Mild Cognitive Impairment Symposium January 19 and 20, 2013

Similar documents
Stephen Salloway, M.D., M.S. Disclosure of Interest

November 16-18, 2017 Hotel Monteleone New Orleans, LA. Provided by

EXPEDITION3: A Phase 3 Trial of Solanezumab in Mild Dementia due to Alzheimer s Disease

AAIC 2018, Chicago, Illinois, US. July 22, 2018

An integrated natural disease progression model of nine cognitive and biomarker endpoints in patients with Alzheimer s Disease

BioArctic announces detailed results of the BAN2401 Phase 2b study in early Alzheimer s disease presented at AAIC 2018

Biogen, Cambridge, MA, USA; 2 Cytel, Cambridge, MA, USA; 3 Neurimmune, Schlieren-Zurich, and University of Zurich, Switzerland

Phase 3 solanezumab trials: Secondary outcomes in mild Alzheimer s disease patients

Custom Intelligence. Alzheimer s Disease Landscape Summary

Biomarkers for Alzheimer s disease

Tammie Benzinger, MD, PhD

Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer s Disease

Alzheimer Clinical Trials: History and Lessons

Two Phase 3 Trials of Bapineuzumab in Mild-to-Moderate Alzheimer s Disease

ISCTM 2014 presentation

Alzheimer s Disease Update: From Treatment to Prevention

The Australian Imaging Biomarkers and Lifestyle Flagship Study of Ageing

Roche satellite symposium/educational session CTAD Asia - China Conference Transforming AD in China: From Diagnosis to Treatment

ALZHEIMER'S DISEASE PREVENTION TRIALS

Il processo diagnostico dell Alzheimer dalla clinica al neuroimaging Giovanni B. Frisoni M.D.

Dementia, Cognitive Aging Services and Support

Issues to Consider in the Clinical Evaluation and Development of Drugs for Alzheimer s Disease. The University of Tokyo Hospital

March 6, 2014 Houston, TX 1:00 PM 2:00 PM

TOWARD EFFECTIVE ALZHEIMER S THERAPY: PROGRESS AND COLLABORATION. Paul S. Aisen, MD Department of Neurosciences University of California, San Diego

The Australian Imaging Biomarkers and Lifestyle Flagship Study of Ageing

42-Lowering Agent, in Alzheimer s s Disease: A Phase 2 Trial of up to 24 Months of Treatment. Gordon K. Wilcock*, University of Oxford, UK

ELND005 for Agitation and Aggression in Alzheimer s Disease (HARMONY-AD Study): Phase 2/3 design and clinical outcomes

BioArctic announces positive topline results of BAN2401 Phase 2b at 18 months in early Alzheimer s Disease

Implementation of fully automated immunoassays for CSF Aβ 1-42, t-tau and p-tau 181 in the Alzheimer s Disease Neuroimaging Initiative

Cognitive Aging: Defining normal vs. disease?

The added value of the IWG-2 diagnostic criteria for Alzheimer s disease

Amyloid and the Vessels. David Weisman, M.D.

FOR IMMEDIATE RELEASE July 26, 2018 Eisai Co., Ltd. Biogen Inc.

Neuroinflammation in preclinical AD: in vivo evidence

Ranjan Duara, MD, FAAN

8/14/2018. The Evolving Concept of Alzheimer s Disease. Epochs of AD Research. Diagnostic schemes have evolved with the research

Alzheimer disease is a progressive neurodegenerative disease. An MRI Rating Scale for Amyloid-Related Imaging Abnormalities with Edema or Effusion

Regulatory Challenges across Dementia Subtypes European View

Supplementary online data

Molecular Imaging Heterogeneity of Clinically Defined AD

Changing diagnostic criteria for AD - Impact on Clinical trials

A perspective on preclinical Alzheimer's disease trials

Dietary Supplements, Caffeine, and Cognitive Aging

Appendix 5. Characteristics of included studies. Study title: NCT

Early Diagnosis of Alzheimer s Disease and MCI via Imaging and Pattern Analysis Methods. Christos Davatzikos, Ph.D.

abstract n engl j med 378;4 nejm.org January 25,

CAMD Annual Regulatory Science Workshop CSF Biomarker Team Refocusing on Low Hanging Fruit

