Download 2009 Program - Society for Magnetic Resonance

Document related concepts

Endovascular aneurysm repair wikipedia , lookup

Nuclear medicine wikipedia , lookup

Positron emission tomography wikipedia , lookup

Image-guided radiation therapy wikipedia , lookup

Medical imaging wikipedia , lookup

Transcript
Dear friends and participants of the
2009 MR Angio Club Meeting
It is with great pleasure that we welcome everyone back to Michigan State University for
the 21st International Conference in MR Angiography. Since being founded by a team
lead by Dr. Jim Potchen in 1989, Michigan State University has been proud to host the
annual meeting on three previous occasions, the last in 1994. The MR Angio Club annual
meeting has spanned the globe from Asia, to Europe, to the USA and Canada as well as
the Far East. This wide diversity in locality has been matched with an ever-growing
diversity and ingenuity to produce ever-improving clinical and research applications of MR
Angiography.
The Local Organizing Team is excited about the excellent quality of the greater than 115
abstracts submitted to the 21st MR Angiography meeting. Both the oral presentations and
the expanded poster section should provide very stimulating discussions typical of the
MRA Club meetings. We are also proud to announce a Student Poster Award as part of
this year’s meeting.
MSU Kellogg Center will be an excellent facility for our scientific program with a large,
comfortable stadium seating auditorium and hotel rooms to accommodate many of the
presenters in one building. The Local Organizing Team has also arranged an exciting
social evening program to highlight the Michigan State University campus and the history
of Michigan that we hope you will enjoy.
We are grateful for the ongoing support of our corporate sponsors who continue to support
such stimulating meetings. The sponsors are listed elsewhere in this program an on the
meeting homepage. We are also indebted to the London, Ontario Team including Janette
Wallace, Johanne Guillemette, and Kellie Griffin.
We hope you enjoy the 21st International Conference on MR Angiography and welcome
back to Michigan State University.
Local Organizing Team
Jim Siebert – Scientific Committee Chairman
Arlene Sierra – MRA Club International Secretary
Stella Cash – MSU Alumni Interim Executive Director
Tom Cooper – Vice Chairman MSU Radiology
Colleen Hammond – Chief MSU MR Technologist
Kevin DeMarco – President MRA Club 2009
MR-Angioclub East Lansing 2009
1
Conference Organization
President
President Elect
J. Kevin DeMarco
Martin Prince
Past Presidents
Executive Board
James Potchen
Georg Bongartz
Dennis L. Parker
Georg Bongartz
Kevin DeMarco
James Potchen
James E. Siebert
Phillipe Douek
Martin Prince
Paolo Pavone
Charles Dumoulin
Stephen J. Riederer
Dennis L. Parker
Christoph Herborn
Brian Rutt
Jorg F. Debatin
John Huston
Kazuhiko (Kaz)Sadamoto
John Huston
Frank R. Korosec
David Saloner
E. Kent Yucel
Gerhard Laub
Klaus Scheffler
Frank R. Korosec
Debiao Li
Stefan Schoenberg
David Saloner
James F.M.
Meaney
James E. Siebert
James F. M. Meaney
Local Organizing
Committee
Stella Cash
Tom Cooper
Colleen Hammond
Kevin Henley
Cheryl Hunley
Arlene Sierra
Graham Wright
Hitoshi Miki
Brian K. Rutt
Charles Mistretta
Debiao Li
Stephen J. Riederer
Program Committee
Congress
Organizing
Team
James Siebert
James Siebert
Conference Coordinator
Kellie Griffin
Johanne Guillemette
Carolyn Pakula
Terri Schneider
Registration Service
www.registrationassistant.com
2
Executive Board
Secretariat
Arlene Sierra
Congress Office @
Robarts Research
Institute
Johanne Guillemette
Janette Wallace
Conference Manager
Secretariat
Janette Wallace
Accommodation
www.hfs.msu.edu/kellogg
MR-Angioclub East Lansing 2009
The 2009 MR Angio Conference Timetable
Tuesday, September 29, 2009
07:30 – 9:00
Registration
09:00 – 9:30
Opening Ceremony
09:30 – 10:30
Selected Non- and Low-Gd Contrast Methods and Applications
10:30 – 11:00
Coffee Break
11:00 – 12:00
Compressed Sensing and HYPR
12:00 – 01:00
Lunch
01:00 – 02:45
Plaque Characterization and Vessel Wall Imaging
02:45 – 03:15
Coffee Break
03:15 – 05:00
Peripheral MRA
Wednesday, September 30, 2009
08:00 – 09:45
Contrast Agents Performance and Safety
09:45 – 10:15
Coffee Break
10:15 – 12:00
Hemodynamics and Perfusion
12:00 – 01:00
Lunch
01:00 – 02:45
Head and Neck MRA
02:45 – 03:15
Coffee Break
03:15 – 05:00
Thoracic MRA
Thursday, October 1, 2009
08:30 – 09:45
Cardiac Imaging / Coronary MRA
09:45 – 10:15
Coffee Break
10:15 – 12:00
Abdominal MRA
12:00 – 01:00
Lunch
01:00 – 02:45
Venous Imaging and Beyond
02:45 – 03:15
Coffee Break
03:15 – 05:00
New Horizons and Challenges in MRA
MR-Angioclub East Lansing 2009
3
MR Angio 2009 Club appreciates the generous support of the
following sponsors:
PLATINUM SPONSORS
GOLD SPONSORS
SILVER SPONSORS
4
MR-Angioclub East Lansing 2009
MR-Angioclub East Lansing 2009
5
1. Registration, Internet Access
2. Speaker Ready Room
3. Main Meeting Room
4. Lunch & Breakfast
5. Exhibits & Posters, Coffee Breaks
6. Board Meeting
Floor Plan for Exhibitors and Posters - Big 10 Room B
(see pages 23-24)
6
MR-Angioclub East Lansing 2009
General Information
Venue
Kellogg Hotel and Conference Center
Michigan State University
East Lansing, Michigan
Registration
Foyer of Kellogg Hotel and Conference Center, Lobby Level, across from Auditorium.
Message Board
There will be a message board near the registration desk.
Coffee Breaks & Lunches
Snacks and drinks will be provided on the Lobby Level of the Kellogg Hotel and Conference
Center in the Big Ten Room C.
Internet Access
Wireless Internet is available throughout the conference center.
Speakers
Speakers are requested to contact the speaker ready room (101, lobby level, right beside
the Auditorium) and to hand in their Power Point presentation (on CD-ROM or data stick) at
least 90 minutes before the start of the session of their presentation. Trained staff will be
available to assist you with equipment. You can retrieve your CD-ROM or USB stick once
your presentation has been uploaded. Your presentation will be deleted from the server
after your talk and thus not made accessible to third parties.
Posters
Posters have to be mounted at the assigned stands in the poster Exhibition Area (Big Ten
Room B) the morning of Tuesday September 29. Please dismount the poster Thursday
October 1 by 5 pm. Remaining posters will be discarded.
MR-Angioclub East Lansing 2009
7
Spousal Program
Tuesday, September 29, 2009
09:00 – 03:30 pm
Bus leaves Kellogg Center at 09:00
am
Tour of the Gerald R. Ford Presidential
Museum
Lunch at Amway Grand
Meijer Sculpture Gardens
Bus leaves at 03:30 pm from Meijer
Gardens back to the Kellogg Center
Wednesday, September 30, 2009
08:30 – 03:30 pm
Bus leaves the Kellogg Center at 08:30
am
Trip to Frankenmuth
Lunch at “Zehnders” Restaurant
Bus leaves at 03:00 pm from
Zehnders back to the Kellogg Center
Thursday, October 1, 2009
Tour of Michigan State University
Campus
Lunch is provided
8
MR-Angioclub East Lansing 2009
Social Events
Monday, September 28, 2009
07:00 – 09:30 pm
Welcome Reception
Radiology Department of
Michigan State University
Tuesday, September 29, 2009
07:00 – 10:00 pm
Dinner at the University Club
of Michigan State University
Wednesday, September 30, 2009
07:00 – 10:00 pm
Evening reception at the State
Capital
Michigan Historical Museum
Dinner catered by Morton’s
Thursday, October 1, 2009
07:00 – 10:00 pm
Casual dining in a lakeside
setting with Motown music.
MR-Angioclub East Lansing 2009
9
Tuesday, September 29, 2009
9:00 am – 9:30 am Opening Ceremony
9:00 – 9:05
J. Kevin DeMarco
MR Angio Club President
9:05 – 9:15
Lou Anna K. Simon
President, Michigan State University
9:15 – 9:20
J. Ian Gray
Vice President for Research and Graduate
Studies
9:20 – 9:25
James Randolph Hillard
Associate Provost for Human Health Affairs
9:25 – 9:30
James E. Siebert
Program Director
9:30 am – 10:30 am Session 1
Selected Non - and Low- Gd Contrast Methods and
Applications
Session Chairs: Stephen J. Riederer, Manuela Aschauer
9:30
Liesbeth Geerts
1.1 Non-CE Imaging of the Pulmonary Arteries
9:42
Kevin Johnson
1.2 Accelerated Time Resolved Inflow with 3D Radial bSSFP
9:54
Gerhard Laub
1.3 Low - Dose 4D MR Angiography
10:06
Samuel Fielden
1.4 Balanced-gradient TSE for Non-contrast Peripheral MRA
10:18
Jing Liu
1.5 Self-gated Free Breathing 3D Cardiac Cine Imaging with Data Acquisition
During Slice Encoding
10:30 am – 11:00 am
10
Coffee Break
MR-Angioclub East Lansing 2009
11:00 am – 12:00 pm Session 2
Compressed Sensing and HYPR
Session Chairs: Mark A Griswold, Dennis L. Parker
11:00
Mark Griswold
2.1 A Simple View of Compressed Sensing and How it Could Change
Everything We Do in MRI and MRA
11:12
Julia Velikina
2.2 Design of Compressed Sensing Reconstruction for Highly Accelerated
Time-Resolved MR Angiography
11:24
Yijing Wu
2.3 Low Dose HYPR FLOW
11:36
Lan Ge
2.4 Myocardial Perfusion MRI in Canines with Improved Spatial Coverage,
Resolution and SNR
11:48
Nicole Seiberlich
2.5 Reconstruction of MR Angiography Images Using Gradient Descent with
Sparsification
12:00 pm – 1:00 pm
Lunch
MR-Angioclub East Lansing 2009
11
1:00 pm – 2:45 pm
Session 3
Plaque Characterization and Vessel Wall Imaging
Session Chairs: J. Kevin DeMarco, Brian K. Rutt
1:00
Chun Yuan
3.1 Carotid Plaque Imaging and Clinical Risk Assessment
1:12
Hideki Ota
3.2 Carotid Intraplaque Hemorrhage Is Associated with Enlargement of Lipid-rich
Necrotic Core and Plaque Volume Over Time: In Vivo 3T MRI Prospective Study
1:24
David Zhu
3.3 The 3D SHINE Sequence Optimizes the Quantification of Carotid Intraplaque
Hemorrhage
1:36
Jinnan Wang
3.4 Improve Intraplaque Hemmorhage Detections by a Phase Sensitive IRTFE
(SPI) Sequence
1:48
William Kerwin
3.5 Fibrous Cap Thickness Assessment: Fact or Fiction?
2:00
Rui Li
3.6 Gradient Echo Based Sequence Provides More Information from Ex Vivo
Carotid Plaque Specimens
2:12
Seong-Eun Kim
3.7 Improved Black Blood Multi-Contrast Protocol for In-vivo Atherosclerotic
Imaging
2:24
Yi Wang
3.8 3D peripheral vessel wall MRI with flow-insensitive blood suppression and
isotropic resolution at 3 Tesla
2:36
Rock Hadley
3.9 A 16 Channel Anterior Neck RF Coil for Cervical Carotid MRA
2:45 pm – 3:15 pm
12
Coffee Break
MR-Angioclub East Lansing 2009
3:15 pm – 5:00 pm
Peripheral MRA
Session 4
Session Chairs: Jeffrey Maki, Tim Leiner
3:15
Tim Leiner
4.1 Gadobenate dimeglumine vs. gadopentetate dimeglumine for peripheral
MR angiography: comparison with DSA
3:27
Jeffrey Maki
4.2 Dose Comparison between Conventional and High Relaxivity Contrast
Agents in Peripheral MRA
3:39
Matthias Voth - NOT ATTENDING
4.3 Periipheral MRA with Continuous Table (CTM) Movement in Combination
with High Temporal and Spatial Resolution TWIST – MRA with 0.1 mmol/kg
Gadobutrol at 3.0T
3:51
John Sheehan
4.4 Flow and Motion-Insensitive Unenhanced MR Angiography of the
Peripheral Vascular System – A pilot study in the lower extremity
4:03
Clifton Haider
4.5 A Comparison of Time-Resolved 3D CE-MRA with Peripheral Run-off
CTA in the Calves
4:15
Kang Wang
4.6 3D Time- Resolved MR Angiography of Lower Extremities using
Cartesian Interleaved Variable Density Sampling and HYPR Reconstruction
4:27
Zhaoyang Fan
4.7 3D Noncontrast MRA Using FSD – Prepared Balanced SSFP
4:39
James Carr
4.8 Non Contrast MRA of the Hand in Patients with Raynauds disease using
Flow Sensitized Dephasing Prepared SSFP
4:51
Casey Johnson
4.9 Two-Station Time-Resolved CE-MRA of the Lower Legs
MR-Angioclub East Lansing 2009
13
Wednesday, September 30th, 2009
8:00 am – 9:45 am
Session 5
Contrast Agents Performance and Safety
Session Chairs: Thomas M. Grist, Martin R. Prince
8:00
Mark Hibberd
5.1 An Update on the Clinical Experience with Gadofosveset
8:12
Edward Parsons
5.2 A Re-analysis of MS-325 (gadofoveset trisodium) Clinical Trial Data in
Support of US-FDA Approval
8:24
Manuela Aschauer
5.3 Gadofoveset Excretion into Human Breast Milk
8:36
Thomas Grist
5.4 Overview of Gd-BOPTA Phase III Trail for CEMRA: What are the results, and
how do we move forward?
