary supplements and specific diets, to genetically engineered designer foods, herbal products, and processed foods such as cereal, soups and beverages.

According to a further aspect of the invention, there is provided a kit for assessing the ageing effect of a test compound comprising a biosensor capable of quantifying the analyte biomarkers as defined herein. In one embodiment, the kit comprises reagents from the Affymetrix Gene-Chip technology platform.

Suitably a kit according to the invention may contain one or more components selected from the group: a ligand specific for the analyte biomarker or a structural/shape mimic of the analyte biomarker, one or more controls, one or more reagents and one or more consumables. Optionally the kit may be provided with instructions for use of the kit in accordance with any of the methods defined herein.

The present invention will now be illustrated by the following studies, and with reference to the accompanying figures, in which:

Figure 1 shows a schematic overview of the use of RNA probe-sets for the development, validation and optimization of the healthy physiological age diagnostics.

Figure 2 provides plots of a cumulative gene score calculated using the 150 genes of Table 2 in ULSAM samples (chronological age = 69-70y) against conventional clinical risk factors Figure 3A shows a plot of a cumulative gene score calculated using the 670 genes of

Table 1 in ULSAM samples (chronological age = 69-70y) against renal function at 82 years. Figure 3B shows a multivariate model for prospective renal function at 82 years in the ULSAM cohort.

Figure 4A shows a Kaplan Meier-plot, with the underlying cox-regression on quarti!es of a cumulative gene-score calculated using the 30 genes of Table 4 in ULSAM samples (chronological age = 69-70y), with the 3 rd and 4 fh quartiles differing from the 1 st quartile (p< 0.04).

Figure 4B shows a logistic regression analysis of a cumulative gene-score

(continuous variable), calculated using the 30 genes of Table 4 in ULSAM samples

(chronological age = 69-70y), versus mortality.

Figure 5:GO p-value distributions. A plot of the distribution of raw p-values from 10,000 hypergeometric tests using randomly sampled probes (n=670) each time (see solid line) and the distribution of the raw p-values from a hypergeometric test using the 670 probes (classifier probes) associated with the genes of Table 1 (see dotted line).

Figure 6: A plot showing median gene score in blood (calculated using the 150 genes of Table 2) for patients with AD or MCI vs control samples.

Figure 7: A graph showing the mean gene score (calculated using the 150 genes of Table 2) for healthy human brain samples from 10 different brain regions with age range across young, middle-aged and old brains.

ABBREVIATIONS

fRMA Frozen Robust Multi-array Analysis

GA Genetic Algorithm

GFR Glomerular filtration Rate

GEO Gene Expression Omnibus

HOCV Hold Out Cross Validation

I PA Ingenuity Pathway Analysis

KNN k- Nearest Neighbour

LOOCV Leave One Out Cross Validation

PGE Positional gene enrichment analysis

RMA Robust Multi-array Analysis

ROC Receiver Operating Characteristic

SNPs Single Nucleotide Polymorphism ULSAM Uppsala Longitudinal Study of Adult Men

AD Alzhiemer's disease

MCI Mild Cognitive impairment METHODS

The following GEO codes represent the source of the raw data used in this project to build and validate the diagnostic/method. STOCKHOLM (GSE59880), DERBY (GSE47881 ), KRAUS (GSE47969), HOFFMAN (GSE38718), TRAPPE (GSE28422), BRAIN (GSE1 1882), CAMPBELL (GSE9419), 10 human brain regions (GSE60862), and human skin (lllumina Human HT-12 V3, Arrayexpress: E-TABM-1 140). The following GEO codes reflect the clinical validation data sets utilized; ULSAM (GSE48264), and for cognitive health

GSE63060 and GSE63061 . Informed consent was obtained from all volunteers and ethical approval received from Institutional Research Ethics Committee as reported in primary clinical publications, all studies were conducted under the auspices of the declaration of Helsinki.

For each microarray data set a unique identifier, often defined as a probe or probeset, represents an equivalent section of gene sequence. To go from the microarray technology identifier (the Gene ID in Tables 1 -5) to the probeset sequences, gene sequence and the gene name, the probeset identifier is entered into one of several readily available databases, e.g. biomart (http://www.biomart.org) or NetAffx

(https://www.affymetrix.com/analysis/index.affx). Alternatively the sequence information from the manufacturer, for each probeset, can be used in BLAST to identify what region of the genome the probeset is complementary too and this also yields identification of the gene name or gene sequence.

Development, validation and optimization of the healthy/physiological age diagnostics

Figure 1 provides a schematic overview of the process by which genes detailed in Tables 1 -5 were identified. 670 unique probe-sets were identified from a possible starting number of -54,000 during step one and these had a variation in classification performance as illustrated. This prototype diagnostic was then developed, evaluating the performance of the entire list, the top-ranked n=150 probe-sets or following an optimization process where a set of n=30 probe-sets were obtained that had improved diagnostic performance when examining a clinical outcome, as illustrated at the end of the work-flow. The process of identification of the probe sets, and the validation of the diagnostic potential of the identified probe sets, is described in more detail below. The healthy-ageing prototype diagnostic was built using 15 young (<28 year) and 15 older subjects free from metabolic diseases and signs of cardiovascular disease (>59 year): the 'Stockholm' data set. Subjects had blood samples taken for glucose measurement and had a fitness test to measure their V02max. This data allowed us to ensure that the young and older subjects were matched for aerobic fitness, as this parameter has been found to be the most powerful predictor of all cause mortality in humans (Wei et al (1999) Jama 282: 1547-1553; Lee et al (201 1 ), supra). RNA was processed and analysed by Affymetrix gene- chip and the probe-set level intensities of these arrays were normalized using the Robust Multi-array Analysis method (RMA) implemented within the R statistical software

environment using the 'affy' package (Bioconductor project) (Gentleman et al (2004)

Genome Biol 5: R80). When samples are prepared in independent laboratories batch effects are introduced (RNA processing and gene-chip processes, technical variation). To limit these batch effects, the data sets were pre-processed using Frozen Robust Multi-array Analysis (fRMA), adjusting using a robust empirical Bayes framework (Leek et a/ (2010) Nat Rev Genet 1 1 : 733-739; Leek and Storey JD (2007) PLoS Genet 3: 1724-1735).

The candidate probe-set lists were created via a nested-loop, holding out two arrays at any one time to estimate two parameters from the data. The first of these was the conventional test set result i.e. is the array correctly classified Yes/No. The second novel parameter was used to calculate a rank order for the useful probe-sets. Two-hundred probe- sets were selected during each of the inner-most computational loops by ranking gene expression differences using an empirical Bayesian statistic (implemented as eBayes in the limma' package) (Smyth (2004) Stat Appl Genet Mol Biol 3: Article 3). All the probe-sets (-800) involved in the most successful inner-loop iteration were then used as the starting point for the prototype classifier. Probe-sets that targeted multiple genomic loci were then removed from the list and then probe-sets that were involved with a correct identification call 70% of the time or more were carried forward into the rest of the validation process. The model built using the Stockholm data yielded a n=670 probe-set and this is referred to as the prototype healthy-age diagnostic and the specific gene lists are provided in Table 1 . An n=150 set was also identified which included probe-sets that were involved in a correct identification call 90% of the time. This set is referred to as the top 150 healthy-age diagnostic and the specific gene lists are provided in Table 2.