The Aging Brain The Aging Brain

Standardization efforts for Alzheimer CSF biomarkers, and Plasma neurofilament light (NFL) as a biomarker for AD

To date, all efforts to develop a disease-modifying treatment

Alzheimer s disease (AD) is the most

Re: National Coverage Analysis (NCA) for Beta Amyloid Positron Emission Tomography in Dementia and Neurodegenerative Disease

Randomized controlled trials in mild cognitive impairment Sources of variability

Known as both a thief and murderer,

RESEARCH AND PRACTICE IN ALZHEIMER S DISEASE VOL 10 EADC OVERVIEW B. VELLAS & E. REYNISH

Neurocognitive Disorders Research to Emerging Therapies

CTAD 2017 Highlights, November 3-4

Outline. Facts and figures Action plans Early / correct diagnosis Conclusions

DISCLOSURES. Objectives. THE EPIDEMIC of 21 st Century. Clinical Assessment of Cognition: New & Emerging Tools for Diagnosing Dementia NONE TO REPORT

Washington University: Setting the Stage for Secondary Prevention Trials in Alzheimer Disease

ADNI PET CORE. Boston April Bill Jagust, Suzanne Baker, Bob Koeppe, Eric Reiman, Kewei Chen, Chet Mathis, Julie Price

HOMOTAURINE INVESTIGATIONAL SUMMARY

Rational Drug Used For Cognitive Treatment of Alzheimer Disease And Related Dementia

Outlines! Current Issue in Management. of Dementia! Cholinesterase Inhibitors! Medications that soon to be used in. Papan Thaipisuttikul, M.D.

Moving Targets: An Update on Diagnosing Dementia in the Clinic

Longitudinal 148-Week Update of ANAVEX 2-73 Phase 2a Alzheimer s Disease Extension Study

Alzheimer s Disease Prevention and Treatment 2014

A phase III randomized trial of gantenerumab in prodromal Alzheimer s disease

Month/Year of Review: September 2013 Date of Last Review: February 2012

Claims & Underwriting. You already told us that story! Maria C. Carrillo, Ph.D. Sr. Director, Medical & Scientific Relations Alzheimer s Association

Progress in Alzheimer s disease diagnostics validation and use of cognitive endpoints and surrogate markers

The Australian Imaging, Biomarkers and Lifestyle Flagship Study of Ageing an example of Australian research on Alzheimer s disease

Dementia is a syndrome composed

XanADu: A Phase II trial of Xanamem TM in mild Alzheimer's disease

Alzheimer s Disease Prevention Clinical Trials: Taking the Next Steps Responsibly. Joshua Grill University of California, Irvine

Indiana University School of Medicine, Indianapolis, IN USA 1

Neuropsychiatric symptoms as predictors of MCI and dementia: Epidemiologic evidence

Genetic Heterogeneity of Clinically Defined AD. Andrew J. Saykin, PsyD Indiana ADC ADC Clinical Core Leaders Meeting April 22, 2017

What s new in dementia?

Oscar L. Lopez, M.D. Departments of Neurology and Psychiatry Alzheimer s Disease Research Center University of Pittsburgh School of Medicine

Neuro degenerative PET image from FDG, amyloid to Tau

Targeting the p75 Receptor to Inhibit Degenerative Signaling and Tau Phosphorylation/Misfolding/ Missorting: Preclinical through Phase 1

Mild Cognitive Impairment (MCI)

AD remains a leading cause of morbidity and mortality without. MR Imaging Features of Amyloid-Related Imaging Abnormalities ORIGINAL RESEARCH BRAIN

A. Personal Statement

Memory Matters: Learning Objectives: Synapses, Age, and Health. Neuronal Synapses DISCLOSURE DECLARATION. Cognition and Normal Aging

Lutz Frölich 1, Glen Wunderlich 2, Claus Thamer 3, Michael Roehrle 3, Miguel Garcia Jr 4 and Bruno Dubois 5*

ADNI PET CORE. Vancouver BC April Susan Landau, Andy Horng, Suzanne Baker, Bob Koeppe, Eric Reiman, Kewei Chen, Chet Mathis, Julie Price

Dementia and Healthy Ageing : is the pathology any different?