8:48
Guenther Schneider
5.5 Safety of Gadobenate dimeglumine (Gd-BOPTA) in Cardiovascular Imaging
of Pediatric Patients
9:00
Giles Roditi
5.6 Retrospective 7 year Study of the Incidence of Nephrogenic System Fibrosis
in Patients Investigated with Gadolinium Contrast-Enhanced Renal Magnetic
Resonance Angiography
Martin Prince
5.7 Risk Factors for NSF: a Meta-analysis
9:12
9:24
James Varani
5.8 Extracellular matrix metabolism in organ-cultured skin from patients with endstage renal disease: Response to gadolinium based MRI contrast agents
9:36
Zheng-Rong Lu - NOT ATTENDING
5.9 Manganese Based Biodegradable Macromolecular MRI Contrast Agents for
Cardiovascular Imaging
9:45 am – 10:15 am
14
Coffee Break
MR-Angioclub East Lansing 2009
10:15 am – 12:00 pm
Session 6
Hemodynamics and Perfusion
Session Chairs: Scott B. Reeder, E. Mark Haacke
10:15
Thorsten Bley
6.1 Non-invasive Trans-Stenotic Pressure Measurements with 3D Phase
Contrast MRA: Validation against Endovascular Pressure Measurements in
Swine
10:27
Alex Frydrychowicz
6.2 Analysis of aortic hemodynamics after treatments for coarctation using
flow-sensitive 4D MRA at 3T
10:39
Christopher Francois
6.3 Flow assessment of arterial dissections using 3D radial phase contrast
MR angiography
10:51
Scott Reeder
6.4 High Temporal and High Spatial Resolution Perfusion Imaging of
Hepatocellular Carcinoma in the Liver
11:03
Steven Kecskemeti
6.5 Stack of Stars 4D Phase Contrast Velocimetry of the Circle of Willis
11:15
Mark Haacke
6.6 High Resolution Perfusion Weighted Imaging
11:27
Luca Marinelli
6.7 Accelerated velocity imaging using compressed sensing
11:39
Michael Markl
6.8 Wall Shear Stress in Normal and Atherosclerotic Carotid Arteries
11:51
Scott McNally
6.9 MR imaging and significance of flow reversal and carotid
atherosclerosis: Initial results
12:00 pm – 1:00 pm
Lunch
MR-Angioclub East Lansing 2009
15
1:00 pm – 2:45 pm
Head and Neck MRA
Session 7
Session Chairs: John Huston, David Saloner
1:00
Winfried Willinek
7.1 4D-MRA in combination with arterial spin labeling for selective and functional
information in patients with AVMs
1:12
Marco Essig
7.2 Intraindividual comparison between multislice CT and 4D TWIST MRA in the
assessment of residual cerebral arteriovenous malformations – a prospective
study protocol
1:24
Keiji Igase
7.3 Our Strategy for the Surgical Planning with 3T MRA in Detecting Unruptured
Cerebral Aneurysms
1:36
Faiza Admiraal-Behloul
7.4 Hybrid of Opposite Contrast MR Angiography of the Brain
1:48
Ek Tsoon Tan
7.5 High Resolution Fast Inversion Recovery MRA (FIR-MRA)
2:00
Nick Zwart
7.6 3D Dual VENC PCMRA using Spiral Projection Imaging
2:12
Samuel Barnes
7.7 High Resolution Simultaneous Angiography and Venography (MRAV) with a
Single Echo
2:24
Bum-soo Kim
7.8 Low Dose 3D Time-Resolved MR Angiography of the Supraaortic Artery:
Correlation to High Spatial Resolution 3D Contrast-Enhanced MRA
2:36
7.9 Tae-Sub Chung
Obstruction of IJV by Asymmetry of Lateral Mass of Atlas on Head and Neck
CEMRA and Contrast CT
2:45 pm – 3:15 pm
16
Coffee Break
MR-Angioclub East Lansing 2009
3:15 pm – 5:00 pm
Thoracic MRA
Session 8
Session Chairs: Winfried A. Willinek, Charles L. Dumoulin
3:15
Dipan Shah
8.1 Evaluation of Gd-DOTA (DOTAREM) enhanced MRA compared to timeof-flight MRA in the diagnosis of clinically significant non-coronary arterial
disease at 1.5 and 3.0 Tesla
3:27
Mark Schiebler
8.2 Pulmonary MRA in 75 patients with dyspnea
3:39
Mark Schiebler
8.3 Origin and Frequency of artifacts in Contrast Enhanced Pulmonary MRA
in 80 patients with dyspnea
3:51
Paul Stein
8.4 Gadolinium Enhanced Magnetic Resonance Angiography for Pulmonary
Embolism: Results of PIOPED III
4:03
Loic Boussel
8.5 4D time-resolved MR angiography for non-invasive pulmonary postembolization AVM patency assessment
4:15
Timothy Carroll
8.6 Radial Sliding Window MRA in Pulmonary Hypertension
4:27
Peng Hu
8.7 Non-Contrast Enhanced Pulmonary Vein MRI with a Spatially Selective
Slab Inversion Preparation Sequence
4:39
Grace Choi
8.8 MRA with the “No Phase Wrap”
4:51
Hitoshi Miki
8.9 Unruptured Intracranial Aneurysms; Detection and Follow-up on 3.0T
MRA
MR-Angioclub East Lansing 2009
17
Thursday, October 1st, 2009
8:00 am – 9:45 am
Session 9
Cardiac Imaging / Coronary MRA
Session Chairs: Debiao Li, Harald H. Quick
8:00
Oliver Wieben
9.1 Comprehensive PC MR Imaging in Congenital Heart Disease
8:12
Gary Liu
9.2 Ultrasound guided cardiac gating for coronary MRA
8:24
Himanshu Bhat
9.3 Contrast-Enhanced Whole-Heart Coronary MRA at 3T Using Gradient Echo
Interleaved EPI (GRE-EPI)
8:36
Jingsi Xie
9.4 Feasibility of Whole-Heart Coronary MRA on 3 Tesla Using Ultrashort-TR
SSFP VIPR
8:48
James Goldfarb
9.5 Cardiac Imaging: Methods for the Detection of Intramyocardial Fat
9:00
Dana Peters
9.6 3D spiral high-resolution late gadolinium enhancement
9:12
Ben Grabow
9.7 Temporal Filtering for Sliding Window Time-resolved Angiography; Beyond
Density Compensation Solutions
9:45 am – 10:15 am
18
Coffee Break
MR-Angioclub East Lansing 2009
10:15 am – 12:00 pm
Abdominal MRA
Session 10
Session Chairs: Franz Ebner, Walter F. Block
10:15
Kevin Johnson
10.1 Angiographic and Hemodynamic Assessment of the Hepatic
Vasculature in Portal Venous Hypertension using High Resolution PC VIPR
10:27
Guenther Schneider
10.2 Renal MR angiography: multicenter intraindividual comparison of
gadobenate dimeglumine and gadofosveset trisodium
10:39
Manojkumar Saranathan
10.3 FINESS (Flow Inversion-prepared Non-contrast Enhancement in the
Steady State): A novel technique for non-contrast renal MRA
10:51
Tiffany Newman
10.4 Magnetic Resonance Angiography of the skin for perforator-based
autologous breast reconstruction
11:03
Isabelle Parienty
10.5 Time-SLIP versus DSA in Patients with Renal Artery Stenosis
11:15
Katherine Wright
10.6 Simultaneous Renal Angiography and Perfusion Measurement Using
Time-Resolved MRA
11:27
Nathan Artz
10.7 Assessing Kidney Perfusion using Arterial Spin Labeling and Radial
Acquisition for Rapid Characterization of Inflow Dynamics
11:39
Walter Block
10.8 Imaging Capabilities for Real-time Guidance and Verification of
Transcatheter Arterial Chemoembolization (TACE) Procedures
12:00 pm – 1:00 pm
Lunch
MR-Angioclub East Lansing 2009
19
1:00 pm – 2:45 pm
Session 11
Venous Imaging and Beyond
Session Chairs: Frank R. Korosec, Yi Wang
1:00
M Louis Lauzon
11.1 Non-Contrast-Enhanced MR identification of DVT
1:12
Mark Haacke
11.2 Susceptibility mapping as a means to image veins
1:24
Petrice Mostardi
11.3 Modified CAPR MRA: Improved Imaging of the Arterial and Venous Phases
1:36
Hyun Jeong
11.4 CAMERA: Contrast-enhanced Angiography with Multi-Echo and Radial kspace
1:48
Philip Robson
11.5 Time-Resolved, Vessel-Selective, Cerebral Angiography Using Arterial Spin
Labelling
2:00
David Steinman
11.6 Quantifying Lumen Geometry from Routine Carotid CEMRA
2:12
Yi Wang
11.7 Magnetic Source MRI for Quantitative Brain Iron Mapping
2:24
Kheireddine El-Boubbou
11.8 Targeted Glyco-Magnetic Fe304 Nanoprobes for Detections and Molecular
Imaging of Atherosclerosis
2:45 pm – 3:15 pm
20
Coffee Break
MR-Angioclub East Lansing 2009
3:15 pm – 5:00 pm
Session 12
New Horizons and Challenges in MRA
Session Chairs: E. James Potchen, Charles A. Mistretta
3:15
James E. Siebert
Presentation of the Best Student Posters Awards
3:20
Charles Mistretta
12.1 4D DSA and Fluoroscopy: A New Challenge for MRA?
3:32
Bas Versluis
12.2 MR Angiography of muscular and collateral arteries in peripheral arterial
disease: reproducibility of morphological and functional vascular status
3:44
Matt Bernstein
12.3 Multicenter Studies: Lessons Learned from ADNI
3:56
David Saloner
12.4 Imaging Considerations in Serial Studies of Vascular Disease
4:08
Mark Ladd
12.5 Towards Abdominal MRA at 7 Tesla
4:20
Harald Quick - NOT ATTENDING: Mark Ladd to present
12.6 7 Tesla Cardiac MRA in Humans
4:32
George Abela
12.7 The Role of Cholesterol Crystals in Acute Cardiovascular Events:
Identifying the Cause for Gender Differences in Clinical Presentation
4:44
James E. Potchen
Perspective on MRA and the MR Angio club (no abstract)
4:56
Kevin DeMarco
Presentation of the new President of the MRA Club
Announcement of the new President Elect
Announcement of the 22nd International MRA Conference 2010 in Seoul, South
Korea
MR-Angioclub East Lansing 2009
21
Posters
MRA Methods
P1
Manuela Aschauer
CE-MRA with tailored 3D random sampling patterns and nonlinear parallel imaging
reconstruction
P2
Jason Mendes
Handling Motion in Sparse MRA with Whiskers
P3
Jordan Hulet
Improved Carotid Imaging with HASTE using a reduced FOV and increased
gradient performance
P4
Randall Stafford
Towards Continuously Moving Table NCE Peripheral MRA
P5
Matthew Latourette
R2* Calibration Phantoms for Cardiovascular Studies
P6
Giles Roditi
Pictorial Review of Supra-Aortic Artery Pathologies as Visualised with MRA using
Blood Pool Contrast Agent
P7
Kristine Blackham
Robust Clinical Application of Time-Resolved MRA
P8
Jonathan Suever
Reproducibility of Aortic Pulse Wave Velocity Measurements Obtained with Phase
Contrast Magnetic Resonance (PCMR) and Applanation Tonometry
P9
Gregory Wilson
Motion-compensated, flow-independent, non-contrast-enhanced renal MR
angiography
MRA Applications
22
P10
Chang-Ki Kang
Vascular response during visual stimulation at 3T MRI: functional phase contrast
angiography (fPCA) study
P11
Jongmin Lee
Optimization of Phase-contrast MR-based Flow Velocimetry and Shear Stress
Measurement
P12
John Oshinski
Blood Flow Patterns in the Abdominal Aorta of Mice: Implications for AAA
localization
MR-Angioclub East Lansing 2009
Posters
Plaque Characterization and Vessel Wall Imaging
P13
Niranjan Balu
3D Vessel Wall imaging of multiple vascular beds
P14
Keigo Kawaji
Feasibility Study of Combining 3D SSFP with T2prep inversion Recovery (T2IR) for
Black Blood Vessel Wall Imaging
P15
Zhaoyang Fan
Identification of Optimal First-Order Gradient Moment for Flow-Sensitive
Dephasing (FSD) Preparation
P16
Rahul Sarkar
Combined Segmentation of Lumen and Intraplaque Hemorrhage in Black-blood
T1-Weighted Carotid Imaging
P17
Rahul Sarkar
Automatic Registration of Multiparametic T1 Weighted Images Using FOVSelective Mutual Information
CFD Modeling and MR-Guided Endovascular
Interventions
P18
Haruo Isoda
MR fluid dynamics using 4D-Flow for intracranial aneurysms with growing blebs
and a ruptured intracranial aneurysm
P19
Charles Dumoulin
Phase-Field Dithering for Active Catheter Tracking
P20
Ethan Brodsky
Interventional Device Tracking and Imaging Using an Extensible Real-Time
System
P21
Mahdi Salmani Rahimi
Simplified Catheter-based Multimode Coil for Active MR Tracking and Intravascular
Imaging
P22
Krishna Kurpad
Transmit Power Optimization for Tracking, Wireless Marker and Imaging
applications of a Multi-mode Endovascular coil
MR-Angioclub East Lansing 2009
23
1.1 Non-CE imaging of the pulmonary arteries
Liesbeth Geerts, Marco van Essen, Gregory Wilson, Tomoyuki Okuaki
Philips Healthcare, Best, The Netherlands
Purpose Bright blood ASL in a single acquisition – without subtraction of tag-on and tagoff acquistions – can be used to depict vascular structures [1].The contrast mechanism
relies on inflow of fresh spins into the imaging region. Therefore, optimal imaging
parameters, such as the time allowed for inflow, may be patient dependant.
The purpose of this study was to 1) evaluate the use of a bright blood ASL approach in a
single acquisition for depiction of the pulmonary vasculature and to 2) evaluate the effect
of different TI times.
Methods A combination of a spatially non-selective and a selective inversion pulse was
implemented on a 1.5T Achieva scanner to obtain a bright blood ASL in a single
acquisition. A respiratory triggered 3D-TSE was used for readout. 12 Healthy volunteers
(mean 54.2 years, range 37 to 71) were scanned, using TI’s of 500, 800 and 1100 ms. In
two volunteers, additional measurements at TI 300, 700 and 900 ms were performed.
Image quality was qualitatively rated.
Results A TI of 800 ms gave the best depiction of a pure arterial filling. At longer TI (1100
ms) filling of the distal segments has progressed, however, also the venous signal
becomes more apparent.
TI 500 ms
TI 800 ms
TI 1100 ms
Conclusion Bright blood ASL in a single acquisition is suitable to depict the pulmonary
vasculature. A TI of 800 ms was found to give good depiction of the pulmonary arteries.
[1] Miyazaki. Radiology 248(1);20-43 (2008)
24
MR-Angioclub East Lansing 2009
1.2 Accelerated Time Resolved Inflow with 3D Radial bSSFP
Kevin M. Johnson, Oliver Wieben, Patrick Turski, Charles Mistretta
Departments of Medical Physics and Radiology, University of Wisconsin, Madison, WI, USA
Purpose: Recently the combination of blood tagging schemes [1] and bSSFP acquisitions
has allowed for sub second resolution of vascular filling dynamics [2]. However, bSSFP
can be sensitive to artifacts from off-resonance banding and flow artifacts. In this work we
investigate the use of 3D radial trajectories for highly accelerated non-contrast enhanced
angiography with reduced flow and banding related artifacts.
Methods: All imaging was performed on a clinical 1.5T scanner. An ecg-triggered,
inversion-recovery prepared, cardiac interleaved, bSSFP, 3D sequence was implemented
with both dual-half echo (non-flow compensated) and 4-half radial trajectories (flow
compensated) [3]. Angiographic images are acquired by subtracting of a pass with nonselective inversion pulse from a pass with a selective inversion just covering the imaging
volume. Typical imaging parameters for 4-half echo (2-half echo) are TR=4.0ms (3.0ms),
readout bandwidth=+/-125kHz, FOV=24x24x14cm, resolution= 0.94x0.94x0.94mm, flip
angle = 45º, RR interval = 2, total scan time =~6min for 45,000 (60,000) total unique
projections.
Results: Representative images from the 4-half echo sequence are shown in Figure 1.
Images show filling of the major vessels without substantial blurring. Signal diminishes as
the signal recovers from inversion and as blood distant from the volume fills the vessels.
Results indicate improved performance of the 4-half echo trajectory, due to flow
compensation and improved sampling efficiency.
Conclusion: The proposed
3D radial sequence
provides significantly
shorter TRs for a given
resolution than their
Cartesian counterparts.
Considerable acceleration
can be achieved allowing
for higher-resolution
imaging or reduced scan time.
References: 1. Kim SG.
MRM 34:293-301 (‘95) 2. Bi et
al. Proc MRA Club 08 pg.31
3. Lu et al. MRM 53:692-699
(05’)
MR-Angioclub East Lansing 2009
25
1.3 Dose 4D MR Angiography
Gerhard Laub, Ph.D.