Each of the 670 genes was down -regulated in the healthy older subjects compared with the young subjects except for the following genes (which were up-regulated): MED13L, TSPYL1 , RBL2, BCKDHB, CUL4A, CAPN1 , C6orf62, GNG10, HMGB1 , TSC22D1 , RAD21 , SFRS1 1 , 236978_at, PTP4A2, HNRNPA1 , TWF1 , PAM, TIA1 , JMJD1 C, DENND5B, H2AFV, 233674_at, SCP2, INTS6, OGFOD3, PRKAA1 , MPDZ, CXorf15, LRRFIP1 , TTC17, GPATCH8, BRD2, ASPH, CEP192, 242425_at, RPS6KA5, TTBK2, LATS1 , PDE7A, ANK3, 229434_at, SLC1 1 A2, SUZ12, NEAT1 , ACSL1 , MCL1 , NBEA, KANSL1 L, TTC3, KRR1 , ETNK1 , LGI1 , PCBP2, 237018_at, FAM76B, FXR1 , PRNP, ARMCX3, MBNL1 , DERL1 , APP, NUCKS1 , CFLAR, 239251_at, MY0Z2, SAV1 , CEP350, CLIP1 , SYNP02, 242467_at, FUS, WSB1 , RBMS3, PPFIBP1 , ZNF638, CD47, IFRD1 , SFRS18, DHX29, GPAM, PCDH9, 228105_at, 213156_at, B3GNT5, 242457_at, MTMR9, KRIT1 , FEZ2, LGR5, NPHP3, MGC24103, PNISR, 229483_at, SKAP2, RUFY3, RP1 1 -271 C24.3, 41929_at, MAN2A1 , ALDH6A1 , LIFR, PFKFB2, ESRRG, TGFBR3, ASH1 L, 233073_at, SCAMP1 , SRD5A2L2, SKAP2, UNCI 3C, UNCI 3C, SPEN, , DUFS1 , 236439_at, SMCHD1 , MALAT1 , CD36, MALAT1 .

Having identified a prototype set of probe-sets (n=670), classification of independent samples was performed using a k-Nearest Neighbour (KNN, n=3) classifier, implemented in the R 'class' package. Leave-One Out Cross Validation (LOOCV) is a specific type of Hold Out Cross Validation (HOCV) which is widely used as a standard procedure to test how well a predictive model is generalized. To implement independent blind validation, we used both independent training and test muscle and brain data sets. That is, we relied on robust external validation methods and not just internal cross validation methods.

To carry out external validation you need two new data sets. In our case the prototype healthy-age diagnostic probe-set list were plotted in multidimensional space, using the Campbell cohort expression values, and this represented the 'expression space' of known old and young samples for the subsequent KNN evaluation of subsequent further independent samples e.g. muscle and brain. For the MuTHER cohort skin data-set, which was produced using the lllumina Human HT-12 V3 Bead chip, log-2 transformed signals were normalised per replicate data set, using the quantile normalisation method. A LOOCV approach was used to predict age of all individuals using the 670 genes of Table 1 of the invention or 150 genes of Table 2 of the invention. Genes were mapped to the lllumina platform (551 from 670 genes were represented in this list). For this set of human skin samples, individuals aged < or = 45 years were pre-defined as young, and those > or = 70 years as old. This was to ensure sufficient numbers of young and old samples existed to fairly assess the classifier performance. Three technical replicates from this skin microarray biobank were analysed separately to establish how reproducible the diagnostic could be in repeated samples from the same clinical sample. Diagnostic performance was judged and optimised using Receiver Operating Characteristic (ROC) analysis (Sing et a/ (2005) Bioinformatics 21 : 3940-3941 ). Examples of how refinement of the prototype healthy-age diagnostic set could be achieved was carried out using a Genetic Algorithm (GA) search and an optimisation process was implemented whereby units of probe-sets (e.g. n=30) were randomly selected from the 670 prototype age probe-set list. Each of these of n=30 'gene' units can be conceptually thought of as chromosomes, and a successive number of 'off-spring' gene-sets (each of n=30) are created following a cross-over event (Srinivas and Patnaik (1994) Syst Man Cybern IEEE Trans 24: 656-667; Lin et a/ (2003) J Inf Sci Eng 903: 889-903), analogous to maternal/paternal DNA recombination. Each set of n=30 was also subjected to 'mutation' events, where a single probe-set is replaced from a pool of probe-sets from the 670 that were not included in the initial sets of n=30 groupings. The resulting n=30 gene-sets are evaluated on the basis of a fitness function/optimisation criterion which determines if the new population generated is better (e.g. improved ROC performance) than the 'parent' gene- sets. Thus, more adaptive chromosomes are kept and less adaptive ones, with lower fitness values, are discarded thereby generating a new population over time. The balance between the rate of the two events, cross-over and mutation, determines the nature of the

optimisation process. In contrast to the strategy of the present invention, application of the GA process to exhaustively examine the entire repertoire of probe-sets on the Affymetrix gene-chip (-54,000) would be extremely protracted and computationally impossible given the computing resources currently available on earth.

Production of new global RNA profiles for clinical validation

Total RNA for the new data sets was extracted from frozen muscle using TRIzol reagent as previously described (Timmons et al (2005) Faseb J 19: 750-760). In vitro transcription (IVT) was performed using the Bioarray high yield RNA transcript labelling kit (P/N 900182, Affymetrix, Inc.). Unincorporated nucleotides from the IVT reaction were removed using the RNeasy column (QIAGEN Inc, USA). Hybridization, washing, staining and scanning of the arrays were performed according to the manufacturer's instructions (Affymetrix, Inc). As a means to control the quality of the individual arrays, all arrays were examined using hierarchical clustering and Normalized Unsealed Standard Error (NUSE, a variance based metric to identify outliers prior to statistical analysis), in addition to the standard quality assessments including scaling factors and chip-housekeeper 573 ' ratios. The data deposited in GEO that did not originate from our laboratory was also quality assessed. In each case a small number of gene-chips (2-3) were identified that had clear evidence of RNA degradation or other technical defects with the gene-chip profile and these were removed from the analysis. ULSAM (Uppsala Longitudinal Study of Adult Men)

This is a cohort of men born in 1920-24 and living in Uppsala, Sweden, who were invited to attend a health examination at the age of 50 years (n= 2322) (Dunder et a/ (2004) Am Heart J 148: 596-601 ). Re-examinations were performed at 60, 70, 77, 82 and 88 years of age. Over the years the cohort has been very well characterized from metabolic and lifestyle perspectives. Of specific importance is that the ULSAM subjects were investigated by DEXA scans at both 82 and 88 years of age. Dual-energy X-ray absorptiometry (DEXA) scan measurements were performed during the last decade of the study at these points and yields a measure of loss of lean body mass. Muscle mass status varied between -15% to +10%. from 70 to 88 years old and was unrelated to physical activity scores. Follow-up of these subjects, which included recording their physical activity and exercise status, has been executed at 82 and 88 years of age. Within the subjects are a range of physical activity levels from completely sedentary (-15%) to recreational-athletic (-10%). Renal function at age 82 was calculated using cystatin C, which is a marker of GFR (Inker et a/ (2012), supra). 129 skeletal muscle biopsies were taken from cohort members at 70 years of age in which DEXA and functional testing was performed at 82 and 88 years of age. Skeletal muscle biopsy tissue, taken in 1992, was processed for RNA, extracted with TRizoi, in 2012. A total of 108 samples provided good RNA and 50ng total RNA was amplified using Ambion's WT expression kit to produce cDNA. The cDNA was fragmented and labeled with GeneChip WT Terminal labeling kit (Affymetrix Inc.). The hybridization of cDNA to exon array was 16h at 45 degrees. The arrays were washed in Affymetrix FS450 wash stations and scanned on an Affymetrix 3000 7G scanner according to the manufacturer's instructions. The array data was processed as detailed above.