Today s Conversation. What's New in Dementia Research and Care? Our vision is a world without Alzheimer s disease

Dementia of the Alzheimer Type: the Drug Treatment Debate

Alzheimer s disease progression by geographical region in a clinical trial setting

ORIGINAL CONTRIBUTION. Mechanism of Amyloid Removal in Patients With Alzheimer Disease Treated With Gantenerumab

#CHAIR2015. Miami, Florida. September 24 26, JW Marriott Miami. Sponsored by

Geriatric Grand Rounds

USE OF BIOMARKERS TO DISTINGUISH SUBTYPES OF DEMENTIA. SGEC Webinar Handouts 1/18/2013

Clinical Policy Bulletin: Alzheimer's Disease: Experimental Treatments

Alzheimer s Disease Neuroimaging Initiative

How can the new diagnostic criteria improve patient selection for DM therapy trials

Baldeiras et al. Alzheimer's Research & Therapy (2018) 10:33

Transcription:

Highlights of Biomarker and Clinical Outcomes in Recent AD Treatment Trials Stephen Salloway, MD, MS Professor of Neurology and Psychiatry Alpert Medical School, Brown University Mild Cognitive Impairment Symposium January 19 and 20, 2013

Stephen Salloway, M.D., M.S. Disclosure of Interest Research Support 1. NIA-ADNI 2. NIA-DIAN 3. Alz Assoc-DIAN Clinical Trials 4. Fain Family Foundation, Champlin Foundation, White Family Foundation 5. Avid Radiopharmeceuticals Speakers Bureau Athena Clinical Trials Elan, Janssen AI, Baxter, BMS, Pfizer, Medivation, Genentech, GE, Avid, Roche, Merck, Biogen Consultant Janssen AI, Baxter, Pfizer, Athena, BMS, Merck, Lilly, Medivation I own no stocks or equity in any pharmaceutical company

Butler Hospital Memory and Aging Program

Current AD passive immunotherapy trials employ sequence-specific anti-aβ antibodies Bapineuzumab (AAB-001) (Elan, Wyeth) Humanized mouse monoclonal Binds first 5 a.a. of N-terminus Clears plaques and vascular Aβ LY2062430 (Lilly) Humanized mouse Monoclonal Binds to amino acids 14-28 Binds soluble Aβ not deposits PF-4360365 (Pfizer) Binds to C-terminus Deglycosylated to decrease Fc effects Clears plaques, less microglial action AAB-001 LY2062430 PF-4360365 N-terminus Central Region C-terminus

Bapineuzumab Trial Design Multi-center randomized double-blind, placebo-controlled, 18-month clinical trials in mild-moderate AD dementia (MMSE 16-26) APOE ε4 carriers: Bapineuzumab 0.5 mg/kg or placebo (ratio 3:2) Non-carriers: Bapineuzumab 0.5 mg/kg, 1.0 mg/kg or placebo (ratio 3:3:4) 2 mg/kg dose terminated early in Phase 3 due to amyloid-related imaging abnormalities (ARIA) Primary Clinical Endpoints: Alzheimer s Disease Assessment Scale Cognitive (ADAS-Cog 11) Disability Assessment for Dementia (DAD) Key Biomarker Secondary Endpoints: Brain amyloid burden on PiB PET CSF phospho-tau MRI brain volume Schedule of Events: 6 infusions every 13 weeks MRI monitoring for ARIA ~6 weeks after each infusion Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, 5 Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 4

Analysis Populations Study 302 APOE ε4 Carriers Total Randomized N = 1121 Population Placebo N (%) Bapineuzumab 0.5 mg/kg N (%) Randomized (Safety population) 448 (100.0) 673 (100.0) mitt 432 (96.4) 658 (97.8) PiB PET 40 (8.9) 75 (11.1) CSF 85 (19.0) 127 (18.9) vmri 238 (53.1) 352 (52.3) Study 301 Non-Carriers Total Randomized N = 1331 Population Placebo N=524 (%) Bapineuzumab 0.5 mg/kg N=337 (%) Bapineuzumab 1.0 mg/kg N=329 (%) Randomized (Safety population) 524 (100.0) 337 (100.0) 329 (100.0) mitt 493 (94.1) 314 (93.2) 307 (93.3) PiB PET 15 (2.9) 12 (3.6) 12 (3.6) CSF 77 (14.7) 47 (13.9) 54 (16.4) vmri 244 (46.6) 169 (50.1) 146 (44.4) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 6