Siemens Healthcare, USA
Time resolved contrast-enhanced MR angiography has been increasingly used to
evaluate the hemodynamic status of normal versus abnormal vasculatures. Fast imaging
sequences, parallel imaging, and view-sharing techniques have been applied to provide
the needed temporal and spatial resolution. The Gadolinium-based contrast agent is
sometimes injected in double dose to enhance the image quality. In light of NSF and the
desire to lower the amount of Gadolinium-based contrast agent to the patient, we have
investigated the use of time resolved TWIST imaging (Time-resolved Imaging with
Stochastic Trajectories) in combination with a small dose of diluted contrast agent for 4D
imaging of the extracranial vasculature.
In this study, we tested the feasibility of using a very low dose of contrast for timeresolved MRA in patients referred to get a clinical MRA examination. For low dose
dynamic MRA, 1-2 ml of Gd-DTPA, diluted with saline at a rate of 1 part Gd and 3 parts of
saline, was injected at a rate of 2 ml/sec. This was compared to routine contrast-enhanced
MRA using a single dose (0.1 mmol/kg) of contrast agent. All imaging was performed at
3T using a combination of a 12-channel head array, 4-channel neck array, and 6-channel
thorax array to extend the FOV and cover the entire aortic arch, the carotid arteries, and
the intracranial arteries all dynamically. Parallel imaging was used in two phase encode
directions with an acceleration factor of up to 9. An additional acceleration factor of 3.8
was achieved using the TWIST dynamic mode. 3D imaging, with 100 slices (slice
resolution = 2.5 mm), was acquired with an in-plane resolution of 1.3 mm x 2.2 mm and
interpolated to isotropic voxels of 1.3 mm.
Using only 2 ml of contrast agent, or less, all time-resolved results were clinically
useful to provide functional information in addition to the anatomical information provided
by the high-resolution, single-phase contrast-enhanced MRA. In this preliminary study,
there was good agreement between the low dose, time-resolved MRA and the routine,
high-resolution contrast-enhanced MRA. By using a combination of parallel imaging and
the TWIST dynamic mode, the temporal update rate was under 2 sec for each 3D
volumetric data set, depending on spatial resolution and vessel coverage.
Time-resolved, three-dimensional MRA with near isotropic resolution and large
coverage is feasible using a small amount of a Gadolinium-based contrast agents. Further
studies involving larger number of patients are needed to determine whether very low
dose time-resolved MRA would lead to any difference in clinical diagnosis.
26
MR-Angioclub East Lansing 2009
1.4 Balanced-gradient TSE for Non-contrast Peripheral MRA
1
1,2
2,3
2,3
1,2
SW Fielden , JP Mugler III , KD Hagspiel , CM Kramer , and CH Meyer
2
Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States, Department of
Radiology, University of Virginia, Charlottesville, Virginia, United States, 3Department of Medicine,
University of Virginia, Charlottesville, Virginia, United States
1
Purpose: To develop a 3D balanced-gradient TSE sequence for generating non-contrast,
non-subtractive, peripheral angiograms.
Methods:
A 3D dataset was acquired on
each of 9 healthy volunteers on a Siemens 3T Trio scanner (Siemens Medical Solutions)
with a TSE sequence modified with (1) a
frequency-encoding
gradient
which
is
rewound between each set of refocusing
pulses and (2) excitation and refocusing RF
pulses applied along the same axis with a
180° phase alternation along the echo train,
analogous to common implementations of
balanced SSFP. Sequence parameters were
as
follows:
TR/TE/Echo
Spacing
=
3000/230/3.1 ms, resolution = 1.4 x 1.4 x 1.5
mm3, acquisition time = 5.38 ± 0.50 min.
Image quality was assessed based on SNR,
CNR, and vessel sharpness.
Results and Conclusion: The incorporation of balanced gradients and RF-pulse phase
alternation in a TSE sequence resulted in T2-weighted images with reduced flow-related
signal loss and blurring, yielding good contrast between arteries and veins without
subtraction1. As seen in the coronal MIP figure to the right, blood-muscle contrast is
similar to that obtained with flow-independent angiography2 and to that obtained with
traditional TSE sequences. The residual fat and edematous signal may be mitigated in
the future via chemically selective pulses and an inversion preparation.
References:
1
Miyazaki M, et al. Radiology, 227(3):890-6, 2Brittain J, et al. Magn Reson
Med, 38(3):343-454
MR-Angioclub East Lansing 2009
27
1.5 Self-Gated Free Breathing 3D Cardiac Cine Imaging With Data
Acquisition During Slice Encoding
1
1
1
Jing Liu , Martin R. Prince , Yi Wang
Weill Medical College of Cornell University, New York, NY
Email: [email protected]
1
Self-gated free-breathing 3D cardiac cine imaging is becoming a promising
technique for heart visualization and functional measurements. The common techniques
for self-gating requires additional scan time for acquiring extra gating data. The purpose of
this study was to develop a cardiac and respiratory self-gated free-breathing 3D pulse
sequence that avoids extra scan time and provides robust self-gating information.
The multi-echo 3D hybrid radial pulse sequence was modified by adding
readout during the slice encoding pulse.
The acquired data during slice encoding
pulse are phase encoded k-space
centers along kz (Fig. 1). Different slice
encoding gives different kz coverage,
while the combination of sequential slice
encodes gives full kz coverage. The 1D
Fourier transform of this combined data
provides the z intensity profile, which
Fig. 1. Samples acquired during sequential slice
encodes (10 slices for example).
contains both cardiac and respiratory
motion information for cardiac and respiratory self gating. Typical imaging parameters
were: TR/TE/FA/BW/FOV=4.5ms/1.3ms/40°/±125kHz/34cm, 256x256 matrix, 10 slices of
slice thickness 10mm. A 1.5 T GE EXCITE 14M4 scanner was used.
Fig. 3 shows short axis heart images acquired within a scan time of 3.4 minutes.
The respiratory gating efficient was 50%.
A self-gating technique
with
during
extra
slice
data
acquisition
encoding
was
demonstrated with 3D hybrid
radial imaging. It can be applied
to other 3D pulse sequences,
such as 3D Cartesian and spiral
pulse
sequences.
Further
validation experiments of the
Fig. 3. Short axis images of five presentative slices at
end diastole (top row) and end systole (bottom row).
proposed self-gating method are underway.
28
MR-Angioclub East Lansing 2009
2.1 A Simple View of Compressed Sensing and How it Could
Change Everything We Do in MRI and MRA.
1
1
Mark A Griswold , Nicole Seiberlich
Dept. of Radiology, Case Western Reserve University, Cleveland, OH
1
MRA provides exquisite depiction of vascular abnormalities without the ionizing radiation
found in conventional DSA or CTA. However, significant drawbacks still exist. This is
primarily due to the limited speed and SNR of MRI, and most important, the fact that they
are linked to each other. Traditionally, any increase in imaging speed has required a loss
in SNR and vice versa. In order to realize any truly dramatic increases in either SNR or
imaging time, some way to break this relationship must be found. The class of newly
described Compressed Sensing (CS) methods promises to revolutionize MRI by breaking
this link and could potentially allow the development of a set of completely new imaging
strategies with dramatic increases in SNR and imaging speed. Unfortunately much of the
field of CS is described in highly mathematical terms, limiting the ability of the average
listener to fully grasp the concepts involved. In this talk, we will review some of these basic
concepts of CS in a non-mathematical, intuitive way. In general, we will demonstrate how
CS will change the focus of an MR acquisition from simply collecting images to directly
collecting information. In particular, we will focus on how somewhat older methods, such
as UNFOLD, BLAST, PARSE, and other related methods can be seen in the framework of
CS, and will also show how methods such as HYPR meet this goal of directly collecting
the important information. Finally, we will highlight several examples where these new
methods have dramatically improved both the SNR and temporal resolution beyond all
previously established limits to provide clinically useful exams in dramatically reduced time
with increased SNR.
MR-Angioclub East Lansing 2009
29
2.2 Design of Compressed Sensing Reconstruction for Highly
Accelerated Time-Resolved MR Angiography
Julia Velikina, Alexey Samsonov
Departments of Medical Physics and Radiology, University of Wisconsin – Madison
Introduction: Many angiographic tasks require high temporal resolution, which results in
data undersampling.
A number of constrained reconstruction methods including
compressed sensing (CS) have been proposed to mitigate problems of conventional
reconstruction (aliasing and low SNR)1,2. However, even natural sparsity of angiographic
images cannot support acceleration factors higher than 4-8 without loss of spatial
resolution and artifacts. In time-resolved imaging, the efficiency of CS can be extended by
promoting joint sparsity in both spatial and temporal dimensions3,4.
In this work, we
evaluate the performance of two CS approaches: k-t FOCUSS5 (sparsity in x-f domain)
and temporal constraining (sparsity of finite differences in x-t domain) in contrastenhanced (CE) and phase contrast (PC) imaging.
Theory and Methods: We adapted spatial/temporal regularization4 into CS framework.
The time series
f
is estimated as follows:
2
f = arg min Ef − m + λΦ( Lf ) 
2
f 

vector m , regularization parameter
Φto be a hybrid  1
acceleration (via
/  2 norm
6
[1], with the encoding matrix E
. L is a 1
λ
st
, data
or 2nd temporal derivative. We chose
to provide both SNR optimization (via
 2) and CS
 1norm).
Results and Conclusions: All approaches were tested on CE and PC data acquired in
healthy volunteers. For Cartesian trajectories acceleration of 8-10 were achieved, while
radials allowed acceleration up to 40 without loss of temporal and spatial resolution. The
developed temporal regularization was found to outperform kt FOCUSS in preservation of
temporal waveforms for high acceleration factors. Additionally, it was found more forgiving
to patient motion than kt FOCUSS. CS methods based on sparsity in x-t domain (Eq. [1])
are promising to accelerate time-resolved angiography.
References:
[1] Çukur T, et al MRM, 2009;61:1122. [2] Mistretta CA, et al.
MRM
2006;55:30. [3] Portniaguine, et al. ISMRM 2003,481. [4] Samsonov, ISMRM 2005, 2311.
[5] Tsao J, et al. MRM 2003;50:1031. [6] Bube KP, et al. Geophysics,1997:62:1183.
30
MR-Angioclub East Lansing 2009
2.3 Low Dose HYPR FLOW
Yijing Wu, Steven Kecskemeti, Kevin Johnson, Charles Mistretta, Patrick Turski
Departments of Medical Physics and Radiology, University of Wisconsin, Madison, WI
INTRODUCTION: Time resolved contrast-enhanced magnetic resonance angiography
has been widely used to evaluate the hemodynamics of the vascular structure. Due to
the recent concern of NSF disease, eliminating or reducing the Gadolinium-based
contrast agent is more desirable than ever. Phase Contrast (PC) HYPR FLOW is able to
decouple the high spatial resolution and SNR, which require relative long scan time,
from high temporal resolution, which demands for fast data acquisitions, and used
HYPR constrained reconstruction to obtain a time series of images with both high
temporal resolution, isotropic high spatial resolution, high SNR and quantitative flow
dynamics from the PC images. Our hypothesis is that the SNR of the HYPR FLOW
images depends on the post contrast PC composite, which requires minimum amount of
contrast agent. High temporal and spatial resolution time resolved contrast-enhanced
MRA can be obtained by using low dose HYPR FLOW method with reduced contrast
agent.
METHODS AND RESULTS: Low dose HYPR FLOW has been tested in three normal
volunteers and one brain AVM patient. Contrast agent was reduced to one half or one
quarter of a single dose (0.1 mmol/kg). Following contrast injection, a CE-MRA
examination of the head is performed using time resolved milt-echo VIPR.
Subsequently, a 5 minute PC VIPR acquisition was acquired and then used as a
composite image for HYPR LR reconstruction. The figure on the right shows a time
series
of
HYPR
FLOW
1
2
4
6
25
35
images of the AVM patient
with
half
dose
contrast
agent (~ 6 cc). Numbers on
8
15
the images are relative time
frames
after
contrast
arrival. Frame rate was 2/s,
with 0.7 mm isotropic spatial resolution. Our preliminary results show that low dose
HYPR FLOW is able to provide high temporal and spatial resolution with adequate SNR
and potential shorter bolus dispersion for intracranial MRA.
MR-Angioclub East Lansing 2009
31
2.4 Myocardial Perfusion MRI in Canines with Improved Spatial
Coverage, Resolution and SNR
1
Lan Ge , Aya Kino1, Daniel Lee1, Rohan Dharmakumar1, Mark Griswold2, Charles Mistretta3, James
Carr1, Debiao Li1
1
Northwestern University, Chicago, IL, USA, 2 Case Western Reserve University, Ohio, USA,
3
University of Wisconsin-Madison, Madison, WI, USA
Purpose: First-pass perfusion MRI is a promising technique for detecting ischemic heart
disease. A combination of sliding window and CG-HYPR (1, 2) methods (SW-CG-HYPR)
have been proposed to increase spatial coverage, resolution, and SNR(3). In this work,
using a controlled animal model, we compare this new method with conventional clinical
protocols and test whether the flow deficits can be detected accurately.
Methods: Five dogs with LCX occlusion were scanned using a 1.5T system during the
first-pass of the contrast agent in stress condition. An ECG-triggered, turbo-FLASH
sequence with radial k-space sampling and saturation recovery (SR) preparation was
used in this study. CG-HYPR method was used to reconstruct the time-resolved images.
The signals from the left ventricle, healthy myocardium, and flow deficits for all of the three
methods were measured and compared.
Results: The SNR of the left ventricle at peak enhancement with SW-CG-HYPR
(32.1±2.32) is significantly higher than turbo-FLASH (20.6±2.62) and EPI (12.9±1.95).
Figure 1 shows examples of the comparison. The defects caused by LCX occlusion can
be clearly delineated in SW-CG-HYPR images. The signal intensity changes of the healthy
myocardium are highly correlated with a correlation coefficient of 0.956.
Conclusions: In conclusion,
SW-CG-HYPR is a
turbo-FLASH vs. SW-CG-HYPR
EPI vs. SW-CG-HYPR
Reference
images
promising method to
improve the
Deficits
SW-CG-
Deficits
myocardial perfusion MRHYPR
imaging with reduced
images
acquisition window
Figure 1. Comparison of conventional method and SW-CG-HYPR
increased spatial coverage,
improved spatial resolution and SNR.
References: 1. Mistretta CA, et. al. MRM, 55: 30-40, 2006. 2. Griswold MA, et. al. Proc
ISMRM, Berlin, 2007: 834. 3. Ge L, et al. Proc ISMRM, Toronto, 2008: 43
32
MR-Angioclub East Lansing 2009
2.5 Reconstruction of MR Angiography Images using Gradient
Descent with Sparsification
Nicole Seiberlich and Mark A. Griswold
Department of Radiology, Case Western Reserve University, Cleveland OH
Purpose: Recently, a new method for generating sparse images from highly
undersampled data, Gradient Descent with Sparsification, has been introduced [1]. This
method iteratively determines x, the sparse image to be reconstructed using the following
formulation:
x ← H ( x + γ ⋅ ΦT ( y − Φx))
where y is the k-space data,Φ
is the
resampling/degridding operation, H is a thresholding operation, and
γcontrols the rate of
convergence. This simple reconstruction algorithm was tested for the reconstruction of
images from highly undersampled MR Angiography data.
Methods: CE-MRA data were acquired on a patient with an arterial-venous malformation
(AVM) with the following parameters: radial GRE acquisition, TR=3ms, TE=1.5ms, total
projections=1344, 75% asymmetric echo, Matrix=192x192, FOV=220x220 FA=20°,
Partitions=15, slice thickness=4mm. Images were reconstructed using Gradient Descent
with Sparsification with 32 projections per frame, with the thresholding value H determined
using the center of k-space and
γ
fixed as suggested in [1].