A gene ranking-based diagnostic methodology was developed and applied to the samples from the ULSAM longitudinal study. The ranking calculation was carried out as follows: for a gene down-regulated with age (in the prototype classifier) subjects were ranked from highest to lowest expression, with the subject with the highest expression assigned 1 . For age up-regulated genes the opposite strategy was used. Each subject was then assigned a gene score which was the median of the individual ranking scores for each gene. Regression analysis was used to study the relationship between 70 year age-related gene score and renal function (as renal function is a marker of future mortality in older subjects). In addition to using the gene-score, clinical features of the subjects at 70 years of age were entered into a multivariate model. Model selection was executed using a forwards selection approach, with p>0.1 as stop criterion (backwards selection yielded the same outcome). Variables, previously reported (Dunder et al (2004), supra), were added to the baseline model one at a time, and selected based on p-value (Hagstrom et a/ (2010) Eur J Heart Fail 12: 1 186-1 192). For baseline characteristics, and results on univariate analysis see Table 6: Table 6

Variable Number of obs. Mean@70y SD P-value

Cystatin C calculated GFR (ml/min) 123 il A ? 0.48 0.110 n .0006

BMI (kg/m2) 128 25.8 2.8 -1 .43 0.052 0.0172 s-Albumin (g/l) 126 59 9 32.1 -0.12 0.045 0.0221

Weight (kg) 128 78.9 9.9 -0.37 0.042 0.0338

OGTT p-gluc 60 min (mmol/l) 128 9 6 2.6 -1 .14 0.028 0.0834 s-Phosphate (mmol/l) 127 43.0 2.3 1 .26 0.025 0.1036

OGTT p-insulin AUC 128 1 4 0.8 -3.38 0.023 0.1 195

OGTT p-gluc 120 min (mmol/l) 128 7.2 2.7 -0.78 0.015 0.2164

Free fatty acids (mmol/l) 128 4 0 1 ,0 2.14 0.014 0.2270

OGTT p-gluc 30 min (mmol/l) 128 9.1 1 .6 -1 .26 0.013 0.2400 lnterleukin-6 (ng/l) 122 3 9 4.9 0.40 0.014 0.2432

HDL cholesterol (mmol/l) 125 0.5 0.2 -8.25 0.015 0.2558 s-Cholesterol (mmol/l) 128 0.3 6.07 0.012 0.2577

Systolic blood pressure supine (mmHg) 145 19 -0.10 0.010 0.2969

Leisure time physical activity 111: 3 * 2.99 0.010 0.3221

-Albumin excretion rate (pg/m 122 1 1 .8 37.1 -0.05 0.009 0.3393 s-Triglycerides (mmol/l) !ii 6.0 1 1 1 .43 0 008 i© 3648 s-lnsulin (pmol/l) 124 45.3 20.7 -0.08 0.008 0.3673

OGTT p-gluc 0 min (mmol/l) 111: 5.5 1 .0 1 .20 0.004 0.5099

Diastolic blood pressure supine (mmHg) 128 84 -0.13 0.004 0.5143

Puis rate (beats/min) 128 65 -0 1 M 0.004 0.51 49

Mini Mental State examination 121 28 * 0.07 0.002 0.6276 s-Creatinine (mol/l) 127 340 64 0 01 0.002 0.6474 s-Uric acid (mol/l) 125 1 .0 0.3 2.04 0.001 0.7157

C-reactive protein (mg/l) 124 2.6 2.7 0.16 0.001 0.7972

LDL cholesterol (mmol/l) 126 80.2 30.8 0.01 0.0005 0.8272

Univariate linear regression on baseline characteristics at 70 years of age versus Cystatin C estimated glomerular filtration rate at 82 years of age. Number of obs denotes the number of complete observations available for each variable. Mean and SD denote mean and standard deviation respectively, variables marked with * are categorical and hence reported using median. R denotes the regression-coefficien of the variable. R2 and P-value denote r-squared and p-value of the univariate analysis.

One of the additional candidate variables, BMI, qualified to the final model in those criteria. The final model had the following format: eGFR@82(ml/min)=18.6+0.65

*GeneScore+0.41 * eGFR70(ml(min)-1 .00 * BMI (kg/m 2 )). For the mortality analysis, both the cox-regression and the logistic regression model were implemented in R. For the cox-model the latest 'survival package' was used whereas the logistic regression model was estimated using the glm (generalized linear model) function and 'logit' model which models the log odds of the outcome as a linear combination of the predictor variables. Over the observation period, 19 mortality events occurred and the relationship with gene-score was analysed with gene-score as a continuous variable. The exponential regression coefficient for optimised gene-score was 0.93 with a p-value of 0.0002. For the Kaplan-Meier plots, gene-score was divided into quartiles and the plot was produced using the 'plot-survfit' function in the survival package. The plot allows overall survival rates to be compared between the four quartiles for gene-score (Figure 4A). The graph from the logistic regression analysis shows the inverse relationship between the probability of death and gene-score with 95% confidence intervals (Figure 4B). Both the KM plot and logistic regression plot demonstrate that a better gene- score at the baseline improves the chances of survival and vice-versa.

A prototype multi-gene molecular classifier that could distinguish between healthy young and healthy old tissue samples was produced and validated in -600 independent tissue samples. Muscle samples were utilised as a starting point as a large number of independent cohorts were possessed with detailed phenotyping of the donor (Keller et a/ (201 1 ), supra; Gallagher et al (2010) Genome Med 2: 9). Theoretically, the genes identified should associate with, or reflect, healthy physiological age rather than disease as older subjects were specifically selected that had good aerobic fitness and glucose tolerance (Timmons et a/ (2010), supra; Gallagher et al (2010), supra). The healthy-age prototype diagnostic was built as previously described, using the following method, with 15 young (-25 years chronological age) and 15 older subjects (-65 years chronological age) and this is referred to as the 'Stockholm' data.

An ensemble of genes were selected using a Leave-One Out Cross Validation (LOOCV) process where the top 200 probe-sets (RNA detection probes equating to 1 gene) were carried forward during each loop, and each of these probe-sets used to 'judge' the age of a second held-out sample, by implementing a k-Nearest Neighbour (KNN, n=3) classifier. Following iterative assessment of all probe-sets on the gene-chip, involving -180,000 permutations during which each one of the 30 samples was held-out of the ranking procedure, a repertoire of the best performing -800 probe-sets was selected (based on the total number of correct judgements during the 180,000 iterations). The 800 probe-sets were manually inspected and those probe-sets that targeted multiple genomic loci were removed from the classification list, and then probe-sets that were involved with a correct identification call 70% of the time or more were carried forward into the rest of the validation process (Figure 1 ). The model built using the Stockholm data yielded n=670 probe-sets and this is referred to as the prototype healthy-age diagnostic and the specific gene lists are provided in Table 1 . An n=150 set was also identified which included probe-sets that were involved in a correct identification call 90% of the time. This set is referred to as the top 150 healthy-age diagnostic and the specific gene lists are provided in Table 2. The 'Stockholm' data set was discarded from the project at this stage, and a fully independent validation process was carried out, as detailed below.

Prior to undertaking an optimisation process (see below) the 'raw' performance of the prototype diagnostic was evaluated, and established if the age of samples obtained could be determined using five independent human muscle cohorts. This was done because an independently validated highly accurate diagnostic of muscle age represents a novel observation in its own right. All the following muscle tissue cohorts were profiled on the same gene-chip platform (Affymetrix U133+2 chip). A new cohort, hereafter named 'Campbell', (n=66 chips (Thalacker-Mercer et a/ (2010) J Nutr Biochem 21 : 1076-1082) was used as the new training data-set, used to evaluate the 'unknown' independent young and old samples from four additional independent clinical cohorts. This included three existing data-sets from GEO ('Trappe' (Raue et al (2012) J Appl Physiol 1 12: 1625-1636) (n=48), 'Hoffman' (Liu et al (2013) J Gerontol A Biol Sci Med Sci: 1-10) (n=22) and 'Derby' (Phillips et a/ (2013), supra) (n=26)) and a fourth gene-chip dataset ('Kraus', n=33) which was produced from proprietary clinical samples (Slentz et a/ (201 1 ) Am J Physiol Endocrinol Metab. 301 :