Baseline Demographics mitt Population Study 302 APOE ε4 Carriers Total Randomized N = 1121 Placebo (N=432) Bapineuzumab (N=658) Age, y (SD) 72.3 (8.4) 72.0 (8.0) Gender (% female) 242 (56.0) 358 (54.4) Race (% Caucasian) 420 (97.2) 624 (94.8) APOE ε4: % heterozygote ε4 % homozygote ε4 325 (75.2) 107 (24.8) 495 (75.2) 163 (24.8) AChEI or memantine use (%) 400 (92.6) 606 (92.1) MMSE total score (SD) 20.7 (3.2) 20.8 (3.1) ADAS-Cog 11 total score (SD) 23.9 (9.5) 23.5 (9.4) DAD total score (SD) 79.4 (18.9) 80.9 (17.3) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 7 6

Baseline Demographics mitt Population Study 301 APOE ε4 Non-Carriers Total Randomized N = 1331* Placebo (N=493) Bapineuzumab 0.5 mg/kg (N=314) Bapineuzumab 1.0 mg/kg (N=307) Age, y (SD) 71.9 (10.1) 73.1 (9.3) 73.5 (9.1) Gender (% female) 248 (50.3) 165 (52.5) 175 (57.0) Race (% Caucasian) 469 (95.1) 298 (94.9) 292 (95.1) AChEI or memantine use, (%) 442 (89.7) 281 (89.5) 278 (90.6) MMSE total score (SD) 21.2 (3.2) 21.2 (3.4) 21.2 (3.3) ADAS-Cog 11 total score (SD) 22.2 (10.1) 22.4 (9.7) 22.2 (10.0) DAD total score (SD) 80.5 (19.2) 80.0 (18.1) 80.4 (18.8) *Bapineuzumab 2.0 mg/kg group (n=141) discontinued early in the course of study; primary cognitive and functional outcomes will not be presented Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 8 www.ctad.fr/12-press/press.asp 7

Solanezumab Trial Design Two, multi-center randomized double-blind, placebo-controlled, 18-month clinical trials in mild-moderate AD dementia (MMSE 16-26), Expedition 1 and 2 Dose: 400 mg IV every 4 weeks Primary Clinical Endpoints: Alzheimer s Disease Assessment Scale Cognitive (ADAS-Cog 11) ADCS Activities of Daily Living (ADCS ADL) ADAS-Cog 14 became the single primary outcome for Expedition 2 based on Expedition 1 data Biomarker Secondary Endpoints: Plasma Ab Brain amyloid burden on Florbetapir PET CSF Ab and phospho-tau MRI brain volume 9 Doody, ANA Annual Meeting 2012

EXP 1 Baseline Demographics Placebo N=506 Solanezumab N=506 Total N=1012 P- Value Age (yrs) 74.5 75.3 74.9 0.15 % Female 287 (57%) 299 (59%) 586 (58%) 0.46 EducaJon (yrs) 12.8 12.6 12.7 0.54 %APOE4+ 288 (61%) 266(57%) 554(59%) 0.23 Screening MMSE 21.0 21.1 21.0 0.93 ADASCOG11 21.8 21.8 21.8 0.64 CDR- SB 5.1 5.0 5.0 0.20 Doody, ANA Annual Meeting 2012

Primary Clinical Outcomes Primary outcomes for bapineuzumab carrier, non-carrier, pooled, mild and moderate were not significant Primary outcomes for solanezumab were negative Mild benefit seen in solanezumab mild group, MMSE > 20, mostly on ADAS-Cog (Exp 1, pooled, and pooled mild) Rate of decline increased by stage in both studies Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp Doody, ANA Annual Meeting 2012 11

Pooled 302/301: Change in ADAS-Cog 11 by Treatment Group Over 78 Weeks (mitt population) Improvement -4-2 0 All Subjects Placebo (n=925) Bap 0.5 mg/kg (n=972) Bap 1.0 mg/kg (n=307) -4-2 0 Mild Subjects (MMSE 21) Placebo (n=497) Bap 0.5 mg/kg (n=521) Bap 1.0 mg/kg (n=176) 2 2 ADAS-Cog Mean (+/-SE) Change From Baseline 4 6 8 10 12 4 6 8 10 12 14 0 13 26 39 52 65 78 Weeks 14 0 13 26 39 52 65 78 Weeks Placebo vs Bap 0.5 mg/kg p=0.793 Placebo vs Bap 1.0 mg/kg p=0.842 Placebo vs Bap 0.5 mg/kg p=0.465 Placebo vs Bap 1.0 mg/kg p=0.513 Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 122012;16:797 www.ctad.fr/12-press/press.asp