Results: Time frames showing the
arrival of contrast into the AVM
reconstructed
Descent
with
using
Gradient
Sparsification
are
shown in Figure 1. The streaking
Figure 1: Gradient Descent with Sparsification
reconstruction depicting flow of contrast into the AVM.
commonly evident in such highly undersampled images has been removed, and the
vessels feeding the AVM are clearly depicted without venous contamination.
Conclusion: Gradient Descent with Sparsification is a simple and fast method to
reconstruct highly undersampled sparse images, including MRA data. Unlike other
reconstruction methods, no composite image is required, reducing reconstruction errors.
Further improvements to the method such as the inclusion of coil sensitivity maps or
constraint on the thresholding images will be considered in the future.
References: [1] Garg R and Khandekar R. Proc. 26th International Conference on
Machine Learning, Montreal, Canada, 2009.
MR-Angioclub East Lansing 2009
33
3.1 Carotid Plaque Imaging and Clinical Risk Assessment
Chun Yuan, PhD, Hunter Underhill, MD, Thomas Hatsukami, MD
Vascular Imaging Laboratory, School of Medicine, University of Washington, Seattle, WA
Carotid MRI has been proven to be able to measure plaque size and characterize tissue
composition. This information provides unique opportunities to study the ‘vulnerable
plaque’ in vivo. This talk aims to summarize the current understanding of vulnerable
plaque features based on longitudinal studies of carotid atherosclerosis, as well as the
relationship between carotid atherosclerosis and neurological symptoms. This talk will also
discuss the current technical and clinical needs of atherosclerosis imaging.
34
MR-Angioclub East Lansing 2009
3.2 Carotid Intraplaque Hemorrhage Is Associated with
Enlargement of Lipid-rich Necrotic Core and Plaque Volume Over
Time: In Vivo 3T MRI Prospective Study
H Ota1, D. Zhu1M, JK DeMarco1
Michigan State University, East Lansing, MI, USA
1
Purpose: To test the hypothesis that intraplaque hemorrhage identified by 3D Inversion
recovery fast SPGR (IR-FSPGR) images contributes to plaque progression over time as
measured by multi-contrast carotid MR imaging at 3.0T.
Methods:
Twenty-six consecutive subjects with known 50-99% carotid stenosis
underwent serial carotid 3T MRI scans with a multicontrast weighted protocol (pre- and
post-contrast T1W, T2W, 3D TOF and 3D IRFSPGR) with intervals of 0.5-2.2 years. Two
reviewers blinded to subject’s clinical information and scan date interpreted images. In
order to insure a similar coverage of the carotid artery for quantitative measurements, only
image locations that could be matched across 2 time points were reviewed. The volumes
of vessel wall, lumen, lipid-rich necrotic core, hemorrhage and calcification were
documented for each study. The changes in each metric adjusted for the period (mean ±
standard deviation/year) were evaluated in the groups with and without intraplaque
hemorrhage at the baseline scans using one sample t-test.
Results: Eight subjects (31%) had intraplaque hemorrhage at baseline. Mean lipid-rich
necrotic core volume at baseline was significantly larger for patients with hemorrhage
(p=0.001) than those without hemorrhage. The mean volumes of lumen, wall and
calcification at baseline were not significantly different. In the eight subjects with
intraplaque hemorrhage at baseline, significant increases of the volumes were found for
wall (23.7± 26.3mm3/year, p=0.038), lipid-rich necrotic core (44.0±30.6 mm3/year,
p=0.005), and intraplaque hemorrhage (24.1±23.4 mm3/year, p= 0.022). The changes in
the volumes of lumen and calcification were not significant. In the remaining 18 subjects
without intraplaque hemorrhage at baseline, the changes in the volumes of wall, lipid-rich
necrotic core were not significant over time.
Conclusion: Intraplaque hemorrhage noted on baseline carotid wall 3T MR imaging is
associated with larger lipid rich necrotic core at baseline as well as significant progression
of overall plaque volume, the size of the lipid-rich necrotic core and intraplaque
hemorrhage over time.
MR-Angioclub East Lansing 2009
35
3.3 The 3D SHINE Sequence Optimizes the Quantification of
Carotid Intraplaque Hemorrhage
1
David C. Zhu , Hideki Ota1, Marina S. Ferguson2, Anthony T. Vu3, J. Kevin DeMarco1
1
Michigan State University, 2University of Washington, 3GE Healthcare
Introduction: Carotid intraplaque hemorrhage (IH) has been shown to promote plaque
progression. A novel optimized 3D SHINE sequence was developed to detect IH based on
high T1 contrast and to characterize hemorrhage into type I (early) and type II (recent)
based on T2* maps (1). With these advantages, algorithms were developed to estimate the
size and composition of intraplaque hemorrhage.
Methods: 3D SHINE images (1) were collected from nine patients with carotid IH at 3T.
At the first TE, the signal intensity ratios of IH versus lumen, wall and surrounding muscle
were 7.94 ± 3.4, 3.20 ± 1.21 and 3.82 ± 0.92. These high contrasts allowed for reliable
computerized segmentation. A target ROI contained IH and a background ROI contained
the surrounding muscle. At each slice, each voxel was normalized by the mean signal
intensity of the background ROI at this slice. Normalization reduced the signal intensity
variation due to the surface coil, and allowed a uniform cutoff of 1.5 on each slice to
identify the probable IH voxels. These voxels were further labeled as “IH” if they were
within a minimal “IH” size of 0.68 mm3 (14 voxels). The IH region was further divided into
type I and type II based a T2* cutoff value of 14 ms (1). The sizes of the segmented
intraplaque hemorrhage and its sub-regions were then calculated.
Results and Discussion: The segmented intraplaque hemorrhage (middle figure,
red/yellow: type I, blue: type II) shows a close match with the original data (left figure) and
a reasonable correspondence with matched histology data (right figure). A semiautomated quantification of intraplaque hemorrhage would allow the monitoring of plaque
progression and could be used as an objective tool in multi-site clinical trials.
Reference:1. Zhu DC, Ota H, Vu AT, DeMarco JK. ISMRM 2009, Honolulu, Hawaii.
36
MR-Angioclub East Lansing 2009
3.4 Improved Intraplaque Hemorrhage Detection by a Phase
Sensitive IRTFE (SPI) Sequence
Jinnan Wang 1, Marina Ferguson 2, Chun Yuan 2, Peter Börnert 3
Affiliations: 1. Philips Research North America, 2. University of Washington, 3. Philips Research Europe
Purpose: Hemorrhage in atherosclerosis plaque has been linked to an increased risk of
plaque progression and rupture [1]. However, only a small portion of the dynamic range
have been utilized [2, 3] by current T1 based hemorrhage detection sequences. This can
lead to inaccurate identification and segmentation of plaque components. In this study, a
Slab-selective Phase-sensitive IRTFE (SPI) sequence is proposed to improve image
contrast of intraplaque hemorrhage.
Methods: The SPI sequence is adapted from a regular IR-TFE sequence by making the
inversion pulse slab selective and phase sensitive. Simulations are used to evaluate the
signal contrast increase by using this novel SPI sequence.
9 carotid endarterectomy (CEA) specimens were scanned with both SPI and regular
IRTFE sequences. The hemorrhage to lumen contrast for each specimen was calculated
and compared between SPI and IRTFE images. Three of the nine CEA specimens were
processed for histology with 1 specimen confirmed to contain intraplaque hemorrhage.
Results: Simulations
indicate that contrast
between hemorrhage
and normal vessel wall
can be increased by
over 50%. The hyper
intense regions on
both images
Fig.1 Ex vivo MR images using routine IRTFE (left) and SPI (middle).
Matching histology (right) confirms the presence of intraplaque
hemorrhage. Note the improved contrast of hemorrhage on the SPI
correspond very well
sequence (arrows).
to the region of hemorrhage on matching histology. An approximate 30% improved tissue
contrast was obtained on the SPI image.
Conclusion: Simulation and ex-vivo experiments confirm that the novel SPI technique
can improve hemorrhage contrast on MR images by utilizing phase information. This
improved tissue contrast can potentially improve the identification and quantification of
intraplaque hemorrhage by carotid plaque MRI.
References: 1. Takaya N, et al. Stroke. 2006; 37:818-23. 2. Moody AR, et al. Circulation
2003;107;3047-3052. 3. Zhu DC, et al. 2008;26:1360-6.
MR-Angioclub East Lansing 2009
37
3.5 Fibrous Cap Thickness Assessment: Fact or Fiction?
William S. Kerwin, Huijun Chen
University of Washington, Department of Radiology, Seattle, WA, USA
Purpose – Determining fibrous cap thickness in carotid atherosclerotic disease is the holy
grail of plaque imaging as cap thickness may be the most important factor in distinguishing
between vulnerable and stable lesions. While histological studies have indicated that the
key break point for vulnerable cap thickness is on the order of 200 microns [1], MRI
resolution in carotid studies is typically >500 microns. Nevertheless, claims of identifying
thin caps (<250 microns) [2] and actual measurement of cap thickness [3] have been
made. The purpose of this study is to assess the extent to which cap thickness can be
assessed at the fringes of MRI resolution.
Methods – A series of physical and computational phantoms that mimicked fibrous cap
geometry were used to probe the relationship between cap thickness, image resolution,
and appearance under MR imaging conditions. Based on these phantoms, a linear Fisher
classifier was constructed that produced a mathematical model for classifying fibrous cap
thickness derived from 11 intensity-related parameters. This classifier was then applied to
26 contrast-enhanced plaque images from 7 individuals with histologically measured cap
thickness.
Results – The experiments indicated that the fibrous cap could not be visualized when its
thickness was less than about half the fundamental resolution, which coincides well with
observations using the absence of a band separating the core from the lumen to define
“thin” caps [2]. Above this threshold, differences in cap appearance that correlated with
cap thickness could be ascertained even below the fundamental resolution; however,
measured thicknesses were heavily biased and sensitive to SNR and physical MR
parameters when the thickness was <1.5 times the fundamental resolution.
The
determinant from the linear Fisher classifier was significantly correlated (p<0.05) with
measured cap thickness for in vivo imaging.
Conclusion – MRI is able to differentiate caps of different thicknesses even below the
fundamental resolution, using intensity criteria. Measurements of cap thickness will be
unreliable, however, unless resolutions of 100-300 microns can be attained in vivo.
[1] Redgrave Stroke 2008; [2] Hatsukami, Circ 2000; [3] Sadat, Atheroscl 2009
38
MR-Angioclub East Lansing 2009
3.6 Gradient Echo Based Sequence Provides More Information
from Ex Vivo Carotid Plaque Specimens
1
1
Rui Li , Chun Yuan
Affiliation: 1. Radiology Department, University of Washington
Purpose: Spin echo (SE) based MRI sequences are generally used for high resolution
plaque imaging. Criterion for vulnerable plaque also uses SE based T1, T2 and PD
weighted images to access the components in the plaque[1]. However gradient echo (GE)
based sequences has rarely been explored for plaque evaluation. This study aimed to find
out whether we can get more information from GE by carotid specimen examination.
Methods: Conventional GE imaging was performed on a 3T MRI system (Philips Achieva)
with the carotid specimens fixed in 70% formalin solution. The main parameters were TR:
721ms, TE: 9.2ms, FA: 60o, SLC: 32, TH: 1mm, FOV: 24*24mm2, and Res: 0.16*0.16mm2.
Four carotid endarterectomy specimens were inspected. Histogram, joint entropy and
conditional entropy[2]
were used to evaluate
the information for the
whole plaque and the
lipid rich necrotic core
(LRNC).
Results:
Ex
vivo
images and their
Figure 1. Cross-sectional image and histogram of different ROI
histograms are shown in
figure 1. The intensity distribution of
GE
and
PD
is
scattered.
Information
measured by joint entropy and conditional
entropy is shown in table 1. PD and GE
images contributed more information to joint
Information
H(T1, T2, PD, GE)
H(T1 | T2, PD, GE)
H(T2 | T1, PD, GE)
H(PD | T1, T2, GE)
H(GE | T1, T2, PD)
Plaque
13.37
0.65
0.80
1.10
0.92
LRNC
11.31
0.29
0.30
0.49
0.47
Table 1. Joint and conditional entropy
entropy. We think this extra information comes from T2* weighting in GE based sequence.
Conclusion: Gradient echo based sequences can provide additional information to
existing plaque imaging protocols.
Reference: [1] C. Yuan, W.S. Kerwin, JMRI, 2004, 19, 710-719. [2] R.G. Gallager,
Information Theory and Reliable Communication, Wiley, 1968.
MR-Angioclub East Lansing 2009
39
3.7 Improved Black Blood Multi-Contrast Protocol for
In-vivo Atherosclerotic Imaging
1,3
Seong-Eun Kim , John Roberts, Gerald S. Treiman2,4, Dennis L. Parker1,3
Utah Center for Advanced Imaging Research, 2VA Salt Lake City Health Care System, 3Radiology,
4
Surgery, University of Utah
1
Purpose: Atherosclerotic plaque characterization by MRI is generally based on the signal
intensities and morphology of plaque in T1, PD and T2 weighted images, but intraplaque
thrombus cannot be detected by conventional contrast imaging. Our multi-contrast
protocol including DWI and 3D MPRAGE may help detect plaque hemorrhage and assist
in the identification of plaque components in the cervical carotid artery.
Methods: DW images (1.0x1.0x2.0 mm3) were acquired using 2D ss-IMIV DWEPI with b
=10 and 300 s/mm2 to create the ADC maps. 2D T1w, T2w and PD images of the same
locations were acquired (0.5x0.5x2.0 mm3) with 2D TSE. (The T1w with our modified
version of the double inversion preparation). 3D MPRAGE images were acquired
(0.5x0.5x1.0 mm3) with non-selective inversion preparation and fat saturation. Total scan
time including 2D and 3D TOF was less than one hour.
Results: Multi-contrast Images of atherosclerotic plaque with hemorrhage from a patient
are shown at the left. The plaque area indicated by white arrows shows a moderate signal
on T1w, T2w and low ADC value (0.29x10-3 mm2/s). 3D MPRAGE images show the hyperintense hemorrhage region in the carotid plaque.
Discussion:
The results obtained indicate that multi-contrast black blood images
including DWI (as a new contrast) and 3D MPRAGE may be of substantial value for
carotid plaque identification.
T1w
40
T2w
ADC
MR-Angioclub East Lansing 2009
3DMPRAGE
3.8 3D peripheral vessel wall MRI with flow-insensitive blood
suppression and isotropic resolution at 3 Tesla
Thanh D. Nguyen, Keigo Kawaji, Pascal Spincemaille, Matthew D. Cham,
Priscilla Winchester, Martin R. Prince, Yi Wang
Weill Medical College of Cornell University, New York, NY
Email: [email protected]
Flow-insensitive T2-prepared inversion recovery (T2IR) has been shown to provide
better blood suppression for 2D arterial vessel wall imaging at 1.5T than double inversion
recovery (DIR) in the lower extremities where blood flow is slow (1). T2IR offers global
blood suppression regardless of flow velocity and direction at the cost of reduced wall
SNR. The purpose of this study was to develop a cardiac triggered 3D black blood T2IR
fast spin echo sequence for vessel wall MRI at 3T.
