23.01.2015 Views

2012 Annual Meeting - American Neurological Association

2012 Annual Meeting - American Neurological Association

2012 Annual Meeting - American Neurological Association

SHOW MORE
SHOW LESS
  • No tags were found...

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

ONSITE PROGRAM<br />

AMERICAN<br />

NEUROLOGICAL<br />

ASSOCIATION<br />

IN PARTNERSHIP WITH<br />

ASSOCIATION<br />

OF BRITISH<br />

NEUROLOGISTS<br />

EXPAND<br />

YOUR NEURAL<br />

NETWORKS<br />

<strong>2012</strong> <strong>Annual</strong> <strong>Meeting</strong><br />

OCTOBER 7-9 | BOSTON<br />

www.aneuroa.org/ANA<strong>2012</strong>


AMERICAN<br />

NEUROLOGICAL<br />

ASSOCIATION<br />

Officers & Councilors<br />

OFFICERS<br />

President: Eva L. Feldman, 2011-2013<br />

President-Elect: Robert H. Brown, Jr., 2011-2013<br />

1st Vice President: Justin C. McArthur, 2010-<strong>2012</strong><br />

2nd Vice President: Karl Kieburtz, 2010-<strong>2012</strong><br />

Secretary: Nina F. Schor, 2010-2013<br />

Treasurer: Steven P. Ringel, 2010-2014<br />

Past President: Robert L. Macdonald, 2011-2013<br />

COUNCILORS<br />

Robert D. Brown, Jr., 2009-<strong>2012</strong><br />

Henry J. Kaminski, 2009-<strong>2012</strong><br />

Daniel H. Geschwind, 2010-<strong>2012</strong><br />

Amy R. Brooks-Kayal, 2010-2013<br />

Barbara G. Vickrey, 2010-2013<br />

Anthony J. Windebank, 2010-2013<br />

Allan I. Levey, 2011-2014<br />

Kenneth L. Tyler, 2011-2014<br />

Allison Brashear, 2011-2014<br />

LOCAL ARRANGEMENTS COMMITTEE<br />

Martin Samuels ‘12<br />

Anne Louise Oaklander ‘12<br />

SCIENTIFIC PROGRAM COMMITTEE<br />

William Mobley, Chair ‘13 J. Tim Greenamyre ‘13<br />

David Fink ‘12<br />

Frances Jensen ‘13<br />

Jaideep Kapur ‘12 Jack Parent ‘13<br />

Alan Pestronk ‘12<br />

James Meschia ‘13<br />

Richard Ransohoff ‘12 Joel Perlmutter ‘13<br />

Ralph Gregory ‘12 Steven Scherer ‘14<br />

Martin Rossor ‘12 Samuel Pleasure ‘14<br />

Hadi Manji ‘12<br />

Ahmet Hoke ‘14<br />

WELCOME REMARKS<br />

Dear Colleagues,<br />

Welcome to Boston and the <strong>American</strong> <strong>Neurological</strong> <strong>Association</strong>’s<br />

<strong>2012</strong> <strong>Annual</strong> <strong>Meeting</strong>! There is plenty to be excited about this year, and we<br />

hope you’ll agree.<br />

First of all we would like to welcome our colleagues from the <strong>Association</strong> of<br />

British Neurologists, who bring with them an extraordinary level of scholarship<br />

and expertise that have helped us craft superb scientific programs on genetics,<br />

Alzheimer’s disease and more.<br />

We also hope you’ll take full advantage of our newer offerings as well, including<br />

daily faculty development courses designed to build the real-world skills that<br />

academic neurologists need to advance their careers, at every level. We’ve<br />

also added daily interactive lunch programs that will put attendees into lively<br />

conversations on common and controversial issues in neurology, as well as<br />

provide outstanding networking opportunities with neurology’s thought leaders,<br />

journal editors and NIH representatives, among others.<br />

And we’re bringing cutting-edge technology into the ANA <strong>Annual</strong> <strong>Meeting</strong> as<br />

well! This year’s onsite program will be available as an app for all mobile devices,<br />

including iPhone, iPad, Android and Blackberry. And the entire <strong>Annual</strong> <strong>Meeting</strong><br />

space will be Wi-Fi enabled!<br />

So please take advantage of all the exciting offerings you’ll find at the <strong>2012</strong><br />

<strong>Annual</strong> <strong>Meeting</strong>, and be sure to meet and talk to as many colleagues as you<br />

can. You’ll see once again why the ANA has been The Home of Academic<br />

Neurology since 1875.<br />

Warm regards,<br />

TABLE OF CONTENTS<br />

Schedule at a Glance .............. 3<br />

Hotel Floorplan .................. 4<br />

General Information ............... 5<br />

Saturday Program Schedule ......... 6<br />

Sunday Program Schedule .......... 6<br />

Monday Program Schedule ......... 12<br />

Tuesday Program Schedule ......... 18<br />

Special Events. ................. 23<br />

Sunday Symposia Speaker Abstracts . . 24<br />

Monday Symposia Speaker Abstracts . . 29<br />

Tuesday Symposia Speaker Abstracts . . 34<br />

Eva L. Feldman, MD, PhD, FAAN<br />

ANA President 2011-2013<br />

William C. Mobley, MD, PhD<br />

Chair, ANA Scientific Program Advisory Committee 2011-2013


PROGRAM AT A GLANCE<br />

Saturday, October 6, <strong>2012</strong><br />

3:00 – 8:00 pm<br />

Registration Hours<br />

Monday, October 8, <strong>2012</strong><br />

6:30 am – 5:45 pm<br />

Registration Hours<br />

Tuesday, October 9, <strong>2012</strong><br />

6:30 am – 5:45 pm<br />

Registration Hours<br />

6:00 – 8:00 pm<br />

AUPN Leadership Lecture & Business <strong>Meeting</strong><br />

Salon H-J (4th Floor)<br />

Junior Faculty<br />

Development<br />

Course<br />

Session I<br />

Salon F<br />

(4th Floor)<br />

Sunday, October 7, <strong>2012</strong><br />

6:00 am – 5:45 pm<br />

Registration Hours<br />

7:00 – 9:00 am 7:00 – 9:00 am 7:00 – 9:00 am<br />

Mid/Senior<br />

Level Faculty<br />

Development<br />

Course<br />

Session I<br />

Salon CD<br />

(4th Floor)<br />

AUPN:<br />

Neurology<br />

Chair<br />

Development<br />

Course<br />

Session I<br />

Salon AB<br />

(4th Floor)<br />

9:00 – 11:30 am<br />

SYMPOSIUM: New Tools to Define the<br />

Genetics of <strong>Neurological</strong> Disorders<br />

Salon E (4th Floor)<br />

11:45 am – 1:00 pm<br />

Interactive Lunch Workshops<br />

Meet the Symposia Presenters – Salon F (4th Floor)<br />

Meet the Editors – Wellesley (3rd Floor)<br />

Meet the Chairs – Dartmouth (3rd Floor)<br />

Meet the Professors – Simmons (3rd Floor)<br />

Meet NIH Institutes – Berkeley/Clarendon (3rd Floor)<br />

Being Heard Clearly – Harvard (3rd Floor)<br />

1:15 – 3:15 pm<br />

SYMPOSIUM: Imaging to Explore Neural Network<br />

Structure and Function<br />

Salon E (4th Floor)<br />

3:30 – 5:30 pm<br />

Special Interest Group Symposia<br />

Cerebrovascular Disease – Salon F (4th Floor)<br />

Sleep Disorders and Circadian Rhythm –<br />

Salon CD (4th Floor)<br />

Education – Provincetown (4th Floor)<br />

Neuro-oncology – Salon B (4th Floor)<br />

Health Services Research – Salon A (4th Floor)<br />

5:30 – 7:00 pm<br />

Poster Stand-by Wine & Cheese Reception<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

7:00 – 9:00 am<br />

Junior Faculty<br />

Development<br />

Course<br />

Session II<br />

Salon F<br />

(4th Floor)<br />

7:00 – 9:00 am<br />

Mid/Senior<br />

Level Faculty<br />

Development<br />

Course<br />

Session III<br />

Salon CD<br />

(4th Floor)<br />

7:00 – 9:00 am<br />

AUPN:<br />

Neurology Chair<br />

Development<br />

Course Session II<br />

Salon AB<br />

(4th Floor)<br />

9:00 – 11:30 am<br />

PRESIDENT’S SYMPOSIUM: Alzheimer’s Disease:<br />

New Perspectives on an Old Disease<br />

Salon E (4th Floor)<br />

11:45 am – 1:00 pm<br />

Interactive Lunch Workshops<br />

MOC for Board-Certified Neurologists –<br />

Berkeley (3rd Floor)<br />

Pharma/Clinical Trials –<br />

Fairfield/Exeter (3rd Floor)<br />

Contemporary Clinical Issues in<br />

Neuromuscular Disease –<br />

Regis (3rd Floor)<br />

Headache & Pain Role of Imaging –<br />

Suffolk (3rd Floor)<br />

Concussion/Trauma/TBI –<br />

Simmons (3rd Floor)<br />

NeuroNEXT –<br />

Clarendon (3rd Floor)<br />

Career Challenges –<br />

Dartmouth (3rd Floor)<br />

11:45 am – 1:00 pm<br />

Women of the<br />

ANA Program<br />

Salon F<br />

(4th Floor)<br />

1:15 – 3:30 pm<br />

SYMPOSIUM: Results of Immune-Based<br />

Trials in <strong>Neurological</strong> Disorders<br />

Salon E (4th Floor)<br />

3:30 – 5:30 pm<br />

Special Interest Group Symposia<br />

Dementia and Aging – Salon F (4th Floor)<br />

Neurocritical Care – Salon D (4th Floor)<br />

Epilepsy – Provincetown (4th Floor)<br />

Neuromuscular Disease – Salon BC (4th Floor)<br />

Case Studies – Salon A (4th Floor)<br />

5:30 – 6:30 pm<br />

Poster Stand-by Wine & Cheese Reception<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

6:30 – 8:00 pm<br />

President’s Reception<br />

Boston Public Library<br />

7:00 – 9:00 am<br />

Junior Faculty<br />

Development<br />

Course<br />

Session III<br />

Salon F<br />

(4th Floor)<br />

7:00 – 9:00 am<br />

Mid/Senior<br />

Level Faculty<br />

Development<br />

Course<br />

Session III<br />

Salon CD<br />

(4th Floor)<br />

7:00 – 9:00 am<br />

AUPN:<br />

Neurology<br />

Chair<br />

Development<br />

Course<br />

Session III<br />

Salon AB<br />

(4th Floor)<br />

9:00 – 11:05 am<br />

SYMPOSIUM: Derek-Denny Brown<br />

New Member Symposium<br />

Salon E (4th Floor)<br />

11:05 am – 12:00 pm<br />

Introduction of New Members and<br />

Executive Session of Membership<br />

Salon E (4th Floor)<br />

12:00 – 1:15 pm<br />

Interactive Lunch Workshops<br />

Decompressive Craniectomy for<br />

Trauma – Dartmouth (3rd Floor)<br />

Continuous EEG Monitoring in the<br />

ICU –Suffolk (3rd Floor)<br />

Neuro-oncology – Berkeley<br />

(3rd Floor)<br />

The Role of Amyloid PET<br />

Imaging in Early Diagnosis of<br />

Alzheimer Disease –<br />

Salon F (4th Floor)<br />

Managing Conflict –<br />

Clarendon (3rd Floor)<br />

12:15 – 1:30 pm<br />

Orientation for<br />

New Members<br />

Salon CD<br />

(4th Floor)<br />

1:30 – 3:15 pm<br />

SYMPOSIUM: Advances in Headache and<br />

Pain Research and Treatment<br />

Salon E (4th Floor)<br />

3:30 – 5:30 pm<br />

Special Interest Group Symposia<br />

Behavioral Neurology – Salon AB (4th Floor)<br />

Autoimmune Neurology – Salon F (4th Floor)<br />

Movement Disorders – Salon CD (4th Floor)<br />

Regulatory Science – Provincetown (4th Floor)<br />

5:30 – 7:00 pm<br />

Poster Stand-by Wine & Cheese Reception<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

3


FLOOR PLANS<br />

3rd Floor<br />

4th Floor<br />

PROVINCE-<br />

TOWN<br />

ATRIUM<br />

AREA<br />

FREIGHT ELEVATORS<br />

SALON K<br />

SALON J<br />

SALON I<br />

SALON H<br />

SALON G<br />

SALON F<br />

SALON E<br />

SALON A<br />

SALON B<br />

SALON C<br />

SALON D<br />

NANTUCKET<br />

HYANNIS YARMOUTH VINEYARD<br />

ORLEANS<br />

FALMOUTH<br />

4


GENERAL INFORMATION<br />

Hotel Information<br />

Boston Marriott Copley Place<br />

110 Huntington Avenue<br />

Boston, Massachusetts 02116<br />

Phone: 1-617-236-5800<br />

Conference Website<br />

www.aneuroa.org/ANA<strong>2012</strong><br />

Registration Hours<br />

4th Floor Registration Area<br />

Saturday, October 6: 3:00 pm – 8:00 pm<br />

Sunday, October 7: 6:00 am – 5:45 pm<br />

Monday, October 8: 6:30 am – 5:45 pm<br />

Tuesday, October 9: 6:30 am – 5:45 pm<br />

Poster Hours<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

Sunday, October 7: 11:30 am – 7:00 pm<br />

(Poster Stand-by 5:30 – 7:00 pm)<br />

Monday, October 8: 11:30 am – 6:30 pm<br />

(Poster Stand-by 5:30 – 6:30 pm)<br />

Tuesday, October 9: 11:30 am – 7:00 pm<br />

(Poster Stand-by 5:30 – 7:00 pm)<br />

Speaker Ready Room<br />

Falmouth (4th Floor)<br />

Saturday, October 6: 6:00 am – 8:00 pm<br />

Sunday, October 7: 6:00 am - 5:30 pm<br />

Monday, October 8: 6:00 am - 5:30 pm<br />

Tuesday, October 9: 6:00 am - 5:00 pm<br />

Wireless Connection<br />

A wireless connection is available for ANA attendees. To connect, join the<br />

wireless network <strong>American</strong> <strong>Neurological</strong> Assoc. When prompted,<br />

you can enter the code 1134ana to access the internet.<br />

Mobile App<br />

The ANA is pleased to announce a new mobile application for the <strong>2012</strong><br />

<strong>Annual</strong> <strong>Meeting</strong>. The Mobile app, powered by Eventlink and created by<br />

Core-apps LLC, is a native application for smartphones (iPhone and<br />

Android), a hybrid web-based app for Blackberry, and there’s also a webbased<br />

version of the application for all other web browser-enabled phones.<br />

Evaluations Online!<br />

Evaluations will be available online only this year. Evaluations can be found<br />

at www.aneuroa.org/<strong>2012</strong>Evaluations. Attendees will also receive<br />

an email with links to the evaluations. Your input is very important to us<br />

in helping plan future <strong>Annual</strong> <strong>Meeting</strong>s, and we urge you to complete the<br />

evaluations on a timely basis.<br />

Accreditation<br />

This live activity has been planned and implemented in accordance with<br />

the Essential Areas and Policies of the Accreditation Council for Continuing<br />

Medical Education (ACCME) through sponsorship of the <strong>American</strong><br />

<strong>Neurological</strong> <strong>Association</strong>. The ANA is accredited by the ACCME to provide<br />

continuing medical education for physicians. The <strong>American</strong> <strong>Neurological</strong><br />

<strong>Association</strong> designates this educational activity for a maximum of 37.25<br />

hours AMA PRA Category 1 Credit. Each physician should claim only<br />

those hours of credit that he/she actually spent in the educational activity.<br />

The <strong>2012</strong> Certificate of Attendance was mailed to U.S. attendees in their<br />

registration packets.<br />

Dress Code<br />

Business Casual<br />

iPosters<br />

ANA is excited to announce that we will again offer iPosters, an online<br />

access to poster presentations found at the <strong>Annual</strong> <strong>Meeting</strong>. Poster<br />

presenters will have the option of uploading their posters to the iPoster<br />

website so attendees can view their posters in advance. Attendees can<br />

search by topic or category, and view research and interact directly with the<br />

presenters online. Computer kiosks will be made available at the <strong>Annual</strong><br />

<strong>Meeting</strong> specifically for viewing iPosters. iPosters are also available<br />

online throughout the year. To view the <strong>Annual</strong> <strong>Meeting</strong> iPosters, please<br />

visit ana.posterview.com.<br />

<strong>2012</strong> New Members<br />

Please go to www.aneuroa.org/<strong>2012</strong>NewMembers for information<br />

on the new active, corresponding, and honorary members of the ANA.<br />

Information on New Members can also be viewed in the Atrium (3rd Floor).<br />

<strong>2012</strong> Award Recipients<br />

Please go to www.aneuroa.org/<strong>2012</strong>Awards to view the <strong>2012</strong> ANA<br />

Award Recipients. Information on Award Recipients can also be viewed in<br />

the Atrium (3rd Floor).<br />

How to Download: for iPhone (plus iPod Touch & iPad) and Android<br />

phones: Visit the App Store or Android Market on your phone and search<br />

for ANA <strong>2012</strong>. For all other phone types (including Blackberry and other<br />

web browser-enabled phones): While on your smartphone, point your<br />

mobile browser to m.core-apps.com/ana_annual<strong>2012</strong>. From there<br />

you will be directed to download the proper version of the app for your<br />

particular device, or, on some phones, you simply bookmark the page for<br />

future reference. We hope this new mobile application makes it even easier<br />

for you to make the most out of your <strong>Annual</strong> <strong>Meeting</strong> experience!<br />

5


SATURDAY, OCTOBER 6 SUNDAY, OCTOBER 7<br />

6:00 – 8:00 pm<br />

Salon H-J (4th Floor)<br />

3:00 – 8:00 pm<br />

Registration<br />

4th Floor Registration Area<br />

AUPN: Neurology Chair Development Course<br />

Leadership Course Kickoff:<br />

Coming Challenges in Health Care:<br />

How Does Leadership Respond<br />

Chair: Henry J. Kaminski, MD, George Washington University,<br />

Washington, DC<br />

Marc J. Roberts, PhD, Harvard University, Boston<br />

The AUPN is pleased to be able to tap into Harvard’s School of Public<br />

Health’s expertise to provide this course on such a timely subject.<br />

The lecture is open to all and is of particular interest to Chairs of<br />

Neurology to assist in strategic decision making.<br />

Marc Roberts is Professor of Political Economy at the Harvard School<br />

of Public Health and is an active consultant in helping organizations<br />

adjusting to changing market conditions. He played a leading role in<br />

the World Bank’s training efforts on health sector reform around<br />

world-having taught courses for senior government officials in nearly<br />

thirty countries. He will apply his knowledge to advise leaders in<br />

neurology what to expect in the coming years from health care reform.<br />

The Leadership Lecture will be preceded by the AUPN Business<br />

<strong>Meeting</strong> and include a reception with wine and appetizers.<br />

7:00 – 9:00 am<br />

Salon F (4th Floor)<br />

6:00 am – 5:45 pm<br />

Registration<br />

4th Floor Registration Area<br />

6:00 – 7:00 am<br />

Coffee and Rolls<br />

Coffee will be available until 10:30 am<br />

Junior Faculty Development Course: Establishing<br />

Yourself in the World of Academic Neurology<br />

Session I: Getting Funded/Grant Writing<br />

Chair: Daniel H. Lowenstein, MD, University of California, San Francisco<br />

Frances E. Jensen, MD, Children’s Hospital, Boston<br />

Randall R. Steward, PhD, NIH/NINDS, Bethesda, Md.<br />

Vicky Holets Whittemore, PhD, NIH/NINDS, Bethesda, Md.<br />

This session will focus on a core need required of everyone who is<br />

pursuing academic neurology with an emphasis on research: getting<br />

funded. Regardless of the funding source, getting funded requires<br />

convincing others that you have the ideas, skills, capacity and track record<br />

to use a research grant in a way that will lead to important discoveries, and<br />

this is accomplished by writing outstanding grant proposals. The session<br />

will focus on the entire process of getting funded, including timelines for<br />

generating a proposal, idea generation, organization of the research plan<br />

(including special tips on effectively conveying your plan), the nature of the<br />

review process, and how to respond to reviewers’ critiques.<br />

7:00 – 9:00 am<br />

Salon CD (4th Floor)<br />

Mid/Senior Level Faculty Development Course:<br />

Negotiations & Conflict Resolution<br />

Session I: Being Heard Clearly<br />

Chair: Lisa M. DeAngelis, MD, Memorial Sloan-Kettering Cancer Center,<br />

New York<br />

Susan Miller, PhD, CCC-SLP, Voice Trainer, LLC, Washington, DC<br />

Whether conversing with patients, presenting to your faculty or delivering a<br />

paper at a national meeting, you want to be heard clearly. In this workshop,<br />

we will investigate how your eye contact, gestures and movement affect the<br />

quality of your daily communications and how to read your listener’s body<br />

language. You will learn how to modulate your vocal tone, breath control,<br />

speaking rate and anxiety to assure pleasant, vocal production. You will<br />

master the basics of developing a clear succinct message about yourself<br />

of a project and delivering it powerfully. In this highly interactive workshop<br />

you will discover the keys to being heard clearly.<br />

6


7:00 – 9:00 am<br />

Salon AB (4th Floor)<br />

AUPN: Neurology Chair Development Course<br />

Session I: Challenges of Faculty Development<br />

and Managing K Awardees<br />

Moderator: Karen C. Johnston, MD, University of Virginia, Charlottesville<br />

Stephen L. Hauser, MD, University of California, San Francisco<br />

This session will focus on the challenges of faculty development and more<br />

specifically on managing K08/K23 and related awards. We hope to have a<br />

vigorous discussion to address the question of how Chairs and Departments<br />

can better support the careers of clinician scientists and help to ensure<br />

their success whenever possible. Although the specific challenges will vary<br />

between institutions, one common area is the management of cost sharing<br />

requirements assumed by departments for recipients of K awards.<br />

This session is not available for AMA PRA Category 1 Credit.<br />

9:00 – 11:30 am<br />

Salon E (4th Floor)<br />

SYMPOSIUM: New Tools to Define the Genetics of<br />

<strong>Neurological</strong> Disorders<br />

Chair: Patrick F. Chinnery, PhD, FRCPath, FRCP, FMedSci,<br />

Newcastle University, Newcastle upon Tyne, UK<br />

Co-Chair: Robert H. Brown, Jr., MD, DPhil, University of<br />

Massachusetts, Worcester<br />

This symposium will describe the current state-of-the-art in neurogenetics,<br />

highlighting key recent findings that influence our understanding of<br />

neurological disease mechanisms, and have a direct role in clinical<br />

neurological practice. Topics covered will include: (i) the role of genomewide<br />

association studies to advance our understanding of common<br />

neurological disorders; (ii) Using next-generation whole exome and whole<br />

genome sequencing to diagnose single-gene disorders, (iii) epigenetic<br />

mechanisms and RNA-regulation in neurological disease; and (iv)<br />

diagnosing mitochondrial DNA diseases. By the end of the symposium,<br />

the attendee will have a broad understanding of current genetic and<br />

epigenetic approaches to understand neurological disease, and new<br />

diagnostic tools for neurogenetic diagnostics.<br />

Learning Objectives: Having completed this symposium, participants<br />

will be able to:<br />

1. Understand the methodological approach, strengths, weaknesses,<br />

successes and failures of GWAS in neurology, and what can we expect<br />

in the future<br />

2. Understand the methodological approach (targeted exon capture vs<br />

whole exome and genome approaches), strengths and weaknesses<br />

(including the “hit rate”), recently identified neurogenetics, and the role<br />

in clinical diagnostics.<br />

3. Understand what epigenetics, dissect epigenetic mechanisms, and<br />

epigenetic mechanisms in neurological disease.<br />

4. Understand what RNA regulation is, how RNA-mediated mechanisms<br />

are studied, and what the implications for understanding neurological<br />

and neurodegenerative disorders are.<br />

5. Understand the genetic basis of mitochondrial disease, principles<br />

of molecular diagnosis in mitochondrial disorders, and diagnostic<br />

algorithm for mitochondrial diseases.<br />

9:00 – 9:05 am<br />

Wolfe Award Presentation<br />

Thomas E. Lloyd, MD, PhD, Johns Hopkins University, Baltimore, Md.<br />

Annals of Neurology Prize Presentation<br />

Michael G. Schlossmacher, MD, FRCPC, Brigham and Women’s Hospital,<br />

Harvard Medical School, Boston and University of Ottawa, Ontario, Canada<br />

9:05 – 9:30 am<br />

Genome-Wide <strong>Association</strong> Studies: Have They Delivered<br />

for Neurology<br />

Stephen Sawcer, PhD, University of Cambridge, Cambridge, UK<br />

9:30 – 10:00 am<br />

Next-Generation Whole Exome and Whole Genome<br />

Sequencing to Diagnose Single-Gene Disorders<br />

Andrew Singleton, PhD, National Institutes of Health, Bethesda, Md.<br />

10:00 – 10:15 am<br />

Coffee Break<br />

10:15 – 10:40 am<br />

Epigenetics and Disorders of the Nervous System:<br />

An Evolving Synthesis<br />

Mark F. Mehler, MD, Albert Einstein College of Medicine, Bronx, N.Y.<br />

10:40 – 11:05 am<br />

Regulatory RNAs in <strong>Neurological</strong> Disease<br />

Claes Wahlestedt, MD, PhD, University of Miami<br />

11:05 – 11:30 am<br />

New Tools to Define the Genetics of <strong>Neurological</strong> Disorders:<br />