E1033-9). Remarkably, each clinical sample, from all of these 4 independent clinical cohorts was classified into the correct group, with a success rate of -83% (Range 70-93%) for the 670 gene set and -93% (Range 70-100%) for the 150 gene set. The 13 gene set (EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2) yielded success rates of 81 % (Derby) and 73% (Trappe). This reproducible result contrasts markedly with methods which study muscle ageing using group mean differential expression analysis (see Phillips et al (2013)). A key feature of the prototype healthy-age diagnostic was that when applied to a group of 'middle-aged' subjects with similar chronological age, a highly variable gene-expression score was observed

demonstrating that the diagnostic score was distinct from chronological age. To evaluate if the prototype healthy-age diagnostic reflected age-related changes in other human tissues it was examined if the prototype sets of genes could accurately identify the age of non-muscle human tissues. While it is much less possible to define the 'health status' of the non-muscle sources it was felt that the genes, which defined healthy older muscle tissue, should also be modulated to some degree in older versus younger samples, in other tissue types - at least sufficient numbers to provide an accurate 'fix' on age - if this was a novel and universal 'ageing' signature. Thus, tissue profiles from both ectodermal (brain) and mesodermal (skin) origin were utilised for this purpose. Global RNA profiles from 120 old and young human brain samples (Berchtold et a/ (2008) Proc Natl Acad Sci U S A 105: 15605-15610) were evaluated using the prototype healthy-age diagnostic. The samples represented four brain regions (Entorhinal Cortex (n=25), Hippocampus (n=31 ), Superior Frontal Gyrus (n=33) and Postcentral Gyrus (n=31 )) all of which were certified to be disease-free by histopathology in the original study. The classification success for these human brain samples, using the 670 gene prototype healthy-age diagnostic and muscle gene-chip expression data from a different laboratory as the external independent training set, was an impressive -76%. When a brain-tissue expression data-set was used to predefine the classification space, this success rate improved to -84% (see Table 7). Thus, without any refinement, the 670-gene prototype healthy-age diagnostic was also able to distinguish between pathology-free old and young brain samples from independent clinical sources, profiles produced under entirely independent laboratory conditions.

Table 7 - Accuracy, sensitivity and specificity of the muscle-derived healthy age classifier when applied to multiple independent data sets. The sensitivity and specificity of the 670 probe-set derived from the STOCKHOLM gene-chip data was determined for multiple human muscle data sets (Campbell, Derby, Hoffman, Trappe and Kraus) and four brain regions derived from the Berchtold et a/ (2008) study, supra, with brain set as the training data, and skin from the MuTHER cohort (Glass et a/ (2013), supra). The majority of data sets demonstrated both high sensitivity and high specificity using the prototype 670 probe-set of Table 1 (shown below in Table 7) or the top-150 prototype list of Table 2. A young sample misclassified as Old' (e.g. in 'Hoffman') is noted as a reduced sensitivity. If an old sample was misclassified as being young, as was the case for some of the Hippocampus region, then this is defined as a reduction in specificity, where young is a true-positive in the model. The contributing factors to these misclassifications include lack of standardisation of a single laboratory gene-chip protocol, variation in RNA quality and in some cases older donors that have not induced the 'healthy ageing' signature to any extent. The Genetic Algorithm (GA)

D C M C

search and optimisation process was DD C C run for 5,000 to 1 million iterations and yielded improved performance, sensitivity and/or specificity in all data sets from only the 670 probe-set as input.

Prototype 670 probe-set performance GA Optimized

Sample Size Accuracy % Sensitivity Specificity Accuracy % Sensitivity Specificity

Muscle (Campbell) 82 0.83 0.80

Muscle (Derby) 93 1 .00 0.88 - - -

Muscle (Trappe) 48 96 0.92 1 .00 111111111111111

Muscle (Hoffman) 22 73 0.79 0.63 >96 >0.93 >0.88

Muscle (Kraus) 33 70 1 .00 0.60 94 >0.88 >0.92

Brain (SFG) 33 88 0.86 0.89 - - -

Brain (PCG) 31 88 0.43 1 .00 >97 >0.86 1.00

Brain (Hippocampus) 31 81 0.33 1 .00 97 >0.83 >0.96

Brain (EC) 25 76 0.43 0.89 >88 >0.71 >0.94

Skin (MuTHER Cohort) 279 79 0.61 0.90 83-88 >0.84 >0.80

The prototype healthy-age diagnostic was then used to evaluate the age of human skin samples ((Sawhney et a/ (2012), supra) and this gene expression data-set originated from a different technology platform: the lllumina Human HT-12 V3 Bead chip. The 670 Affymetrix probe-sets were mapped to gene names, and then to 551 probes on the lllumina chip. There were 279 skin samples for classification analysis, and many of these samples also had two additional technical replicates (n=131 replicate 1 ; n= 124 replicate 2; n=24 replicate 3). The prototype healthy-age classifier gene-list demonstrated good classification success in sets of human skin profiles (79%, see Table 7), confirming that the muscle- derived gene-expression signature appears to be a universal diagnostic of human tissue age and able to operate across technology platforms. This was achieved because of the robust and novel feature selection 2-step process we implemented to build the prototype healthy- age diagnostic and the fact that we uniquely used disease-free older tissue samples.

Assessment of diagnostic performance was achieved using Receiver Operating Characteristic (ROC) analysis ((Sing et a/ (2005), supra) where both sensitivity and specificity are considered rather than just raw success rates. In fact, the prototype healthy- age signature had excellent sensitivity to specificity ratios in many human clinical cohorts, despite the technical variation and post-mortem processing e.g. brain tissue. However, as access to multiple independent data-sets was possible and promising classification performance was demonstrated, an optimisation process was undertaken to improve ROC performance.

Optimisation of age classifier performance

Optimisation was undertaken by selecting sub-sets of genes using only the original 670 probe-sets to yield optimal ROC performance for data-sets where sensitivity or specificity could be shown to be further improved (see Table 7). Refinement of the prototype was carried out using a Genetic Algorithm (GA) search and optimisation process was implemented whereby units of probe-sets (e.g. n=30) were randomly selected from the 670 prototype age probe-set list. Each of these of n=30 'gene' units can be conceptually thought of as chromosomes, and a successive number of 'off-spring' gene-sets (each of n=30) are created following a cross-over event (Srinivas and Patnaik (1994), supra; Lin et a/ (2003), supra), analogous to maternal/paternal DNA recombination. Each set of n=30 was also subjected to 'mutation' events, where a single probe-set is replaced from a pool of probe- sets from the 670 that were not included in the initial sets of n=30 groupings. The GA process was set to run through a number of recombination events lasting up to 1 million iterations and classifier performance was guided to yield greater specificity or sensitivity depending on which parameter was being improved. This self-adapting process allows the search of the 670 probe-set data to optimise diagnostic performance.

Applying the GA process first to muscle, the 'Campbell' data was used as the independent training data-set, and the sensitivity and specificity for n=30 gene-sets to demonstrate improved classification performance of the 'Hoffman' and 'Kraus' cohorts was determined. For these two cohorts, several n=30 gene-sets were noted which exceeded the prototype performance, where each n=30 probe-set list is largely distinct from each other. For Hoffman, classification success was now 96-100% with near perfect specificity and sensitivity, while a similar result was achieved for the Kraus data set (see Table 7). Similar improvements in performance could be obtained in both brain and skin, such that a number of n=30 gene-sets could be identified using only the original age-classifier prototype gene list that contained sufficient information to determine human tissue age with near perfect success (see Table 7). No single gene was common to all subsets and this is likely to be a key feature of the diagnostic of the invention, as one that successfully operates across numerous diverse tissues and clinical sources should not be driven by a single or small number of biological features.