For comparison, 4 normal volunteers (27 ± 8 years) were imaged at both 1.5T and 3T
(GE HDxt) with parameters: TR=1RR, TE=20 ms, FOV=28 cm, matrix=384x384, coronal
slice thickness=1.4 mm (1.5T, interpolated to 1.5 mm) and 0.7 mm (3T), number of
slices=100/200 (1.5/3T), NEX=2/1
(1.5/3T),
bandwidth=±62.5
.
kHz,
ETL=64 (variable flip angles to
increase sampling efficiency), selfcalibrated parallel imaging (ARC)
factor R=1.9, partial kz factor=0.75,
fat suppresion, scan time~8 min, 8channel cardiac coil. For T2IR
preparation, T2PREP time=120/150
ms (1.5/3T), and TI~250/300 ms
(1.5/3T).
a
Figure 1 shows images of
distal
superficial
femoral
and
b
Figure 1. Curved reformatted vessel wall images
Obtained at a) 1.5T b) 3T.
proximal popliteal arteries obtained at 1.5T and 3T, demonstrating extended bilateral
coverage, excellent arterial blood suppression across large FOV, and good wall
visualization. Note that 3T images offer true 0.7x0.7x0.7 mm3 isotropic resolution with
similar wall SNR. In conclusion, the global blood suppression of T2IR and the higher SNR
afforded by 3D acquisition and higher field strength allow efficient imaging of vessel wall
with sub-millimeter isotropic resolution.
References. Brown R. ISMRM 2009. p3844.
MR-Angioclub East Lansing 2009
41
3.9 A 16 Channel Anterior Neck RF Coil for Cervical Carotid MRA
Quinn Tate, Emilee Minalga, Laura C. Bell, J. Rock Hadley
Utah Center for Advanced Imaging Research, Dept of Radiology, University of Utah
Purpose: Increased speed in cervical carotid imaging is highly desired to reduce the
artifacts from occasional motion such as swallowing.
Methods: We have developed a 16 channel receive only RF coil array with overlapping
elements placed on a form fitting fiberglass former as shown in Figure 1.
Figure 1: A 16 channel (8 channels on
each side) RF coil array for the neck,
with emphasis on the cervical carotid
artery. The array was fabricated based
on an overlapping element philosophy.
A second coil array is being fabricated
based upon evaluations of this coil
array.
Below are shown example
images from the 16 channel coil for (left)
R=1 (no reduction, 6:44min), (middle)
R=2 (3:25min) and (right) R=3 (2:43min)
using
2D TSE with DIR prep TE/TR=8/885ms
matrix=256x256
ETL=9
TI=600ms 2 AVG, FOV=130mm
Results: Images shown in Figures 1 and 2 illustrate the improvement for parallel
imaging over the 4 channel bilateral RF coil that we currently use.
Figure 2: (left) Head/neck positioner with 4 channel (2 channel bilateral) carotid coil.
(right) Comparison 2D DIR TSE images (TE/TR=8.8/885ms, TI= 600ms, 0.5x0.5mm2
inplane, 2mm slice thickness) obtained using the 4 channel coil shown at left (bottom
row) and the 16 channel coil shown in Figure (top row). With a reduction factor of 2, the
16 channel coil images are less noisy.
Conclusion: The new 16 channel coil shows great potential for rapid carotid MRI.
42
MR-Angioclub East Lansing 2009
4.1 Gadobenate dimeglumine vs gadopentetate dimeglumine for
peripheral MR angiography: comparison with DSA
SC Gerretsen1, T le Maire2, S Miller3 , SA Thurnher4, CU Herborn5, H Michaely 6,
7
8
9
10
1
H Kramer , A Vanzulli , J Vymazal , M Wasser , T Leiner
1. Maastricht University Hospital, Maastricht, The Netherlands; 2 Catharina Hospital, Eindhoven,
Netherlands; 3. Eberhardt Karls University, Tuebingen, Germany; 4. Hospital Brothers of St. John of
God, Vienna, Austria; 5. University Medical Center, Hamburg-Eppendorf, Germany, 6. University
Hospital, Mannheim, Germany, 7. Ludwig Maximilians University, Munich, Germany, 8. Ospedale
Niguarda Ca' Granda, Milan, Italy, 9. Na Homolce Hospital, Prague, Czech Republic, 10. Leiden
University Medical Center, Leiden, Netherlands
Purpose: To prospectively compare equivalent 0.1 mmol/kg doses of gadobenate
dimeglumine and gadopentetate dimeglumine in patients undergoing contrast-enhanced
MR angiography (CE-MRA) of the peripheral arteries.
Methods: 96 adult subjects with suspected moderate-to-severe peripheral arterial
occlusive disease (PAOD) were enrolled at 7 sites. Patients underwent 2 identical 1.5-T,
3-station, CE-MRA examinations from the aortic bifurcation to the lower leg with
randomized
0.1
mmol/kg
bodyweight
doses
of
gadobenate
dimeglumine
and
gadopentetate dimeglumine. Diagnostic performance (sensitivity, specificity, accuracy,
positive predictive value [PPV], negative predictive value [NPV]) was determined in a
subset of patients (n=31) that also underwent conventional DSA. The presence and extent
of steno-occlusive disease on DSA images was determined on a segmental basis using a
4-point scale (1=stenosis ≤25%; 2=stenosis >25–≤50% 3=stenosis >50–99%; and
4=occlusion). Statistical analyses were performed using the Wilcoxon Signed Rank,
McNemar, and Wald tests.
Results: A total of 397 segments were evaluated by DSA. Of these 397 segments, 270
(68.0%) had stenoses of ≤50% while 127 (32.0%) had hemodynamically-relevant (>50%)
stenoses. All 3 blinded readers reported significantly (p≤0.0017) better diagnostic
performance with gadobenate dimeglumine compared to gadopentetate dimeglumine, with
increases of 11.0–18.1% in sensitivity, 4.4–9.3% in specificity, and 7.8–10.1% in overall
accuracy (Table 1). Readers also reported significantly (p≤0.0028) higher PPV and NPV
with gadobenate dimeglumine, with differences ranging from 12.7–19.3% for PPV and
5.5–7.9% for NPV.
Conclusion: In patients with suspected PAOD referred for CE-MRA, use of 0.1 mmol/kg
bodyweight gadobenate dimeglumine results in significantly better diagnostic performance
than use of an equivalent dose of gadopentetate dimeglumine.
MR-Angioclub East Lansing 2009
43
4.2 Dose Comparison between Conventional and
High Relaxivity Contrast Agents in Peripheral MRA
Jeffrey H. Maki1, George R. Oliveira1, Gregory J. Wilson2
1 - University of Washington Dept of Radiology, Seattle, WA. 2 - Philips Medical Systems, Cleveland, OH.
Purpose Evaluate the utility of standard dose (~0.1 mmol/kg) high relaxivity (SDHR)
gadolinium contrast for peripheral MRA (pMRA), and compare this to high dose high
relaxivity (~0.2 mmol/kg) (HDHR) and high dose conventional relaxivity (HDCR)
gadolinium contrast.
Methods 60 patients undergoing 3 station moving table pMRA for suspected peripheral
vascular occlusive disease received one of three contrast agents/doses (20 each): 17 cc
gadobenate dimeglumine (MultiHance; Bracco Diagnostics), 34 cc gadobenate
dimeglumine, or 34 cc gadoteridol (ProHance; Bracco Diagnostics).
Imaging was
performed on a 1.5T system (Gyroscan NT, Philips Medical Systems) using a prototype
18 channel coil and a SNR/timing optimized pulse sequence designed to maximize SNR
while avoiding venous contamination1. Upper/middle scan times ranged from 5-12.5 sec,
with lower station 45-60 sec. Arterial contrast ratios (CR: arterial SI minus muscle SI
divided by muscle SI) were calculated for each station and corrected for differences in
TR/TE. Subjective image quality and venous enhancement were evaluated. Statisitical
analysis was performed using the Student t-test and Mann Whitney U tests.
Results All studies were diagnostic and high quality. Contrast ratio was uniformly higher
for HDHR contrast in all stations, but only significant in the lower station. Interestingly,
upper station HDHR image quality was rated significantly worse than the others despite
its higher CR. This trend has been noted in other studies, where high dose gadobenate
dimeglumine appears to decrease MRA image quality/diagnostic efficacy2. There was no
significant contrast ratio or image quality difference between SDHR and HDCR, however
lower station venous enhancement was significantly less than the others for SDHR.
Conclusion
Excellent quality peripheral MRA can be performed with standard dose
gadobenate dimeglumine, as overall quality is equivalent to or better than double dose
conventional contrast. Increasing HR contrast dose improves CR, but may not improve
image quality. Further investigation into this phenomenon is underway.
References
1.
Potthast et al. J Magn Reson Imag 29:1106-1115, 2009.
2.
Schneider et al. J Magn Reson Imaging. 26(4):1020-32, 2007.
44
MR-Angioclub East Lansing 2009
4.3 Peripheral MRA With Continuous Table (CTM) Movement in
Combination with High Temporal and Spatial Resolution TWISTMRA With 0.1 mmol/kg Gadobutrol at 3.0 T
1
Voth M , Haneder S1, Gutfleisch A1, Schoenberg SO1, Michaely HJ1
Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical
Faculty of Mannheim, University of Heidelberg, Germany
1
Purpose: To prove the concept of peripheral CTM-MRA in combination with high spatial
and temporal resolution time-resolved TWIST-MRA in a single MR-exam at 3.0T with a
single dose (0.1 mmol/kg) of gadobutrol in total.
Methods: 22 consecutive patients (15m/7f, mean age 64 years) referred for peripheral
MRA with clinical symptoms of peripheral arterial occlusive disease (PAOD) Fontaine
stages II–IV underwent both CTM-MRA (TR 2.4ms/ TE 1.0ms/ flip angle 21°) of the run-off
vessels and TWIST-MRA (TR 2.8ms/ TE 1.1ms/ flip angle 20°) of the calf station during a
single MR-exam at 3.0T (Siemens Tim Trio). Spatial resolution of the CTM-MRA datasets
was 1.2mm isotropic. The TWIST-MRA was acquired with 1.1x1.1x1.35mm³ and
reconstructed to 1.1mm isotropic with a temporal resolution of 5.5 s. A total of 0.1 mmol/kg
BW gadobutrol diluted 1:1 with saline was injected at a flow rate of 1.5 mL/s of which
0.07 mmol/kg was administered for the CTM-MRA and 0.03 mmol/kg for the TWIST-MRA.
CTM-MRA run off datasets were qualitatively assessed using a four point scale (4excellent, 1-non-diagnostic) followed by TWIST-MRA datasets. Additional relevant findings
only visible in the TWIST-MRA were documented.
Results: All datasets could be evaluated with a total of 397 assessable segments. CTMMRA was diagnostic in 99% (393/397) with image quality judged as excellent in 54%
(213/397), good in 42% (14/397), and moderate in 4% (14/397) respectively. Non
diagnostic image quality was seen in 1% (4/397). TWIST-MRA was diagnostic in 100%
(115/115) with good or excellent image quality. In 14 of 22 patients additional relevant
findings were detected by TWIST-MRA.
Conclusion: Single-dose gadobutrol CTM-MRA in combination with a high spatial and
temporal resolution TWIST-MRA at 3.0 T is a reliable technique with good image quality.
Despite the use of single dose contrast agent large field of view coverage and dynamic
images can be acquired. Due to its robustness, this imaging approach of the vasculature
has great potential for a broad clinical use.
MR-Angioclub East Lansing 2009
45
4.4 Flow and Motion-Insensitive Unenhanced MR Angiography of
the Peripheral Vascular System –A pilot study in the lower
extremity
John J. Sheehan 1,2, Ioannis Koktzoglou1, James C. Carr2, Eugene Dunkle1, Robert R. Edelman1
1
Department of Radiology, NorthShore University HealthSystem, 2650 Ridge Ave., Evanston, IL
2
Cardiovascular Imaging, Northwestern University, 737 N. Michigan Ave, Ste1600, Chicago, IL
Introduction: Current limitations of unenhanced magnetic resonance angiography
techniques include sensitivity to flow velocity, cardiac rhythm, and patient motion. We
developed an alternative unenhanced method for peripheral magnetic resonance
angiography (MRA) with the potential for robust performance over a wide range of
physiological conditions.
Materials and Methods: Flow-insensitive single shot (FISS) MRA acquires data with a
specially modified single shot two-dimensional (2D) balanced steady-state free precession
(bSSFP) pulse sequence. A key feature is the use of a quiescent inflow time period
(QITP), coincident with systole, which is sandwiched between a saturation module and the
bSSFP readout. The QITP provides the opportunity for maximal inflow of unsaturated
arterial spins while ensuring suppression of venous signal. The combination of a
saturation magnetization preparation with a single shot acquisition minimizes sensitivity to
heart rate variations and arrhythmias. In order to test the clinical feasibility of the
technique, we performed a pilot study of FISS MRA using contrast-enhanced MRA as the
reference standard. A series of 4 healthy subjects (4 male, ages 28-45) and 8 patients (8
male, ages 56-90) with documented peripheral vascular disease were studied.
Results: In all subjects, FISS MRA demonstrated the entire length of the peripheral
vascular tree from aorta to pedal vessels. Considering CE-MRA as the standard of
reference examination and excluding stented arterial segments, the sensitivity, specificity,
PPV, and NPV values of FISS MRA for arterial narrowing greater than 50% or occlusion
were 92.2%, 94.9%, 83.9% and 97.7% respectively. FISS MRA provided robust depiction
of normal arterial anatomy and peripheral vascular disease, irrespective of disease
severity, in scan times on the order of eight minutes for the entire peripheral vascular tree.
In no subject was there substantial degradation of image quality due to bulk motion or
variation in cardiac rhythm.
Conclusion: FISS MRA is a fast, flow-insensitive and easy-to-use method for depicting
the peripheral arteries. In a small group of patients, the unenhanced technique had
excellent negative predictive value and image quality was consistent irrespective of the
severity of PVD. Unlike subtraction-based unenhanced 3D MRA, the technique does not
need to be tailored for each patient and initial results demonstrate reliable image quality
for the pelvic vessels despite the presence of respiratory motion.
46
MR-Angioclub East Lansing 2009
4.5 A Comparison of Time-Resolved 3D CE-MRA with
Peripheral Run-off CTA in the Calves
CR Haider, JF Glockner, TJ Vrtiska, TA Macedo, EA Borisch, SJ Riederer
MR Laboratory, Mayo Clinic, Rochester MN USA
PURPOSE
To perform a comparison of time-resolved 3D CE-MRA of the calves using Cartesian
Acquisition with Projection-Reconstruction-like sampling (CAPR) with peripheral CTA.
METHODS
Nine patients who underwent clinically indicated peripheral run-off CTA were recruited for
MRA within 48 hours of their CTA. CTA was performed using the standard protocol of our
vascular CT clinical practice on a 64-detector row CT scanner (Sensation 64; Siemens
Medical Solutions, Forschheim, Germany) resulting in 0.6 mm in-plane resolution with 2
mm thick axial sections and 1.2 mm increment. The CAPR acquisitions were performed
on a 3.0 T MRI system (GE Healthcare, Milwaukee, WI.). 3D image sets with 1 mm
isotropic spatial resolution were generated for a field of view of 40 cm S/I, 32 cm L/R, and
13.2 cm A/P using a 5 sec frame time and 20 sec temporal footprint [1, 2]. Using CTA as
the reference, the MR data sets were evaluated with respect to depiction of any pathology,
relative prominence of the luminal signal, and any added value of the time-resolved
information.
RESULTS
The diagnostic image quality of the MRA
results was rated very competitively vs.
CTA (example, Figure 1). The CAPR
sequence provided clear arterial frames in
the case of rapid arterial to venous transit
in patients with ulceration of the feet. In no
MRA study was any reconstruction artifact
observed that adversely affected image
quality.
(A)
(B)
(C)
CONCLUSION
These studies suggest that time-resolved
MRA of the calves using CAPR can
provide similar information to that
generated using CTA.
References: [1] Haider et al., MRM 60:749
(2008); [2] Haider et al., Radiology (in
press).