Mitochondrial Disorders<br />

Patrick F. Chinnery, PhD, FRCPath, FRCP, FMedSci, Newcastle University,<br />

Newcastle upon Tyne, UK<br />

11:30 am – 7:00 pm<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

All Day Poster Presentations<br />

Poster Stand-by time will be from 5:30 – 7:00 pm<br />

11:30 am – 1:00 pm<br />

Lunch<br />

Pick up your lunch in the 4th Floor Foyer.<br />

7


SUNDAY, OCTOBER 7<br />

11:45 am – 1:00 pm<br />

Interactive Lunch Workshops:<br />

Networking Roundtables<br />

Sunday’s Interactive Lunch Workshops are designed to connect attendees<br />

with experts in the field in a fast-paced series of informal discussions.<br />

The room will be set up with five to six round tables – each assigned to one<br />

expert in the identified topic and each allowing for 15-20 attendees per table.<br />

Because of this informal format, attendees are encouraged to move between<br />

experts, tables and workshops as they wish.<br />

Meet the Symposia Presenters<br />

Salon F (4th Floor)<br />

Speakers from the New Tools to Define the Genetics of <strong>Neurological</strong><br />

Disorders symposium will be available to further discuss their<br />

symposium presentations.<br />

Faculty: Andrew Singleton, PhD, National Institutes of Health,<br />

Bethesda, Md.<br />

Stephen Sawcer, PhD, University of Cambridge, UK<br />

Claes Wahlestedt, MD, PhD, University of Miami<br />

Patrick F. Chinnery, PhD, FRCPath, FRCP, FMedSci, Newcastle University,<br />

Newcastle upon Tyne, UK<br />

Mark F. Mehler, MD, Albert Einstein College of Medicine, Bronx, N.Y.<br />

Meet the Editors<br />

Wellesley (3rd Floor)<br />

Editors from the Annals of Neurology, Brain, Archives of Neurology,<br />

Neurology, Stroke and the New England Journal of Medicine will be<br />

available to discuss the submission process, publishing tips and other<br />

key topics of interest.<br />

Moderator: Nilufer Ertekin-Taner, MD, PhD, Mayo Clinic,<br />

Jacksonville, Fla.<br />

Faculty: Stephen L. Hauser, MD, University of California, San Francisco<br />

Alastair Compston, MBBS, PhD, FmedSci, University of Cambridge, UK<br />

Allan H. Ropper, MD, Brigham and Women’s Hospital, Boston<br />

Roger N. Rosenberg, MD, University of Texas Southwestern, Dallas<br />

Karen L. Furie, MD, MPH, Rhode Island Hospital and Brown Medical<br />

School, Providence<br />

David S. Knopman, MD, Mayo Clinic, Rochester, Minn.<br />

Moderator: Joachim Baehring, MD, DSc, Yale School of Medicine,<br />

New Haven, Conn.<br />

Faculty: Justin C. McArthur, MBBS, MPH, FAAN,<br />

Johns Hopkins University, Baltimore, Md.<br />

Richard P. Mayeux, MD, MSc, Columbia University, New York<br />

Martin A. Samuels, MD, DSc(hon), FAAN, MACP, FRCP, Brigham and<br />

Women’s Hospital, Boston<br />

David A. Hafler, MD, Yale University, New Haven, Conn.<br />

Robert H. Brown Jr., MD, DPhil, University of Massachusetts, Worcester<br />

Merit E. Cudkowicz, MD, Massachusetts General Hospital, Boston<br />

Meet the Professors<br />

Simmons (3rd Floor)<br />

This is a place to connect with seasoned academic neurology professors to<br />

discuss tried and true practices for teaching and a free exchange of ideas.<br />

Moderator: Rebecca F. Gottesman, MD, PhD, Johns Hopkins University,<br />

Baltimore, Md.<br />

Faculty: David A. Drachman, MD, University of Massachusetts, Worcester<br />

J.P. Mohr, MD, Columbia University, New York<br />

William M. Landau, MD, Washington University, St. Louis, Mo.<br />

Anne B. Young, MD, PhD, Massachusetts General Hospital, Boston<br />

Guy M. McKhann, MD, Johns Hopkins University, Baltimore, Md.<br />

Meet NIH Institutes<br />

Berkeley/Clarendon (3rd Floor)<br />

This is your chance to get your questions answered by representatives from<br />

the National Institute of <strong>Neurological</strong> Disorders and Stroke (NINDS).<br />

Moderator: Anne Louise Oaklander, MD, PhD, Massachusetts General<br />

Hospital, Boston<br />

Faculty: Story C. Landis, PhD, NINDS, Bethesda, Md.<br />

Stephen Korn, PhD, NINDS, Bethesda, Md.<br />

Walter J. Koroshetz, MD, NINDS, Bethesda, Md.<br />

Being Heard Clearly: A Follow-up Discussion<br />

Harvard (3rd Floor)<br />

Continue the conversation on using the power of voice and body language<br />

to get your message heard with Susan Miller following her Mid/Senior Level<br />

Faculty Development Course talk.<br />

Faculty: Susan Miller, PhD, Voicetrainer LLC, Washington, DC<br />

Meet the Chairs<br />

Dartmouth (3rd Floor)<br />

Prominent Chairs of neurology will discuss how they have handled their<br />

position, what’s involved with being a Chair, what is the process for attaining<br />

their position, how to interact with Chairs, etc.<br />

8


1:15 – 3:15 pm<br />

Salon E (4th Floor)<br />

SYMPOSIUM: Imaging to Explore<br />

Neural Network Structure and Function<br />

Chair: Kirk A. Frey, MD, PhD, University of Michigan, Ann Arbor<br />

Co-Chair: James B. Brewer, MD, PhD, University of California, San Diego<br />

This symposium will discuss ways in which the structure and function<br />

of the network of biological neurons or neural networks can be explored<br />

using different imaging practices. Much research is currently centered<br />

on detecting pre-disease or molecular states that occur before typical<br />

symptoms of a disease are detected using molecular imaging. Due to its<br />

increasing popularity, a large number of investigators are also collecting<br />

imaging data from healthy and clinical subjects during rest. Diffusion<br />

tensor imaging (DTI) uses MR imaging to map the brain’s white matter<br />

tracts and perform fiber-tracking. By the end of this symposium, attendees<br />

will have a broad understanding of how molecular imaging, resting state<br />

connectivity and diffusion tensor imaging (DTI) can be used to explore<br />

neural network structure and function.<br />

Learning Objectives: Having completed this symposium, participants<br />

will be able to:<br />

1. Understand the basic underpinnings of the BOLD signal and what it<br />

means in functional MRI.<br />

2. Understand the application of MR-based resting state functional<br />

connectivity and how this can be applied to investigate the<br />

pathophysiology of neurologic disorders.<br />

3. Understand how MR-based diffusion weighted imaging can be used<br />

to measure regional diffusivity and reveal neural connections via tract<br />

tracing methods.<br />

4. Understand the ability of molecular imaging approaches to investigate<br />

brain pathways, the importance of validation of these methods and the<br />

integration of these methods with MR-based methods.<br />

1:15 – 1:20 pm<br />

Distinguished Neurology Teacher Award Presentation<br />

Carl E. Stafstrom, MD, PhD, University of Wisconsin, Madison<br />

1:20 – 1:45 pm<br />

Decoding the BOLD Signal in Functional MR Imaging<br />

Anna Devor, PhD, University of California, San Diego<br />

1:45 – 2:10 pm<br />

Resting State Connectivity<br />

Michael D. Greicius, MD, MPH, Stanford University, Stanford, Calif.<br />

2:10 – 2:35 pm<br />

Diffusion Tensor Imaging (DTI)<br />

Lawrence L. Wald, PhD, Massachusetts General Hospital, Boston<br />

2:35 – 3:00 pm<br />

Molecular Imaging<br />

Joel S. Perlmutter, MD, Washington University, St. Louis, Mo<br />

3:00 – 3:15 pm<br />

Question and Answer<br />

3:30 – 5:30 pm<br />

3:15 – 3:45 pm<br />

Coffee Break<br />

Special Interest Group Symposia (SIG)<br />

Please note the color of your SIG’s signage, as they have been color<br />

coded to match the posters related to the SIG topic.<br />

Cerebrovascular Disease<br />

Salon F (4th Floor)<br />

Co-Chairs: Argye E. Hillis, MD, MA, Johns Hopkins University,<br />

Baltimore<br />

E. Steve Roach, MD, Ohio State University, Columbus<br />

Leaders in the Field Presentations:<br />

Early to Middle Phase Trial Designs in Acute Stroke:<br />

One Size Does Not Fit All<br />

Elliott Clarke Haley, MD, University of Virginia, Charlottesville<br />

Wake Up, Little Susie: Extending the Window of IV Thrombolysis<br />

Lee H. Schwamm, MD, FAHA, Massachusetts General Hospital, Boston<br />

Populations at Risk: Stroke and Diabetes<br />

Kerstin Bettermann, MD, PhD, Penn State University, Hershey, Pa.<br />

Applying the Lessons from 20 Years of Sickle Cell<br />

Stroke Research<br />

E. Steve Roach, MD, Ohio State University, Columbus<br />

Data Blitz Presentations:<br />

Lipid Measurements and Risk of Ischemic Vascular Events:<br />

Framingham Study<br />

Aleksandra Pikula, MD, Boston University, Boston<br />

Genetic Risk in Cerebral Small Vessel Disease (SVD):<br />

17q25 Locus Associates with White Matter Lesions but<br />

Not Lacunar Stroke<br />

Poneh Adib-Samii, MBBS, St. George’s University, London, UK<br />

Quality of Life after Lacunar Stroke: The Secondary<br />

Prevention of Small Subcortical Strokes (SPS3)<br />

Mandip Dhamoon, MD, MPH, Mount Sinai School of Medicine,<br />

New York<br />

Q&A/Wrap Up/Adjourn<br />

9


SUNDAY, OCTOBER 7<br />

Sleep Disorders and Circadian Rhythm<br />

Salon CD (4th Floor)<br />

Co-Chairs: Clifford B. Saper, MD, PhD, Beth Israel Deaconess<br />

Medical Center, Boston<br />

Phyllis C. Zee, MD, PhD, Northwestern University, Chicago<br />

Leader in the Field Presentation:<br />

Molecular Dissection of Narcolepsy Signs and Symptoms<br />

Thomas Scammell, MD, Harvard Medical School, Beth Israel<br />

Deaconess Medical Center, Boston<br />

Data Blitz Presentations:<br />

Sleep and Circadian Rhythm Disruption in Parkinson’s Disease<br />

David Breen, MD, University of Cambridge, Cambridge, UK<br />

Rest-Activity Fragmentation and Risk of Alzheimer’s Disease<br />

Andrew Lim, MD, University of Toronto, Canada<br />

TSC-mTOR Pathway and Circadian Rhythms<br />

Jonathan Lipton, MD, PhD, Children’s Hospital, Boston<br />

Medical and Psychiatric Conditions Associated with Narcolepsy<br />

Maurice Ohayon, MD, DSc, PhD, Stanford University, Palo Alto, Calif.<br />

REM Sleep without Atonia and Freezing Gait in<br />

Parkinson’s Disease<br />

Aleksandar Videnovic, MD, MSc, Northwestern University, Chicago<br />

Sleep Research Society Reception<br />

Education: The ACGME Milestones Project:<br />

The Next Step in Program Accreditation<br />

Sponsored by the AUPN<br />

Provincetown (4th Floor)<br />

Moderator and Chair: Ralph F Józefowicz, MD, University of<br />

Rochester, N.Y.<br />

Co-Moderator: Steven L. Lewis, MD, Rush University, Chicago<br />

The Neurology Milestones: What They Are and What<br />

They Are Not<br />

Steven L. Lewis, MD, Rush University, Chicago<br />

The Program Director’s Perspective: The Devil is in the Details<br />

Ralph F Józefowicz, MD, University of Rochester, N.Y.<br />

The Chair’s Perspective: Getting Faculty on Board<br />

David Lee Gordon, MD, University of Oklahoma, Oklahoma City<br />

The Resident’s Perspective: Views from the Trenches<br />

Sarah Wahlster, MD, Harvard University, Boston<br />

The ABPN Perspective: Strengthening the Credentialing<br />

Process for Board Certification<br />

Larry Faulkner, MD, <strong>American</strong> Board of Psychiatry and Neurology, Chicago<br />

The International Perspective: Credentialing Neurologists<br />

in the UK<br />

Geraint Fuller, MD, <strong>Association</strong> of British Neurologists, London, UK<br />

Panel – Audience Discussion<br />

Neuro-oncology<br />

Salon B (4th Floor)<br />

Co-Chairs: Jeremy N. Rich, MD, Cleveland Clinic, Cleveland, Oh.<br />

Benjamin W. Purow, MD, University of Virginia, Charlottesville<br />

Leaders in the Field Presentations:<br />

Targeted Molecular Therapies for GBM; Lessons Learned<br />

and Future Directions<br />

Patrick Wen, MD, Harvard Medical School, Brigham and Women’s<br />

Hospital, Boston<br />

An Update on the Benefits and Limitations of Anti-Angiogenic<br />

Therapy for Glioblastoma<br />

David Reardon, MD, Dana-Farber Cancer Institute, Boston<br />

The Search for Predictive Markers for Glioblastoma Response<br />

Versus Resistance to Anti-Angiogenic Therapies<br />

Tracy Batchelor, MD, Dana-Farber Cancer Institute, Boston<br />

Current Challenges in Primary CNS Lymphoma<br />

Lisa DeAngelis, MD, Memorial Sloan-Kettering Cancer Center, New York<br />

Data Blitz Presentations:<br />

Genetic Modifiers Affecting Neurofibromatosis (GMAN):<br />

Cutaneous Tumor Burden in Neurofibromatosis Type 1<br />

Fawn Leigh, MD, Massachusetts General Hospital, Boston<br />

Bevacizumab for NF2-related Vestibular Schwannoma<br />

Scott Plotkin, MD, PhD, Massachusetts General Hospital, Boston<br />

Leveraging Expression of the GABA-A Receptor, alpha 5 in<br />

Medulloblastoma as a Novel Therapeutic Target<br />

Soma Sengupta, MD, Children’s Hospital, Boston<br />

Cerebrospinal Fluid and MRI Analysis in Leptomeningeal<br />

Carcinomatosis<br />

Joachim Baehring, MD, DSc, Yale School of Medicine, New Haven, Conn.<br />

Ataxia, Ophthalmoplegia and Areflexia: What Would You Think<br />

Nazia Karsan, MBBS, BSc, MRCP, St. George’s Hospital, London, UK<br />

Question and Answer<br />

10


Health Services Research — NEW!<br />

Salon A (4th Floor)<br />

Chair: Barbara G. Vickrey, MD, MPH, University of California, Los Angeles<br />

Leaders in the Field Presentations:<br />

Community Partnered Research to Improve Outcomes &<br />

Eliminate Disparities in <strong>Neurological</strong> Care<br />

Barbara G. Vickrey, MD, MPH, University of California, Los Angeles<br />

Can Corporate America Solve Health Disparities<br />

Lewis B. Morgenstern, MD, University of Michigan, Ann Arbor<br />

Data Blitz Presentations:<br />

What Patient Factors Associate with Inaccuracies in Reporting<br />

of Parkinsonian Signs<br />

Nabila Dahodwala, MD, MS, University of Pennsylvania, Philadelphia<br />

The <strong>Association</strong> of Non-Clinical Factors with Head CT<br />

Use in Emergency Department Dizziness Visits:<br />

A Population-Based Study<br />

Kevin Kerber, MD, University of Michigan, Ann Arbor<br />

5:30 – 7:00 pm<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

Poster Stand-by Wine & Cheese Reception<br />

Poster Categories<br />

Cerebrovascular Disease<br />

Education<br />

Neuro-oncology<br />

Sleep Disorders and Circadian Rhythm<br />

Neurogenetics<br />

Neuroinfectious Disease<br />

Neuro-ophthalmology<br />

Pediatric Neurology<br />

Rehabilitation and Regeneration<br />

Trauma/Injury<br />

HEALS (Healthy Eating And Lifestyle after Stroke): A Pilot Trial<br />

of a Multidisciplinary Lifestyle Intervention Program<br />

Amytis Towfighi, MD, University of Southern California and Rancho Los<br />

Amigos National Rehabilitation Center, Los Angeles<br />

Racial and Ethnic Differences in Post-Stroke Depression among<br />

Community Dwelling Adults<br />

Lesli Skolarus, MD, University of Michigan, Ann Arbor<br />

Improving Stroke Symptom Recognition and Response in<br />

Elderly Korean-<strong>American</strong>s<br />

Sarah Song, MD, MPH, Rush University, Chicago<br />

Wrap-up/Summary/Implications for Neurology<br />

11


MONDAY, OCTOBER 8<br />

7:00 – 9:00 am<br />

Salon F (4th Floor)<br />

6:30 am – 5:45 pm<br />

Registration<br />

4th Floor Registration Area<br />

6:30 – 7:30 am<br />

Coffee and Rolls<br />

Coffee will be available until 10:30 am<br />

Junior Faculty Development Course: Establishing<br />

Yourself in the World of Academic Neurology<br />

Session II: Where Does All the Money Go<br />

Chair: Daniel H. Lowenstein, MD, University of California, San Francisco<br />

Clifford B. Saper, MD, PhD, Beth Israel Deaconess Medical Center, Boston<br />

Justin C. McArthur, MBBS, MPH, FAAN, Johns Hopkins University,<br />

Baltimore, Md.<br />

This course will provide an overview of financial management as it affects<br />

a typical faculty member within an academically oriented neurology<br />

department. Specific topics will include: funds flow, clinical revenues and<br />

expenses, construction and implementation of compensation and bonus<br />

plans, philanthropy, and research budgets, indirect costs of research, and<br />

grants management. Case studies (examples) will be used to illustrate best<br />

practices, and lessons learned.<br />

7:00 – 9:00 am<br />

Salon CD (4th Floor)<br />

Mid/Senior Level Faculty Development Course:<br />

Negotiations & Conflict Resolution<br />

Session II: Negotiations<br />

Chair: Lisa M. DeAngelis, MD, Memorial Sloan-Kettering Cancer Center,<br />

New York<br />

Career Challenges for Mid/Senior Level Physicians<br />

Ranna I. Parekh, MD, MPH, Massachusetts General Hospital and McLean<br />

Hospital, Boston<br />

This course is designed for mid-senior level faculty interested in<br />

collaborative negotiations. For most successful people at this point in<br />

their careers, they are already skilled at negotiations. Hence, one hope of<br />

this course is to build confidence that many us already employ important<br />

negotiations strategies and to perhaps explain some of the theories why<br />

they have been effective.<br />

At the core of collaborative negotiations is achieving the best outcome for<br />

oneself and others while building trusting relationships.<br />

There are 5 major objectives for participants:<br />

1. Describe major work/life situations and the appropriate negotiation<br />

strategies to employ<br />

2. Define interest based or win:win or Collaborative Negotiations.<br />

3. Understand the five stages of Collaborative Negotiations and how each<br />

stage predicts effective outcomes and longterm partnerships<br />

4. Guide participants through the most challenging stage of negotiations:<br />

the brainstorming or creative problem stage<br />

5. Learn to build trust and rapport during the negotiations process.<br />

Throughout the course, there will be many work and interpersonal examples<br />

and the audience will be encouraged to bring their own negotiation “cases”<br />

for small group discussion.<br />

7:00 – 9:00 am<br />

Salon AB (4th Floor)<br />

AUPN: Neurology Chair Development Course<br />

Session II: Avoiding Senior Faculty and Chair Burnout<br />

Steven T. DeKosky, MD, University of Virginia, Charlottesville<br />

Sharon L. Hostler, MD, University of Virginia, Charlottesville<br />

Chairs of Departments of Neurology and senior faculty who have been<br />

working for several decades are at risk of fatigue and burnout with the<br />

changes in medical care, pressures regarding NIH grants, and requirements<br />

for increased productivity. In contrast to most departments’ experience<br />

with orienting and mentoring young faculty, efforts to provide development<br />

and resilience in older faculty are much less, and the faculty themselves<br />

are reluctant to “ask for help” to appear needy, inadequate, or be seen<br />

as no longer being independent, “triple threats,” or able to “do it all.”<br />

In this session we will discuss the phenomenon as well as programs and<br />

techniques to identify and characterize burnout, and methods to prevent<br />

or combat it.<br />

This session is not available for AMA PRA Category 1 Credit.<br />

9:00 – 11:30 am<br />

Salon E (4th Floor)<br />

President’s Symposium: Alzheimer’s Disease:<br />

New Perspectives on an Old Disease<br />

Chair: Alastair Compston, MBBS, PhD, FmedSci, University of<br />

Cambridge, UK<br />

Co-Chair: Karen Ashe, MD, PhD, University of Minnesota, Minneapolis<br />

The symposium will highlight development in the genetics of Alzheimer’s<br />

disease, the identification of biomarkers for the molecular neuropathology<br />

and its serial impact on regional brain structure and function, and emerging<br />

concepts on the initiation and evolution of the disease processes.<br />

12


Steady progress has been made in identifying Mendelian genes for familial<br />

Alzheimer’s disease but less is known about risk factors for sporadic<br />

disease although genome wide association studies have now prioritized<br />

several possible new genes implicating novel disease pathways. Improved<br />

understanding of the genetic basis for Alzheimer’s disease is expected to<br />

expand our understanding of mechanisms involved early in the disease<br />

process; and to provide guidelines for cost-effective genetic screening in<br />

clinical practice.<br />

The current focus is on early detection of Alzheimer’s disease through<br />

the recognition of mild cognitive impairment; but there is a need to<br />

identify individuals without symptoms in whom the disease process of<br />

Alzheimer’s disease is nevertheless present, at a stage when brain structure<br />

and function are minimally impaired and interventions are likely to be<br />

most effective.<br />

This pre-supposes that work on the mechanisms of tissue injury will<br />

progress beyond understanding the accumulation of plaques and tangles<br />

to provide an account of propagation and dissemination of the molecular<br />

pathology, or its multiple sites of origin so that mechanism-based<br />

therapeutic strategies are used to improve on the existing limited efficacy<br />

of Aricept, memantine and vitamin E.<br />

Learning Objectives: Having completed this symposium, participants<br />

will be able to:<br />

1. Compare the clinical features and mechanisms of familial and sporadic<br />

Alzheimer’s disease.<br />

2. Understand the mechanisms whereby the risk allele for ApoE confers<br />

susceptibility to Alzheimer’s disease.<br />

3. Explore current ideas on the extent to which knowledge of prion disease<br />

informs ideas on the possibility of transmission and propagation of the<br />

molecular pathology of Alzheimer’s disease.<br />

4. Provide insights into early disease mechanisms from detecting<br />

biomarkers for the neuropathology in blood and cerebrospinal fluid.<br />

5. Provide insights into disease mechanisms from correlating alteration<br />

in the structure and function of brain regions early and throughout the<br />

course of Alzheimer’s disease.<br />

9:00 – 9:05 am<br />

Welcome from the President; Presentation to the <strong>Association</strong> of<br />