Applying the age classifier to determine long-term health in the ULSAM cohort

The primary hypothesis of the invention was that a validated diagnostic of healthy physiological age could be used to predict health outcomes in a longitudinal study, where subjects were all the same chronological (calendar) age at the point of assessment. When a median rank score was calculated (see below) for twenty middle-aged subjects (Phillips et al (2013), supra), the prototype age-diagnostic gene expression score demonstrated -10 times more variation than the chronological age-range, however this in itself does not establish if the information contained within the age signature (the 'additional' variance) would be useful for predicting health outcomes. To assess if the prototype healthy-age diagnostic was indeed prognostic, in a longitudinal study, RNA profiles were produced from healthy tissue samples taken and frozen two decades ago from members of the ULSAM cohort (Dunder et al (2004), supra). Each subject was profiled on the Affymetrix EXON 1 .0 gene-chip platform and the 670 probe-sets were mapped to the equivalent new probe-sets (yielding 575 probe- sets) so testing the diagnostics ability to work on yet another technology type. The pattern of changes in gene expression between young and healthy old subjects in the prototype age diagnostic was ~2/3 rd down regulated and ~1/3 rd up regulated. Thus, a gene-ranking based diagnostic was calculated taking the direction of gene expression change into account, as described above. The gene-score was, as hoped, unrelated to physical activity levels, the closest surrogate identified herein for physical fitness in the ULSAM cohort so further demonstrating the unique nature of the age diagnostic from conventional clinical tests.

Prior to full optimization (see below) a typical approach to evaluating classification success (Knudsen S (2004) Guide to analysis of DNA microarray data. 2nd ed. Hoboken, N.J.: Wiley-Liss) was taken and used the top 150 healthy-age classifier genes from the prototype list (see Table 2). We generated a cumulative gene-score from the median rank order for all 150 genes for each ULSAM subject. Clinical variables were determined as previously reported (Huang et a/ (2014) J Intern Med 275(1 ), 71 -83; Zethelius et a/ (2008) N Engl J Med 358: 2107-21 16). Linear regression was used to examine the relationship between the cumulative gene-score of a sample and the respective clinical parameter. As can be observed from plots A-C of Figure 2 there was no relationship between rank-order for cumulative gene-score and baseline renal function (cystatin-c), blood pressure or total cholesterol (score was unrelated to resting heart rate or physical activity questionnaire scores either). Thus the cumulative gene-score could not be substituted by any of these conventional risk factors (or others listed in Table 6) to predict health-outcomes over the following 20y. Note that at the point of assessment (1992), when the muscle biopsy was taken for subsequent gene-chip profiling, all subjects would be considered in good health for their age and remained physically active.

At 70 years, three subjects had Cystatin C > 1 .5 mg/l, while by 82 years 36 of the subjects studied in the present analysis had Cystatin C > 1 .5 mg/L. A 1 .5 mg/L Cystatin C corresponds to an estimated GFR of -45 mL/min which is borderline for a moderately (30-45 mL/min) elevated risk for all-cause mortality (Zethelius et a/ (2008), supra). Renal function using Cystatin C was estimated to calculate eGFR, and demonstrated that the baseline healthy-age diagnostic ranking score was related to renal function 12 years later (age 82, p=0.009). An optimized healthy age diagnostic was generated using the GA search and optimisation process (60,000 iterations) yielding an optimised n=30 gene diagnostic

(r 2 =0.203, p<0.000001 , Regression Coefficient = 0.4504, Figure 3A and Table 3) for gene- score versus renal function at 82 years. As before, those subjects that 'switched on' the healthy-ageing gene expression pattern had superior renal function at age 82 years.

The potential for the healthy-age diagnostic to be combined with clinical variables to provide enhanced prognosis of impaired renal function was investigated using multivariate modeling. In addition to the optimized gene-score, clinical features of the subjects at 70 years of age were considered in the multivariate model. Model selection was executed using a forwards selection approach, with p > 0.1 as stop criterion. Variables, previously reported (Dunder et a/ (2004), supra), were added to the baseline model of gene-score and cystatin C estimated renal function at 70 years of age. A final model utilizing gene-score, eGFR (Estimated Glomerular Filtration Rate) and BMI at a chronological age of 70 years, yielded a model with r 2 =0.329 (p<0.00001 , Figure 3B). Thus, the gene-score derived from an RNA profile of healthy skeletal muscle (and validated across multiple tissues) was able to combine with two simple clinical measures to capture 33% of the total variance of renal function at 82 years.

The cumulative gene-score was calculated from 670 genes of Table 1 for the USLAM subjects at 70 years of age. While renal function is not sufficiently powerful to predict mortality in disease-free older subjects from the ULSAM cohort (Zethelius et a/ (2008), supra), it was found that the top 150 healthy age diagnostic was able to predict 20 year survival (p=0.025) in a cox-regression model, with gene-score as a continuous variable.

For those subjects who died during a 20 year follow-up observation period the score was significantly lower than those subjects who remained alive (Wilcoxon test p=0.02). Furthermore, following optimizing of the protoype healthy age diagnostic (GA optimization leading to the 30 genes of Table 4) the baseline gene-score could distinguish between those that had died or not with greater significance (Wilcoxon test p=0.00072).

The GA optimized subset of 30 probes (Table 4) from the prototype (n=670) yielded a strong diagnostic of mortality as demonstrated by logistic regression analysis of gene- score (continuous variable) versus mortality, where the four-fold range in gene-score related to up to a 70% probability of death during the 20 year follow-up period (p=0.00085, Figure 4B). Further, when dividing this GA optimized gene-score into quartiles, there was a significant difference in survival between the first versus the third and fourth quartiles (p=0.049 and p=0.024) in this cox-regression model (Figure 4A). Thus, those subjects who died during the observation period started the period with the least induction of the 'healthy ageing' expression pattern at chronological age 70 years. The prediction of mortality in the ULSAM 20 year follow-up study is of course preliminary, but it provides further support that induction of the age signature, by the 6 th decade of life, represents a positive event since the directional shift in gene-expression and better 'health' was consistent for the renal and mortality analysis. A biological analysis of the healthy physiological age diagnostic

The RNA signature was evaluated for pathway and gene ontology analysis using both Ingenuity pathway analysis and R-based ontology analysis. There were no significant pathways noted in the Ingenuity analysis, either when using the entire n=670 gene list or when using the sub-set optimised gene lists. While it has previously been demonstrated (Gallagher et a/ (2010), supra) that applying gene ontology analysis to transcriptome data is problematic due to imprecise knowledge of the true background transcriptome (both tissue specific biases and technology biases mean that certain ontologies can be artificially enriched) it is unusual that a large gene list (n=670 gene), linked to a strong physiological phenotype, is not enriched for specific biological processes. This does however prove that our diagnostic list could not be selected from the literature using prior knowledge. To confirm this observation, 10,000 random 670 gene-set samples were measured from the entire population of genes measured in the present experiment, and the gene ontology p-value distribution of the random samples was compared with the 670 gene prototype healthy-ageing diagnostic. In Figure 5 the distribution of raw p-values from 10,000 hypergeometric tests using randomly sampled probes are plotted in black sold lines, while distribution of the raw p-values from a hypergeometric test using the prototype healthy- ageing diagnostic genes are plotted in a dotted line. The analyses clearly demonstrate that the ontological profile of the prototype healthy-ageing diagnostic is not different from a random sample of the starting 54,000 probe-sets, while >98% of the 54,000 probe-sets have no ability to discriminate tissue age.

The inclusion of some previously identified ageing related genes was noted; LMNA (linked with Hutchinson-Gilford Progeria Syndrome), Unc-13 homolog (UNC13C) which is linked with beta-amyloid biology and COL1 A1 (thought to change in skin-ageing). It was also examined whether the age-related genes were over represented at genomic loci using Positional enrichment analysis (De Preter et al (2008), supra). The genes from the prototype classifier (the 670 genes claimed herein) found to be over-represented at 7q22 and 1 1 q13. The results were consistent in positional gene enrichment analysis and ToppGene algorithm, both identified 3, 12 and 3 genes at each loci with p<0.001 or less. 1 1 q13 and 1 1 q23 in particular were most significant, and contained genetic variants proven to influence the age of onset of human age-related disease e.g. cancer.