Figure 1. CTA vs. CAPR MRA. Vessel
noted in CTA (A, arrow) is seen to fill
retrograde in consecutive 5 sec frames in
MRA (B-C).
MR-Angioclub East Lansing 2009
47
4.6 3D Time-Resolved MR Angiography of Lower Extremities using
Cartesian Interleaved Variable Density Sampling and HYPR
Reconstruction
1a
K. Wang, 2R. Busse, 1aY. Wu, 1aL. Keith, 2J. Holmes, 1a,bF. Korosec
1b
Medical Physics, Radiology, University of Wisconsin-Madison, Madison, WI
2
Applied Science Lab, GE Healthcare, Madison, WI
1a
Purpose: To simultaneously improve spatial and
temporal resolution of 3D time-resolved MR
angiography (TR-MRA) in lower extremities using
Cartesian interleaved variable density sampling
(VDS) [1] and HYPR [2-4].
Methods: Each time frame, a
subset of k-space views are
acquired with sampling density
proportional to 1/kr as shown in
Fig. 1 and described further in Ref.
[1]. The sub-sampling patterns for
a series of timeframes are
designed to interleave, such that
the data over N timeframes is fully
sampled and HYPR methods in Ref. [3,4] can be used. Imaging parameters include: 0.94
x 0.94 x 1.5mm3 voxel size with matrix size of 512x282x72, 30 time frames with 5.8
sec/frame.
Results: Fig. 2 shows coronal MIP images at arterial (a) and venous (b) phases.
Compared with zero-filling, HYPR reduces the spatial blurring and increases the SNR,
while preserving the temporal fidelity; View-sharing techniques yield early enhancement
(thin arrow) and temporal blurring (thick arrow), as shown in Fig. 2(d).
Conclusion: It is feasible to improve the spatial and temporal resolution in 3D TR-MRA
using Cartesian interleaved variable density sampling and HYPR reconstruction.
References: [1] Busse et al., ISMRM 2009, p4534. [2] Mistretta et al. MRM 55:30 (2006).
[3] Wang et al. ISMRM 2009, p3884 [4] Busse et al., ISMRM 2009, p2834
in B being slightly different. After a short pause, a conventional high spatial resolution
MRA (0.1 mmol/kg Gadovist®) was acquired using a 3D spoiled gradient-echo sequence
48
MR-Angioclub East Lansing 2009
4.7 3D Noncontrast MRA Using FSD-Prepared Balanced SSFP
1,2
1
3
1,2
3
1
1,2
Z. Fan , J. Sheehan , X. Bi , T. J. Carroll , R. Jerecic , J. Carr , and D. Li
Radiology, 2Biomedical Engineering, Northwestern University, Chicago, IL, USA;
3
Siemens Medical Solutions USA, Inc., Chicago, IL, USA
1
Purpose: To develop a 3D noncontrast MRA (NC-MRA) method for peripheral arterial
system using flow-sensitive dephasing (FSD)-prepared balanced SSFP (bSSFP).
Methods: The FSD preparative module consists of a 90ox-180oy-90o-x pulse series and
symmetric gradients around the 1800 pulse, which imparts flow sensitization measured by
the first-order gradient moment (m1) [1]. An optimal m1 can selectively suppress the fastflow arterial blood signal during systole while having little effect on the slow-flow venous
blood and static tissues. Subtraction between a bright-artery scan using bSSFP and a
dark-artery scan using FSD-bSSFP will result in an artery-only dataset. The new
technique was tested in multiple peripheral arterial territories of healthy volunteers and
patients at 1.5-T. Contrast-enhanced (CE) MRA was performed in patient studies for
reference. Parameters: 3D coronal acquisition, FSD gradients applied in the readout
direction, isotropic high spatial-resolution (0.8 -1.2 mm), TE/TR =1.5-1.9/ 3.1-3.8 ms, flip
angle = 70-90o, centric phase encoding, GRAPPA factor =2, m1 range 17-87 (leg), 58-156
(hand), 195-390 (foot) mT.ms2/m.
Results: Proof of principle was obtained from the lower extremities and hands (Fig. 1-3).
The value of m1 was shown to be critical for suppressing blood signal and achieving
excellent MRA quality. Comparable depiction of stenosis/occlusion and superior
visualization of patent segments were achieved using NC-MRA vs. CE-MRA.
Conclusion: The feasibility of this NC-MRA approach has been successively
demonstrated. Systematic optimization of m1 is warranted for clinical applications.
References: [1] Koktzoglou I, et al. JCMR 9:33 (2007).
a
a
b
Fig.1 Both CE-MRA (a) and NC-MRA (b)
demonstrate significant calf artery occlusion in a
patient with PAD. The large collateral vessels are
better depicted by NC-MRA. However, image
artifacts due to metallic clip (arrow) and field
inhomogeneity (arrowhead) are also appreciable.
b
Fig.2 NC-MRA (b)
demonstrated more
arterial vessels (in
red) and detail than
CE-MRA (a) in a
patient with
Raynauds disease.
Fig.3 NC-MRA of the
feet in a healthy
volunteer.
MR-Angioclub East Lansing 2009
49
4.8 Non Contrast MRA of the Hand in Patients with Raynauds
disease using Flow Sensitized Dephasing Prepared SSFP
1,2
1
2
2
2
J. C. Carr , J. J. Sheehan , Z. Fan , A. Davarpanah , and D. Li
Cardiovascular Imaging, Northwestern Memorial Hospital, Chicago, IL, United States, 2Cardiovascular
Imaging, Northwestern University, Chicago, IL, United States
1
Introduction: Raynauds disease is vasospastic disorder of the digital arteries. 3D
contrast-enhanced (CE) MRA is increasingly utilized for patients with Raynauds. Safety
concerns with contrast administration in patients with renal insufficiency have led to a
renaissance of non-contrast MRA (NC-MRA). NC-MRA strategies employing 3D halfFourier FSE [1] or SSFP [2] have shown great promise but various challenges remain.
The aim of this study was to retrospectively assess the diagnostic quality and accuracy of
a new NC-MRA method for hand MRA based on flow-sensitized dephasing (FSD)prepared SSFP.
Materials and Methods: The proposed NC-MRA method acquires a bright-artery scan
using ECG-triggered SSFP and a dark-artery scan using ECG-triggered, FSD-prepared
SSFP [3]. Subtraction of the two scans results in bright arteries and suppression of the
background and veins. 8 patients with clinically established Raynauds disease who have
been imaged at 1.5T (Avanto, Siemens) using a 16-element peripheral matrix coil and
spine coils, were retrospectively reviewed with IRB approval. Phase-contrast flow imaging
was first performed to derive the arterial flow peak time T. Each patient subsequently
underwent a time resolved TWIST MRA (TR MRA) and a high resolution MRA (HR MRA)
with Gadolinium-BOPTA. The FSD subtraction images along with the TR MRA and HR
MRA were reviewed and compared. Diagnostic quality was assessed by giving a per
vessel score for the 17 segments per hand (1, poor; 2, fair; 3, good; 4, excellent) and
adding them together for each hand (full score: 36). The degree of stenosis for each
vascular segment was characterized using a four-point scale (grade 0, normal; grade 1,
luminal narrowing <50%; grade 2, luminal narrowing >50%; grade 3 occlusion).
Results: The mean qualitative score for TR MRA, HR MRA and FSD were similar: 3.8, 3.4
and 3.5, respectively (t-test, P>0.05). Differences were more apparent when comparing
the distal digital arteries. Of 90 possible arterial segments, 6 (7%) were not adequately
depicted with all techniques because of severe venous overlay. FSD identified 95% of
luminal narrowings ≤50% and ≥50% that were identified on contrast enhanced MRA. Noncontrast FSD identified all of the arterial occlusions identified on contrast enhanced MRA.
The mean quantitative scores were similar for the degree of stenosis for TR MRA, HR
MRA and FSD were: 1.9, 1.9 and 2.0, respectively (t-test, P>0.05).
Conclusion: Hi-resolution non contrast FSD MRA of the hand in patients with Raynauds
compares favorably with contrast enhanced time resolved TWIST and high resolution
static MRA and in many cases demonstrates improved resolution and visualization of
normal and vasospastic vessels.
References: 1. Miyazaki M, et al. Radiology 227:890 (2003). 2. Stafford R, et al. MRM
59:430 (2008). 3. Koktzoglou I, et al. JCMR 9:33 (2007).
50
MR-Angioclub East Lansing 2009
4.9 Two-Station Time-Resolved CE-MRA of the Lower Legs
CP Johnson, CR Haider, EA Borisch, RC Grimm, PJ Rossman, SJ Riederer
Department of Radiology, Mayo Clinic, Rochester MN 55902 USA
Purpose: Contrast-enhanced MRA of the calves with high spatiotemporal resolution has
recently been demonstrated [1,2]. The goal of this work was to demonstrate the technical
feasibility of acquiring time-resolved extended field-of-view arteriograms of the thighs and
calves, nearly doubling the longitudinal coverage of the calf studies while maintaining
similar quality.
Methods: Acquisition was based on the CAPR method [3] on a GE 3.0T MRI system.
Two stations covering the thighs and calves of a volunteer were imaged with identical
imaging parameters using a stepping table approach. 8x 2D SENSE was achieved with
two high-performance eight-channel receive arrays
[4], one placed at each station. A 2 mL timing bolus
was used to determine the bolus transit time to the
calves.
The diagnostic scan was then acquired
using 1.0 mm isotropic resolution with a 5.0 second
image update time. 18 mL of Multihance followed by
a 20 mL flush were injected intravenously at 3
mL/sec. Prior to the time-resolved sequence, a twostation calibration scan was acquired.
Results: A maximum intensity projection of select
arterial frames overlapped using a weighted sum is
shown in Figure 1, demonstrating good image
quality.
Conclusion: Time-resolved CE-MRA of the lower
legs using CAPR may allow for extended field-ofview arteriograms with diagnostic quality comparable
Figure 1: MIP of select
arterial frames.
to that achieved in single-station studies.
References: [1] Haider CR, ISMRM 2009, #2734. [2] Wu Y, JMRI 2009, 29:917-23.
[3] Haider CR, MRM 2008, 60:749-60. [4] Johnson CP, ISMRM 2008, #1079.
MR-Angioclub East Lansing 2009
51
5.1 An Update on the Clinical Experience with Gadofosveset
1
2
1
Stephen Schmitz ; Edward Parsons ; Mark G. Hibberd
1
Lantheus Medical Imaging, N. Billerica, MA, USA;
2
Independent Consultant, Burlington, MA
Purpose: The clinical use of gadofosveset trisodium is reviewed in light of over three
years’ use in Europe and Canada, the recent US-FDA approval of gadofosveset trisodium,
and the establishment of a new clinical registry in the United States.
Methods: The post-marketing registry that samples the clinical experience of roughly
70,000 exposures of gadofosveset trisodium in Europe was reviewed. The adverse event
profile was summarized and compared with the data found in clinical trials and scientific
literature describing gadofosveset and gadolinium contrast agents in general.
The
physiochemical and pharmacokinetic properties of gadofosveset were reviewed in
consideration of leading hypotheses on the mechanism of NSF, and a new clinical registry
in the US was proposed.
Results: To date, only one blood-pool contrast agent (gadofosveset) has been registered
for use in MRA. The utility of gadofosveset in vascular MRI has been detailed both by
well-controlled clinical trials and by investigator-initiated studies. The adverse event profile
reported in post-market assessments is consistent with that reported in clinical trials. The
properties and pharmacokinetics suggest a relatively low risk of NSF, and no cases of
NSF have been associated with gadofosveset to date. A new prospective registry will
assess ongoing adverse event risks.
Conclusion: Over three years of experience has expanded knowledge of the safety and
efficacy of gadofosveset since its introduction in Europe and Canada. This information
should benefit the decision-making of clinicians considering its use.
The impeding
introduction of gadofosveset to the US market will bring many of these advances to a large
new group of physicians and patients.
52
MR-Angioclub East Lansing 2009
5.2 A Re-analysis of MS-325 (gadofosveset trisodium) Clinical Trial
Data in Support of US-FDA Approval
1
Edward Parsons , Neil Rofsky2, Gary Stevens3, Margaret Uprichard1, Kirsten Overoye-Chan1, Andrew
Uprichard1.
1
EPIX Pharmaceuticals, Inc.;
2
3
Beth Israel Deaconess Medical Center, Harvard Medical School; Dynastat, Inc.
Purpose: To verify the clinical efficacy of MS-325 (gadofosveset trisodium) in terms of
sensitivity and specificity for detection of stenotic lesions in MRA, as demonstrated in
phase 3 clinical trials [1,2], fulfilling current US-FDA regulatory requirements for efficacy.
Methods: Image data collected in support of registration of gadofosveset was re-read
according to a rigorous interpretation protocol. The protocol was developed in consultation
with the FDA and included additional steps for specific training of blinded MRA readers,
use of a consistent paradigm for image assessment, strict categorization of any image
artifacts that precluded diagnostic assessment, and a conservative statistical analysis with
precise conditions on unanimous gains in sensitivity and non-inferiority of specificity.
Three readers assessed non-contrast and gadofosveset-enhanced MRA data from two
trials of patients with suspected aorto-iliac arterial occlusions. Assessments were
compared to DSA results as a gold standard.
Results: The sensitivity endpoints were similar to those of the prior reads, with smaller
gains in specificity. All three readers showed statistically significant (p<0.001) gains in
sensitivity (+20.4%, +12.2%, +14.9%%) with at least non-inferior specificity (+0.4%,
+7.9%, +0.2%).
Significantly fewer contrast-enhanced MRAs were considered
uninterpretable when compared with non-contrast images (average of 8.6% vs 1.4%).
Conclusion: The efficacy of gadofosveset was affirmed, resulting in US-FDA marketing
approval [3]. More standardized reading guidelines produced more consistent results.
References: [1] Rapp et al. Radiology. 2005 Jul; 236(1):71-8.
Radiology.
2005 Sep;
236(3):825-33.
[3]
Drug Approval
[2]. Goyen et al.
Package: Vasovist.
http://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/021711s000TOC.cfm
MR-Angioclub East Lansing 2009
53
5.3 Gadofosveset Excretion into Human Breast Milk
Malene S. Thomsen1, Walter Gössler2, Uwe Lang3 Manuel Aigner1,
and Manuela Aschauer1
1
Division of Vascular and Interventional Radiology, Department of Radiology, Medical University of
Graz, Auenbruggerplatz 9, 8036 Austria, 2 Institute of Chemistry, Analytical Chemistry, Karl-Franzens
3
University, Universitaets-platz 1, 8010 Graz, Austria, Department of Obstetrics & Gynaecology,
Medical University of Graz
Purpose - Gadolinium (Gd) based contrast agents are generally accepted as
pharmaceutical agents with few adverse effects. However, for the contrast media
gadopentetate (Magnevist®) it is common practice that nursing mothers interrupt
breastfeeding, and discard the milk the first 24 - 48 hours after injection of gadopentetate
[1]
. The aim of this current investigation was to evaluate if this guideline can be applied as
well when using the contrast agent gadofosveset (Vasovist®).
Methods - The Gd-content of breast milk was monitored up to 51 hours after injecting
gadofoveset to a lactating patient (indication: objective evaluation of pulmonary artery
embolism and deep venous thrombosis to plan the cava filter implantation) by collecting
the breast milk with intervals of 2-4 hours and analyzing samples for the Gd-content
applying inductively coupled plasma mass spectrometry (ICP-MS)
Results - A significant result found was that gadofosveset was excreted from the body
with a half-life time of around 26 hours, which was twice that of gadopentetate
[2,3]
. That
gadofosveset stays longer in the blood circulation can most likely be explained by the fact
that gadofosveset reversibly bind to proteins[4] whereas gadopentetate is highly water
soluble and has low binding to proteins.