British Neurologists<br />

Eva L. Feldman, MD, PhD, FAAN, ANA President, University of Michigan<br />

9:05 – 9:25 am<br />

Lessons for Alzheimer’s Disease from Early Detection of<br />

Familial Cases<br />

Martin N. Rossor, MD, Institute of Neurology, University College<br />

London, UK<br />

10:00 – 10:15 am<br />

Coffee Break<br />

10:15 – 10:40 am<br />

Mechanisms of Neurodegeneration: Lessons from Prion Disease<br />

John Collinge, MD, Institute of Neurology, University College London, UK<br />

10:40 – 11:05 am<br />

Biomarkers for the Diagnosis and Course of Alzheimer’s Disease<br />

Randall J. Bateman, MD, Washington University, St. Louis, Mo.<br />

11:05 – 11:30 am<br />

Imaging Structure and Function in Alzheimer’s Disease<br />

William J. Jagust, MD, University of California, Berkeley<br />

11:30 am – 6:30 pm<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

All Day Poster Presentations<br />

Poster Stand-by time will be from 5:30 – 6:30 pm<br />

11:45 am – 1:00 pm<br />

Salon F (4th Floor)<br />

11:30 am – 1:00 pm<br />

Lunch<br />

Pick up your lunch in the 4th Floor Foyer.<br />

12th <strong>Annual</strong> Women of the ANA Lunch Program:<br />

34 Years in Academic Neurology<br />

Co-Chairs: Kathleen B. Digre, MD, University of Utah, Salt Lake City<br />

Shirley H. Wray, MD, PhD, Massachusetts General Hospital, Boston<br />

Faculty: Anne B. Young, MD, PhD, Massachusetts General<br />

Hospital, Boston<br />

Dr. Young will reflect on her life, including her scientific contributions and<br />

years leading a neurology department. She will share advice for others<br />

following a similar path.<br />

The Women of the ANA program is tailored to address the concerns and<br />

view of women. Lunch will be provided.<br />

9:25 – 10:00 am<br />

Presentation of Raymond D. Adams Lectureship Award and<br />

Neurobiology of APOE and its Impact on Alzheimer’s Disease<br />

David M. Holtzman, MD, Washington University, St. Louis, Mo.<br />

13


MONDAY, OCTOBER 8<br />

11:45 am – 1:00 pm<br />

Interactive Lunch Workshops:<br />

Common <strong>Neurological</strong> Issues<br />

Monday’s Interactive Lunch Workshops are designed to offer information and<br />

informal discussions on key issues facing academic neurology. Because of<br />

the informal format, attendees are encouraged to move between workshops if<br />

they wish.<br />

Maintenance of Certification (MOC) for Board-Certified<br />

Neurologists<br />

Berkeley (3rd Floor)<br />

This session will provide an overview of the ABPN MOC Program.<br />

The background and rationale for MOC will be discussed. The specific<br />

requirements for the four parts of MOC will be presented and the new<br />

Continuous MOC Program will be described. Participants will learn how<br />

to establish their own ABPN folios for identifying their personalized MOC<br />

requirements and for recording their progress in MOC.<br />

Moderator: Ralph F. Józefowicz, MD, University of Rochester,<br />

Rochester, N.Y.<br />

Faculty: Janice Massey, MD, Duke University, Durham, N. Car.<br />

Larry R. Faulkner, MD, Buffalo Grove, Ill.<br />

Pharma/Clinical Trials<br />

Fairfield/Exeter (3rd Floor)<br />

Despite intensive efforts, success in clinical trials for neurodegenerative<br />

disorders has been elusive. Panelists will discuss the challenges, progress,<br />

and promising new approaches to clinical development in Alzheimer’s<br />

disease, Parkinson’s disease, and ALS.<br />

This session is not available for AMA PRA Category 1 Credit.<br />

Moderator: Volney Sheen, MD, PhD, Beth Israel Deaconess<br />

Medical Center, Boston<br />

Faculty: Eric Siemers, MD, Eli Lilly and Co., Indianapolis, Ind.<br />

Genevieve A. Laforet, MD, PhD, Biogen Idec, Boston<br />

Bernard Ravina, MD, Biogen Idec, Cambridge, Mass.<br />

Contemporary Clinical Issues in Neuromuscular Disease:<br />

Role of Genetic Testing<br />

Regis (3rd Floor)<br />

This symposium will include a discussion of the utility of genetic testing in<br />

the evaluation of suspected inherited peripheral neuropathies and muscle<br />

disorders. The discussants will present an overview of indications for genetic<br />

testing coupled with testing strategies, against a backdrop of the need to<br />

provide cost effective health care. Controversies and the uses and abuses<br />

of genetic testing will be covered. Advances in genetic techniques that may<br />

change the landscape and approach to genetic diagnosis in neuromuscular<br />

disorders will be addressed. Brief presentations by Dr. David Herrmann<br />

(genetic testing in suspected inherited neuropathies) and Dr. Anthony Amato<br />

(genetic testing in muscle disorders) will be followed by an interactive<br />

discussion encouraging audience participation.<br />

Moderator: Juliann M. Paolicchi, MD, Weill Cornell Medical Center,<br />

New York<br />

Faculty: David N. Herrmann, MBBCh, University of Rochester, New York<br />

Anthony A. Amato, MD, Brigham and Women’s Hospital, Boston<br />

Headache & Pain Role of Imaging<br />

Suffolk (3rd Floor)<br />

What is the role of neuroimaging and funcitional MRI in migraine What does<br />

it tell us about migriane chronification, drug effects, and the effect of gender<br />

on migraine When is neuroimaging absolutely required Which modality<br />

should be used<br />

Faculty: Stephen D. Silberstein, MD, Thomas Jefferson University,<br />

Philadelphia<br />

David Borsook, PhD, MBBCh, Children’s Hospital Boston<br />

Concussion/Trauma/TBI<br />

Simmons (3rd Floor)<br />

Traumatic brain injury (TBI) is associated with a variety of pathophysiologic<br />

events and clinical consequences that are typically labeled on a continuum<br />

ranging from mild to severe. The preponderance of cases of TBI is on the mild<br />

side of the continuum and mild TBI has been receiving increasing attention<br />

in part because of a recent focus on sports concussion and blast injury.<br />

The other extreme on the continuum, those with very severe TBI causing<br />

prolonged disorders of consciousness (DOC), has also received increasing<br />

attention. Interest in this group of patients is largely related to concerns about<br />

distinguishing patients who are unconscious, in a vegetative state (VS), from<br />

those who have small, inconsistent signs of consciousness, recently defined<br />

as the minimally conscious state (MCS). It has also been a fertile area of<br />

investigation because it contributes to our understanding of one of the key<br />

questions in neuroscience, the nature of consciousness. In the last decade,<br />

investigations of patients with DOC after severe TBI have probed differences in<br />

brain network activity using fMRI and other physiologic modalities in patients<br />

in a VS, MCS and higher levels of consciousness, have demonstrated better<br />

than expected outcomes in patients with prolonged DOC, and have established<br />

potentially effective treatments such as deep brain stimulation of the thalamus<br />

to improve purposeful behavior and pharmacological agents (i.e., amantadine)<br />

to speed the pace of recovery. This workshop will include discussions of the<br />

problems of DOC after severe TBI and recent studies on assessment, outcome<br />

and treatment.<br />

Moderator: Thomas P. Bleck, MD, Rush Medical College, Chicago<br />

Faculty: Ann C. McKee, MD, Boston University, Boston<br />

Doug I. Katz, MD, Boston University, Boston<br />

14


NeuroNEXT<br />

Clarendon (3rd Floor)<br />

The Network for Excellence in Neuroscience Clinical Trials, or<br />

NeuroNEXT, was created by NINDS to conduct studies of treatments<br />

for neurological diseases through partnerships with academia, private<br />

foundations, and industry. This session will feature the NeuroNEXT<br />

Scientific Program Director and the Clinical and Data Coordinating<br />

Center’s Principal Investigator.<br />

Moderator: Anne Louise Oaklander, MD, PhD, Massachusetts General<br />

Hospital, Boston<br />

Faculty: Elizabeth McNeil, MD, MSc, NINDS, Bethesda, Md.<br />

Merit E. Cudkowicz, MD, Massachusetts General Hospital, Boston<br />

Christopher Coffey, PhD, University of Iowa, Iowa City<br />

Career Challenges: A Follow-up Discussion<br />

Dartmouth (3rd Floor)<br />

Continue the conversation on the principles of negotiation with Rana Parekh<br />

following her Mid/Senior Level Faculty Development Course talk.<br />

Faculty: Ranna I. Parekh, MD, MPH, Massachusetts General Hospital,<br />

Boston<br />

1:15 – 3:30 pm<br />

Salon E (4th Floor)<br />

SYMPOSIUM: Results of Immune-Based Trials in<br />

<strong>Neurological</strong> Disorders<br />

Chair: Edward H. Koo, MD, University of California, San Diego<br />

Co-Chair: Kevin Talbot, DPhil, FRCP, University of Oxford, UK<br />

This symposium will discuss the knowledge of theoretical and mechanistic<br />

basis for Alzheimer ’s disease. Attendees will learn about the science of<br />

immunotherapy in the context of neurological disorders. Lastly, attendees<br />

will gain a new awareness of current trials, designs and findings so that<br />

they are updated on the most recent scientific discoveries surrounding the<br />

identified disorders. The program will include talks on the pathogenetic<br />

context for immunotherapy, a Phase III Study of Solaneuzumab for<br />

Alzheimer’s disease, two Phase III Study Results of Bapineuzumab for<br />

Alzheimer’s Disease, and a Phase II Study of Immunotherapy in Multiple<br />

Sclerosis followed by a panel discussion with audience participation.<br />

1:15 – 1:25 pm<br />

The Pathogenetic Context for Immunotherapy &<br />

ACCME Overview<br />

Edward H. Koo, MD, University of California, San Diego<br />

1:25 – 1:50 pm<br />

Phase 3 Studies of Solanezumab for Mild to Moderate<br />

Alzheimer’s Disease<br />

Rachelle S. Doody, MD, PhD, Baylor College of Medicine, Houston<br />

1:50 – 2:15 pm<br />

Phase III Studies of Bapineuzumab for Mild to<br />

Moderate Alzheimer’s Disease Dementia<br />

Reisa A. Sperling, MD, Brigham and Women’s Hospital, Boston<br />

2:15 – 2:40 pm<br />

Panel Discussion of Immunotherapeutic Trials in<br />

Alzheimer Disease<br />

2:40 – 3:10 pm<br />

Soriano Lectureship: Phase III Trials of Alemtuzumab in<br />

Relapsing-Remitting Multiple Sclerosis<br />

Alastair Compston, MBBS, PhD, FmedSci, University of Cambridge, UK<br />

3:10 – 3:20 pm<br />

Panel Discussion MS<br />

Panelists: Carole Ho, MD, Genentech, San Francisco<br />

Susanne Ostrowitzki, MD, F. Hoffmann-La Roche, Ltd., Basel, Switzerland<br />

Norman R. Relkin MD, PhD, Weill Cornell Medical College, New York<br />

John C. Morris MD, Washington University, St. Louis, Mo.<br />

Jody Corey-Bloom, MD, PhD, University of California, San Diego<br />

Robert O. Messing, MD, University of California, San Francisco<br />

Stephen Salloway, MD, MS, Brown University, Providence, R.I.<br />

3:20 – 3:30 pm<br />

General Discussion of Symposium<br />

3:15 – 3:45 pm<br />

Coffee Break<br />

Learning Objectives: Having completed this symposium, participants<br />

will be able to:<br />

1. Understand the biological basis for immunotherapy use in neurological<br />

disorders.<br />

2. Convey most recent findings on this trial: A Phase III Study of<br />

Solaneuzumab for Alzheimer disease.<br />

3. Convey most recent finding on this trial: A Phase III Study of<br />

Immunotherapy in Multiple Sclerosis.<br />

4. Convey most recent finding on this trial: A Phase III Study of<br />

Bapineuzumab.<br />

15


MONDAY, OCTOBER 8<br />

3:30 – 5:30 pm<br />

Special Interest Group Symposia (SIG)<br />

Please note the color of your SIG’s signage, as they<br />

have been color coded to match the posters related to the SIG topic.<br />

Dementia and Aging<br />

Salon F (4th Floor)<br />

Chair: Richard P. Mayeux, MD, MSc, Columbia University, New York<br />

Leaders in the Field Presentations:<br />

Parkinson’s Disease with Dementia and Lewy<br />

Body Dementia: Genetics and Biomarkers<br />

Karen Marder, MD, MPH, Columbia University, New York<br />

Alzheimer’s Disease: Implications from GWAS<br />

Christiane Reitz, MD, PhD, Columbia University, New York<br />

Data Blitz Presentations:<br />

Gephyrin Plaques Identified in Frontal Cortex of Alzheimer’s<br />

Disease Brains<br />

Chadwick Hales, MD, PhD, Emory University, Atlanta<br />

Relationship between Beta-Amyloid Retention and Ischemia in<br />

the Patients with Subcortical Vascular Cognitive Impairment<br />

Young Noh, MD, Sungkyunkwan University School of Medicine,<br />

Samsung Medical Center, Seoul, Republic of Korea<br />

Brain Imaging and Cognitive Predictors of Incident Stroke,<br />

Dementia and Alzheimer’s Disease<br />

Galit Weinstein, PhD, Boston University, Framingham, Mass.<br />

Genetic Susceptibility for Amyloid Pathology in<br />

Alzheimer’s Disease<br />

Joshua Shulman, MD, PhD, Baylor College of Medicine, Houston<br />

Retinal Degeneration in FTLD Patients and PGRN-Deficient<br />

Mice Preceded by TDP-43 Mislocalization<br />

Ari Green, MD, MCR, University of California, San Francisco<br />

Question & Answer/Discussion<br />

Neurocritical Care — NEW!<br />

Salon D (4th Floor)<br />

Chair: Thomas P. Bleck, MD, Rush University, Chicago<br />

Leaders in the Field Presentations:<br />

Refractory Status Epilepticus<br />

Thomas Bleck, MD, Rush University University, Chicago<br />

Intracerebral Hemorrhage<br />

Jonathan Rosand, MD, MSc, Massachusetts General Hospital Center for<br />

Human Genetic Research, Boston<br />

Infectious Causes of Encephalitis<br />

Alan Aksamit, Jr., MD, Mayo Clinic, Rochester, Minn.<br />

Data Blitz Presentations:<br />

Fatal Hyperammonemic Brain Injury from Valproic Acid Exposure<br />

Danny Bega, MD, Brigham and Women’s Hospital, Boston<br />

Rapid High Volume CSF Loss – A New Cause of Coma<br />

Minjee Kim, MD, Brigham and Women’s Hospital, Boston<br />

A Population-Based Study of Aetiology and Outcome in<br />

Acute Neuromuscular Respiratory Failure<br />

Aisling Carr, MD, PhD, Royal Victoria Hospital, Belfast, Northern Ireland, UK<br />

Risk Factors for Intracranial Haemorrhage in Acute<br />

Ischaemic Stroke Patients Treated with rtPA<br />

Peter Fernandes, MRCP, University of Edinburgh, UK<br />

Incidence and Impact of Medical and <strong>Neurological</strong> ICU<br />

Complications on Moderate-Severe Traumatic Brain Injury<br />

Susanne Muehlschlegel, MD, MPH, University of Massachusetts Medical<br />

School, Worcester<br />

Question & Answer/Discussion<br />

Epilepsy<br />

Provincetown (4th Floor)<br />

Co-Chairs: Kevin J. Staley, MD, Massachusetts General Hospital, Boston<br />

Sydney S. Cash, MD, PhD, Massachusetts General Hospital, Boston<br />

Leaders in the Field Presentations:<br />

Using Dravet Syndrome Patient-Derived Cells to Study<br />

Epilepsy and SUDEP Mechanisms<br />

Jack Parent, MD, University of Michigan, Ann Arbor<br />

Optogenetic Treatment of Focal Epilepsy<br />

Laura Mantoan Ritter, MD, Institute of Neurology, University College<br />

London, UK<br />

16


Identification of the Gene for Paroxysmal Kinesigenic<br />

Dyskinesia with Infantile Convulsions<br />

Louis Ptacek, MD, University of California, San Francisco<br />

Ketamine Use in Refractory Status Epilepticus<br />

Andrea Synowiec, DO, Allegheny General Hospital, Pittsburgh, Pa.<br />

EEG Based Functional Networks: A Robust Biomarker<br />

Across Time<br />

Catherine Chu, MD, Massachusetts General Hospital, Boston<br />

Monogenic and “Polygenic” Epilepsy Mutations Alter<br />

GABAA Receptor Biogenesis and Function<br />

Robert Macdonald, MD, PhD, Vanderbilt University, Nashville, Tenn.<br />

Neuromuscular Disease<br />

Salon BC (4th Floor)<br />

Chair: Jon Ravits, MD, FAAN, University of California, San Diego<br />

Co-chair: Tahseen Mozaffar, MD FAAN, University of California, Irvine<br />

Leaders in the Field Presentations:<br />

Updates on ALS<br />

Pamela Shaw, MBBS, MD, FRCP, University of Sheffield, UK<br />

Using Antisense Oligonucleotides in Neuromuscular Disease<br />

Charles Thornton, MD, University of Rochester, N.Y.<br />

Updates on Neuropathies<br />

Jun Li, MD, PhD, Vanderbilt University, Nashville, Tenn.<br />

Data Blitz Presentations:<br />

Infections in Myasthenia Gravis<br />

Raghav Govindarajan, MD, Cleveland Clinic, Weston, Fla.<br />

The Survival Motor Neuron (SMN) Gene as a Therapeutic Target<br />

in Amyotrophic Lateral Sclerosis<br />

Kevin Talbot, MBBS, DPhil, University of Oxford, UK<br />

Case Studies — NEW!<br />

Salon A (4th Floor)<br />

Two Weeks on the Consult Service: An Experience in<br />

Medical Neurology<br />

Chair/Presenter: Martin A. Samuels, MD, DSc(hon), FAAN, MACP, FRCP,<br />

Brigham and Women’s Hospital, Boston<br />

An important aspect of neurology involves the neurological aspects of<br />

general medical diseases. This session will review some real recent cases<br />

from two weeks on the consult service in July <strong>2012</strong>. Selected aspects of<br />

the interface between neurology and general medicine will be addressed.<br />

Audience participation will be encouraged.<br />

5:30 – 6:30 pm<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

Poster Stand-by Wine & Cheese Reception<br />

Poster Categories:<br />

Dementia and Aging<br />

Epilepsy<br />

Neurocritical Care<br />

Neuromuscular Disease<br />

6:30 – 8:30 pm<br />

President’s Reception at the Boston Public Library<br />

Boston Public Library | Central Library | McKim Building<br />

700 Boylston St., Boston, MA 02116<br />

See page 23 for details and directions.<br />

Blockade of Matrix Metalloproteinase-3 after Traumatic Nerve<br />

Injury Preserves the Motor End Plate<br />

Ranjan Gupta, MD, University of California, Irvine<br />

A Multicenter Phase II Open-Label Trial of Valproic Acid and<br />

L-Carnitine in Infants with SMA Type I<br />

Kathryn Swoboda, MD, University of Utah, Salt Lake City<br />

Peripheral Nerve Function Following Treatment with Tanezumab<br />

Mark Brown, Pfizer, Inc., Groton, Conn.<br />

Question and Answer/Discussion<br />

17


TUESDAY, OCTOBER 9<br />

7:00 – 9:00 am<br />

Salon F (4th Floor)<br />

6:30 am – 5:45 pm<br />

Registration<br />

4th Floor Registration Area<br />

6:30 – 7:30 am<br />

Coffee and Rolls<br />

Coffee will be available until 10:30 am<br />

Junior Faculty Development Course: Establishing<br />

Yourself in the World of Academic Neurology<br />

Session III: Getting Published<br />

Chair: Daniel H. Lowenstein, MD, University of California, San Francisco<br />

Getting Published and Increasing Your Visibility<br />

Stephen Hauser, MD, University of California, San Francisco,<br />

Alastair Compston, MBBS, PhD, FmedSci, University of Cambridge, UK<br />

Conveying your ideas and discoveries in writing, and getting these out<br />

in the public domain so they can be scrutinized and valued as important<br />

contributions to the field, constitute yet another essential skillset in<br />

academics. This session, led by two of the most eminent editors in neurology<br />

today, will focus on how to be successful in getting papers published.<br />

The discussion will include a review of the essential characteristics of<br />

outstanding papers, as well as an inside look into the review process,<br />

including the initial “in-house” assessment of manuscripts, the selection of<br />

reviewers, the editors’ consideration of the reviews, and best approaches for<br />

addressing editorial decisions on your manuscript.<br />

7:00 – 9:00 am<br />

Salon CD (4th Floor)<br />

Mid/Senior Level Faculty Development Course:<br />

Negotiations & Conflict Resolution<br />

Session III: Managing Conflict: How to Talk About the Tough Stuff<br />

Chair: Lisa M. DeAngelis, MD, Memorial Sloan-Kettering Cancer Center,<br />

New York<br />

Catherine J. Morrison, JD, Johns Hopkins Carey Business School,<br />

Baltimore, Md.<br />

The environment in which research and clinical faculty practice is one in<br />

which the need for negotiation and conflict management abounds. The ability<br />

to foster agreements and manage conflict within and between workgroups<br />

can strengthen relationships, transform ideas into initiatives, and move<br />

parties beyond stuck places. This interactive workshop is designed to help<br />

participants understand and apply an effective framework when engaging in<br />

conflict conversations.<br />

7:00 – 9:00 am<br />

Salon AB (4th Floor)<br />

AUPN: Neurology Chair Development Course<br />

Session III: Tricks to Financial Success<br />

Martin A. Samuels, MD, DSc(hon), FAAN, MACP, FRCP, Brigham and<br />

Women’s Hospital, Boston<br />

Neurology departments are traditionally strong in the research arena, but<br />

have faced challenges from hospital and medical school leadership with<br />

regard to financial performance. This session is meant to examine some of<br />

the possible approaches that serve to mitigate this chronic problem. Major<br />

topics include: creating an incentive system that balances academic and<br />

clinical requirements, handling cross-subsidy among divisions, maintaining<br />

adequate compensation to encourage recruitment and retention, dealing with<br />

increasing disparities in salary for procedural neurologists, collaborating<br />

with neurosurgery, neuroradiology and psychiatry and maintaining academic<br />

strength in the face of fiscal challenges.<br />

This session is not available for AMA PRA Category 1 Credit.<br />

9:00 – 11:05 am<br />

Salon E (4th Floor)<br />

SYMPOSIUM: Derek Denny-Brown<br />

New Member Symposium<br />

Chair: William C. Mobley, MD, PhD, FRCP, University of California,<br />

San Diego<br />

This symposium will include a presentation by the ANA’s Derek<br />

Denny-Brown <strong>Neurological</strong> Award recipients, along with seven additional<br />

new members of the ANA. Their talks will highlight their work – either<br />

clinical or lab-based investigation. New Member Induction and the <strong>Annual</strong><br />