Doody, ANA Annual Meeting 2012

Secondary Clinical Endpoints Bapineuzumab In the overall study population, no differences were seen in NTB, MMSE or CDR-SB in either study 302 or 301 Analyses in mild and moderate subgroups are ongoing Solanezumab MMSE-trend or significant for mild group in Expedition 1 and 2 and pooled mild and pooled all No difference seen in CDR-SB for pooled and mild NPI-no difference Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp, Aisen CTAD 2012 14

Biomarker Outcomes

Amyloid PET 93% of ApoE4 carriers met the amyloid cut-off for both studies 36% and 33% of ApoE4 non-carriers in bapineuzumab and solanezumab did not meet the amyloid cut-off Bapineuzumab-mild significant difference in amyloid load in carriers, pooled all and pooled mild, with no difference in noncarriers Amyloid lowering less than phase 2 (Rinne 2010) Solanezumab-no difference in florbetapir pooled Increase in plasma Aβ 42 Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp, Aisen CTAD 2012 16

Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and 17 Aging 2012;16:797 www.ctad.fr/12-press/press.asp Distribution of PIB PET Global Cortical Average SUVr 3 Heterozygotes Homozygotes Noncarrier Baseline PiB PET GCA SUVr 2.5 2 1.5 1.35 1 8/123 (6.5%) Below threshold 22/61 (36.1%) for inclusion APOE ε4 carriers 302 Study N=123 Non-carriers 301 Study N=61 Carrier GCA SUVr Non-Carrier GCA SUVr P-value All PiB PET analysis population 2.07 1.72 p<0.0001 PiB PET analysis population 2.14 2.05 p=0.18

Change in Amyloid Burden as assessed by [ 11 C] PiB-PET at Week 71 APOE ε4 Carriers (PiB PET analysis population) Reduction 0.2 0.16 0.12 APOE ε4 Carriers Placebo (n=40) Bap 0.5 mg/kg (n=75) PiB PET 0.08 0.04 Global Cortical Average SUVr Mean (+/-SE) 0-0.04-0.08-0.12-0.16 Bap 0.5 mg/kg p=0.004 0 45 71 Weeks Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 18

Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, 19 Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp Change in Amyloid Burden as assessed by [ 11 C] PiB-PET at Week 71 APOE ε4 Non-Carriers (PiB PET analysis population) Reduction PiB PET 0.2 0.16 0.12 0.08 0.04 APOE ε4 Non-Carriers Placebo (n=15) Bap 0.5 mg/kg (n=12) Bap 1.0 mg/kg (n=12) Global Cortical Average SUVr Mean (+/-SE) 0-0.04-0.08-0.12-0.16 Bap 0.5 mg/kg p=0.193 Bap 1.0 mg/kg p=0.466 0 45 71 Weeks Pre-specified primary analyses of pooled bapineuzumab doses was not significant, p=0.724 Post hoc exploratory analysis showed a within cohort trend for reduction in PiB PET at 1.0 mg/kg dose (nominal p = 0.057)

Pooled 302/301: Change in Amyloid Burden as assessed by [ 11 C] PiB-PET at Week 71 (PiB PET analysis population) Reduction 0.25 0.2 0.15 All Subjects Placebo (n=55) Bap 0.5 mg/kg (n=87) Bap 1.0 mg/kg (n=12) 0.25 0.2 0.15 Mild Subjects (MMSE 21) Placebo (n=25) Bap 0.5 mg/kg (n=46) Bap1.0 mg/kg (n=9) PiB PET 0.1 0.1 0.05 0.05 Global 0 Cortical Average -0.05 SUVr Mean (+/-SE) -0.1-0.15-0.2 0 45 71 Weeks Placebo vs Bap 0.5 mg/kg p=0.027 Placebo vs Bap 1.0 mg/kg p=0.028 0-0.05-0.1-0.15-0.2 0 45 71 Weeks Placebo vs Bap 0.5 mg/kg p=0.009 Placebo vs Bap 1.0 mg/kg p=0.020 No significant effect in moderate group Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp

Change in C11 PIB in Bapineuzumab 202 Rinne Lancet Neurology 2010 Gantenerumab -15.6% 60 mg and -35.7% 200 mg Ostrowitzki, 2011

CSF Phospho-tau, Total tau and Aβ 42 Phospho-tau Bapineuzumab-mild significant decrease in phospho-tau in carriers, 1.0 mg non-carriers, pooled all and pooled mild Solanezumab-no difference in phospho-tau Total tau Bapineuzumab-no differences seen in carriers or pooled studies, decrease only observed in non-carriers only at 1.0 mg/kg dose Solanezumab-no difference in total tau Aβ 42 Bapineuzumab-no difference Solanezumab-increase in total Aβ 42, no change in free Aβ 42 Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp, Aisen CTAD 2012 22

Change in CSF Phospho-tau by Treatment Group at Week 71 APOE ε4 Carriers (CSF analysis population) Reduction 8 6 4 APOE ε4 Carriers Placebo (n=85) Bap 0.5 mg/kg (n=127) CSF P-tau 181P 2 0-2 Mean (+/-SE) Change From Baseline (pg/ml) -4-6 -8-10 -12 Bap 0.5 mg/kg p=0.005 0 71 Weeks Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 23 www.ctad.fr/12-press/press.asp

Change in CSF phospho-tau by Treatment Group at Week 71 APOE ε4 Non-Carriers (CSF analysis population) Reduction CSF p-tau 181P 8 6 4 2 0-2 APOE ε4 Non-Carriers Placebo (n=77) Bap 0.5 mg/kg (n=47) Bap 1.0 mg/kg (n=54) Mean (+/-SE) Change From Baseline (pg/ml) -4-6 -8-10 -12 Bap 0.5 mg/kg p=0.984 Bap 1.0 mg/kg p=0.009 0 71 Weeks *Pre-specified primary analyses of pooled bapineuzumab doses was not significant, p=0.106 Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, 24 Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp

Pooled 302/301: Change in CSF phospho-tau by Treatment Group at Week 71 (CSF analysis population) Reduction 12 8 All Subjects Placebo (n=162) Bap 0.5 mg/kg (n=174) Bap 1.0 mg/kg (n=54) 12 8 Mild Subjects Placebo (n=97) Bap 0.5 mg/kg (n=110) Bap 1.0 mg/kg (n=35) 4 4 CSF p-tau 181P Mean (+/-SE) Change From Baseline (pg/ml) 0-4 -8-12 -16 0-4 -8-12 -16-20 0 71 Weeks Placebo vs Bap 0.5 mg/kg p=0.014 Placebo vs Bap 1.0 mg/kg p=0.002 Significant effect at both doses in moderate group 25-20 0 71 Weeks Placebo vs Bap 0.5 mg/kg p=0.185 Placebo vs Bap 1.0 mg/kg p=0.041 Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp

Examples of CSF Outcomes in Other AD Clinical Trials CSF AN1792-decreased CSF tau but no change in Aβ42 in antibody responders (Gilman, 2005) Scyllo-inositol-decreased Aβ42 but no difference in tau or p-tau (Salloway, 2011) Avagasestat phase 2-decrease in CSF Aβ1-42 at highest dose only (Coric, 2012) Solanezumab phase 2-12 weekly doses, dose-dependent increase in plasma and CSF total Aβ1-40 and Aβ1-42 (bound and unbound) and increase in unbound CSF Aβ1-42 (Farlow, 2012) Bapineuzumab phase 2-decreased p-tau and trend for decreased tau but no difference in Aβ42 (Blennow 2012)

Volumetric MRI Rate of cortical atrophy Bapineuzumab-no difference in carriers and non- carriers, slight increase in pooled 1.0 mg and pooled mild Solanezumab-no difference Increase in ventricular volume Bapineuzumab-increase in ventricular volume in carriers, 1.0 mg noncarriers, pooled all and pooled mild Solanezumab-no difference Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 27 www.ctad.fr/12-press/press.asp, Aisen CTAD 2012