There were in fact a number of significant findings. In particular, 1 1 q13 made a significantly greater contribution (adjusted p-value=0.005-0.007) to the prototype classifier than would be expected by proportionality, while there were a total of 15 genes from the 1 1 q13 and 1 1 q23 over-represented genomic locations (1 1 q13 (ALDH3B1 , CAPN1 ,

CDC42EP2, COR01 B,LTBP3, NRXN2, PPP1 R14B, RCE1 , RCOR2, SART1 , SYT12 and ZDHHC24, P=0.0005) and 1 1 q23 (FXYD2, SCN2B and TMPRSS13, P=0.0009)). Interestingly, 1 1 q23 is the location for age-related genetic interactions, namely the apolipoprotein A family (Garasto et a/ (2003) Ann Hum Genet 67: 54-62; Feitosa et a/ (2014) Front Genet 5: 159) as well as a region containing genetic association single nucleotide polymorphisms (SNP) which substantially modify for the age of onset of colorectal cancer (Talseth-Palmer et al (2013) Int J Cancer 132: 1556-1564; Lubbe et al (2012) Am J

Epidemiol 175: 1-10). Further, 1 1 q13 harbours SNP's associated with age of onset of renal cell carcinoma and prostate cancer and modulating age-related disease emergence by ~5yrs (Audenet et a/ (2014) J Urol 191 : 487-492; Lange et a/ (2012) Prostate 72: 147-156; Jin et al (2012) Hum Genet 131 : 1095-1 103).

Healthy aging signature and cognitive health

A study was carried out of the activation status of the healthy aging signature in blood samples from two large case-control studies of Alzheimer's disease (AD) (publication embargoed GEO data GSE63060 and GSE63061 ) and it was found that AD patients, and those with early signs of dementia, had a lower median healthy age gene score. The AD cohort has been previously used to study disease pathway changes (Hodges, J. Alzheimers. Dis. 33, 737-53 (2013), Hodges, 30, 685-710 (2012)). 1 13 subjects aged 75 years or younger in cohort 1 and 1 12 subjects aged 75 years or younger in cohort 2 were utilised. Using the very oldest subjects in each trial, retrospectively, did not change the outcome of our analysis. Each case-control data-set was ranked for gene-score using only genes selected from the prototype healthy age diagnostic (670 genes, Table 1 ) and selected from the top 150 healthy age diagnostic (150 genes, Table 2). There is no more than random chance levels of overlap between the healthy aging gene markers, and previously published genomic and genetic disease markers of AD. AD is a multi-factorial disease (8) with around 22 genetic loci associated with disease risk but no DNA marker is useful in the clinic, as a modifier of risk. Removal of the 7 genes (SKAP2, CEP192, RBM17, NPEPL 1, PDLIM7, APP and BIN1) common to the 'healthy aging gene 670 list' and previously published genomic markers of AD ((Hodges, J. Alzheimers. Dis. 33, 737-53 (2013), Hodges, 30, 685-710 (2012), Fillit, Alzheimers. Dement. 10, 109-14 (2014); Barmada, Transl. Psychiatry 2, e1 17 (2012); Amouyel Nat. Genet. 45, 1452-8

(2013); Vellas , J. Alzheimers. Dis. 32, 169-81 (2012); Federoff, Nat. Med. 20, 415-8 (2014) did not alter our results.

Blood RNA from the AD case-control cohort 1 was profiled on lllumina HT-12 V3 bead-chips and lllumina HT-12 V4 for cohort 2. Control subjects were matched in a manner which retained the same chronological age and gender as the AD or MCI subjects. Venous blood for the RNA analysis was collected from the subjects who had fasted 2 hours prior to collection using a PAXgene™ Blood RNA tube (Becton & Dickenson, Qiagene Inc.,

Valencia,CA). The tubes were frozen at - 20 ° C overnight prior to long-term storage at - 80 ° C. After thawing samples overnight at room temperature, RNA was extracted using PAXgene™ Blood RNA Kit (Qiagen), according to the manufacturer's instructions. The whole genome expression was analyzed using lllumina Human HT-12 v3 Expression BeadChips (lllumina) for the first case-control study and lllumina Human HT-12 v4

Expression BeadChips for the second, independent, case-control study used in our analysis. The expression data was first transformed using variance-stabilization and then quantile normalized using the LUMI package in R. The appropriate probes were mapped from Affymetrix based healthy ageing prototype to lllumina. We calculated a gene-ranking based score in the same manner as for ULSAM data set. Wilcoxon rank sum test from the R stats package was used to test if the median gene score ranks between the two groups, control and AD and control and MCI were significantly different or not.

In cohort 1 , the median rank score for AD patients versus chronologically matched controls was highly significantly different (p=0.00089) for 308 genes from the prototype 670 gene list. This confirms the directionality observed for both renal function and mortality in the ULSAM study. Blood RNA from the second AD case-control cohort blood was profiled and in this case 284 genes were common to the prototype 670 gene list. As before, the median rank healthy aging gene-score for AD patients in cohort 2 was significantly lower than the control group (p=0.0099). Furthermore, for both cohort 1 and cohort 2, the median rank healthy ageing gene-score for subjects diagnosed with mild cognitive impairment was lower than that of the chronological age-matched controls (p=0.00000034 and p=0.00055).

When applying the top 150 prototype the probes were mapped from Affymetrix to lllumina yielding 128 genes from the original 150-gene list. The relative median rank score for AD patients was significantly lower than the age and gender matched controls (p=0.004, Figure 6), based on Wilcoxon rank sum test. Blood RNA from the second AD case-control cohort was profiled on the lllumina HT-12 V4 platform and in this case 122 genes were common to the 150-gene healthy ageing gene score. As before, the median rank healthy ageing gene-score for AD patients in Batch 2 was significantly lower than in the control group (p=0.009, Figure 6). Furthermore, for both Batch 1 and Batch 2, the median rank healthy aging gene-score for subjects diagnosed with mild cognitive impairment was lower than that of the age-matched controls (MCI, Figure 6 p=0.00005 and p=0.003 respectively). When applying the 13 gene set (EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2) the median rank healthy ageing gene-score for AD patients (Batch 1 , p=0.043, Batch 2, p=0.051 ) and MCI patients (Batch 2, p =0.0006) was also significantly lower than in the control group. It is important to note that the control samples used for comparison with MCI overlapped with those used for comparison with AD and that the MCI analysis cannot therefore be considered a fully independent observation. Nevertheless, the greater performance at detecting MCI supports the claim that the age signature in blood can predict disease at least 10yr in advance.

We also evaluated if the healthy aging signature could act as a diagnostic for AD or MCI when combined with disease biomarkers, and found it exceed current state of the art blood AD diagnostics (when judged using independent data). For example, a combination of a previously published whole blood RNA diagnostic consisting of 48 genes (J. Alzheimer's Disease 33 (2013) 737-753) and the 150-gene healthy aging diagnostic was evaluated using batch 2 samples. The performance of the combined test as a diagnostic for Alzheimer's disease was assessed using a receiver operator characteristic curve yielding an AUC=0.73- 0.86. Our healthy aging prototype diagnostic can therefore be combined with disease- specific biomarkers to improve the accuracy of clinical diagnosis or prognosis of age related diseases.

The age diagnostic has allowed the demonstration that patients diagnosed with AD or mild cognitive impairment (many on the cusp of AD), when compared with controls of the same chronological age, had less induction of the healthy aging expression signature in their blood. This diagnostic is the first OMIC signature able to identify AD from controls based entirely on an independently developed research hypothesis that does not include feature selection using disease cohorts.