Business <strong>Meeting</strong> will follow this session.<br />

9:00 – 9:20 am<br />

Presentation of Derek Denny-Brown <strong>Neurological</strong> Scholar<br />

Award: Clinical Science and Temporal Trends in Acute<br />

Stroke Management<br />

Dawn Kleindorfer, MD, University of Cincinnati, Oh.<br />

9:20 – 9:35 am<br />

Presentation of Derek Denny-Brown <strong>Neurological</strong> Scholar Award:<br />

Basic Science and A Large Repeat Expansion in the C9ORF72<br />

Gene is a Common Cause of ALS and FTD<br />

Bryan J. Traynor, MB, MD, PhD, MMSc, MRCPI,<br />

National Institute on Aging, National Institutes of Health, Bethesda, Md.<br />

9:35 – 9:50 am<br />

Pilot Clinical Trial of Eculizumab in AQP4-IgG-Positive NMO<br />

Sean J. Pittock, MD, Mayo Clinic, Rochester, Minn.<br />

18


TUESDAY, OCTOBER 9<br />

9:50 – 10:05 am<br />

Iatrogenic Mitochondrial Disease Emerging Years after<br />

Stopping Anti-HIV Treatment: The Tip of the Iceberg in <strong>2012</strong><br />

Patrick F. Chinnery, PhD, FRCPath, FRCP, FMedSci, Newcastle University,<br />

Newcastle upon Tyne, UK<br />

10:05 – 10:20 am<br />

SMN Controlled MiR-183 Regulates Neuronal Morphology<br />

Via mTOR and Akt1<br />

Mustafa Sahin, MD, PhD, Children’s Hospital Boston<br />

10:20 – 10:35 am<br />

Presentation of The Grass Foundation – ANA Award in<br />

Neuroscience Award and Multiscale Investigations of Epilepsy,<br />

Sleep and Cognition<br />

Sydney S. Cash, MD, PhD, Massachusetts General Hospital, Boston<br />

10:35 – 10:50 am<br />

Congenital Myasthenic Syndrome (CMS), Autophagic Myopathy,<br />

and Cognitive Dysfunction Caused by Mutations in DPAGT1<br />

Duygu Selcen, MD, Mayo Clinic, Rochester, Minn.<br />

10:50 – 11:05 am<br />

Anti-NMDA Receptor Encephalitis, a Series of 212 Children<br />

Maarten Titulaer, MD, PhD, IDIBAPS/University of Barcelona<br />

11:05 – 11:30 am<br />

Salon E (4th Floor)<br />

Introduction of New Members<br />

11:30 am – 12:00 pm<br />

Salon E (4th Floor)<br />

Executive Session of Membership<br />

12:00 – 7:00 pm<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

All Day Poster Presentations<br />

Poster Stand-by time will be from 5:30 – 7:00 pm<br />

12:00 – 1:15 pm<br />

Lunch<br />

Pick up your lunch in the 4th Floor Foyer.<br />

12:15 – 1:30 pm<br />

Salon CD (4th Floor)<br />

Orientation for New Members<br />

For new ANA members only.<br />

12:00 – 1:15 pm<br />

Interactive Lunch Workshops: Controversies<br />

in Neurology<br />

Tuesday’s Interactive Lunch Workshops are designed to offer information<br />

and informal discussions on key issues facing academic neurology.<br />

Because of the informal format, attendees are encouraged to move between<br />

workshops if they wish.<br />

Decompressive Craniectomy for Trauma<br />

Dartmouth (3rd Floor)<br />

Decompressive craniectomy has become more common in the past<br />

decade in circumstances of threatened herniation or increased intracranial<br />

pressure. The recently published DECRA trial raises questions about the<br />

utility of at least one form of this procedure. The speakers, a neurologist<br />

and a neurosurgeon who are both neurointensivists, will discuss the<br />

controversy.<br />

Moderator: Thomas P. Bleck, MD, Rush University, Chicago<br />

Faculty: Allan H. Ropper, MD, Brigham and Women’s Hospital, Boston<br />

William B. Gormley, MD, MPH, Brigham and Women’s Hospital, Boston<br />

Continuous EEG Monitoring in the ICU: When and Why is it<br />

Needed and How Much is Enough<br />

Suffolk (3rd Floor)<br />

Dr. Herman and Dr. Hirsch will discuss and debate controversies in the<br />

use of continuous EEG monitoring in the ICU, including indications for<br />

monitoring, optimal duration of monitoring, impact on clinical care and<br />

outcome, and models for staffing and technology. They will outline<br />

current recommendations for monitoring, gaps in current knowledge,<br />

and directions for future research.<br />

Moderator: Nathan Crone, MD, Johns Hopkins, Baltimore, Md.<br />

Faculty: Lawrence J. Hirsch, MD, Yale University, New Haven, Conn.<br />

Susan T. Herman, MD, Beth Israel Deaconess Medical Center, Boston<br />

Neuro-oncology: Treatment of Low-Grade Gliomas<br />

Berkeley (3rd Floor)<br />

Low-grade infiltrative astrocytoma, oligodendroglioma and mixed<br />

astrocytic-oligodendroglial tumors (World Health Organization grade II)<br />

account for approximately 10 % of neuroepithelial tumors and 3 % of all<br />

primary nervous system neoplasms. With only about 2,000 cases per year,<br />

they are outnumbered by their malignant counterparts (anaplastic glioma,<br />

glioblastoma multiforme). Recent discoveries as part of the Cancer Genome<br />

Atlas project and other whole genome analyses and gene expression<br />

profiling studies have refined the multistep paradigm of gliomagenesis.<br />

As a result, new diagnostic and prognostic markers have been established<br />

(for example, somatic mutations of isocitrate dehydrogenase, translocation<br />

between chromosome 1p and 19q). Technological advances including<br />

magnetic resonance spectroscopy, functional MRI and fiber tractography<br />

have led to our ability to diagnose these tumors earlier and determine their<br />

spatial relationship to important cortical structures and white matter tracts.<br />

Traditional management strategies have been challenged in prospective<br />

clinical trials. Novel multidisciplinary protocols incorporating surgery,<br />

19


TUESDAY, OCTOBER 9<br />

radiation, and chemotherapy have been developed to treat gliomas more<br />

effectively and with reduced morbidity. This workshop aims to provide<br />

an up-to-date and concise review of the molecular pathogenesis,<br />

diagnostic strategies, current standards of care, and novel therapies of<br />

low-grade gliomas. In an interactive format, remaining controversies,<br />

mainly with respect to timing of diagnostic and therapeutic interventions,<br />

will be discussed.<br />

Faculty: Joachim M. Baehring, MD, MSc, Yale University,<br />

New Haven, Conn.<br />

Helen A. Shih, MD, Massachusetts General Hospital, Boston<br />

The Role of Amyloid PET Imaging in Early Diagnosis of<br />

Alzheimer Disease<br />

Salon F (4th Floor)<br />

Amyloid PET imaging is emerging as a preclinical marker of AD that may<br />

be valuable for early (prodromal and pre-symptomatic) diagnosis and<br />

thus for targeting appropriate candidates for prevention drug trials. It may<br />

also prove useful for judging the efficacy of certain AD disease-modifying<br />

therapies. This session will discuss both the exciting possibilities and the<br />

potential for misuse of this emerging modality.<br />

Prof. Sperling will briefly highlight the history of and evidence for this<br />

imaging biomarker, current recommendations and practice in research and<br />

clinical settings within the US. She will also discuss the role of amyloid<br />

PET in risk prognostication and for improving accuracy of differential<br />

diagnosis in certain clinical situations. Prof. Breteler will present the<br />

caveats that need to be considered in the interpretation of amyloid PET<br />

imaging and its use in clinical settings, drug trials and population-based<br />

epidemiological research. For instance that a biomarker needs to be<br />

validated in the same clinical or population setting in which it will be<br />

utilized. She will discuss the use of this imaging in Europe, the required<br />

bayesian perspective of pre-test probabilities modifying the interpretation<br />

of results, and the uncertainties regarding the significance of a positive<br />

PET in a population setting. She will discuss limitations in assuming that<br />

deposition of amyloid on PET is equivalent to a diagnosis of AD and the<br />

need for additional evidence to ascertain the time line and significance of<br />

various imaging and laboratory biomarkers in the course of development<br />

of early AD. The integration of amyloid PET with the other imaging and<br />

laboratory markers and the relative advantages, risks and disadvantages<br />

of the various amyloid tracers such as Pittsburgh Compound-B (PiB) and<br />

florbetapir as well as all other questions raised by the audience will be<br />

discussed by both speakers.<br />

Moderator: Sudha Seshadri, MD, Boston University, Boston<br />

Faculty: Reisa A. Sperling, MD, Brigham and Women’s Hospital, Boston<br />

Monique M.B. Breteler, MD, PhD, German Center for Neurodegenerative<br />

Disease, Bonn, Germany<br />

Managing Conflict: A Follow-up Discussion<br />

Clarendon (3rd Floor)<br />

Continue the conversation on resolving conflict and creating effective<br />

change with Catherine Morrison following her Mid/Senior Level Faculty<br />

Development Course talk.<br />

Faculty: Catherine J. Morrison, JD, Johns Hopkins University,<br />

Baltimore, Md.<br />

1:30 – 3:15 pm<br />

Salon E (4th Floor)<br />

SYMPOSIUM: Advances in Headache and Pain<br />

Research and Treatment<br />

Chair: Robert O. Messing, MD, University of California, San Francisco<br />

Co-Chair: Clifford Woolf, MB, BCh, PhD, Children’s Hospital, Boston<br />

This symposium will highlight new developments in the pathogenesis<br />

of migraine headache and chronic neuropathic pain that inform new<br />

approaches to treatment.<br />

Migraine is a recurring, episodic, often unilateral headache. It can be<br />

preceded by abnormal visual, sensory, motor and/or speech functions<br />

(migraine with aura), perceived as throbbing, and be associated with<br />

nausea and vomiting, hypersensitivity to light (photophobia), noise<br />

(phonophobia) and smell (osmophobia), as well as muscle tenderness and<br />

cephalic and extracephalic cutaneous allodynia. This lecture will summarize<br />

current understanding of (a) the mechanism by which aura activates<br />

peripheral nociceptors in the cranial meninges, (b) the mechanisms by<br />

which repeated activation of meningeal nociceptors produces central<br />

sensitization and renders migraineurs allodynic, (c) the mechanisms<br />

by which light exacerbates the headache, (d) the mechanisms by which<br />

thalamo-cortico-thalamic circuits produce many of the migraine-specific<br />

associated symptoms, and (e) the implications of our current understanding<br />

to therapeutic choices of the individual patient.<br />

The development of neuropathic pain therapeutics faces several<br />

challenges. Less than 50% of patients have 50% pain relief with the<br />

most effective agents and many patients experience intolerable side<br />

effects. Therefore, there is great need for the development of new<br />

treatments. Peripheral nerve injury results in profound changes in gene<br />

expression in sensory neurons resulting both in adaptive responses, like<br />

regeneration, and maladaptive ones that contribute to neuropathic pain. Use<br />

of model organisms like Drosophila and rodents has identified candidate<br />

“pain genes” which have been validated in patient cohorts both as pain<br />

biomarkers and as targets for novel therapeutics. Studies of pain signaling<br />

in peripheral sensory neurons have identified specific ion channels and<br />

kinases as additional targets for developing new therapeutic agents.<br />

Animal models of neuropathic pain, while providing substantial insight<br />

into neuropathic pain mechanisms, have not been reliable predictors of<br />

therapeutic effectiveness in people. These clinical trials are frequently<br />

negative due to a large positive therapeutic response in the placebo group,<br />

which presents challenges for clinical investigators. Recent experience<br />

in novel approaches to therapy will be highlighted by discussion of<br />

20


preclinical justification and results of ongoing trials testing the effectiveness<br />

of anti-NGF antibodies in chronic pain, including risk of adverse effects and<br />

their management.<br />

Learning Objectives: Having completed this symposium, participants<br />

will be able to:<br />

1. Understand the unique relationship between visual aura and the delayed<br />

onset of migraine headache, the progression of disease and the impact<br />

of early treatment, how light can make the headache worse. Participants<br />

will also be able to recognize signs of peripheral and central sensitization<br />

during migraine and they will learn about thalamocortical pathways that<br />

modulate cortical activity relevant to migraine headache.<br />

2. Provide insights into maladaptive responses in the CNS that provide<br />

targets for new therapeutics to treat chronic pain.<br />

3. Understand how studies of pain signaling in peripheral sensory neurons<br />

can reveal novel therapeutic targets.<br />

4. Understand limitations of current medications and clinical trials design<br />

issues in developing pain therapeutics.<br />

5. Provide an update on the Tanezumab trial.<br />

1:35 – 1:50 pm<br />

The Pathophysiology of Migraine Headache and its<br />

Modulation by Light<br />

Rami Burstein, PhD, Beth Israel Deaconess Medical Center, Boston<br />

1:50 – 2:15 pm<br />

F.E. Bennett Memorial Lectureship: Finding and Exploiting<br />

Neuropathic Pain Genes<br />

Clifford Woolf, MD, PhD, Children’s Hospital of Boston<br />

2:15 – 2:35 pm<br />

Mining Nociceptor Signaling for Pain Therapeutics<br />

Robert O. Messing, MD, University of California, San Francisco<br />

2:35 – 2:55 pm<br />

The Bench to Bedside Challenges in Neuropathic Pain<br />

Therapeutics<br />

Roy L. Freeman, MD, Beth Israel Deaconess Medical Center, Boston<br />

2:55 – 3:10 pm<br />

Anti-NGF: A Saga in Drug Development<br />

David R. Cornblath, MD, Johns Hopkins University, Baltimore, Md.<br />

3:10 – 3:15 pm<br />

Questions and Answers<br />

3:15 – 3:45 pm<br />

Coffee Break<br />

3:30 – 5:30 pm<br />

Special Interest Group Symposia (SIG)<br />

Please note the color of your SIG’s signage, as they have been color<br />

coded to match the posters realated to the SIG topic.<br />

Behavioral Neurology<br />

Salon AB (4th Floor)<br />

Chair: Bruce L. Miller, MD, University of California, San Francisco<br />

Leaders in the Field Presentations:<br />

Understanding the Nature and Neuroanatomy of Consciousness<br />

Prof. Adam Zeman, University of Exeter, UK<br />

Chronic Traumatic Encephalopathy<br />

Ann McKee, MD, Boston University, Bedford, Mass.<br />

Executive Control and the Frontal Lobes<br />

Kirk R. Daffner, MD, Harvard Medical School, Boston<br />

Misdiagnosis in Dementia<br />

Bruce L. Miller, MD, University of California, San Francisco<br />

Data Blitz Presentations:<br />

Evolution of Psychiatric Co-Morbidity in the Transition Phase<br />

in a Cohort of Patients with <strong>Neurological</strong> Perinatal Disability<br />

Andrea Martinuzzi, MD, PhD, E. Medea Scientific Institute,<br />

Conegliano, Italy<br />

Inflammation and Cognitive Decline in Normal Elderly<br />

Joel Kramer, PsyD, University of California, San Francisco<br />

Changes in Cortical Functional Connectivity Introduced<br />

with Intermittent Theta Burst TMS to the Cerebellum<br />

Assessed with fMRI<br />

Mark Halko, PhD, Harvard Medical School, Boston<br />

Modulation and Assessment of the Cerebello-Cortical<br />

Connectivity through Transcranial Magnetic Stimulation (TMS)<br />

Combined with Electroencephalography (EEG)<br />

Faranak Farzan, PhD, Harvard Medical School, Boston<br />

Effects of the Dopamine Agonist Rotigotine on Hemispatial<br />

Neglect Following Stroke<br />

Masud Husain, DPhil, FMedSci, UCL Institute of Neurology, London, UK<br />

Increased Phosphorylation of the MAPK/ERK Pathway Is<br />

Associated with Social Impairment in BTBR Mice<br />

Aliereza Faridar, MD, PhD, University of California, San Francisco<br />

Discussion<br />

21


TUESDAY, OCTOBER 9<br />

Autoimmune Neurology<br />

Salon F (4th Floor)<br />

Chair: Vanda A. Lennon, MD, PhD, Mayo Clinic, Rochester, Minn.<br />

Leaders in the Field Presentations:<br />

Autoimmune Encephalitis: The Antigens and Guidelines<br />

to Interpretation<br />

Josep Dalmau, MD, PhD, University of Pennsylvania, Philadelphia<br />

<strong>Neurological</strong> Autoimmunity: From Cortex to Colon<br />

Sean J. Pittock, MD, Mayo Clinic, Rochester, Minn.<br />

Pathogentic Insights from Comparisons of NMO and Multiple<br />

Sclerosis Lesions<br />

Bogdan Popescu, MD, PhD, University of Saskatchewan, Saskatoon,<br />

Canada<br />

Autoimmune Pain<br />

Christopher Klein, MD, Mayo Clinic, Rochester, Minn.<br />

Data Blitz Presentations:<br />

Faciobracial Dystonic Seizures are Immunotherapy-Responsive<br />

and Treatment may Prevent Amnesia<br />

Sarosh R. Irani, DPhil, MRCP, University of Oxford, UK<br />

Clinical Information of Four Men with Anti-N-Methyl D-Aspartate<br />

Receptor Encephalitis<br />

Hirano Makito, MD, Kinki University, Osaka, Japan<br />

Unusual Presentation of Paraneoplastic Stiff-Person Syndrome<br />

with Underlying Breast Cancer<br />

Alvin Shrestha, MBChB, University Hospitals Birmingham, UK<br />

Myositis with Autoantibodies to / Sarcoglycan: A New Disease<br />

Russell Lane, MBBS, Imperial College, Healthcare NHS Trust, London, UK<br />

Discussion<br />

Movement Disorders<br />

Salon CD (4th Floor)<br />

Co-Chairs: William T. Dauer, MD, University of Michigan, Ann Arbor<br />

Ming Guo, MD, PhD, University of California, Los Angeles<br />

Leaders in the Field Presentations:<br />

Update on Spinocerebellar Ataxias (SCAs)<br />

Stefan Pulst, MD, University of Utah, Salt Lake City<br />

Basal Ganglia Dysfunction in a Model of Cerebellar-induced<br />

Dystonia<br />

Kamran Khodakhah, PhD, Albert Einstein College of Medicine, Yeshiva<br />

University, Bronx, New York<br />

22<br />

Common Pathways in Neurodegeneration: Focus<br />

on Lysosomal Dysfunction in Parkinson’s Disease<br />

Dimitri Krainc, MD, PhD, Harvard Medical School, Boston<br />

Data Blitz Presentations:<br />

What Issues Face Parkinson’s Patients at Ten Years<br />

and Beyond<br />

Anhar Hassan, MBBCh, FRACP, University of Florida, Gainesville, Fla.<br />

Predictors of Cognitive Performance Following Bilateral<br />

Subthalamic Nucleus Deep Brain Stimulation<br />

Angela Costello, D ClinPsych, King’s College Hospital,<br />

NHS Foundation Trust, London, UK<br />

Inhibitory Neurons in the Ventral Medial Medulla Modulate<br />

Gait and Tone<br />

Veronique VanderHorst, MD, PhD, Harvard Medical School, Boston<br />

Discussion<br />

Regulatory Science — NEW!<br />

Provincetown (4th Floor)<br />

Chair: Ira Shoulson, MD, Georgetown University, Washington, DC<br />

Regulatory Science Informing the Review and Approval of<br />

Innovative Neurology Products<br />

Russell Katz, MD, US Food & Drug Administration (FDA),<br />

Silver Spring, Md.<br />

Translational and Clinical Neuroscience Contributions to<br />

Regulatory Science<br />

D. Elizabeth McNeil, MD, National Institute of <strong>Neurological</strong> Disorders<br />

and Stroke (NINDS), National Institutes of Health, Bethesda, Md.<br />

How Industry and Translational Neurology Can Contribute<br />

to and Benefit from Regulatory Science<br />

Bernard Ravina, MD, Biogen Idec, Cambridge, Mass.<br />

Centers of Excellence in Regulatory Science & Innovation<br />

and Applications to Academic Neurology<br />

Ira Shoulson, MD, and Erin Wilhelm MPH, Center of Excellence in<br />

Regulatory Science and Innovation (CERSI), Georgetown University,<br />

Washington, DC<br />

Panel and General Discussion<br />

5:30 – 7:00 pm<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

Poster Stand-by Wine & Cheese Reception<br />

Poster Categories<br />

Behavioral Neurology<br />

Headache and Pain/Neuro-otology<br />

Movement Disorders<br />

Neuroimmunology


SPECIAL EVENTS<br />

President’s Reception at the Boston Public Library<br />

Monday, October 8, <strong>2012</strong>, 6:30 – 8:30 pm<br />

Boston Public Library<br />

Central Library, McKim Building, 700 Boylston St., Boston, MA 02116<br />

Everyone is invited to attend the President’s Wine and Cheese Reception at the Boston Public Library’s McKim<br />

Building. The Boston Public Library is within walking distance of the Boston Marriott Copley Place, but shuttles will<br />

be provided for those who wish to use them. The Boston Public Library’s McKim Building was built in 1895 and is<br />

on the National Register of Historic Buildings and is a National Historic Landmark. Light hors d’oeuvre selections,<br />

along with wine and beer, will be served at the library. The President’s Reception is complimentary for paid meeting<br />

attendees, but a name badge is required for entry. Guests may purchase a ticket for $50 at the registration desk.<br />

Walking Directions<br />

From the Marriott Copley Place Hotel lobby, go Northeast on Huntington Avenue (take a right out of the lobby)<br />

Take a sharp left to stay on Huntington Avenue (Westin Copley will be on your right hand side)<br />