Rate of Change in MRI Brain Volume (BBSI) by Treatment Group at Week 71 (vmri analysis population) 24 APOE ε4 Carriers 24 Non-Carriers Decreasing Rate of Change 20 p=0.128 20 p=0.725 p=0.132 16 BBSI: Brain Boundary Shift Integral 12 16 12 Mean (+/-SE) Annualized Rate of Change from Baseline to Week 71 (ml/year) 8 4 8 4 0 Placebo (n=238) Bap 0.5 mg/kg (n=352) 0 Placebo (n=244) Bap 0.5 mg/kg (n=169) Bap 1.0 mg/kg (n=146) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 282012;16:797 www.ctad.fr/12-press/press.asp

Pooled 302/301: Rate of Change in MRI Brain Volume (BBSI) by Treatment Group at Week 71 (vmri analysis population) 28 All Subjects 28 Mild Subjects Decreasing Rate of Change 24 20 p=0.323 p=0.034 24 20 p=0.018 p=0.030 BBSI: 16 Brain Boundary Shift Integral 12 16 12 Mean (+/-SE) 8 Annualized Rate of Change From Baseline to Week 71 4 (ml/year) 8 4 0 Placebo (n=482) Bap 0.5 mg/kg (n=521) No significant effect in moderate group Bap 1.0 mg/kg (n=146) 0 Placebo (n=275) Bap 0.5 mg/kg (n=278) Bap 1.0 mg/kg (n=87) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 29 2012;16:797 www.ctad.fr/12-press/press.asp

Rate of Change in MRI Ventricular Volume (VBSI) by Treatment Group at Week 71 (vmri analysis population) Decreasing Rate of Change VBSI: Ventricular Boundary Shift Integral 8 6 4 APOE ε4 Carriers p<.001 Mean (+/-SE) Annualized Rate of Change from Baseline to Week 71 (ml/year) 2 0 Placebo (n=238) Bap 0.5 mg/kg (n=352) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 30

Rate of Change in MRI Ventricular Volume (VBSI) by Treatment Group at Week 71 (vmri analysis population) Decreasing Rate of Change VBSI: Ventricular Boundary Shift Integral Mean (+/-SE) Annualized Rate of Change from Baseline to Week 71 (ml/year) 8 6 4 2 0 Placebo (n=244) APOE ε4 Non-Carriers p=0.362 Bap 0.5 mg/kg (n=169) p=0.001 Bap 1.0 mg/kg (n=146) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health 31 and Aging 2012;16:797 www.ctad.fr/12-press/press.asp

Pooled 302/301: Rate of Change in MRI Ventricular Volume (VBSI) by Treatment Group at Week 71 (vmri analysis population) 10 All Subjects 10 Mild Subjects Decreasing Rate of Change 8 VBSI: 6 ntricular Boundary Shift Integral p=0.0005 p=0.0002 8 6 p<0.0001 p=0.0013 4 4 Mean (+/-SE) Annualized 2 Rate of Change from Baseline to Week 71 (ml/year) 0 Placebo (n=482) Bap 0.5 mg/kg (n=521) Bap 1.0 mg/kg (n=146) 2 0 Placebo (n=275) Bap 0.5 mg/kg (n=278) Bap 1.0 mg/kg (n=87) No significant effect in moderate group Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 32 2012;16:797 www.ctad.fr/12-press/press.asp

Examples of vmri Outcomes in Other AD Clinical Trials MRI AN1792-antibody responders had greater loss of brain volume and larger ventricles but no difference in Hc and no correlation with cognitive decline (Fox, 2005) Bapineuzumab phase 2-no difference in brain volume for all Rx groups combined. Less volume loss in ApoE 4 non-carriers and increased ventricular size in ApoE4 carriers (Salloway, 2009) Scyllo-inositol-No difference in cortical volume but increase in ventricular volume in the 250 mg group (Salloway, 2011) Semagasestat phase 3-n=229, 4.3% decrease in hc volume and 1% decrease in WBV with treatment (Siemers, AAIC 2011)

Amyloid Related Imaging Abnormalities Multi-focal gray and white matter edema (ARIA-E) Microhemorrhages (ARIA-H) Sulcal effusion (ARIA-E) Subtle leptomeningeal involvement (ARIA-E) 34 Sperling et al. Alz & Dementia 2011 24