The induction of the healthy aging expression signature in brain regions with age was also investigated using the BrainEac.org gene-chip resource (GSE60862) which comprises 10 post-mortem brain samples from 134 subjects representing 1 ,231 samples. Using the 150 genes of Table 2 and same ranking approach as applied to the ULSAM cohort, the median sum of the rank score was calculated for each anatomical brain region (Figure 7). As before, in healthy older individuals the 'age' signature was 'switched on' (yielding a greater ranking score). Regulation of the healthy age gene score increased across individual healthy brain regions with chronological age, especially in the hippocampus (p=0.00000002), as well as other regions (putamen, thalamus, substantia nigra and the occipital, frontal and temporal cortex regions (all at least p<0.002 by Holm adjusted Mann-Whitney test). Using the 13 genes (EIF3H, JMJD8, CDK13, TNK2, TNP02, CALR, CARM1 , NRXN2, RAB3A, SIN3A, TFRC, TGFBR3 and U2AF2) the median sum of the rank score increased between young and old brain samples in the hippocampus (p=0.00004). DISCUSSION

A change in population age demographics has resulted in an increased prevalence of age-related medical conditions, including cardiovascular and neurodegenerative diseases. It is presumed that successful ageing reflects positive gene-environment interactions that slow the emergence of chronic disease during the 4 th to 7 th decades of life. Many of the molecular mechanisms which extend the lifespan of laboratory animals have been reported to also positively impact on disease-free lifespan (Kenyon (2010) Nature 464: 504-512). Many of these longevity molecules belong to developmental and growth pathways that impact on important physiological pathways. Nevertheless, it has been difficult to establish if any of these are reliably modulated during human ageing (Phillips et al (2013), supra; Glass et al (2013), supra; Beltran Vails et a/ (2014) J Gerontol A Biol Sci Med Sci DOI:

10.1093/gerona/glu007). Even if ageing-related molecular mechanisms are conserved across species, such molecules still may not represent reliable clinical biomarkers. In humans, aerobic fitness has been found to be a powerful but limited 'biomarker' of all-cause mortality (Blair et al (1989), supra; Wei et al (1999) Jama 282: 1547-1553; Myers et al

(2002) N Engl J Med 346: 793-801 ; Church et al (2005) Arch Intern Med 165: 21 14-2120), reflecting genetics (Timmons et a/ (2010), supra), co-morbidity and behavior (e.g. people who feel better may choose to be more physically active). Since the present aim was to develop a RNA diagnostic that when applied to any RNA tissue expression profile, would yield an accurate prediction of healthy physiological age and forecast long-term health, the younger and older samples used in the prototype development were matched for aerobic fitness in an attempt to reveal a novel underlying biomarker.

Molecular diagnostics of human ageing

Genome-wide association analysis has identified DNA variants associated with human longevity; a trait associated with good long-term health. Sebastinani et al identified 281 DNA variants which collectively explained -17% of exceptional longevity in humans (Sebastiani et a/ (2012), supra) and had a ROC value of only 0.6. Indeed, long-lived humans appear to have a similar genetic burden for common DNA disease variants, suggesting the exceptional longevity model may be the clinical equivalent of the 'knock-out' mouse; yielding data that is ultimately difficult to translate to out-bred subjects of 'normal' longevity. A recent 27-SNP DNA-based diagnostic (in the Malmo Preventive Project study; 45 year olds) correlated with 23 year blood-pressure increases (Fava et a/ (2013) Hypertension 61 : 319- 326). However ROC analysis yielded a poor score of 0.66 (0.5 = zero ability) with the established 'non-genetic' correlates, and this was not improved using DNA-based data. Thus data with interesting biological association does not always translate into a useful prognostic tool. Thus, while an ageing diagnostic which relies on DNA holds some practical attraction, based on first principles a RNA-based diagnostic is likely to yield superior explanatory power ((Timmons et a/ (2010), supra).

There have also been several attempts to yield linear models that define the molecular features of chronological age ((Passtoors et al (2012), supra; Phillips et al (2013), supra; Horvath (2013), supra; Hannum et a/ (2013), supra). In the case of Horvath et al, a methylation based model of chronological age was developed, whereby age was

transformed in a unique manner for ages less than and greater than 20 (log and linear transformation respectively). The divergence from chronological age was minimal and thus it is unclear how this can be utilized to identify successful ageing. There was no overlap between the genes in the present healthy-ageing RNA classifier and that of the quasi-linear methylation model derived by Horvath (2013), supra. For the two gene-lists identified by Hannum et al (n=94 and n=326) 4 genes were found to be in common: 1 gene from his primary model (PKM2) and 3 genes from his RNA Methylation association analysis

(ANKRD13B, RUNX3 and TCF3) (Hannum et al (2013), supra). It is felt that there will be a fundamental problem with models built on a linear association with chronological age, as such models will not easily distinguish between 'age' and the accumulation of molecular features of disease and drug treatment. For this reason, neither RNA nor DNA methylation models, built around linear changes with chronological age, are going to be sufficiently independent of disease variables to be a useful independent diagnostic for predicting long- term health outcomes. In contrast, the present study was able to identify a robust molecular diagnostic of 'healthy age' in human tissue, and one that worked in samples of both mesodermal and ectodermal origin. In a study from Passtoors et al, a set of 21 RNA molecules were reported to 'mark out' familial longevity in blood RNA (Passtoors et a/ (2012), supra) but these correlates had no classification capacity. Further, none of these age-related blood RNA changes replicated in the recent analysis of human brain or muscle (Phillips et a/ (2013), supra); Glorioso et al (201 1 ) Neurobiol Dis 41 : 279-290) indicating that they do not represent a starting point for a multi-tissue diagnostic. It is also true that a novel diagnostic may not supersede

chronological age or traditional clinical risk factors for providing prognostic advice. For example, a recent large-scale metabolomic analysis (Fischer et a/ (2014) PLoS Med 1 1 : e1001606) found that the addition of a significant 4-metabolite signature for mortality did not actually improve risk stratification and the metabolites merely co-varied with age. Strict independent validation is often neglected and in one recent example an RNA diagnostic with excellent ROC performance was reported, but it transpires that the validation data-set used the same control samples as the training-data set invalidating the claim (Ramos et a/ (2013) Ann Rheum Dis doi: 10.1 136/annrheumdis-2013-203405). In fact all published work fails to utilise appropriate independent data to validate their models.

It is perhaps important to explain the primary reasons why it was possible to discover such a robust set of marker genes for healthy physiological age. One major feature of the present research strategy was to build a prototype diagnostic using tissue samples obtained from 65 year subjects who had demonstrated successful ageing i.e. they were selected to have excellent metabolic and cardiovascular health (Keller et a/ (201 1 ), supra; Gallagher et al (2010), supra). The use of skeletal muscle as a source of high quality RNA for production of a prototype reflects the fact that such material is easily collected from humans (Gallagher et al (2010), supra; Timmons et al (2005), supra) where the functional status of the precise tissue being profiled is readily established. The muscle derived prototype RNA expression pattern was unrelated to several life-style related influences known to impact on muscle phenotype, and the exceptionally high ROC performance in independent muscle, skin and brain tissue profiles, obtained from several countries, demonstrates that a systemic diagnostic of ageing status in humans has been discovered. There was a lack of association between the prototype age diagnostic and various muscle RNA-disease interactions (Keller et a/ (201 1 ), supra; Fredriksson et a/ (2008) PLoS One 3: e3686; Stephens et a/ (2010) Genome Med 2: 1 ). For example none of the genes modulated in muscle cancer cachexia, wasting or diet-induced muscle atrophy (Thalacker-Mercer et al (2010), supra; Fredriksson et a/ (2008), supra; Gallagher et a/ (2012) Clin Cancer Res 18: 2817-2827) appear in the age-diagnostic. Furthermore, the excellent performance in human brain and skin tissue allows us to conclude that it has been possible to identify a robust diagnostic that is not tissue specific and thus is less likely to be related to any tissue-specific environmental interactions or disease processes.