Turn left on Dartmouth Street<br />

Enter the Boston Public Library (McKim Building)<br />

Please Note: Use the entrance on Dartmouth Street and not Boylston St.<br />

Poster Stand-by Wine & Cheese Receptions<br />

Back Bay Hall: Gloucester and Arlington (3rd Floor)<br />

Sunday, October 7, <strong>2012</strong>, 5:30 – 7:00 pm<br />

Monday, October 8, <strong>2012</strong>, 5:30 – 6:30 pm<br />

Tuesday, October 9, <strong>2012</strong>, 5:30 – 7:00 pm<br />

What would the best<br />

of medicine look like<br />

A heritage built upon helping protect and preserve vision.<br />

A commitment to advancing the Science of Rejuvenation.<br />

A resolve to confront the global obesity epidemic head-on.<br />

A devotion to help improve patients’ lives through urologics.<br />

Helping people with skin conditions face the world.<br />

A dedication to enabling patients to live life without compromise and to their fullest potential.<br />

It would look like Allergan.<br />

Allergan is a multi-specialty health care company established more than 60 years ago with a commitment to<br />

uncover the best of science and develop and deliver innovative and meaningful treatments to help people<br />

reach their life’s potential. Today, we have approximately 10,000 highly dedicated and talented employees,<br />

global marketing and sales capabilities with a presence in more than 100 countries, a rich and ever-evolving<br />

portfolio of pharmaceuticals, biologics, medical devices and over-the-counter consumer products, and stateof-the-art<br />

resources in R&D, manufacturing and safety surveillance that help millions of patients see more<br />

clearly, move more freely and express themselves more fully. From our beginnings as an eye care company to<br />

our focus today on several medical specialties, including eye care, neurosciences, medical aesthetics, medical<br />

dermatology, breast aesthetics, obesity intervention and urologics, Allergan is proud to celebrate 60 years<br />

of medical advances and proud to support the patients and physicians who rely on our products and the<br />

employees and communities in which we live and work.<br />

To find out more about Allergan, visit www.allergan.com<br />

Copyright 2011 Allergan, Inc. Irvine, CA 92612. TM and ® marks owned by Allergan, Inc. APC16NB10<br />

23


SUNDAY SPEAKER ABSTRACTS<br />

SYMPOSIUM: New Tools to Define the Genetics<br />

of <strong>Neurological</strong> Disorders<br />

Genome-Wide <strong>Association</strong> Studies: Have They Delivered<br />

for Neurology<br />

Stephen Sawcer, PhD, University of Cambridge, Cambridge, UK<br />

Genome-Wide <strong>Association</strong> Studies (GWAS) have resulted in a veritable<br />

explosion in knowledge about all manner of complex traits (http://www.<br />

genome.gov/gwastudies/), 1 and have revolutionised the genetic analysis of<br />

multiple sclerosis. 2,3 Each robustly associated variant provides the potential<br />

for a novel insight into aetiology and thereby improves the prospects for<br />

the development of rational therapy. However, for many of these loci the<br />

potential insights and benefits are not immediately obvious and seem<br />

set to require extensive fine mapping and functional analysis before they<br />

are revealed. In most cases associated variants are neither necessary nor<br />

sufficient and effects measured in the context of mathematical models are<br />

not expected to reflect underlying biology or to indicate the importance of<br />

implicated genes.<br />

In considering the differences between this type of complex genetics and<br />

the more familiar Mendelian genetics we might draw an analogy with the<br />

difference between quantum theory and Classical mechanics; in each<br />

case seemingly incontrovertible determinism is replaced by mathematical<br />

descriptions of uncertainty and counterintuitive behaviour is common<br />

place. In this probabilistic environment purely mathematical issues can<br />

profoundly influence the perception of results and need to be properly<br />

considered if interpretation is not to be misleading. 4 In my talk, as well as<br />

describing what has been discovered, I will also try and describe some of<br />

the relevant mathematical issues in the hope of aiding in the interpretation<br />

of these exciting new data.<br />

References:<br />

1. Hindorff et al. Potential etiologic and functional implications of<br />

genome-wide association loci for human diseases and traits. PNAS.<br />

2009 106: 9362-7<br />

2. IMSGC. Risk Alleles for Multiple Sclerosis Identified by a Genomewide<br />

Study. N Engl J Med. 2007;357(9):851-62.<br />

3. IMSGC and WTCCC2. Genetic risk and a primary role for<br />

cell-mediated immune mechanisms in multiple sclerosis. Nature.<br />

2011;476(7359):214-9.<br />

4. Sawcer and Wason. Risk in complex genetics: “All models are wrong<br />

but some are useful” Ann Neurol. <strong>2012</strong> ePub April.<br />

Next-Generation Whole Exome and Whole Genome<br />

Sequencing to Diagnose Single-Gene Disorders<br />

Andrew Singleton, PhD, National Institutes of Health, Bethesda, Md.<br />

Participants will be provided with an update on the use of second<br />

generation sequencing approaches in the identification of disease causing<br />

genetic variants. This will center on the strengths and weaknesses of<br />

these approaches, and in particular how such methods will facilitate<br />

the identification of disease-linked mutations that were resistant to<br />

identification using traditional linkage and positional cloning approaches.<br />

There will be a discussion of considerations such as family structure,<br />

sample collection, data handling, and data analysis, with an emphasis on<br />

when to use such methods, and a review of the problems and pitfalls that<br />

can lead to false positives. In addition there will be discussion of when this<br />

approach is most useful in identifying the genetic causes of disease in the<br />

context of genetic testing, particularly how this may be a time and resource<br />

efficient method in genetically heterogeneous disorders [1].<br />

References:<br />

1. Singleton AB. Exome sequencing: a transformative technology.<br />

Lancet Neurol. 2011 Oct;10(10):942-6.<br />

Epigenetics and Disorders of the Nervous System:<br />

An Evolving Synthesis<br />

Mark F. Mehler, MD, Albert Einstein College of Medicine, Bronx, N.Y.<br />

Epigenetics and epigenomic medicine encompass an evolving science<br />

of brain and behavior that are providing revolutionary insights into the<br />

mechanisms underlying brain development and evolution, neuronal and<br />

neural network homeostasis, plasticity connectivity, stress responses,<br />

bioenergetics, senescence, neuropsychiatric disease pathogenesis and<br />

dynamic tissue remodeling and cellular reprogramming. Epigenetic<br />

processes include DNA methylation, histone modifications, nucleosome<br />

repositioning, higher order chromatin remodeling events, deployment of<br />

non-protein-coding RNAs and RNA editing and DNA recoding. RNA is<br />

centrally involved in orchestrating these processes, thereby suggesting<br />

that the evolving transcriptional and post-transcriptional output of the cell<br />

is the primary determinant of epigenetic memory states. These contextspecific<br />

processes can be modulated by: (1) metabolic, homeostatic<br />

and environmental cues that control profiles of gene expression and<br />

post-transcriptional processing, (2) RNA/DNA editing events via activitydependent<br />

intracellular transport and regulation of RNA processing and<br />

RNA regulatory complexes, and (3) intricate intercellular neural, systemic<br />

and even germ line trafficking of diverse classes of RNAs and transposable<br />

elements with short- and long-range effector functions. These interrelated<br />

processes promote dynamic nuclear reorganization to continuously<br />

modulate individual gene expression profiles and functional gene networks<br />

with complex temporal and spatial trajectories. Such gene regulatory<br />

trajectories are required for enhancing the fidelity of neural signaling<br />

events linked to complex cognitive and behavioral repertoires. Epigenetics<br />

therefore represents the long sought after molecular interface mediating<br />

gene-environmental interactions along the entire neuraxis during critical<br />

periods throughout the lifecycle. The emerging field of environmental<br />

epigenomics has begun to identify combinatorial profiles of environmental<br />

stressors modulating the latent phase, time of initiation, and progression<br />

24


of neurological disease states, as well as defining novel biomarkers<br />

and molecular indicators of graded responses to therapeutic initiatives.<br />

Next-generation pharmacoepigenomic therapies will likely promote<br />

accelerated recovery of seemingly irrevocably lost cognitive, behavioral and<br />

sensorimotor functions, without exhibiting potentially deleterious off-target<br />

effects, through epigenetic reprogramming of endogenous neural stem cell<br />

fates, dynamic tissue remodeling and restoration of neural network integrity,<br />

plasticity and connectivity.<br />

References:<br />

1. Qureshi, IA, Mehler, MF. <strong>2012</strong>. Emerging Roles of Non-Coding RNAs<br />

in Brain Evolution, Development, Plasticity and Disease. Nat Rev<br />

Neuroscience 13: 528-541.<br />

2. Qureshi, IA, Mehler, MF. 2011. Non-Coding RNA Networks<br />

Mediating Cognitive Disorders Across the Lifespan. Trends Mol Med<br />

17: 337-346.<br />

3. Abrajano, JJ, Quershi, IA, Gokhan, S, Molero, AE, Zheng, D, Bergman,<br />

A, Mehler, MF. 2010. Corepressor for Element-1-Silencing Factor<br />

Preferentially Mediates Gene Networks Underlying Neural Stem Cell<br />

Fate Decisions. Proc Natl Acad Sci USA 107: 16685-16690.<br />

4. Mehler, MF. 2008. Epigenetic Principles and Mechanisms Underlying<br />

Nervous System Functions in Health and Disease. Prog Neurobiol<br />

86: 305-341.<br />

Regulatory RNAs in <strong>Neurological</strong> Disease<br />

Claes Wahlestedt, MD, PhD, University of Miami<br />

Much of the mammalian genome is transcribed (1) into non-coding<br />

RNAs of different categories which will be described. This lecture will be<br />

concerned with microRNAs (2) as well as natural antisense transcripts<br />

(NATs) most of which are long noncoding RNAs (3). NATs are found in<br />

most gene loci and regulate gene expression through several distinct<br />

mechanisms including chromatin modifications. Inhibition/perturbation of<br />

endogenous NATs by modified oligonucleotides called AntagoNATs (4), in<br />

vitro or in vivo, reveals concordant or discordant regulation and results in<br />

down- or up-regulation of conventional (protein-coding) gene expression,<br />

respectively. Evidence will be presented that AntagoNATs can potently<br />

induce locus-specific and reversible up-regulation of gene expression,<br />

such as BDNF, associated with alterations in chromatin marks (4).<br />

References:<br />

1. Carninci P, Wahlestedt C, et al. The transcriptional landscape of the<br />

mammalian genome. Science 309: 1559-1563, 2005.<br />

2. Miller, B.H., Zeier, Z., Xi, L., Deng, S., Strathmann , J., Willoughby, D.,<br />

Kenny, P.J., Elsworth, J.D, Lawrence, M.S., Roth, R.R., Edbauer, D.,<br />

Kleiman, R., Wahlestedt, C. MiR-132 dysregulation in schizophrenia<br />

has implications for both neurodevelopment and adult brain function.<br />

Proc Natl Acad Sci (USA) 109(8):3125-30, <strong>2012</strong>.<br />

3. Katayama S … and Wahlestedt C. Antisense transcription in the<br />

mammalian transcriptome. Science 309:1564-1566, 2005.<br />

4. Modarresi F, Faghihi MA, Lopez-Toledano MA, Fatemi RP, Magistri<br />

M, Brothers SP, Van der Brug MP, Wahlestedt C. Natural antisense<br />

inhibition results in transcriptional de-repression and gene upregulation.<br />

Nature Biotechnology, doi: 10.1038/nbt.2158, <strong>2012</strong>.<br />

New Tools to Define the Genetics of <strong>Neurological</strong> Disorders:<br />

Mitochondrial Disorders<br />

Patrick F. Chinnery, PhD, FRCPath, FRCP, FMedSci, Newcastle University,<br />

Newcastle upon Tyne, UK<br />

Clinical and genetic heterogeneity are the hallmark of mitochondrial<br />

disorders. Ultimately due to a biochemical defect of the respiratory chain,<br />

mitochondrial disorders can be caused by mutations in both the nuclear<br />

genome, and the maternally inherited 16.5Kb mitochondrial genome<br />

(mtDNA). As a result, mitochondrial disorders can be inherited maternally,<br />

or as an autosomal dominant, recessive, or X-chromosome-linked trait.<br />

On the one hand, the same clinical phenotype can be caused by mutations<br />

in mtDNA or an expanding array of different nuclear genes. On the other<br />

hand, the same genetic defect can cause a wide range of different clinical<br />

phenotypes. This presents a major diagnostic challenge in routine<br />

neurological practice. This is compounded by the fact that mitochondrial<br />

disease enters the differential diagnosis of many common diseases seen<br />

in the general neurology clinic. However, making a genetic diagnosis is<br />

important, because it can have a profound implications for the recurrence<br />

risks within the family, and because new therapies are emerging for<br />

specific genetic subgroups.<br />

The traditional clinical approach to the investigation of mitochondrial<br />

disease involves targeted molecular genetic testing in patients<br />

with “classical” mitochondrial syndromes, such as mitochondrial<br />

encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS),<br />

or Leber hereditary optic neuropathy (LHON), but these form the minority.<br />

The next step involves a biopsy of an affected tissue, followed by highlyspecialised<br />

histochemical and biochemical studies, which are usually<br />

carried out in specialized laboratories, and are sometimes difficult to<br />

interpret. The results of these investigations guide further genetic analysis<br />

of the mitochondrial and nuclear genomes. Finally, despite an often<br />

protracted and costly series of investigations, in ~30% of patients it is not<br />

possible to reach a molecular diagnosis, even in a specialist centre.<br />

In principle, next generation sequencing provides an opportunity to side<br />

step much of this complexity. A growing number of new mitochondrial<br />

disease genes have been identified within the last 18 months using<br />

targeted and whole-exome capture systems, and mitochondrial DNA can<br />

be sequenced at great depth, potentially circumventing the problem of<br />

detecting low-level heteroplasmy levels in blood. Combing the two in a<br />

single “office-based” blood test is on the horizon, but there are potential<br />

pitfalls at present. This presentation will provide a guide to these pitfalls,<br />

and how they can be avoided.<br />

References:<br />

1. Koopman WJ, Willems PH, Smeitink JA. Monogenic mitochondrial<br />

disorders. N Engl J Med <strong>2012</strong>;366:1132-41.<br />

2. Haack TB, Danhauser K, Haberberger B, et al. Exome sequencing<br />

identifies ACAD9 mutations as a cause of complex I deficiency.<br />

Nat Genet;42:1131-4.<br />

3. Taylor RW, Turnbull DM. Mitochondrial DNA mutations in human<br />

disease. Nat Rev Genet 2005;6:389-402.<br />

4. Calvo SE, Compton AG, Hershman SG, et al. Molecular diagnosis<br />

of infantile mitochondrial disease with targeted next-generation<br />

sequencing. Science translational medicine <strong>2012</strong>;4:118ra10.<br />

25


SUNDAY SPEAKER ABSTRACTS<br />

SYMPOSIUM: Imaging to Explore Neural Network<br />

Structure and Function<br />

Decoding the BOLD Signal in Functional MR Imaging<br />

Anna Devor, PhD, University of California, San Diego<br />

Understanding how neuronal activity drives changes in blood flow and<br />

energy metabolism is critical for laying a solid physiological foundation<br />

for interpreting functional neuroimaging studies in humans. In particular,<br />

functional magnetic resonance imaging (fMRI) based on the blood<br />

oxygenation level dependent (BOLD) response has become a widely<br />

used tool for exploring brain function, and yet the physiological basis<br />

of this technique is still poorly understood. The primary physiological<br />

phenomenon that leads to the BOLD effect is that cerebral blood flow (CBF)<br />

increases much more than the cerebral metabolic rate of oxygen (CMRO 2<br />

),<br />

increasing local blood oxygenation and causing a slight increase of the MRI<br />

signal due to different magnetic properties of oxy- and deoxyhemoglobin.<br />

Although the goal of fMRI studies is usually to assess changes in neuronal<br />

activity, the signal measured depends on the relative changes in CBF and<br />

CMRO 2<br />

. For this reason, it is difficult to interpret the magnitude of the<br />

BOLD response as a quantitative reflection of the magnitude of the change<br />

in neuronal activity without a deeper understanding of how neuronal<br />

activity, CBF and CMRO 2<br />

are connected.<br />

The simplest picture one could imagine for this physiological coupling is<br />

that changes in neuronal activity drive changes in energy metabolism which<br />

then drive changes in blood flow. However, a large body of work over the<br />

last decade indicates that this simple picture is almost certainly wrong.<br />

While molecules produced by increased energy metabolism (CO 2<br />

, lactate,<br />

etc.) do have a vasoactive effect, much of the acute CBF response under<br />

healthy conditions appears to be driven by molecules related to neuronal<br />

signaling. In short, rather than a linear chain of events, blood flow and<br />

energy metabolism appear to be driven in parallel by neuronal activity.<br />

With the growing recognition of the complexity of neurovascular coupling,<br />

research has focused on the “neurovascular unit”, a close association<br />

between neurons, astrocytes and blood vessels. A number of experimental<br />

tools have been developed for probing the neurovascular unit in animal<br />

models, providing the potential for a much deeper understanding of these<br />

fundamental physiological mechanisms. In this talk, we review recent<br />

advances in imaging technology with microscopic resolution applicable<br />

to in vivo animal studies, discuss our working hypotheses regarding the<br />

regulation of blood flow and neurophysiological correlates of fMRI signals,<br />

and provide an outlook for the future directions in neurovascular research.<br />

References:<br />

1. Devor A, Sakadzic S, Srinivasan VJ, Yaseen MA, Nizar K, Saisan PA,<br />

Tian P, Dale AM, Vinogradov SA, Franceschini MA, Boas DA. Frontiers<br />

in optical imaging of cerebral blood flow and metabolism. J Cereb<br />

Blood Flow Metab. <strong>2012</strong>;32:1259-76.<br />

2. Tian P, Teng IC, May LD, Kurz R, Lu K, Scadeng M, Hillman EM,<br />

De Crespigny AJ, D’Arceuil HE, Mandeville JB, Marota JJ, Rosen<br />

BR, Liu TT, Boas DA, Buxton RB, Dale AM, Devor A. Cortical depthspecific<br />

microvascular dilation underlies laminar differences in blood<br />

oxygenation level-dependent functional MRI signal. Proc Natl Acad Sci<br />

U S A. 2010;107:15246-15251.<br />

3. Devor A, Tian P, Nishimura N, Teng IC, Hillman EM, Narayanan SN,<br />

Ulbert I, Boas DA, Kleinfeld D, Dale AM. Suppressed neuronal activity<br />

and concurrent arteriolar vasoconstriction may explain negative blood<br />

oxygenation level-dependent signal. J Neurosci. 2007;27:4452-4459.<br />

4. Cauli B, Hamel E. Revisiting the role of neurons in neurovascular<br />

coupling. Front Neuroenergetics. 2010;2:9.<br />

Resting State Connectivity<br />

Michael D. Greicius, MD, MPH, Stanford University, Stanford, CA<br />

Resting-state fMRI is a relatively novel imaging modality that allows<br />

for the detection and characterization of large-scale, distributed brain<br />

networks. Like standard task-activation fMRI the method relies on the<br />

intrinsic contrast provided by the blood oxygen level-dependent (BOLD)<br />

signal. During quiet rest, brain regions are characterized by low-frequency<br />

BOLD signal fluctuations. Resting-state fMRI leverages the fact that these<br />

low-frequency BOLD signal fluctuations are strongly correlated across<br />

brain regions known to constitute networks. The first demonstration of<br />

the neurobiological plausibility and potency of this method was by Biswal<br />

and colleagues at the Medical College of Wisconsin, who showed that<br />

the left motor cortex was functionally connected to the contralateral motor<br />

cortex, the bilateral sensory cortices, and the bilateral premotor cortices.<br />

Subsequent work has shown that a large array of networks, supporting<br />

a host of sensory, motor, and cognitive functions can be delineated with<br />

resting-state fMRI.<br />

Among the 15-20 networks that have been reliably demonstrated with<br />

resting-state fMRI one of them, known as the default-mode network (DMN),<br />

appears critical for episodic memory retrieval and has been strongly<br />

linked to Alzheimer’s disease (AD). In 2004, our group first demonstrated<br />

that the DMN shows reduced connectivity in AD patients compared to<br />

healthy controls. This finding has been replicated numerous times and<br />

advanced up the clinical spectrum. Sorg and colleagues showed in<br />

2007 that DMN connectivity is reduced in patients with mild cognitive<br />

impairment (MCI) compared to healthy controls. Two groups have shown<br />

that healthy older controls with positive amyloid PET scans have reduced<br />

DMN connectivity compared to older controls with negative amyloid PET<br />

scans. The APOE story is a bit murkier with some groups showing reduced<br />

DMN connectivity in healthy older E4 carriers compared to E3 carriers<br />

and some groups showing increased connectivity or no difference. Our<br />

group has demonstrated that the predicted reduction in DMN connectivity<br />

among healthy older E4 carriers is robust and detectable in women but<br />

not in men (in keeping with the often overlooked interaction between<br />

gender and APOE). Recent work from Sperling and colleagues in the<br />

Dominantly Inherited Alzheimer’s Network (DIAN) study has shown that<br />

DMN connectivity begins to decline in carriers of autosomal dominant AD<br />

mutations about 5-10 years before predicted symptom onset.<br />

DMN connectivity has substantial support, therefore, as a candidate<br />

imaging biomarker in AD. A number of critical questions and limitations<br />

remain before this approach can be reliably applied at the single subject<br />

level. At the group level, however, resting-state fMRI has already yielded<br />

some valuable new insights into AD. The hope is that with continued<br />

refinement in the acquisition and analysis parameters, resting-state fMRI<br />

26


will prove useful as a diagnostic aid in individual patients. For the time<br />

being, it has the potential to play a valuable role, at the group-level, as an<br />

early, objective marker of treatment response in clinical trials.<br />

References:<br />

1. Biswal B, Yetkin FZ, Haughton VM, Hyde JS. Functional connectivity<br />

in the motor cortex of resting human brain using echo-planar MRI.<br />

Magn Reson Med. 1995 Oct;34(4):537-41.<br />

2. Greicius MD, Srivastava G, Reiss AL, Menon V. Default-mode<br />

network activity distinguishes Alzheimer’s disease from healthy aging:<br />

evidence from functional MRI Proc Natl Acad Sci U S A. 2004 Mar<br />

30;101(13):4637-42. Epub 2004 Mar 15.<br />

3. Sorg C, Riedl V, Mühlau M, Calhoun VD, Eichele T, Läer L, Drzezga<br />

A, Förstl H, Kurz A, Zimmer C, Wohlschläger AM. Selective changes<br />

of resting-state networks in individuals at risk for Alzheimer’s disease.<br />

Proc Natl Acad Sci U S A. 2007 Nov 20;104(47):18760-5. Epub 2007<br />

Nov 14<br />

4. Damoiseaux JS, Seeley WW, Zhou J, Shirer WR, Coppola G,<br />

Karydas A, Rosen HJ, Miller BL, Kramer JH, Greicius MD; for the<br />

Alzheimer’s Disease Neuroimaging Initiative. Gender Modulates<br />

the APOE {varepsilon}4 Effect in Healthy Older Adults: Convergent<br />

Evidence from Functional Brain Connectivity and Spinal Fluid Tau<br />

Levels.J Neurosci. <strong>2012</strong> Jun 13;32(24):8254-8262.<br />

Diffusion Tensor Imaging (DTI)<br />

Lawrence L. Wald, PhD, Massachusetts General Hospital, Boston<br />

MRI diffusion imaging has emerged as our principle method for the study<br />

of anatomical connectivity in the living human brain (human connectome).<br />

It achieves this by utilizing the relatively free diffusion of endogenous<br />

brain water along the axon bundles compared to the restricted diffusion<br />

perpendicular to a fascicle. But, perhaps more than any other “-omics”<br />

endeavor, the accuracy and level of detail obtained from mapping the major<br />

whitematter pathways in the living human brain depends on the capabilities<br />

of the imaging technology used. The current tools are remarkable; for<br />

example allowing the formation of an “image” of the water diffusion<br />

probability distribution in regions of complex crossing fibers at each of half<br />

a million voxels in the brain. Nonetheless our ability to map the complex<br />

organization of connection pathways in vivo is acquisition limited. This talk<br />

will review the methods in use for studying fiber architecture with diffusion<br />