Conclusion - From the study it was discovered that gadofosveset stays in the body twice
as long as gadopentetate. Therefore the guideline might not be directly applicable. We
recommended no brest feeding in this case for 48 hr.
References -
1) Lin & Brown, J. Magn. Reson. Imaging, 2007 25, 884-899
2) Schmiedl et al, Am. J. Roentgenol., 1990, 154, 1305-6
3) Rofsky et al, J. Magn. Reson. Imaging, 1993, 3, 131-132
4) Goyen, Vasc. Health Risk Manag., 2008, 4, 1-9
54
MR-Angioclub East Lansing 2009
5.4 Overview of Gd-BOPTA Phase III Trial for CERMA: What are the
results, and how do we move forward?
Thomas M. Grist, M.D. University of Wisconsin – Madison
PURPOSE
Contrast-enhanced magnetic resonance angiography (CE-MRA) is widely used for
imaging atherosclerotic disease in the carotid, renal, and peripheral arteries yet few multicenter trials have been reported that document the safety and efficacy of modern CE-MRA
techniques. Studies were recently initiated to assess the safety and efficacy of a single
0.1 mmol/kg bw dose of gadobenate dimeglumine (Gd-BOPTA; MultiHance) for MRA in
these three vascular territories. The objective of this presentation is to review the results
of a multi-center trial and highlight the potential impact of the salient findings on MRA
practice in the future.
MATERIALS AND METHODS
A total of 825 patients from Europe (63%), North America (27%), and South America
(10%) were evaluated. Imaging of the carotid (N=247), renal (N=291) and peripheral
(N=287) vessels was performed. 2D TOF and CE MRA was performed, and compared to
intra-arterial DSA as a reference standard. Assessment of MRA images was performed at
an independent core imaging laboratory by 3 experienced, fully-blinded radiologists per
study unaffiliated with any study center. DSA images were reviewed in a similar fashion
by a single reader. McNemar’s test was used to compare sensitivity, specificity, and
accuracy of TOF and CE-MRA for detection of clinically significant steno-occlusive
disease (>50% or 60% for carotid).
RESULTS AND DISCUSSION
The diagnostic accuracy of CE-MRA was significantly greater than TOF-MRA for most
vessel territories and readers (p<0.05). Likewise, inter-observer agreement for CE-MRA
was greater than 2D-TOF MRA. However, lower sensitivity values than anticipated were
observed for CE-MRA due to methodological issues encountered in the study. The results
and potential impact of these findings on acceptance of CE-MRA techniques will be
discussed.
MR-Angioclub East Lansing 2009
55
5.5 Safety of Gadobenate dimeglumine (Gd-BOPTA) in
Cardiovascular Imaging of Pediatric Patients
Guenther Schneider, Hellmut Schürholz, Arno Bücker, Peter Fries
Homburg University Hospital, Homburg/Saar, Germany
Purpose: The advent of nephrogenic systemic fibrosis (NSF) has brought the safety of
Gd-based contrast agents into sharp focus. Nowhere is safety of greater concern than
among pediatric patients who frequently require multiple contrast-enhanced (CE) MR
examinations over an extended period of time. Gadobenate dimeglumine (MultiHance) is a
contrast agent that has proven extremely safe among adult subjects for a variety of
indications. Due to its increased relaxivity and the stability of the complex it seems to be
well suited for cardiovascular imaging in pediatric patients. The present retrospective
analysis was performed to determine the safety of gadobenate dimeglumine in pediatric
subjects referred for routine cardiovascular MRI.
Material and Methods: Gadobenate dimeglumine is routinely used off-label with IRB
approval at our center for this specific patient population because of its beneficial
properties. A total number of 88 pediatric patients (134 studies; 0 years - 15 years)
underwent CE-MRA studies with a gadobenate dimeglumine dose of 0.1 mmol / kg
bodyweight. Since only in-patients were studied, monitoring for adverse events was
performed up to at least 24 hours post injection. Laboratory measurements, vital signs and
ECG determinations were made before and after CE-MRA.
Results: No severe or serious adverse events were noted in our series. No significant
changes of creatinine, bilirubin, vital signs or ECG parameters were noted. No patients
exhibited symptoms of NSF even after repeat doses of gadobenate dimeglumine. Image
quality was excellent especially in patients that underwent CE-MRA for evaluation of CHD.
Conclusion: Gadobenate dimeglumine at a dose of 0.1 mmol/kg bodyweight is a safe and
effective contrast agent for cardiovascular imaging of pediatric patients. Concerning the
safety of Gd-based MR contrast agents, the lower required dose of gadobenate
dimeglumine seems to be beneficial for vascular imaging.
56
MR-Angioclub East Lansing 2009
5.6 Retrospective 7 year Study of the Incidence of Nephrogenic
Systemic Fibrosis in Patients Investigated with Gadolinium
Contrast-Enhanced Renal Magnetic Resonance Angiography
Collidge T A, Brown, M, Rao, A, Thomson P C, & Roditi G Glasgow Royal Infirmary, Alexandra
Parade, Glasgow G31 2ER
Purpose: There is now well documented association between the development of the
condition Nephrogenic Systemic Fibrosis (NSF) and high dose gadolinium contrast MRI
examinations in patients with severe renal failure. We set out to assess the incidence of
NSF in patients with differing degrees of renal impairment investigated by contrastenhanced renal MRA.
Methods & Materials:
Patients from 1998 to 2005 assessed through retrospective
analysis of electronic patient record (EPR). Patients excluded if follow-up < 90 days.
Analysis included renal function (CKD stage) at time of scan, time to first follow-up, total
follow-up time, number of follow-up episodes, EDTA diagnoses, diagnoses of NSF, reports
of pruritus or other skin disorder, episodes of gadolinium-enhanced MRI and cumulative
gadolinium doses.
Results: Of 1200 patients who underwent gadolinium-enhanced renal MRA studies 500
had follow-up documented > 90 days with average follow-up of 1326 days.
CKD
eGFR
MRA
5
<15
67
NSF
1
4
15-29.9
169
2
3
30-60
222
0
1&2
>60
31
0
Normal
>90
11
0
(potential live renal donors)
The 3 patients who developed NSF during the course of follow-up had estimated GFRs of
<1, 16 & 18. One patient with initial CKD stage 4 at time of renal MRA developed NSF at
a significantly later stage following a peripheral run-off MRA when clinically unwell in CKD
stage 5. 5 other patients were noted at clinic follow-up to be complaining of pruritus but
none of these was ever diagnosed as suffering from NSF.
Conclusion:
This study confirms that NSF is not seen in patients undergoing contrast-
enhanced MRA with less severe degrees of renal failure than CKD stages 4 & 5
MR-Angioclub East Lansing 2009
57
5.7 Risk Factors for NSF: a Meta-analysis
1,
Martin R. Prince 2, MD, PhD, Hong Lei Zhang1, MD, Giles H. Roditi3, MD
4
5
Tim Leiner , MD, Walter Kucharczyk , MD
From Departments of Radiology at Weill Medical College of Cornell University1, Columbia College of
Physicians and Surgeons2, New York, Glasgow Royal Infirmary, Scotland3, Maastricht University
4
5
Hospital, Maastricht, the Netherlands , and University of Toronto, Toronto, Canada
Purpose: To investigate who can safely undergo Gd MRA with minimal risk of NSF.
Method: Pubmed was searched for ‘Nephrogenic Systemic Fibrosis’ to identify papers
with detailed clinical information from January, 2003 to June, 2009. Patient age, gender,
race, type of Gd enhanced imaging, Gd type and dose, interval between Gd and NSF
onset, GFR, dialysis, interval between Gd and dialysis, acuteness of renal failure, kidney
transplant, serum phosphorus, acidosis, epoetin, pro-inflammatory events and NSF
symptoms, were recorded and analyzed. Corresponding authors were contacted for
corroboration and to provide as much missing data as possible.
Results: 77 papers with detailed information on 290 patients were included in the final
analysis. The gender distribution was approximately equally weighted. Very young (<10
yrs) and old (> 60 yrs) patients are at less risk. In 244 patients for whom MR exam history
was investigated, 217 (89%) were noted to have GBCA injections prior to NSF symptom
onset including use of gadodiamide (n = 167), Gadopentetate dimeglumine (n = 11),
gadoversetamide (n = 5), multiple agents (n = 8) and unspecified/unknown (n = 26). All
NSF patients had renal dysfunction with highest prevalence for dialysis and for GFR < 15
mL/min. Acute renal failure was an NSF risk factor with odds ratio of 13:1. In 182 cases
for which data on GBCA dose were available or could be estimated, 90% (n = 163)
received high dose. Other risk factors include pro-inflammatory events, epoetin, acidosis
and hyperphosphatemia. Risk of NSF is small compared to the risk of iodinated contrast
induced nephropathy and the risk death from NSF was small compared to the risk of death
from allergic reactions.
Conclusion: Order of magnitude reductions in NSF risk can be attained by 1) avoiding
high dose, 2) avoiding nonionic linear chelates, 3) dialyzing within 24 hours of Gd
administration for patients already on dialysis, 4) avoiding injecting acute renal failure
patients while serum creatinine is rising.
58
MR-Angioclub East Lansing 2009
5.8 Extracellular matrix metabolism in organ-cultured skin from
patients with end-stage renal disease: Response to gadolinium
based MRI contrast agents
James Varani, Marissa DaSilva, Monica O’Brien Deming, Kent Johnson and 1Richard Swartz;
1
Departments of Pathology and Medicine, University of Michigan,
Ann Arbor, Michigan, 48109
Purpose:
Nephrogenic systemic fibrosis (NSF) is a clinical syndrome linked with
exposure in renal failure patients to gadolinium - based MRI contrast agents (GBCAs).
The present study addresses potential patho-physiological mechanisms.
Methods: Here we have examined human skin from normal subjects and individuals with
end stage renal disease for response to GBCA stimulation in organ culture.
Results:
Omniscan, one of the clinically used GBCAs, had no effect on type I
procollagen production, but increased levels of both matrix metalloproteinase-1 (MMP-1)
and tissue inhibitor of metalloproteinases-1 (TIMP-1).
The level of TIMP-1 was
significantly higher than the level of MMP-1 and there was no detectable collagenolytic
activity. Qualitatively, there were no differences in the responses of skin from renal failure
patients as compared to controls. However, basal responses were higher in skin from
subjects in renal failure as compared to control.
Conclusion:
These data suggest that GBCA exposure does not directly stimulate
collagen production but, rather, modulates an enzyme-inhibitor system responsible for
regulation of collagen turn-over in the skin. We speculate that heightened responses in
subjects with end-stage renal disease may reflect elevated basal activity.
MR-Angioclub East Lansing 2009
59
5.9 Manganese Based Biodegradable Macromolecular MRI
Contrast Agents for Cardiovascular Imaging
1
1
1
1,2
Zhen Ye , Eun-Kee Jeong , Dennis L. Parker , Zheng-Rong Lu
Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah;
2
Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
1
Purpose:
This
study
was
intended
to
develop
Mn(II)
based
biodegradable
macromolecular contrast agents for cardiovascular MRI.
Methods: The polymeric contrast agents (CAs), Mn(II)-DTPA cystamine copolymers
(MDCC) and Mn(II)-EDTA cystamine copolymers (MECC), were synthesized and
characterized(1). Their in vivo contrast enhanced MR cardiovascular imaging was
evaluated in mice on a Siemens 3T MRI scanner with MnCl2 as a control.
Results: The number average molecular weight of MDCC and MECC was 30.5 and 60.8
KDa, respectively. The T1 relaxivity of MDCC and MECC was 4.7 and 6.41 mM-1s-1 at 3T,
respectively, in the same range of that of MnCl2 (5.2 mM-1s-1)[2]. Contrast enhancement
was observed in the vasculature with MDCC and MECC in the initial period post injection.
Significant enhancement in the myocardium was also observed for MECC and MnCl2, not
for MDCC. The difference in myocardium enhancement between MDCC and MECC might
be attributed to the different coordination chemistry and the stability of two contrast
agents[3].
-
OOC
N
O
O
N
N
Mn2+
COO -
COO N
H
N
S
S
n
MDCC
-OOC
-OOC
Mn2+
O
N
H
N
H
O
N
N
H
H
N
S
S
n
pre
MECC
MnCl
MDCC
MECC
2
(30 minutes after i.v injection)
Conclusion: The Mn(II) based biodegradable macromolecular contrast agents MDCC and
MECC are promising for contrast enhanced cardiovascular imaging with MRI.
Reference: [1] Intl. J. Nanomed. 2007, 2, 191-9. [2] Chem. Rev. 1987.87.901-927; [3]
Cur.Pharm.Biotech, 2004, 5, 539-549
60
MR-Angioclub East Lansing 2009
6.1 Non-invasive Trans-Stenotic Pressure Measurements with 3D
Phase Contrast MRA: Validation against Endovascular Pressure
Measurements in Swine
1, 3
T Bley, 2K Johnson, 1C François, 1,2S Reeder, 1M Schiebler, 2O Wieben, 1T Grist
1
2
Departments of Radiology and Medical Physics, UW Madison, WI, USA
Department of Radiology, University Medical Center Hamburg-Eppendorf, Germany
3
Purpose
To evaluate trans-stenotic pressure gradients (TSPG) in renal artery stenosis (RAS)
noninvasively utilizing a 3D phase contrast acquisition with vastly under sampled isotropic
projection reconstruction (PC-VIPR) MRA in a porcine study.
Methods
Respiratory gated PC-VIPR MRA (dual echo, 18,000 projection angles, 10˚ flip, TR/TE
(first echo) = 11.4/3.7 msec, BW = ±62.5kHz, imaging volume: 260x260x160mm3,
acquired isotropic spatial resolution: 1.0x1.0x1.0mm3, venc = 150cm/s) of renal arteries
with surgically created RAS in 12 swine was performed on a 1.5T clinical system (Signa
HDx, GE Healthcare, Waukesha, WI). NCE MRA and TSPG were calculated directly from
the
magnitude
and
velocity
measurements
using
the
Navier-Stokes
equation.
Endovascular pressure measurements were used as the gold standard for pressure
gradient quantification, performed under DSA guidance.
Results
In 19 renal arteries, the TSPG analysis demonstrated excellent correlation between the
non-invasive TSPG utilizing PC-VIPR and endovascular pressure measurements (average
stenosis = 62%, r = 0.977; 95% CI: 0.931, 0.998; p < 0.001). In 5 arteries with severe RAS
(mean 86%), the residual lumen within the stenosis was so small that TSPG could not be
determined using PC-VIPR and they were excluded from the statistical analysis. However,
the angiographic reformats from those data readily revealed severe, hemodynamically
significant RAS.
Conclusion
Noninvasive assessment of hemodynamic significance of RAS in swine was feasible
utilizing non contrast material enhanced PC-VIPR MRA. Excellent correlation between
TSPG measurements by PC-VIPR and endovascular guide wires was found. PC-VIPR
has the potential to become a major advance in the noninvasive evaluation of RAS and,
as a result, in the management of patients with renal hypertension.
MR-Angioclub East Lansing 2009
61
6.2 Analysis of aortic hemodynamics after treatment for
coarctation using flow-sensitive 4D-MRT at 3T
A. Frydrychowicz1, D. Hirtler2, R. Arnold2, A. Berger1, A.F. Stalder1, J. Bock1, M. Langer1, J. Hennig1,
M. Markl1
1
Department of Diagnostic Radiology, Medical Physics, University Hospital Freiburg, Germany,
2
3
Department of Pediatric Cardiology, University Hospital Freiburg, Germany Department of
Cardiology, University Hospital Freiburg, Germany\
Purpose: To evaluate the hemo-dynamic alterations in aortic blood flow after coarctation
repair by flow-sensitive,
Fig. 1: Vortical flow patterns (white
time-resolved 3D MRI at
arrows) observed in 15/24 patients
3T and to compare
after treatmen of aortic coarctation.
findings to acquisitions in
This finding could be observed
volunteers.
irrespective of the treatment
Methods: Flow-sensitive
methodology. This patient also
4D MR was performed in
shows flow acceleration over a
28
patients
after
mild re-stenosis (openwhite
coarctation
repair
arrow).