ARIA-E ARIA-E occurred at low rates in the placebo groups (0.5-1.1%) Bapineuzumab Rate of ARIA-E related to dose and ApoE carrier status New cases detected on Final Read Most cases asymptomatic and transient Symptomatic ARIA-E higher in 2.0 mg dose Most cases occurred with first 3 doses ARIA E associated with an increased rate of incident microhemorrhage Treating through ARIA-E not associated with clinical decline Solanezumab Low rate of ARIA-E-1% solanezumab vs. 0.5% placebo Onset distributed throughout the 18 months of the trial, all but one case asymptomatic All ARIA-E cases in the placebo group were ApoE4 carriers, while cases in the solanezumab group were carriers and non-carriers Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp, Aisen CTAD 2012 35

Treatment Emergent ARIA-E on MRI by Safety Read and Final Read Analysis Group Analysis Group Placebo N=448 (%) Bapineuzumab 0.5 mg/kg N=673 (%) Safety Read 1 (0.2) 103 (15.3) Final Read 5 (1.1) 143 (21.2) Placebo N=524 (%) APOE ε4 Carriers Non-Carriers Bapineuzumab 0.5 mg/kg N=337 (%) Bapineuzumab 1.0 mg/kg N=329 (%) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 36 2012;16:797 www.ctad.fr/12-press/press.asp Bapineuzumab 2.0 mg/kg N=141 (%) Safety Read 1 (0.2) 14 (4.2) 31 (9.4) 20 (14.2) Final Read 3 (0.6) 19 (5.6) 44 (13.4) 28 (19.9) Reasons for additional cases of ARIA-E in Final Read: 1. Not detected by local radiologist (central reads implemented during study) 2. Not detected by central neuroradiologist 3. Site PI did not acknowledge ARIA-E finding at safety read

Pooled 302/301: ARIA-E by APOE ε4 Copy Number (Final Read) Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health 37 and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 45 40 35 Non-carrier ε4 heterozygote ε4 homozygote 57/165 30 Incidence Proportion (%) 25 20 15 86/508 10 5 0 3/524 4/337 1/111 Placebo 19/337 Bap 0.5 mg/kg ε4 heterozygote: RR=3.0 (95% CI: 1.9 4.8; p<0.0001) ε4 homozygote: RR=6.1 (95% CI: 3.8 9.9; p<0.0001)

Questions Is Aβ 42 the right target for mild-moderate AD? Compelling genetic data supporting role of amyloid but unclear which part of the amyloid cascade to target Evidence of mild bapineuzumab anti-amyloid treatment effects on a downstream marker of neurodegeneration (CSF p-tau) without clear clinical benefit What is the mechanism of action for solanezumab? Is a biomarker effect necessary to see a clinical benefit? Too little? Higher doses limited by ARIA-E with bapineuzumab, possible room to increase dose of solanezumab Amyloid lowering on PIB PET probably insufficient to alter clinical course What is the clinical impact of mild cognitive improvement only in mild AD pt? Too late? AD stage may be too far advanced for these drugs to demonstrate a major clinical benefit Will anti-amyloid therapies may be more efficacious at earlier stages and what effect size might we see? Combination therapies may be required for maximizing clinical benefit Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health 38 and Aging 2012;16:797 www.ctad.fr/12-press/press.asp

Issues Regarding Biomarker Outcomes Amyloid PET Include amyloid cut-offs in future AD trials, especially in noncarriers Ensures sample more likely to have AD Avoids exposing individuals to the risks treatments with little chance of benefit Determine the diagnosis and rate of progression in amyloidnegative subjects Measure outcomes for amyloid + subjects (CSF and amyloid PET) Compare PIB and CSF for subjects who participated in both studies Reasons for decreased amyloid lowering in phase 3 Lower doses, separate cohorts for carriers and non-carriers Rate of change in biomarkers may vary by disease stage Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health 39 and Aging 2012;16:797 www.ctad.fr/12-press/press.asp

Issues Regarding Biomarker Outcomes vmri Risks in choosing vmri as outcome measure Uncertainty regarding direction of change Effect on sample size calculation Possible mechanisms for increased rate of cortical atrophy and ventricular enlargement Increased neurodegeneration Amyloid removal Reduction in amyloid-associated inflammation Changes in CSF absorption or other fluid shifts Salloway, Sperling, Fox, CTAD 2012,. J Nutrition, Health and Aging 2012;16:797 www.ctad.fr/12-press/press.asp 40