MRI as well as the limitations of the technique, which include sensitivity,<br />

image resolution, and efficiency of encoding the diffusion probability<br />

distribution for diffusion tractography.<br />

Fortunately, recent advances in MR technology have advanced the level<br />

of white matter structure that can be interrogated in the living human<br />

brain. These include High Angular Resolution Diffusion Imaging (HARDI)<br />

techniques as well as improved scanner hardware and more efficient<br />

acquisition sequences (by 3x). These tools have provided rich engineering<br />

challenges but are found to be enabling technology for improved structural<br />

connectomics with MRI.<br />

References:<br />

1. Wedeen VJ, Rosene DL, Wang R, Dai G, Mortazavi F, Hagmann P,<br />

Kaas JH, Tseng WY. The geometric structure of the brain fiber<br />

pathways. Science <strong>2012</strong>; 335(6076);1628-34<br />

2. Hagmann P, Cammoun L, Gigandet X, Gerhard S, Grant PE, Wedeen V,<br />

Meuli R, Thiran JP, Honey CJ, Sporns O. MR connectomics: Principles<br />

and challenges. J. Neurosci Methods. 2010 194(1); 34-45.<br />

3. Schmahmann JD, Smith EE, Eichler FS, Filley CM. Cerebral white<br />

matter: neuroanatomy, clinical neurology, and neurobehavioral<br />

correlates. Ann N Y Acad Sci. 2008 1142: 266-309.<br />

4. Johansen-Berg H, and Behrens T.E.J. Diffusion MRI: from quantitative<br />

measurement to in-vivo neuroanatomy. Academnic Press 2009.<br />

Molecular Imaging<br />

Joel S. Perlmutter, MD, Washington University, St. Louis, Mo.<br />

Molecular imaging with either PET or SPECT agents has provided new<br />

insights into the function of brain networks. These methods have revealed<br />

surprising findings about the pathophysiology of Parkinson disease (PD),<br />

yet some imaging studies have raised more questions than answers. This<br />

talk will focus on several examples of how molecular imaging has revealed<br />

new findings about cognitive impairment in PD; describe the applications<br />

and limitations of imaging methods to quantify nigrostriatal pathways and<br />

finally discuss the integration of molecular imaging with other imaging<br />

modalities to investigate brain networks.<br />

Dementia associated with PD has been thought to be caused by a<br />

combination of underlying alpha-synuclein pathology in cortical area or<br />

co-existing Alzheimer’s disease. The development of the amyloid imaging<br />

agent Pittsburgh Compound B (PIB) has permitted in vivo measurement of<br />

brain deposition of ABeta42, which could reveal whether Alzheimer’s was<br />

a contributing factor. Initial studies revealed that a subset of people with<br />

PD and cognitive impairment, whether starting early in the course of PD<br />

or later, had positive PIB PET scans. Since PIB is based upon thioflavin T<br />

which binds to beta-pleated sheet structures such as ABeta42 or alphasynuclein,<br />

the first step in interpretation of these studies was to determine<br />

whether the PIB also bound to alpha-synuclein in vivo. Postmortem follow<br />

up of those PD patients with dementia that had PIB scans revealed that PIB<br />

did not mark alpha-synuclein. However, the surprise was that those with<br />

positive PIB scans had abnormal ABeta42 but only rarely had abnormal<br />

tau deposition, both of which are commonly associated with dementia due<br />

to Alzheimer’s disease. Thus, application of PIB PET to PD has different<br />

implications than to those with Alzheimer’s disease. Most importantly, this<br />

also demonstrates the importance of careful validation of a biomarker for<br />

each application.<br />

A similar lesson can be learned from application of several different<br />

molecular imaging methods to measure presynaptic nigrostriatal neurons.<br />

The basic approaches include fluorodopa (FD) that reflects decarboxylase<br />

and storage; dopamine transporter (DAT) ligands and radioligands that<br />

bind to vesicular monoamine transporter type 2 (VMAT2) on presynpatic<br />

vesicles. Striatal uptake of these tracers has been used to either indicate<br />

degenerative parkinsonism or measure its progression. Yet, studies using<br />

these tracers have found discordant results when compared to clinical<br />

27


SUNDAY SPEAKER ABSTRACTS<br />

measures of PD progression. Recent studies to validate these measures<br />

in nonhuman primates have revealed rather surprising findings. First,<br />

nigrostriatal reserve, the degree of loss of dopaminergic neurons needed<br />

to cause parkinsonism, is less than previously thought. Second, the degree<br />

of parkinsonism correlates with loss nigral neurons but not with striatal<br />

dopamine. All of the PET measures of striatal uptake correlate with striatal<br />

dopamine but only correlate with nigral neurons when loss of those neurons<br />

does not exceed 50%. All of the PET measures change in synch with each<br />

other providing no support for differential regulation, at least in this animal<br />

model. The striatal PET uptakes measures do correlate strongly with in vitro<br />

quantitatively autoradiography demonstrating that these findings are not<br />

likely due to specific characteristics of the PET tracers used. These findings<br />

may explain much of the discordant results in past studies. New approaches<br />

to these PET measures may reveal approaches that can directly measure<br />

nigral neurons and obviate some of the past limitations.<br />

Finally, molecular imaging studies can be used to drive analysis of other<br />

methods to investigate brain networks. For example, abnormalities found<br />

on PET can be used in conjuntion with resting state functional connectivity<br />

studies with MR to investigate the functional consequences of selected<br />

regional abnormalities.<br />

References:<br />

1. Burack M, Hartlein J, Flores H, Taylor-Reinwald L, Perlmutter JS,<br />

Cairns NE. In vivo amyloid imaging in autopsy confirmed Parkinson<br />

disease with dementia. Neurology, 74:77-84, 2010.<br />

2. Kotzbauer PT, Cairns NJ, Campbell MC, Willis AW, Racette BA,<br />

Tabbal S, Perlmutter JS. Pathological accumulation of -synuclein and<br />

A in Parkinson disease with dementia. Arch Neurol. 23:1-6, <strong>2012</strong><br />

[Epub ahead of print].<br />

28


MONDAY SPEAKER ABSTRACTS<br />

President’s Symposium: Alzheimer’s Disease:<br />

New Perspectives on an Old Disease<br />

Lessons for Alzheimer’s Disease from Early Detection of<br />

Familial Cases<br />

Martin N. Rossor, MD, Institute of Neurology, University College<br />

London, UK<br />

A major challenge in understanding the early natural history of Alzheimer’s<br />

disease (AD) has been that the disease is likely to be well established by the<br />

time patients present with memory complaints, even if they do not fulfill the<br />

criteria for a diagnosis of dementia. The identification of at risk individuals<br />

by virtue of age or mild symptoms are inevitably limited by either the<br />

numbers needed to study or by the late presentation. The elucidation of<br />

the genetics of autosomal dominant familial AD offers an opportunity of<br />

studying in detail the offspring of affected parents many years prior to the<br />

anticipated age at onset. The similarity of the clinical and pathological<br />

features of familial to sporadic AD suggests that the insights gained can be<br />

generalised to sporadic cases. Moreover, non-carrier siblings provide the<br />

ideal control group. Due to the relative rarity of autosomal dominant AD<br />

only small cohorts of APP and presenilin AD have previously been studied<br />

but have been able to show early changes in PiB binding and MRI atrophy<br />

before the onset of cognitive change. The analysis of longitudinal changes<br />

suggest a progression of tissue loss that starts in the medial temporal<br />

lobe and early cingulate cortex and spreads to involve association areas<br />

in a pattern similar to that in the Braak and Braak classification. Recently<br />

the multi-national study of dominantly inherited AD (DIAN: PI J. Morris)<br />

has provided the increased number of patients with cognitive imaging and<br />

CSF measures to establish a sequence of events in disease progression<br />

and to explore the heterogeneity across different mutations. DIAN also<br />

offers the prospect of prevention trials. The methodology of studying at<br />

risk familial dementias is also being applied to other disorders such as the<br />

frontotemporal dementias.<br />

References:<br />

1. Fox NC, Warrington EK, Seiffer AL, Agnew SK, Rossor MN.<br />

Presymptomatic cognitive deficits in individuals at risk of familial<br />

Alzheimer’s Disease. A longitudinal prospective study. Brain<br />

1998;121:1631-1639<br />

2. Scahill RI, Schott JM, Stevens JM, Rossor MN, Fox NC. Mapping the<br />

evolution of regional atrophy in Alzheimer’s disease: Unbiased analysis<br />

of fluid-registered serial MRI. Proceedings of the National Academy of<br />

Science 2002; 99(7): 4703-4707<br />

3. Klunk WE, Price JC, Mathis CA, Tsopelas ND, Lopresti BJ, Ziolko<br />

SK, Bi W, Hoge JA, Cohen AD, Ikonomovic MD, Saxton JA, Snitz BE,<br />

Pollen DA, Moonis M, Lippa CF, Swearer JM, Johnson KA, Rentz DM,<br />

Fischman AJ, Aizenstein HJ, DeKosky ST Amyloid deposition begins<br />

in the striatum of presenilin-1 mutation carriers from two unrelated<br />

pedigrees. J Neurosci. 2007 Jun 6;27(23):6174-84.<br />

4. Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC,<br />

Marcus DS, Cairns NJ, Xie X, Blazey TM, Holtzman DM, Santacruz A,<br />

Buckles V, Oliver A, Moulder K, Aisen PS, Ghetti B, Klunk WE, McDade<br />

E, Martins RN, Masters CL, Mayeux R, Ringman JM, Rossor MN,<br />

Schofield PR, Sperling RA, Salloway S, Morris JC; the Dominantly<br />

Inherited Alzheimer Network. Clinical and Biomarker Changes in<br />

Dominantly Inherited Alzheimer’s Disease N Engl J Med. <strong>2012</strong> Jul 11.<br />

[Epub ahead of print]<br />

Presentation of Raymond D. Adams Lectureship Award and<br />

Neurobiology of APOE and its Impact on Alzheimer’s Disease<br />

David M. Holtzman, MD, Washington University, St. Louis, Mo.<br />

The apolipoprotein E gene (APOE) is the strongest genetic risk factor for<br />

Alzheimer’s disease (AD) and also cerebral amyloid angiopathy (CAA). It<br />

also appears to play a role in outcome following head trauma and possibly<br />

other neurological disorders. APOE is located on chromosome 19 and in<br />

humans has 3 common isoforms APOE2, APOE3, and APOE4 which only<br />

differ from each other by a single amino acid (E2: cys 112, cys 158; E3<br />

cys 112, arg 158; E4 arg 112, arg 158). In regard to AD, relative to APOE3<br />

the most common allele, one copy of APOE4 increases risk for AD ~3.7<br />

fold while 2 copies increases risk by ~ 12-fold. Relative to APOE3, one<br />

allele of APOE2 decreases risk for AD by ~40%. APOE is an apolipoprotein<br />

expressed at highest levels in the liver but is also expressed at high levels<br />

in the CNS. In the periphery, APOE plays an important role in plasma<br />

cholesterol metabolism. It is the most abundant apolipoprotein expressed<br />

in the CNS where it secreted predominantly by astrocytes in high-density<br />

lipoprotein (HDL)-like particles. There are several ways that APOE has<br />

been proposed to influence the CNS both normally and in diseases states<br />

including effects on plasticity, inflammation, and cell signaling. However,<br />

there is abundant evidence that a major mechanism via which it influences<br />

both AD and CAA is by interacting with the amyloid- (A) peptide to<br />

affect both A clearance and aggregation. In humans, APOE4 results in a<br />

dose-dependent earlier onset of brain amyloid deposition and an increase<br />

in CAA while APOE2 results in a delay of amyloid deposition. A variety<br />

of transgenic mouse models that express human forms of the amyloid<br />

precursor protein (APP) develop amyloid deposition, neuritic plaques, and<br />

CAA with associated hemorrhages. APP transgenic mice in which apoE<br />

is knocked out develop very little to none of these changes. Expression<br />

of human APOE isoforms in APP transgenic mice results in restoration<br />

of amyloid deposition dose-dependently with the amount of amyloid<br />

pathology being in the order E4>E3>E2. In vivo microdialysis and other<br />

studies suggest that E4 slows soluble A clearance vs. E3 and E2 which<br />

may account for this differential effect. In some recent immunotherapy trials<br />

for AD involving passive administration of anti-A antibodies, E4-positive<br />

individuals have been found to have increased risk for development of<br />

amyloid related imaging abnormalities (ARIA) including vasogenic edema<br />

and cerebral microhemorrhages. The etiology of this complication is not<br />

completely clear but it is probably due to an interaction between anti-A<br />

antibodies and CAA which is present to a significantly greater extent in<br />

E4-positive individuals with AD. APOE may also be a therapeutic target for<br />

AD in that decreasing its levels in different ways results in less amyloid<br />

deposition and increasing its lipidation state also decreases amyloid<br />

deposition. Further studies are needed to better understand both the normal<br />

neurobiology of APOE, its exact role in neurological diseases, and its ability<br />

to be therapeutically targeted.<br />

References:<br />

1. Strittmatter, W.J., Saunders, A.M., Schmechel, D., Pericak-Vance, M.,<br />

Enghild, J., Salvesen, G.S., and Roses, A.D. (1993). Apolipoprotein E:<br />

high avidity binding to ß-amyloid and increased frequency of type 4<br />

allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA<br />

90, 1977-1981.<br />

2. Kim, J., Basak, J.M., and Holtzman, D.M. (2009). The role of<br />

apolipoprotein E in Alzheimer’s disease. Neuron 63, 287-303.<br />

29


MONDAY SPEAKER ABSTRACTS<br />

3. Verghese, P.B., Castellano, J.M., and Holtzman, D.M. (2011).<br />

Apolipoprotein E in Alzheimer’s disease and other neurological<br />

disorders. Lancet Neurology 10, 241-252.<br />

4. Castellano, J.M., Kim, J., Stewart, F.R., Jiang, H., DeMattos, R.B.,<br />

Patterson, B.W., Fagan, A.M., Morris, J.C., Mawuenyega, K.G.,<br />

Cruchaga, C., et al. (2011). Human apoE isoforms differentially<br />

regulate brain amyloid-beta peptide clearance. Science Translational<br />

Medicine 3, 89ra57.<br />

Mechanisms of Neurodegeneration: Lessons from Prion Disease<br />

John Collinge, MD, Institute of Neurology, University College London, UK<br />

Prions are lethal pathogens causing neurodegenerative diseases such<br />

as Creutzfeldt-Jakob disease in humans but which also affect a range of<br />

other animals. They appear devoid of nucleic acid and composed of<br />

polymerised conformational isomers of host-encoded cellular prion protein<br />

(PrP) 1 . Their unique biology, allied with the risks to public health posed by<br />

prion zoonoses such as bovine spongiform encephalopathy (BSE), has<br />

focussed much attention on understanding the molecular basis of prion<br />

propagation and the “species barrier” which controls cross-species<br />

transmission. Both are intimately linked to understanding how multiple<br />

prion “strains” are encoded by a protein-only agent, a challenging question<br />

in molecular biology which raises intriguing questions in evolution.<br />

It is increasingly clear that the underlying mechanisms, involving<br />

aggregation of a misfolded host protein, may be of much wider significance<br />

in commoner neurodegenerative diseases and contribute to their aetiology<br />

or to the spreading of pathology. Recent advances suggest that prions<br />

themselves are not directly neurotoxic, but rather their propagation leads<br />

to production of toxic species which may be uncoupled from infectivity.<br />

A general protein-only model has been proposed for prion propagation<br />

and neurotoxicity which may also be relevant to mechanisms of<br />

neurotoxicity in other neurodegenerative diseases associated with<br />

accumulation of misfolded proteins 1,2 . Therapeutic proof of principle has<br />

been achieved 3 and the development of effective treatment for prion<br />

neurodegeneration appears realistic; fully humanised anti-PrP monoclonal<br />

antibodies have been produced for clinical trial. In addition to a wider role<br />

for the general protein-only model, the proposal that prion protein may<br />

have a direct role in mediating aspects of neurotoxicity in Alzheimer’s<br />

disease is being investigated which might allow common therapeutic<br />

approaches and development 4 .<br />

References:<br />

1. Collinge, J. & Clarke, A. A general model of prion strains and their<br />

pathogenicity. Science 318, 930-936 (2007).<br />

2. Sandberg, M. K., Al Doujaily, H., Sharps, B., Clarke, A. R., & Collinge,<br />

J. Prion propagation and toxicity in vivo occur in two distinct<br />

mechanistic phases. Nature 470, 540-542 (2011).<br />

3. Mallucci G et al. Depleting neuronal PrP in prion infection prevents<br />

disease and reverses spongiosis. Science 302, 871-874 (2003).<br />

4. Freir, D. B. et al. Interaction between prion protein and toxic amyloid<br />

beta assemblies can be therapeutically targeted at multiple sites.<br />

Nature Communications 2, 336 (2011).<br />

Biomarkers for the Diagnosis and Course of<br />

Alzheimer’s Disease<br />

Randall J. Bateman, MD, Washington University, St. Louis, Mo.<br />

The order and magnitude of Alzheimer’s disease (AD) pathologic processes<br />

are not well understood, partly because AD develops over many years.<br />

Autosomal dominant Alzheimer’s disease (ADAD) has a predictable<br />

age at onset, and provides an opportunity to determine the sequence<br />

and magnitude of pathologic changes which culminate in symptomatic<br />

disease. Results of the in vivo metabolic production, exchange, and<br />

clearance of amyloid-beta isoforms in Presenilin mutation carriers<br />

will be presented. In addition, results from the Dominantly Inherited<br />

Alzheimer Network (DIAN) clinical, cognitive, MRI, FDG-PET, PIB-PET,<br />

CSF and blood tests will be reviewed and compared to sporadic AD.<br />

Cross sectional data analyzed using the estimated years to symptom<br />

onset will be reviewed to determine the relative order and magnitude of<br />

pathophysiological changes. Pathogenic mutations causing ADAD result in<br />

a series of changes beginning with systemic increased soluble amyloidbeta<br />

(A)42, followed by brain amyloidosis, increased CSF tau, decreased<br />

brain glucose metabolism, decreased brain volume, and subtle cognitive<br />

impairment before dementia is manifest. The sequence of events in AD<br />

pathophysiology offers the opportunity for predictive testing and secondary<br />

prevention trials. Because the clinical and pathological phenotypes of<br />

dominantly inherited AD are similar in many respects to those of the far<br />

more common late-onset AD, the nature and sequence of brain changes in<br />

ADAD are likely to be relevant for late-onset AD. (Clinicaltrials.gov number,<br />

NCT00869817)<br />

References:<br />

1. Bateman RJ, Xiong C, Benzinger TLS, Fagan AM, Goate A, Fox<br />

NC, et al. Clinical and Biomarker Changes in Dominantly Inherited<br />

Alzheimer’s Disease. New England Journal of Medicine.0(0):null. doi:<br />

doi:10.1056/NEJMoa1202753.<br />

2. Bateman RJ, Aisen PS, De Strooper B, Fox NC, Lemere CA, Ringman<br />

JM, et al. Autosomal-dominant Alzheimer’s disease: a review and<br />

proposal for the prevention of Alzheimer’s disease. Alzheimers Res<br />

Ther. 2011;3(1):1. Epub 2011/01/08. doi: alzrt59 [pii]10.1186/alzrt59.<br />

PubMed PMID: 21211070; PubMed Central PMCID: PMC3109410.<br />

3. Jack CR, Jr., Knopman DS, Jagust WJ, Shaw LM, Aisen PS,<br />

Weiner MW, et al. Hypothetical model of dynamic biomarkers of the<br />

Alzheimer’s pathological cascade. Lancet neurology. 2010;9(1):119-28.<br />

Epub 2010/01/20. doi: 10.1016/S1474-4422(09)70299-6. PubMed<br />

PMID: 20083042; PubMed Central PMCID: PMC2819840.<br />

4. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM,<br />

et al. Toward defining the preclinical stages of Alzheimer’s disease:<br />

recommendations from the National Institute on Aging-Alzheimer’s<br />

<strong>Association</strong> workgroups on diagnostic guidelines for Alzheimer’s<br />

disease. Alzheimers Dement. 2011;7(3):280-92. Epub 2011/04/26.<br />

doi: 10.1016/j.jalz.2011.03.003. PubMed PMID: 21514248; PubMed<br />

Central PMCID: PMC3220946.<br />

30


Imaging Structure and Function in Alzheimer’s Disease<br />

William J. Jagust, MD, University of California, Berkeley<br />

Imaging studies of patients with Alzheimer’s disease (AD), as well as<br />

studies of patients with Mild Cognitive Impairment (MCI) and normal<br />

aging, have informed our understanding of the role of -amyloid (A)<br />

in the pathogenesis of dementia. A conceptual model of dementia<br />

development postulates an early effect of A on a variety of “downstream”<br />

processes that probably involve the microtubule-associated protein tau,<br />

as well as a number of phenomena that can be measured in vivo. These in<br />

vivo measures, or biomarkers, include regionally specific brain atrophy,<br />

alterations in the connectivity of large scale brain networks, and changes<br />

in brain glucose metabolism that all reflect neurodegeneraion. In theory,<br />

by imaging A and these other biomarkers of neurodegeneration, one<br />

may not only detect evidence of AD in individuals with manifest dementia<br />

symptoms, but also detect AD in individuals in pre-dementia and even<br />

presymptomatic stages.<br />

This talk will explore the current evidence for this disease model. In<br />

particular, data on the prevalence of A deposition in the brain in normal<br />

aging, MCI, and AD will be reviewed, along with evidence that this A<br />

is associated with alterations in neuronal structure and function. The<br />

prognostic implications of A deposition in normal aging and MCI will<br />

also be reviewed, as well as the implications of brain atrophy, resting<br />

state functional connectivity, and glucose hypometabolism. In particular,<br />

evidence for the concept that AD exists in presymptomatic stages with<br />

various degrees of neurodegeneration will be presented. The application<br />

of PET amyloid imaging to the evaluation of patients with dementia,<br />

particularly those with unusual presentations, will also be discussed,<br />

contrasting effects related to A and the brain-behavior associations<br />

revealed with glucose metabolism. The application of these imaging<br />

modalities permits the in vivo examination of the cascade of pathological<br />

events that occurs in the progression of AD from presymptomatic stages<br />

to overt dementia. These data will indicate the extensive damage present<br />

in those with dementia symptoms, and the possibilities for selecting and<br />

treating individuals at presymptomatic disease stages.<br />

References:<br />

1. Perrin RJ, Fagan AM, Holtzman DM. Multimodal techniques for<br />

diagnosis and prognosis of Alzheimer’s disease. Nature,<br />

461:916-922, 2009<br />

2. Jack CR, Knopman DS, Jagust WJ, Shaw 461: LM, Aisen PS, Weiner<br />

MW, Petersen RC, Trojanowski JQ. Hypothetical model of dynamic<br />

biomarkers of the Alzheimer’s pathological cascade. Lancet Neurology,<br />

9:119-128. 2010.<br />

3. Rabinovici GD, Rosen HJ, Alkalay A, Kornak, J, Furst AJ, Agarwal N,<br />

Mormino EC, O’Neil JP, Janabi M, Karydas A, Growdon ME, Jang,<br />

JY, Huang EJ, DeArmond SJ, Trojanowski JQ, Grinberg LT, Gorno-<br />

Tempini ML, Seeley, WW, Miller BL, Jagust WJ. Amyloid versus<br />

FDG-PET in the differential diagnosis of AD and FTLD. Neurology,<br />

77: 2034-2042, 2011.<br />

4. Reiman EM, Jagust WJ. Brain imaging in the study of Alzheimer’s<br />

Disease. Neuroimage 61:505-516, <strong>2012</strong>.<br />

SYMPOSIUM: Results of Immune-Based Trials in<br />

<strong>Neurological</strong> Disorders<br />

Phase 3 Studies of Solanezumab for Mild to Moderate<br />

Alzheimer’s Disease<br />

Rachelle S. Doody, MD, PhD, Baylor College of Medicine, Houston<br />

Immune-based therapies in Alzheimer’s disease were initiated after<br />

seminal studies showed that immunization with aggregated A42 reduced<br />

development of and stimulated clearance of amyloid plaques in amyloid<br />

precursor protein (APP) transgenic mice which overproduce beta amyloid<br />

in the brain (1). A trial of this approach (which also employed the adjuvant<br />