While exceptional longevity (e.g. 100 years or more) is driven by a strong genetic contribution (Sebastiani et a/ (2012), supra; Puca et a/ (2001 ) Proc Natl Acad Sci U S A 98: 10505-10508), being fit and healthy at age 65 year is a more common occurrence and likely to reflect complex molecular factors (Kenyon (2010), supra; Sabia et al (2012) CMAJ 184: 1985-1992). The ultimate aim of the invention is to be able to predict long-term health outcomes in middle-aged subjects to facilitate personalization of prevention programs.

Ideally, to validate such a new healthy age diagnostic, it would have been desirable to analyze global 'healthy' RNA profiles (non-tumorous) from middle-aged subjects with the appropriate 40 year clinical follow-up data. However, no such materials apparently exists. Instead, healthy members of the ULSAM cohort at age 70 years were profiled, and 20 year follow-up data was analysed. In 1992, these 70 year Swedish men were very healthy and physically active for their chronological age, by European or North American standards, while longevity to 90 year of age is not exceptional in the Swedish population (Danielsson and Talback (2012) Scand J Public Health 40: 6-22). The age diagnostic score

demonstrated a 4-fold range at 70 years, while chronological age varied by no more than 1 year across the group. Using both the 'raw' 670 prototype and the optimised diagnostics, the model of the invention was able to predict health over the following 20 years.

Renal function is an important determinant of all cause mortality (Zethelius et al (2008), supra) and while only 3 from 108 subjects had mild impairment of renal function at 70 years, a clinical model was generated that captured 33% of the variance in renal function at 82 years. The majority of this was driven by the novel healthy-ageing RNA diagnostic of the invention (see Figure 3B). Despite the small sample size (relative to epidemiological studies) for predicting mortality the fact that the healthy-ageing diagnostic also predicted renal function, is consistent with renal function associating with mortality and morbidity in a number of large epidemiological studies (Zethelius et al (2008) N Engl J Med 358: 2107- 21 16; Swindell et al (2012) Rejuvenation Res 15: 405-413). The fact that renal function can be diagnosed from a 'healthy' muscle RNA profile could be considered remarkable, but the excellent multi-tissue performance of the classifier indicates that the diagnostic should be applicable to any RNA sample, including human blood samples. It is notable that the healthy age diagnostic included genes originating from significantly enriched genomic regions at 1 1 q23 and 1 1 q13 and both regions contain SNPs influencing the age of onset of colorectal, renal and prostate cancer (Garasto et a/ (2003), supra; Feitosa et a/ (2014), supra; Talseth- Palmer et a/ (2013), supra; Lubbe et a/ (2012), supra; Audenet et a/ (2014), supra; Lange et al (2012), supra; Jin et al (2012), supra). This is precisely what would be expected if the healthy age diagnostic of the invention was a measure of successful ageing and reflected a set of molecular responses which favoured health in older adults.

Molecular features of the healthy Physiological age diagnostic

In a global DNA analysis by Sebastinani et al, the nearest genes to the 281 longevity- related SNPs were related to a number of chronic disease networks (Sebastiani et al (2012), supra), yet in contrast to this link between disease pathways and longevity, long-lived family lines appear to have a similar number of risk alleles for the common age-related chronic diseases (Beekman et a/ (2010) PNAS 107(42):18046-9). In the present study three genes in the present RNA classifier (erythrocyte membrane protein band 4.1 like 4B (EPB41 L4B), calmodulin binding transcription activator 1 (CAMTA1 ) and the "ageing gene" lamin A/C

(LMNA)) relate to three SNPs (rs10512392, rs2032563 and rs915179) from the Sebastinani et al analysis. This provides independent support for two of these previously unvalidated longevity associated genes (EPB41 L4B and CAMTA1 ), while LMNA is a well established component of ageing like disease (Jiang (2013) Nat Med 19: 515). Nevertheless the degree of overlap between these genomic markers of extreme longevity and the present healthy age diagnostic is very limited supporting the idea that these are two distinct phenomena. As noted earlier, the genetic classifier built by Sebastiani et al (2012; supra) yielded an age diagnostic that had a classification sensitivity of 61 %, during the validation step, while the present RNA based diagnostic substantially exceeded this performance (>90%).

Furthermore, no DNA diagnostic has been shown to capture enough information to be prognostic of long-term health in populations that demonstrate 'normal' longevity.

Identification of the molecular processes that contribute to ageing could provide new ideas to tackle age-related functional decline in humans (Curtis et a/ (2005) Nat Rev Drug Discov 4: 569-580). It has been argued that the natural ageing process reflects a gene- environment interaction whereby genomic variants evolved to enhance early life success impact negatively on health during the transition into older adulthood. The present data suggests that a multi-organ molecular program is induced in those that successfully respond during adulthood and that this process is beneficial. It was noted that a very limited number of young samples have the 'healthy physiological age' profile already at 25 years of chronological age (miss-classification equating to reduced sensitivity in Table 7). Whether these are stochastic events or represent true examples of younger subjects with induction of the healthy physiological age profile is unclear. Further, whether induction at an early chronological age reflects a beneficial characteristic or greater exposure to the molecular mediators of ageing would require 40 year longitudinal trials to unravel. For related reasons the majority of ageing mechanisms identified so far have derived from non-primate biological models (Kenyon (2010), supra) and there has been limited ability to validate such

mechanisms in humans.

The search for ageing related genes directly in humans has relied on an experimental design that focuses on nonagenarian, centenarians and their siblings or offspring. To this end, differential gene-chip comparisons of human tissue samples (Lu et al (2004) Nature

429: 883-891 ) and molecular analysis of case-control or cohort studies have been employed to describe some of the gene expression pathways regulated by ageing (Lu et al (2004), supra; McCarroll et al (2004) Nat Genet 36: 197-204). Other strategies for discovering age- related genes such as multi-species RNA expression comparisons, combined with gene ontology analysis, have also been attempted. However, such analysis is compromised by incomplete knowledge of the population of expressed genes utilised as the statistical background for generation of the ontology enrichment scores (Keller et a/ (201 1 ), supra; Gallagher et a/ (2010), supra). This renders inter-tissue or inter-species comparisons currently challenging to interpret, as not all genes have an equal probability of appearing in the regulated RNA list. This latter issue relates to both biology (divergence of the molecular characteristics across organisms) and divergent technology (gene-chip performance), factors that no current approach can solve easily.

With these caveats in mind, no significant ontology pathway enrichment was noted within the present 670 prototype (or sub-set) healthy-ageing diagnostic gene lists. In fact, when the ontology profile of the 670 prototype was compared with 10,000 randomly selected 670 gene-sets the distribution of p-values were identical (Figure 5). The healthy age prototype diagnostic did however demonstrate some linkage with specific genomic regions. The 3 genes from 1 1 q23, also the location for the apolipoprotein A family (Garasto et al (2003), supra; Feitosa et a/ (2014), supra), originate at a region where single nucleotide variants substantially modify the age of onset of colorectal cancer (Talseth-Palmer et al (2013), supra; Lubbe et al (2012), supra), while at 1 1 q13 several single nucleotide variants modify the age of onset of renal cell carcinoma and prostate cancer (Audenet et al (2014), supra; Lange et a/ (2012), supra; Jin et a/ (2012), supra). Thus, while it cannot neatly place the healthy physiological age diagnostic genes into convenient canonical signalling pathways, the technical performance, prediction of human health over 20 years and the association with age-of-onset modifying regions in large human cohort studies, combine to argue that these molecules are genuine markers of human ageing.

In summary, in the present body of work a novel tool has been provided that should enable the future translation of basic science into clinical advances, namely a robust diagnostic of healthy physiological age. A link has been established between induction of the gene expression signature and renal function and mortality in humans over a 20 year follow- up period, which suggests that it may be possible to facilitate healthy ageing in humans through manipulation of the gene-expression networks. The present technology could be used to facilitate the evaluation of anti-ageing related treatment strategies in humans, screen for long-term safety during drug development or augment clinical decision-making that currently inputs chronological age into treatment algorithms.