QS-21) in patients with mild to moderate AD was stopped early due to the<br />

adverse occurrence of meningoencephalitis in six percent of the immunized<br />

patients (2). Subsequently, both active and passive immunization strategies<br />

have been pursued, with the therapeutic agents differing in their methods of<br />

delivery, antigenic targets, binding properties, and the degree to which they<br />

enter the central nervous system.<br />

Solanezumab is a humanized anti--amyloid monoclonal antibody that<br />

binds to soluble A species and can foster its clearance from brain (3).<br />

The antigenic target for solanezumab is the mid-region (epitopes 16-23) or<br />

central domain of A, and it preferentially binds to the soluble forms of A<br />

(3). A pre-clinical study comparing passive A antibodies with N-terminal<br />

versus central domain antigenic targets suggested that solanezumab may<br />

be less likely to induce vasogenic edema compared to N-terminally directed<br />

antibodies (4). Phase 1 and phase 2 studies supported the safety of this<br />

agent for further testing in AD patients and showed changes in peripheral<br />

and central amyloid levels as a result of treatment (5,6).<br />

Eli Lilly and Company has completed two identical phase 3 studies of<br />

solanezumab in patients with mild to moderate AD (NINCDS-ADRDA<br />

criteria). A total of 2052 patients were randomized to receive 400 mg<br />

solanezumab IV or placebo every four weeks for 80 weeks. Co-primary<br />

outcomes were the Alzheimer’s Disease Assessment Scale-cognitive<br />

subscale (ADAS-cog11) and the Alzheimer’s Disease Cooperative<br />

Study-Activities of Daily Living Scale (ADCS-ADL). All subjects underwent<br />

magnetic resonance imaging at weeks 12, 28, 52, and 80; and subsets<br />

underwent Florbetapir PET imaging/lumbar puncture at baseline and<br />

endpoint. We will present results for the co-primary efficacy measures,<br />

safety, and some biological markers and will discuss how the results of<br />

these studies influence our understanding of the amyloid hypothesis and<br />

AD therapeutics.<br />

References:<br />

1. Schenk D, etal Nature 1999;400-173-177 (Letter)<br />

2. Gilman S, etal Neurology 2005;64:1553-1562<br />

3. Samadi H and Sultzer D Expert Opin Biol Ther 2011:1-12<br />

4. Racke M, etal Neurobiol Disease 2005;25(3):629-636<br />

5. Siemers E, etal Clin Neuropharm 2010;33(2):67-73<br />

6. Farlow M, etal Alzheimer’s & Dem <strong>2012</strong> do1:10.1016/j.jalz2011.09.224<br />

31


MONDAY SPEAKER ABSTRACTS<br />

Phase III Studies of Bapineuzumab for Mild to<br />

Moderate Alzheimer’s Disease Dementia<br />

Reisa A. Sperling, MD, Brigham and Women’s Hospital, Boston<br />

Amyloid- accumulation is one of the hallmark pathologic processes<br />

that defines Alzheimer’s disease (AD). Bapineuzumab is an N-terminus,<br />

anti-amyloid- monoclonal antibody being evaluated in a Phase 3 clinical<br />

trial program for the treatment of AD. The Phase 3 studies enrolled AD<br />

patients with mild to moderate dementia severity (MMSE 16-26) into four<br />

78 week double-blind, placebo-controlled trials. Results of the first two of<br />

these studies are presented here.<br />

Separate Phase 3 trials, with differing dose levels of bapineuzumab,<br />

were designed for APOE 4 allele carriers and non-carriers as Phase 2<br />

data suggested possible differences between these populations. The<br />

non-carrier (301) study was initially designed to test 3 dose levels of<br />

bapineuzumab (0.5, 1.0, and 2.0 mg/kg) vs. placebo. The 2.0mg/kg dose<br />

was stopped approximately one year into the study due to the incidence<br />

of amyloid-related imaging abnormalities (ARIA) thought to represent<br />

vasogenic edema. The carrier (302) study tested only 1 dose level<br />

(0.5mg/kg) vs. placebo.<br />

Bapineuzumab or saline placebo was administered intravenously every<br />

13 weeks over the 78 week trial. Safety MRIs to monitor for ARIA were<br />

performed 6 weeks after each infusion. The co-primary clinical endpoints<br />

are change in the Alzheimer’s Disease Assessment Scale – Cognitive<br />

subscale (ADAS-Cog, a composite cognitive measure) and the Disability<br />

Assessment for Dementia (DAD, functional measure of activities of daily<br />

living). Biomarker substudies assessing effects of bapineuzumab on PiB-<br />

PET imaging estimates of brain amyloid burden, cerebrospinal fluid (CSF)<br />

phospho-tau, and brain volume on structural MRI, were included. Statistical<br />

analyses were performed to estimate treatment differences at study endpoint<br />

through mixed models for repeated measures and analysis of covariance.<br />

The APOE 4 non-carrier study enrolled and dosed 1,331 AD patients,<br />

and the APOE 4 carrier study randomized and dosed 1,121 AD patients,<br />

including 4 hetero- and homozygotes. The primary efficacy outcomes<br />

(ADAS-Cog and DAD) for both carrier and non-carrier studies will be<br />

presented. Biomarker endpoints (PiB PET, CSF phospho-tau, volumetric<br />

MRI) will also be presented, as well as safety data, including ARIA findings<br />

on MRI and other adverse events.<br />

These two Phase 3 clinical trials were designed to provide a robust<br />

evaluation of the efficacy and safety of bapineuzumab treatment in AD<br />

patients with mild-moderate dementia as a potential disease-modifying<br />

therapy, based on a combination of clinical and biomarker evidence. The<br />

implications of findings across the APOE 4 carrier and non-carrier studies<br />

will be discussed. The results will also be discussed in the context of the<br />

plans for earlier intervention studies with anti-amyloid immunotherapeutic<br />

agents, including secondary prevention trials in asymptomatic genetic atrisk<br />

and biomarker at-risk populations.<br />

Sperling R, Salloway S, Raskind M, Ferris S, Honig L, Porsteinsson A,<br />

Sabbagh M, Liu E, Yuen E, Lull J, Miloslavsky M, Reichert M, Ketter N,<br />

Lu Y, Wang D, Nejadnik B, Guenzler V, Grundman M, Black R, Brashear HR<br />

References:<br />

1. Black R, Sperling RA, Safirstein B, Motter RN, Pallay A, Nichols A,<br />

Grundman M. A single ascending dose study of bapineuzumab in<br />

patients with Alzheimer’s disease. Alzheimer’s Disease and Associated<br />

Disorders 2010 Apr-Jun; 24(2):198-203. PMID: 20505438<br />

2. Salloway SP, Sperling RA, Gilman S, Fox NC, Blennow K, Raskind M,<br />

Sabbagh M, Honig LS, Doody R, van Dyck CH, Mulnard R,Barakos J,<br />

Gregg K, Liu E, Lieberberg I, Schenk D, Black R and Grundman M. A<br />

Phase 2 multiple ascending dose trial of bapineuzumab in mild to<br />

moderate Alzheimer’s disease. Neurology 2009, Dec 15; 73(24):2061-<br />

70. PMCID: PMC2790221<br />

3. Sperling RA, Salloway S, Brooks, DJ, Tampieri D, Barakos J, Fox NC,<br />

Raskind M, Sabbagh M, Honig LS, Porsteinsson AP, Lieberburg I,<br />

Arrighi HM, Morris KA, Lu Y, Liu E, Gregg KM, Brashear RH, Kinney<br />

GG, Black R, Grundman M. Amyloid-related imaging abnormalities<br />

(ARIA) in Alzheimer’s disease patients treated with bapineuzumab:<br />

A retrospective analysis. Lancet Neurology <strong>2012</strong> Mar; 11(3):241-49<br />

PMID: 22305802<br />

Phase III Trials of Alemtuzumab in Relapsing-Remitting<br />

Multiple Sclerosis<br />

Alastair Compston, MBBS, PhD, FmedSci, University of Cambridge, UK<br />

Lymphocyte depletion has been used over many years to treat multiple<br />

sclerosis. Campath-1H (alemtuzumab), the first humanized monoclonal<br />

antibody, targets CD52 and rapidly depletes circulating lymphocytes. It<br />

was first used in the context of multiple sclerosis in 1991. After it became<br />

clear that lymphocyte depletion is not clinically effective once patients have<br />

moved to the progressive phase of the disease, open-label studies and a<br />

phase 2 clinical trial with an active comparator, involving 334 previously<br />

untreated patients with early-stage relapsing-remitting multiple sclerosis,<br />

showed that alemtuzumab reduces the number of episodes by 74 per cent,<br />

and reduces the risk of sustained accumulation of disability by 71 per cent<br />

compared to interferon beta-1a. Many individuals receiving alemtuzumab<br />

were less disabled after three years than at the beginning of the study, in<br />

contrast to worsening disability in the interferon beta-1a treated patients.<br />

The main adverse effect of treatment was a 25% risk of developing another<br />

autoimmune disease as the immune system reconstitutes following<br />

exposure to alemtuzumab. A further period of observation, now extended<br />

to at least 5 years for all participants, confirms the superior efficacy of<br />

alemtuzumab in this study. Two phase 3 trials are also now completed.<br />

In CARE-MS1 treatment-naïve patients with relapsing-remitting multiple<br />

sclerosis were randomized to pulsed treatment with alemtuzumab (n=376)<br />

at baseline and 12 months, or continuous interferon beta-1a (n=187), and<br />

assessed at two years for clinical and MRI outcomes. CARE-MS2 used<br />

the same protocol but recruited patients with relapsing-remitting multiple<br />

sclerosis who had already experienced 1 relapse on interferon beta or<br />

glatiramer. In CARE-MS1, alemtuzumab reduced annualized relapse rate<br />

by 55% compared to interferon beta-1a; 8% of alemtuzumab patients<br />

experienced sustained accumulation of disability versus 11% of interferon<br />

beta-1a patients. Alemtuzumab reduced the proportions of patients with<br />

gadolinium-enhancing lesions and new or enlarging T2-hyperintense<br />

32


lesions and reduced brain volume loss by 40%, all compared to interferonbeta.<br />

In CARE-MS2, alemtuzumab reduced relapse rate by 49% compared<br />

to interferon beta-1a, risk of sustained accumulation of disability by<br />

42%, new gadolinium or T2 lesions by 60% and 32% respectively, and<br />

brain volume loss by 0.20%. Sustained reduction in disability was more<br />

likely after alemtuzumab. In both studies the frequency of secondary<br />

autoimmunity was c20%; no new safety signals were observed.<br />

Inflammation and axon degeneration are each involved in the pathogenesis<br />

of multiple sclerosis. The evaluation of alemtuzumab has revealed the<br />

relative contribution of inflammatory and neurodegenerative events<br />

underlying the natural history of multiple sclerosis as this evolves from the<br />

relapsing-remitting to progressive phase of the disease; identified novel<br />

mechanisms of symptom production reflecting altered axonal structure and<br />

function; provided a ‘human’ model of lymphocyte reconstitution that has<br />

explained general principles of autoimmunity; demonstrated the critical<br />

importance of timing in order to achieve high efficacy for immunological<br />

treatments in multiple sclerosis; shown that – at least over a period of 10<br />

years – lymphocyte depletion early in the course of relapsing-remitting<br />

multiple sclerosis stabilises the natural history of the disease; demonstrated<br />

that fixed disability in multiple sclerosis may recover; and provided<br />

evidence for the mechanisms and effect of protective autoimmunity in the<br />

human central nervous system.<br />

References:<br />

1. International CAMMS223 Trial Study Group. A randomized, raterblinded,<br />

trial of alemtuzumab versus interferon beta-1a in early,<br />

relapsing-remitting multiple sclerosis. New England Journal of<br />

Medicine 2008: 359; 1786-1801<br />

2. Coles AJ, Fox EJ, Vladic A, Gazda SK, Brinar VV, Selmay K, Skoromets<br />

AA, Stolyarov I, Bass A, Sullivan H, Margolin DH, Lake SL, Moran<br />

S, Palmer J, Smith MS, Compston DAS Alemtuzumab treatment<br />

of multiple sclerosis: five-year follow-up of the CAMMS223 trial.<br />

Neurology <strong>2012</strong>: 78; 1069-78<br />

3. Coles AJ, Twyman CL, Arnold DL, Cohen JA, Confavreux C, Fox E,<br />

Hartung H-P, Havrdova E, Selmaj K, Weiner HL, Miller T, Fisher E,<br />

Sandbrink R, Lake SL, Margolin DH, Oyuela P, Panzara MA, Compston<br />

DAS for the CARE-MS 2 investigators. Alemtuzumab in multiple<br />

sclerosis relapsing on disease-modifying therapy (submitted)<br />

4. Cohen JA, Coles AJ, Arnold DL, Confavreux C, Fox EJ, Hartung H-P,<br />

Havrdova E, Selmaj KW, Weiner HL, Fisher E, Brinar VV, Giovannoni G,<br />

Stojanovic M, Ertik BI, Lake SL, Margolin DH, Panzara MA, Compston<br />

DAS for the CARE-MS I investigators. Alemtuzumab versus interferon<br />

beta as initial multiple sclerosis treatment (submitted).<br />

33


TUESDAY SPEAKER ABSTRACTS<br />

SYMPOSIUM: Derek Denny-Brown New<br />

Member Symposium<br />

Presentation of Derek Denny-Brown <strong>Neurological</strong><br />

Scholar Award: Clinical Science and Temporal Trends in<br />

Acute Stroke Management<br />

Dawn Kleindorfer, MD, University of Cincinnati, Cincinnati, Oh.<br />

There is no doubt that the approval of rt-PA has revolutionized the field<br />

of stroke. However, it is not clear how often stroke patients are actually<br />

receiving rt-PA. This deceptively simple question is actually very difficult<br />

to answer. There is no perfect database to answer these types of questions.<br />

Administrative data lacks clinical detail, and represents a sampling of<br />

stroke patients. Clinical databases lack generalizability, and typically have<br />

significant referral biases. However, utilizing an administrative database<br />

with linked pharmacy data, we estimated that the rates of rt-PA utilization<br />

were initially very low and flat, but finally started increasing around 2004 1 .<br />

This timing of this coincided with the institution of primary stroke centers<br />

accreditation in 2004, and did not appear to be as influenced by changes<br />

in hospital reimbursement rates for rt-PA patients as we had hypothesized.<br />

In 2009, we estimate that 4.5% of hospitalized ischemic stroke patients<br />

received rt-PA.<br />

The next logical question becomes, “Why are the rates of rt-PA use so low”.<br />

To answer that, one must understand the eligibility of stroke patients for<br />

rt-PA, which requires more detailed clinical data from an entire population. It<br />

turns out that only 6-8% of ischemic stroke patients are eligible for rt rt-PA<br />

PA within a population 2 , mostly driven by delayed arrival times of patients<br />

to medical attention 3 . Recent attempts to expand the limited time window in<br />

which rt-PA can be given likely will not significantly improve treatment rates,<br />

as patients either arrive very early or very late. Other targets, such as treating<br />

infarcts with milder severity, may be better targets for improvement.<br />

We also began exploring the impact hospital-level factors and their impact<br />

on rates of rt-PA use. We were especially interested in the role of primary<br />

stroke centers and how they impacted hospitallevel rates of rt-PA utilization.<br />

Many had thought that those centers choosing to certify would already be at<br />

a high level of treatment, and the certification process would not change this,<br />

or that the effect would “wear off” over time. We found exactly the opposite,<br />

that centers seeking certification had dramatically increased rates of rt-PA<br />

use that was relatively constant for the three years pre- and post-certification.<br />

Finally, the Center for Medicare Services has become interested in measuring<br />

and publicly ranking hospitals regarding the quality of stroke care. By<br />

working with the incredibly productive Get With the Guidelines team, we were<br />

able to prove that comparisons between hospitals must account for initial<br />

stroke severity, or else those hospitals providing the highest quality care to<br />

the sickest patients will likely be unfairly portrayed. 4<br />

In order to improve our public’s health for stroke, the fourth leading cause<br />

of death, we should make every attempt to understand the true quality of<br />

care provided and the proportion of patients eligible for new therapeutic<br />

approaches. Without accurate and precise measurement of how often and to<br />

whom acute stroke treatments are given, we cannot measure our success nor<br />

advance the implementation of these proven therapies.<br />

References:<br />

1. Adeoye O, Hornung R, Khatri P, Kleindorfer D. Recombinant tissue-type<br />

plasminogen activator use for ischemic stroke in the united states:<br />

A doubling of treatment rates over the course of 5 years. Stroke. 2011<br />

2. Kleindorfer D, Kissela B, Schneider A, Woo D, Khoury J, Miller R,<br />

Alwell K, Gebel J, Szaflarski J, Pancioli A, Jauch E, Moomaw C,<br />

Shukla R, Broderick JP. Eligibility for recombinant tissue plasminogen<br />

activator in acute ischemic stroke: A population-based study. Stroke.<br />

2004;35:e27-29<br />

3. Kleindorfer D, Alwell, K, Khoury, J, Ewing, I, Schneider, A, Flaherty,<br />

ML, Moomaw, C, Khatri, P, Stettler, B, Broderick JP. Temporal trends in<br />

emergency department arrival times for acute ischemic stroke:<br />

A population-based study. Stroke. 2005;36:494<br />

4. Fonarow G PW, Saver JL, Smith EE, Reeves MJ, Broderick JP, Kleindorfer<br />

DO, Sacco RL, Olson DM, Hernandez AF, Peterson ED, Schwamm<br />

LH. Comparison of 30-day mortality models for proflining hospital<br />

performance in acute ischemic stroke with vs. Without adjustment<br />

for stroke severity. Journal of the <strong>American</strong> Medical <strong>Association</strong>.<br />

<strong>2012</strong>;308.:257-264<br />

Presentation of Derek Denny-Brown <strong>Neurological</strong> Scholar Award:<br />

Basic Science and A Large Repeat Expansion in the C9ORF72<br />

Gene is a Common Cause of ALS and FTD<br />

Bryan J. Traynor, MB, MD, PhD, MMSc, MRCPI, National Institute on Aging,<br />

National Institutes of Health, Bethesda, Md.<br />

Amyotrophic lateral sclerosis (ALS; Lou Gehrig’s disease) is a fatal<br />

neurodegenerative disorder that leads to rapidly progressive paralysis<br />

and respiratory failure. ALS is the third most common neurodegenerative<br />

disease in the Western world, and there are currently no effective therapies.<br />

Frontotemporal dementia (FTD) is the most common form of dementia in<br />

the population under the age of 65. An overlap between these two clinically<br />

distinct neurological diseases has long been recognized, but the molecular<br />

basis of this intersection was unknown.<br />

Recently, the Neuromuscular Diseases Research Unit (NDRU), a part of the<br />

Laboratory of Neurogenetics at the National Institute on Aging, identified the<br />

major genetic cause of both ALS and FTD. To do this, Dr. Traynor (chief of<br />

NDRU) organized a worldwide consortium, bringing together groups that<br />

had previously been competitors to focus their efforts towards identifying<br />

this gene. This was made possible by the next generation sequencing<br />

technologies available at the NIH. This innovative approach worked, and his<br />

group published the cause of chromosome 9-linked ALS/FTD in the journal<br />

Neuron in September 2011. 1,2 In these cases, the disease is caused by a six<br />

base pair segment of DNA that is pathologically repeated over and over again,<br />

up to several thousand times. This so-called large hexanucleotide repeat<br />

disrupts the C9ORF72 gene located on chromosome 9, and is the most<br />

common genetic cause of both ALS and FTD identified to date, accounting<br />

for approximately 40% of all familial cases of ALS and FTD in European and<br />

North <strong>American</strong> populations. Further, Dr. Traynor’s group has shown that<br />

this mutation underlies about 8% of cases of sporadically occurring ALS and<br />

FTD that lack a family history. 3 This represents the first time that a common<br />

genetic cause has been identified for the sporadic form of these diseases. In<br />

a separate publication in The New England Journal of Medicine, they have<br />

34


also shown that the same large hexanucleotide repeat expansion underlies<br />

~1% of cases clinically diagnosed with Alzheimer’s Disease. 4 A one percent<br />

reduction in the number of AD cases would represent approximately $1<br />

billion in healthcare cost savings annually.<br />

The discovery of the C9ORF72 hexanucleotide repeat expansion is a<br />

landmark discovery in our understanding of neurodegenerative disease.<br />

It has already greatly effected how these diseases are diagnosed,<br />

investigated and perceived, and provides a mechanistic link between<br />

two clinically distinct disorders, ALS and FTD. It also provides a distinct<br />

therapeutic target for gene therapy efforts aimed at ameliorating the disease,<br />

and such efforts are already well underway.<br />

References:<br />

1. Renton AE, et al. A hexanucleotide repeat expansion in C9ORF72 is<br />

the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011;<br />

72: 257–68.<br />

2. DeJesus-Hernandez M, et al. Expanded GGGGCC hexanucleotide<br />

repeat in noncoding region of c9orf72 causes chromosome 9p-linked<br />

FTD and ALS. Neuron 2011; 72: 245–56.<br />

3. Majounie E, et al. Frequency of the C9orf72 hexanucleotide repeat<br />

expansion in patients with amyotrophic lateral sclerosis and<br />

frontotemporal dementia: a cross-sectional study. Lancet Neurol. <strong>2012</strong>;<br />

11:323–30<br />

4. Majounie E, et al. Repeat expansion in C9ORF72 in Alzheimer’s<br />

Disease. N Engl J Med. <strong>2012</strong>; 366:283–4<br />

Pilot Clinical Trial of Eculizumab in AQP4-IgG-Positive NMO<br />

Sean J. Pittock, MD, Mayo Clinic, Rochester, Minn.<br />

Objective: Investigate safety and efficacy of blocking terminal complement<br />

activation in reducing frequency of neuromyelitis optica (NMO) relapses.<br />

We conducted an open-label study of eculizumab, an inhibitor of C5<br />

cleavage, in patients with aggressive NMO.<br />

Methods: 14 adults with NMO spectrum disorder ( 2 relapses in the<br />

preceding 6 months or 3 in the preceding 12 months) were treated<br />

with eculizumab for 1 year. 7 had failed standard immunosuppressant<br />

treatments.<br />

Results: After 12 months treatment, 12 of 14 were relapse-free. The<br />

median annualized attack rate declined from 3 (pre-eculizumab) to 0 (oneculizumab;<br />

p


TUESDAY SPEAKER ABSTRACTS<br />

direct miR-183 binding to their 3’UTRs. Moreover, elevated levels of miR-<br />

183 can block Akt1 and mTOR translation in growing axons. Taken together,<br />

these observations suggest that microRNAs can regulate local translation of<br />

specific genes directly and control protein synthesis broadly by modulating<br />

the activity of mTORC1 and mTORC2 and highlight a new molecular<br />

mechanisms contributing to SMA pathology.<br />

Min J. Kye, PhD 1 , Mary H. Wertz 1 , Bikem Akten, PhD 1 , Pierre Neveu, PhD 2 ,<br />

Kenneth S. Kosik, MD 3 and Mustafa Sahin, MD, PhD 1 . 1 Neurology, Boston<br />

Children’s Hospital, Boston, MA, 02115; 2 Cell Biology and Biophysics Unit,<br />

European Molecular Biology Laboratory, Heidelberg, Germany, 60117;<br />

3<br />

Neuroscience Research Institute, University of California at Santa Barbara,<br />

Santa Barbara, CA 93106.<br />

Presentation of The Grass Foundation – ANA Award in<br />

Neuroscience Award and Multiscale Investigations of Epilepsy,<br />

Sleep and Cognition<br />

Sydney S. Cash, MD, PhD, Massachusetts General Hospital, Boston<br />

Both normal and abnormal activity in the brain involves actions at<br />

multiple different scales. From ion channels, to individual neurons, to<br />

widespread neuronal interactions encompasses cortical and subcortical<br />

structures, activities at different levels interact to culminate in particular<br />

behaviors. Unraveling the mechanisms at these different scales, and the<br />

nature of interactions across scales remains a tremendous challenge in<br />

the neurosciences and neurology. Our group has focused on making use<br />

of the unprecedented spatiotemporal information obtainable from patients<br />

undergoing invasive physiological monitoring for epilepsy to explore the<br />

different scales of neuronal activity during normal cognitive processing,<br />

sleep and epileptic activity. We augment the already substantial capabilities<br />

of routine, clinical recordings with different forms of microelectrodes<br />

which record the activity from small groups of neurons and even<br />

individual neurons.<br />

In this brief talk we will start with a summary of the different techniques<br />

that we are currently using with an emphasis and the different scales of<br />

information that can be obtained from each system. These modalities include<br />

non-invasive approaches of EEG and MEG as well as routine invasive,<br />

intracranial recordings techniques. They also include microelectrodes<br />

which are either placed on the pial surface or within the parenchyma of<br />

the brain. These later devices include microgrids and microwires, laminar<br />

microelectrode arrays and the NeuroPort microelectrode array.<br />

Using these systems we have found that epileptic activity in the brain<br />

involves a complex interplay of inhibitory and excitatory neuronal activity<br />

which does not strictly demonstrate pure hypersynchrony or hyperexcitability<br />

[1-2]. Analysis of individual neuronal firing during both interictal discharges<br />

and seizures themselves reveals a wide variety of different neuronal firing<br />

patterns. Furthermore, there are hints that some neurons may change their<br />

firing activity well in advance of the event itself. Examination of larger scale<br />

dynamics, at the level of the cortex as a whole, also shows complex behavior<br />

of the entire network with periods of the seizure (primarily mid-seizure)<br />

in which there is less synchrony than might be expected [3]. From these<br />

combined, multi-scalar, data emerges a model of the seizure as a process<br />

which is heterogeneous in space and temporally dynamic.<br />

36<br />

Similar investigations of typical sleep activity also reveal more complexity in<br />

both space and time than is expected from early analysis of scalp EEG alone.<br />

Sleep spindles, for example, show a variety of different localizations across<br />

the brain during sleep. The combined microscale and macroscale data has<br />

led to revisions of existing models of thalamocortical interactions with a new<br />

emphasis on differences between core and matrix projections accounting for<br />

different aspects of the spindle and carrying implications for processing of<br />

information during sleep .<br />

Finally, multiscale analysis of language processing suggests widespread<br />

cortical involvement in even simple semantic tasks – even in areas outside<br />

traditional language cortex [4]. At the level of the single neuron this aspect<br />

is revealed in individual neuronal responses to particular word sounds in the<br />

superior temporal gyrus.<br />

Together, these studies exemplify the power and possibilities of detailed<br />

neurophysiological examination which takes into account and tries to<br />

understand the interplay of activity across scales. While in their early<br />

stages, these types of investigation will hopefully shed substantial light on<br />

both normal and abnormal processing in the brain in a way which can be<br />

leveraged for novel therapeutic benefit.<br />

References:<br />

1. Keller, C.J., et al., Heterogeneous neuronal firing patterns during<br />

interictal epileptiform discharges in the human cortex. Brain, 2010.<br />

133(Pt 6): p. 1668-81.<br />

2. Truccolo, W., et al., Single-neuron dynamics in human focal epilepsy.<br />

Nat Neurosci, 2011.<br />

3. Kramer, M.A., et al., Coalescence and fragmentation of cortical networks<br />

during focal seizures. J Neurosci, 2010. 30(30): p. 10076-85.<br />

4. Chan, A.M., et al., Decoding word and category-specific spatiotemporal<br />

representations from MEG and EEG. Neuroimage, 2011. 54(4):<br />

p. 3028-39.<br />

Congenital Myasthenic Syndrome (CMS), Autophagic Myopathy,<br />

and Cognitive Dysfunction Caused by Mutations in DPAGT1<br />

Duygu Selcen, MD, Mayo Clinic, Rochester, Minn.<br />

We recently identified two CMS patients with CNS involvement. Patient 1<br />

is a16-year-old mentally retarded girl with severe generalized CMS since<br />

infancy. One of her siblings is also affected and has autistic features.<br />

Patient 2 is a 14-year-old girl with mild cognitive deficits and progressive<br />

limb-girdle CMS since infancy. Both respond poorly to anti-AChE therapy.<br />

Intercostal muscle specimens in both show small tubular aggregates in<br />

type 2 fibers, type1 fiber atrophy, and a vacuolar myopathy with autophagic<br />

features. Endplate studies reveal that quantal release, postsynaptic response<br />

to acetylcholine quanta, and endplate AChR content are reduced to ~50%<br />

of normal. Quantitative EM of 65 endplate regions shows hypoplastic<br />

endplates, very small nerve terminals, and poorly differentiated postsynaptic<br />

regions. Neither patient harbors mutations in currently recognized CMS<br />

disease genes but exome sequencing in each identifies two heteroallelic<br />

mutations in DPAGT1 coding for dolichyl-phosphate N-acetylglucosamine<br />

phosphotransferase, an enzyme subserving protein N-glycosylation.<br />

Immunoblots of muscle extracts probed by two different antibodies<br />

demonstrates reduced to absent glycosylation of ~70 kDa proteins. We<br />

hypothesize that hypoglycosylation of synapse-specific proteins causes<br />

defects in motor as well as central synapses.


Duygu Selcen, MD1, XinMing Shen, PhD1, Ying Li, PhD2, Eric Wieben,<br />

PhD3 and Andrew G. Engel, MD1. 1Neurology, Mayo Clinic, Rochester,<br />

MN, 55905; 2Biomedical Statistics and Informatics, Mayo Clinic, Rochester<br />

and 3Biochemistry and Molecular Biology, Mayo Clinic, Rochester.<br />

Anti-NMDA Receptor Encephalitis, a Series of 212 Children<br />

Maarten Titulaer, MD, PhD, IDIBAPS/University of Barcelona<br />

Since its discovery in 2007, anti-NMDAR encephalitis has been<br />

recognized as one of the most frequent autoimmune encephalitis. We<br />

report a longitudinal cohort study of 212 children, focusing on symptom<br />

presentation, treatment, and outcome at 24 months, and provide<br />

treatment guidelines.<br />

73% of patients were girls. 41% of girls 12 years had ovarian teratoma(s)<br />

compared with 6%


TUESDAY SPEAKER ABSTRACTS<br />

3. Burstein R, Jakubowski M, Garcia-Nicas E, Kainz V, Bajwa Z, Hargreaves<br />

R, et al. Thalamic sensitization transforms localized pain into widespread<br />

allodynia. Ann Neurol. 2010;68(1):81-91.<br />

4. Burstein R, Jakubowski M, Collins B. Defeating migraine pain with<br />

triptans: A race against the development of cutaneous allodynia. Ann<br />

Neurol. 2004;55(1):19-26.<br />

5. Noseda R, Kainz V, Jakubowski M, Gooley JJ, Saper CB, Digre K, et al.<br />

A neural mechanism for exacerbation of headache by light. Nat Neurosci.<br />

2010;13(2):239-45.<br />

F.E. Bennett Memorial Lectureship: Finding and Exploiting<br />

Neuropathic Pain Genes<br />

Clifford Woolf, MD, PhD, Children’s Hospital of Boston<br />

Loss function recessive mutations in certain key “pain genes” like Nav1.7, can<br />

produce a large phenotype, such as congenital insensitivity to pain, but only in<br />

populations where consanguinity is present, and therefore, such probands are<br />

extremely rare. Both acute pain sensitivity and the risk of developing chronic<br />

pain are, however heritable, with an estimated ~50% genetic contribution. The<br />

challenge is identifying the common genetic polymorphisms responsible, which<br />

will require genome-wide screens in large, well phenotyped cohorts. In the<br />

meanwhile, candidate gene studies based on unbiased preclinical genome-wide<br />

profiling in Drosophila and rodents have revealed that the while the relative risk<br />

produced by each pain gene in most cases is likely to be small, cumulatively<br />

they will progressively help identify individuals at high risk. Furthermore,<br />

identifying pain-related genes provides a powerful means of validating in<br />

patients, targets for the development of novel analgesics.<br />

Costigan M, Belfer I, Griffin RS, Dai F, Barrett LB, Coppola G, Wu T, Kiselycznyk<br />

C, Poddar M, Lu Y, Diatchenko L, Smith S, Cobos EJ, Zaykin D, Allchorne A,<br />

Shen PH, Nikolajsen L, Karppinen J, Männikkö M, Kelempisioti A, Goldman D,<br />

Maixner W, Geschwind DH, Max MB, Seltzer Z, Woolf CJ. Multiple chronic pain<br />

states are associated with a common amino acid-changing allele in KCNS1.<br />

Brain. 2010 133:2519-27<br />

Neely GG, Hess A, Costigan M, Keene AC, Goulas S, Langeslag M, Griffin RS,<br />

Belfer I, Dai F, Smith SB, Diatchenko L, Gupta V, Xia CP, Amann S, Kreitz S,<br />

Heindl-Erdmann C, Wolz S, Ly CV, Arora S, Sarangi R, Dan D, Novatchkova<br />

M, Rosenzweig M, Gibson DG, Truong D, Schramek D, Zoranovic T, Cronin<br />

SJ, Angjeli B, Brune K, Dietzl G, Maixner W, Meixner A, Thomas W, Pospisilik<br />

JA, Alenius M, Kress M, Subramaniam S, Garrity PA, Bellen HJ, Woolf CJ,<br />

Penninger JM. A genome-wide Drosophila screen for heat nociception identifies<br />

23 as an evolutionarily conserved pain gene. Cell. 2010 143:628-38<br />

Sorge RE, Trang T, Dorfman R, Smith SB, Beggs S, Ritchie J, Austin JS, Zaykin<br />

DV, Meulen HV, Costigan M, Herbert TA, Yarkoni-Abitbul M, Tichauer D,<br />

Livneh J, Gershon E, Zheng M, Tan K, John SL, Slade GD, Jordan J, Woolf CJ,<br />

Peltz G, Maixner W, Diatchenko L, Seltzer Z, Salter MW, Mogil JS. Genetically<br />

determined P2X7 receptor pore formation regulates variability in chronic pain<br />

sensitivity. Nature Medicine. <strong>2012</strong> 18:595-599<br />

Tegeder I, Costigan M, Griffin RS, Abele A, Belfer I, Schmidt H, Ehnert C,<br />

Nejim J, Marian C, Scholz J, Wu T, Allchorne A, Diatchenko L, Binshtok<br />

AM, Goldman D, Adolph J, Sama S, Atlas SJ, Carlezon WA, Parsegian<br />

A, Lotsch J, Fillingim RB, Maixner W, Geisslinger G, Max MB, Woolf CJ.<br />

GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and<br />

persistence. Nature Medicine. 2006 12:1269-1277.<br />

38<br />

Mining Nociceptor Signaling for Pain Therapeutics<br />

Robert O. Messing, MD, University of California, San Francisco<br />

Recent evidence indicates that acute activation of pain pathways can provoke<br />

long-lasting changes in signal transduction within peripheral sensory<br />

pain neurons (nociceptors) that render these neurons hyper-responsive.<br />

This phenomenon, termed “hyperalgesic priming”, has been modeled in<br />

animals where inflammatory mediators, such as prostaglandin E2, which<br />

acutely signal via protein kinase A, acquire the capacity to activate additional<br />

signaling pathways that produce a prolonged hyperalgesic state. Critical to<br />

this process is activation of the enzyme protein kinase C epsilon (PKC).<br />

Studies in rats and mice have demonstrated that activation of PKC is<br />

important for hyperalgesia due to inflammation, to chemotherapeutic,<br />

alcoholic, or diabetic neuropathy, and to stress-induced generalized pain.<br />

Confirmed downstream targets of PKC that regulate the excitability of<br />

nociceptors include the polymodal receptor TRPV1 and the sodium channel<br />

Nav1.8; additional targets remain to be identified and validated. Given the<br />

apparent key role of PKC as a hub for nociceptor signaling, there has<br />

been growing interest in developing PKC inhibitors to treat inflammatory,<br />

neuropathic, and generalized pain disorders. Our recent work has identified<br />

a series of potent kinase inhibitors that are relatively selective for PKC over<br />

other PKC isozymes and cross the blood brain barrier. Additional animal<br />

studies indicate that PKC inhibition is likely to be anxiolytic and possibly<br />

anti-inflammatory, providing a favorable therapeutic profile for a new class of<br />

drugs to treat pain.<br />

References:<br />

1. Reichling DB, Levine JD. Critical role of nociceptor plasticity in chronic<br />

pain. Trends Neurosci. Dec 2009;32(12):611-618.<br />

2. Aley KO, Messing RO, Mochly-Rosen D, Levine JD. Chronic<br />

hypersensitivity for inflammatory nociceptor sensitization mediated<br />

by the epsilon isozyme of protein kinase C. J Neurosci.<br />

2000;20(12):4680-4685.<br />

3. Wu DF, Chandra D, McMahon T, et al. PKCepsilon phosphorylation<br />

of the sodium channel NaV1.8 increases channel function and produces<br />

mechanical hyperalgesia in mice. J Clin Invest. Apr 2 <strong>2012</strong>;122(4):<br />

1306-1315.<br />

4. Hodge CW, Raber J, McMahon T, et al. Decreased anxiety-like behavior,<br />

reduced stress hormones, and neurosteroid supersensitivity in mice<br />

lacking protein kinase C epsilon. J. Clin. Invest. Oct 2002;110(7):<br />

1003-1010.<br />

The Bench to Bedside Challenges in Neuropathic Pain<br />

Therapeutics<br />

Roy L. Freeman, MD, Beth Israel Deaconess Medical Center, Boston<br />

Neuropathic pain therapeutics faces several challenges. There are currently<br />

seven drugs approved by the FDA for the treatment of neuropathic pain:<br />

lidocaine delivered topically via a patch, gabapentin in three different<br />

formulations, pregabalin, duloxetine and topical capsaicin administered<br />

as an 8% concentration patch. Despite the availability of these drugs and<br />

the widespread use of numerous unapproved drugs from different classes,<br />

current pharmacological approaches do not provide satisfactory pain relief<br />

in many patients. In clinical trials, even the most effective of these agents<br />

provides less than a 50% improvement in less than 50% of subjects.<br />

Furthermore, many patients experience intolerable adverse events with


these therapies. Inarguably, the availability and widespread use of these<br />

agents has improved patient quality of life, but there remains a need for<br />

more potent agents with a superior adverse event profile.<br />

It is not yet known whether a response to one intervention is generalizable.<br />

Specifically, it is not known whether individuals who respond to one<br />

medication would also respond to a second medication with a different<br />

mechanism of action or whether response characteristics vary among<br />

patients. There is preliminary evidence that the pain phenotype and genotype<br />

may influence individual response characteristics. The rationale behind<br />

the role of the pain phenotype in determining therapeutic response is<br />

based on the hypothesis that the phenotype is a specific manifestation of<br />

the mechanism that underlies the pain and that, if so, the pain phenotype<br />

predicts responsiveness to a medication with a specific mechanism of action.<br />

Detailed phenotypic studies have been performed in multicenter clinical trials<br />

and by multinational academic consortia. These may provide the basis of<br />

studies to predict therapeutic responsiveness and a more rational approach<br />

to pain therapeutics.<br />

The animal models of neuropathic pain continue to provide insights into<br />

the molecular mechanisms whereby nerve injury results in chronic pain<br />

and all of the approved agents are effective in models of neuropathic pain.<br />

However, a successful response in these models is not a consistent predictor<br />

of success in clinical trials; many agents appear effective in preclinical pain<br />

models but fail in subsequent clinical trials. Further, none of the approved<br />

agents originated from preclinical studies. All showed initial clinical<br />

effectiveness and were subsequently tested in animal models, following a<br />

bedside-to-bench route. Yet success in the animal neuropathic pain model<br />

remains the gateway to the clinical arena. There are no data as to whether<br />

agents, ineffective or partially effective in the animal model show success in<br />

the clinic. As an alternative, several human models of neuropathic pain exist<br />

but the predictive value of these is not established.<br />

References:<br />

1. Freeman R. Pharmacotherapy of Neuropathic Pain. In: Simpson DM,<br />

McArthur JCM, Dworkin R, Editors. Neuropathic Pain: Mechanisms and<br />

Management. Oxford University Press. New York, NY. <strong>2012</strong><br />

2. Klein T, Magerl W, Rolke R, Treede RD. Human surrogate models of<br />

neuropathic pain. Pain. 2005 Jun;115:227-33.<br />

3. Maier C, Baron R, Tölle TR, Binder A Birbaumer N, Birklein F,<br />

Gierthmühlen J,et al. Quantitative sensory testing in the German<br />

Research Network on Neuropathic Pain (DFNS): somatosensory<br />

abnormalities in 1236 patients with different neuropathic pain<br />

syndromes. Pain. 2010;150:439-50.<br />

4. Mogil JS. Animal models of pain: progress and challenges. Nat Rev<br />

Neurosci. 2009;10:283-94.<br />

Anti-NGF: A Saga in Drug Development<br />

David R. Cornblath, MD, Johns Hopkins University, Baltimore, Md.<br />

Prof. Rita Levi-Montalcini and Prof. Stanley Cohen discovered nerve growth<br />

factor (NGF) in the 1950s while faculty members at Washington University<br />

in St Louis initially working in the laboratory of Prof. V. Hamburger. For this<br />

discovery, they won the Nobel Prize in Physiology or Medicine in 1986.<br />

NGF is a small secreted protein that is critical for the growth, survival and<br />

maintenance of sympathetic and sensory neurons during a critical period<br />

of development. Without it, these neurons undergo apoptosis during<br />

this critical period. NGF causes axonal growth and axonal branching.<br />

NGF prevents or reduces neuronal degeneration in animal models of<br />

neurodegenerative diseases. NGF has also been shown to promote peripheral<br />

nerve regeneration. The expression of NGF is increased in inflammatory<br />

diseases where it suppresses inflammation. NGF appears to promote myelin<br />

repair. These basic science results lead to the first-in-human clinical trials<br />

in humans (Petty et al. 1994). In that study, it was discovered that there was<br />

a dose-limiting toxicity: with systemic administration, subjects developed<br />

a myalgic syndrome while with local administration, subjects developed<br />

hypersensitivity at the site of injection. These observations lead to the idea<br />

that blocking NGF may block pain transmission.<br />

Several companies have now developed anti-NGF drugs. The initial<br />

concern from a safety perspective was that these would cause a peripheral<br />

neuropathy as NGF is known to be important in the maintenance of small<br />

sensory neurons. Thus, considerable thought was given to potential<br />

peripheral neurologic toxicity. Monitoring schemes were developed and<br />

implemented. Extensive neurologic review of possible toxicity was in place.<br />

Then the unexpected occurred. There appeared to be an excess of cases<br />

of osteonecrosis leading to total joint replacement. The company furthest<br />

along in development notified the DSMB and the FDA. Further review lead<br />

to the discovery of additional cases although expert adjudication opined that<br />

the adverse events were not ON but rather a rare condition called rapidly<br />

progressive osteoarthritis (RPOA). Prior to complete expert adjudication<br />

of the cases, the FDA placed the clinical trials on hold for one company<br />

and began close monitoring of the other companies clinical development<br />

programs. Later all clinical development programs were placed on hold.<br />

The mechanism of how anti-NGF drugs leads to this adverse event<br />

remains unknown.<br />

The clinical trials of the several anti-NGF drugs have suggested considerable<br />

clinical promise in the treatment of pain – from osteoarthritis to diabetic<br />

polyneuropathy to potentially visceral pain syndromes. One hopes that<br />

clinical development can resume so that we can learn about the full risk:<br />

benefit profile of these drugs.<br />

References:<br />

1. Levi-Montalcini R (2004). “The nerve growth factor and the neuroscience<br />

chess board”. Prog. Brain Res. 146: 525–7.<br />

2. The Nobel Prize in Physiology or Medicine 1986 Stanley Cohen,<br />

Rita Levi-Montalcini http://www.nobelprize.org/nobel_prizes/medicine/<br />

laureates/1986/press.html<br />

3. Petty BP et al. The effect of systemically administered recombinant<br />

human nerve growth factor in healthy human subjects. Ann Neurol.<br />

1994;36:244-6.<br />

39


The <strong>2012</strong> ANA <strong>Annual</strong> <strong>Meeting</strong> is supported by generous educational grants from:<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

This activity is supported by an educational grant from Lilly USA, LLC.<br />

For further information concerning Lilly grant funding visit www.lillygrantoffice.com.<br />

The <strong>American</strong> <strong>Neurological</strong> <strong>Association</strong> wishes to<br />

thank the following <strong>2012</strong> <strong>Annual</strong> <strong>Meeting</strong> sponsors:<br />

Bronze Level — Resident/Fellow Travel Awards:<br />

USB Flash Drives: Mayo Clinic<br />

Overall Mobile App Sponsor: Intermountain Healthcare<br />

Mobile App Sponsors: Novartis Pharmaceuticals<br />

Accera<br />

JOIN US NEXT YEAR!<br />

138 th <strong>Annual</strong> <strong>Meeting</strong><br />

New Orleans, LA | October 13-16, 2013

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!