(16.5±8.0years, range 436) and 19 volunteers
(35.3±18.1 years, range
20-75) from a previous
analysis were included
as a reference [3].
Results: The operative
site showed a relative restenosis with a diameter
of 13.3 ± 4.0mm (range
6-24) and a post-stenotic
dilatation of 18.8 ±
6.5mm (range 7-43).
Next to accelerated flow
in all patients, there were
additional helices, an
Fig. 2: Different stages of aneurysm development were
increased rate of vortices
detectable. (A) represents flow patterns in a 10yo boy
and most noticeably, in
before treatment of an otherwise not symptomatic
the majority
of all
coarctation. Clearly, the elongated aortic arch, the
patients
unexpected
vortical flow acceleration (white arrowheads) before
vortices
in
the
the stenosis (white arrow, moderate flow acceleration
supraaortics that might
during late systole) can be apreciated. In (B), a
attribute
to
intimal
hypoplastic arch (white arrow) gives rise to a flow
thickening were found.
acceleration taht enters a highly vortical flow through a
Quantitative
analysis
small aneurysm that distributes greatly helical flow in
showed
significantly
the Dao (yellow arrows).
elevated
wall
shear
stress levels and decreased oscillatory shear indices in the patients that hint towards an
influence concerning arterial remodeling.
Conclusion: Flow-sensitive MRI revealed marked changes in the hemodynamics after
CoA repair. Follow-up examinations have to clarify, whether a predictive value can be
attributed to hemodynamics or derived vessel wall parameters.
References: 1. Oliver JM J Am Coll Cardiol 2004, 44:1641-1647. 2. Wigström L, et al.
MRM 1999;4:793-799, 3. Frydrychowicz A, ISMRM 2008
62
MR-Angioclub East Lansing 2009
6.3 Flow assessment of arterial dissections using 3D radial phase
contrast MR angiography
Christopher J François, Kevin M Johnson, Benjamin Landgraf, Mark L Schiebler, Scott B Reeder,
Thomas M Grist, Oliver Wieben
University of Wisconsin, Madison, WI, USA
Purpose: The ability to predict the progression and complications of dissections (eg.
aneursymal dilatation and rupture) is a limitation of current diagnostic methods.
Differences in the flow patterns in the true and false lumina may be important in the
outcomes of dissections1-3. Our aim was to develop a 3D phase contrast (PC) radial pulse
sequence that can be peformed in less than 10 minutes to assess the hemodynamics of
dissections, as a potential prognosticator of dissection
A
evolution.
Methods: 3D PC VIPR was performed in patients with
arterial dissections. Parameters for PC VIPR were: ±62.5
kHz receiver bandwidth, 1.00-1.25mm3 isotropic spatial
resolution, 8-10 min of free breathing with 50% respiratory
gating efficiency, imaging volume: 32x32x16 cm3, VENC of
80-100 cm/s, retrospective cardiac gating with a temporal
filter for radial acquisitions. PC VIPR data were acquired after obtaining patient consent
according to our IRB protocol. Datasets were analyzed using EnSight (CEI, Inc., Cary,
NC).
Results: Flow patterns in true (arrows) and false (open arrows) lumina were laminar and
non-laminar, respectively. In a patient with celiac and superior mesenteric artery
dissections (A), the flow in the false lumina was vortical. In a
B
patient with chronic descending thoracic aortic dissection
(B), flow in the false lumen was very slow and turbulent.
Conclusion: Using PC VIPR it is possible to detect
differences in flow patterns between true and false lumina of
arterial
dissections,
which
should
permit
a
more
comprehensive quantitative hemodynamic assessment and
could have prognostic implications in addition to improving our understanding of the
underlying pathophysiology.
References: 1. Markl M, et al. JCAT 2004;28:459. 2. Strotzer M, et al. Acta Radiol
2000;41:594. 3. Mohri M, et al. Clin Cardiol 1985;8:225.
MR-Angioclub East Lansing 2009
63
6.4 High Temporal and High Spatial Resolution
Perfusion Imaging of Hepatocellular Carcinoma in the Liver
Scott B. Reeder, MD, PhD; Ethan K. Brodsky, PhD; Eric Bultman, BS;
William Schelman, MD, PhD; Yin Huang, PhD; Sean F Fain, PhD; Walter F. Block, PhD
Departments of Radiology, Medical Physics, Biomedical Engineering and Medicine
University of Wisconsin, Madison, WI, USA
Introduction: The field of cancer therapeutics has been shifting from traditional “cytotoxic”
agents to targeted anti-angiogenic therapies. This trend requires biomarkers of early tumor
response. Here we aim to develop high spatial and temporal resolution imaging methods
for quantitative perfusion imaging as a biomarker of tumor response.
Methods: Patients with known hepatocellular carcinomas (HCC) were imaged on a 3.0T
scanner (MR750, GE Healthcare) with a 32 channel abdominal coil, after single dose
injection of gadobenate dimeglumine (Bracco, Princeton, NJ). A time-resolved 3D radial
sequence collected whole abdomen data using one interleaved sub-frame/sec,
reconstructed in real-time1, allowing fluoroscopic monitoring of the contrast bolus. Real
time bolus tracking permits timing of breath-holding during 1) contrast arrival during the
arterial phase, 2) portal venous phase (50-70s), and 3) delayed phase from 1:40-2:00
minutes. Using temporal filtering through density compensation filters2, time-resolved
image volumes at full spatial resolution (1.6x1.6x1.6mm3) were reconstructed with an
effective temporal resolution of 8s. Using the high temporal resolution images, quantitative
perfusion modeling can be performed using a dual input model of the hepatic blood flow
that includes the portal vein and hepatic arterial input3.
Results and Discussion:
Figure 1 shows images of a 1.5cm HCC, identified on
surveillance MRI. Arterial phase shows brisk enhancement and rapid washout during the
portal venous phase, highly characteristic of HCC. Spatial resolution is 1.6mm isotropic
with 8s temporal resolution. High spatial and temporal resolution will allow clinicians to
accurately capture contrast uptake in the tumor contrast kinetics, the portal vein and
hepatic artery, necessary for dual input modeling of perfusion to the HCC3.
Figure 1: Pre-contrast, arterial
phase, and portal venous phase
images through a 1.5cm HCC
(arrow). Brisk arterial
enhancement and rapid
washout is highly characteristic
of HCC. Spatial resolution is
1.6mm isotropic with 8s
temporal resolution.
References: 1. Brodsky et al, MRM, 2006 2. Barger et al, MRM, 2002 3. Materne et al, MRM, 2002
64
MR-Angioclub East Lansing 2009
6.5 Stack of Stars 4D Phase Contrast Velocimetry of the Circle of
Willis
Steven Kecskemeti, Kevin Johnson, Yijing Wu,Warren Chang, Charles Mistretta, Patrick Turski
Departments of Medical Physics and Radiology, University of Wisconsin, Madison, WI
INTRODUCTION: In addition to angiograms, cardiac gated phase contrast (PC) MR
velocimetry provides hemodynamic information such as pulsatility, flow streamlines,
relative pressure, and estimates of wall shear stress. In certain cases, such as
aneurysms in the circle of Willis (COW), high resolution over a moderate excitation slab
(40mm) can be achieved using a stack of stars hybrid PR acquisition.
METHODS AND RESULTS: A cardiac gated (both retrospective and prospectively)
gated PC stack of stars (SOS) sequence was developed with radial readout in the xyplane and traditional phase encoding in the z-direction. A bitreversed projection ordering
allows for retrospective viewsharing in multiples of the minimum temporal width of 4TR
for four point velocity encoding. Exams have been performed on a 3T clinical scanner
with
8-channel
head
coil.
Parameters
were
FOV:
220x220x50mm,
resolution:0.43x0.43x1.0mm, reconstructed voxel size: 0.43x0.43x0.43mm, TR/TE =
9.3/4.1ms, tip angle = 20, BW = 62.5kHz, scantime 7:03+30s calibration scan. Full MIPs
have been shown for the axial (a) and coronal(d) views, while limited MIPS of sagittal
are displayed to distinguish left (c) and right (d) hemispheres.
We conclude that 4D cardiac gated PC SOS a practical method to study flow normal and
pathological flow conditions within the Circle of Willis.
Mean Flow
2.8
flow [ml/s]
2.6
2.4
2.2
High
Resolution
Perfusion Weighted Imaging
2
0
200
400
600
800 1000
time [ms]
MR-Angioclub East Lansing 2009
65
6.6 High Resolution Perfusion Weighted Imaging
Meng Li, MS and E. Mark Haacke, PhD
Wayne State University, Detroit, MI, USA
Purpose: Perfusion-weighted imaging (PWI) using dynamic susceptibility contrast (DSC)
is a useful tool to evaluate various diseases of the brain. But current low resolution PWI is
unable to easily show differentiation between gray matter and white matter. We plan to
use susceptibility weighted imaging (SWI) and MR angiography (MRA) to remove major
vessel information from a high resolution PWI approach to better reveal gray matter
perfusion without the confounding major vessel effects.
Methods: HR PWI images of 1 x 1 x 4 mm3 were acquired on a 1.5T Siemens Sonata with
an
8-channel
head
coil.
The
imaging
parameters
TR/TE/FA/Resolution/BW=2200ms/98ms/60°/1x1x4mm3/752Hz/pixel,
used
the
were:
acquisition
matrix was 256x256 (interpolated to 512x512). Both MRA and SWI data were acquired.
MRA was obtained pre-contrast with TE/TR/FA = 7ms/37ms/25°, SWI was acquired pre
and post-contrast with TE/TR/FA = 40ms/49ms/20°.
Results: High resolution PWI was successfully obtained with parallel imaging. Compared
to the usual 2mm in-plane resolution, the following advantages were observed on CBF
and CBV maps: the blurring of blood vessels was minimal, fine details of blood vessels
were observed, gray matter was clearly separated from blood vessels, and small vessels
like the medullary veins could be seen. Using the HR PWI data it was possible to remove
the blurring associated with the major blood vessels. This is accomplished by using both
the MRA and SWI data. The major vessels were then removed from the HR PWI CBV
map in an attempt to create a segmented vessel free image. This made it possible to
focus on just gray matter or white matter and made it easier to filter the gray matter data
without interference from major vessels.
Conclusion: The separation of gray matter and white matter in PWI can be achieved by
increasing the in-plane resolution of conventional PWI data to 1mm. Combination of high
resolution maps of PWI, SWI and MRA can provide additional information about macro
and micro circulations and vasculature of the brain.
66
MR-Angioclub East Lansing 2009
6.7 Accelerated velocity imaging using compressed sensing
1
1
2
1
1
L. Marinelli , K. Khare , K. F. King , R. Darrow , T. K. F. Foo1, and C. J. Hardy
1
2
GE Global Research, Niskayuna, NY, GE Healthcare, Waukesha, WI
Purpose: Accurate measurement of blood velocity in complex flows can improve the
diagnosis and characterization of a variety of cardiovascular diseases. We have
developed a 2D Fourier velocity encoding (FVE) M-mode MRI pulse sequence to probe
multi-dimensional velocity distributions. Unlike in conventional MRI, parallel imaging
cannot be used to shorten FVE scan time. We have instead developed a compressed
sensing (CS) approach to accelerate 2D FVE, which exploits the sparsity of the blood
velocity distribution.
Methods: Following localization of the mitral valve, 2D FVE M-mode MRI was performed
with a VENC of 64 cm/s. A 2-cm pencil of spins was excited and 16 velocity-phaseencoding steps were applied in each velocity-encoding direction. The 16x16 velocity
encodings were undersampled by various factors and we compared uniform random,
Gaussian random, and Poisson disk distributions.
Results and Conclusions: Figure 1 shows a typical temporal evolution of the blood
velocity distribution near the mitral valve. Frame (a) was acquired 20ms after the QRS
complex and in frame (c) we note that there was some flow towards the aortic valve.
During diastole, the aortic valve closes and the mitral valve opens; flow near the mitral
valve becomes more directional towards the valve (frame (e)) and then relaxes back to
zero velocity sweeping an arc in velocity space (frame (f) and (a)). This is the only
technique that provides multi-dimensional velocity distribution data that may have impact
in the assessment of valvular disease and regional intravascular wall pressure.
vx 1/32
a vz
3/32
b
8/32
c
18/32
11/32
d
e
21/32
g
f
Figure 1. (a)-(f)Time evolution of the 2D blood velocity distribution
near the mitral valve (green arrow in (g)). vx (resp. vz) is the velocity
component parallel (perpendicular) to the pencil beam. (g) Left
h
ventricular outflow tract view with M-mode pencil prescribed through
the mitral valve. (h) 2D FVE pulse sequence diagram.
MR-Angioclub East Lansing 2009
67
6.8 Wall Shear Stress in Normal and Atherosclerotic
Carotid Arteries
M. Markl, S. Bauer, J. Bock, A. F. Stalder, A. Frydrychowicz, A. Harloff
1 Diagnostic Radiology, Medical Physics, University Hospital Freiburg, Germany
Introduction: Hemodynamic conditions
in the carotid bifurcation resulting in low
wall shear stress (WSS) and high
oscillatory shear index (OSI) can predict
plaque development1,2. The normal
distribution of segmental WSS and OSI
was evaluated in 32 normal volunteers
and compared to findings in 6 patients
with moderate internal carotid artery
(ICA) stenosis < 55%.
Methods: Flow-sensitive 4D-MRI3,4
(a=15°,
venc=150cm/s,TRes=45.6ms,
1.1x0.9x1.4mm³) was performed to
estimate
time-averaged
absolute
WSSmag and OSI in 7 analysis planes
(figure 1)5. To identify areas at risk for
plaque
development
segments
representing the individual upper 15%
of OSI and lower 15% WSSmag were
determined.
Results: 64 normal carotid bifurcations
(figure 2) revealed high inter-individual
consistency and a high incidence of low
WSSmag and high OSI in segments
corresponding to the proximal ICA bulb.
Patients showed a more heterogeneous
distribution and a spatial relocation of
critical wall parameters to proximal
regions distal to the ICA stenosis.
Discussion: Atherogenic low WSSmag
and high OSI in the normal ICA bulb
may explain why ICA plaques often
develop at this site. The presence of
ICA stenosis can alter wall parameter
distributions which may help to predict
the direction and extent of progression
of the disease.
Fig. 1: Assessment of wall parameters in 7 analysis
planes (left) and extracted systolic velocity profile
and wall shear stress vectors (WSS) for analysis
plane 3 in the ICA bulb (right).
Fig. 2: Distribution of critical wall parameters for
n=64 normal carotid arteries and n=6 patients with
moderate ICA stenoses. In normal controls critical
wall parameters were predominately located in the
posterior ICA bulb while patients showed critical
WSS in more distal segments.
References: 1. Cheng C et al. Circulation. 2006;113:2744-2753; 2. Lee SW et al. Stroke.
2008;39:2341-7; 3. Harloff A, et al. MRM 2009, 61:65-74; 4. Markl M, et al. JMRI 2007,
25:824-831; 5. Stalder AF et al MRM. 2008;60:1218-1231.
68
MR-Angioclub East Lansing 2009
6.9 MR imaging and significance of flow reversal and carotid
atherosclerosis: Initial results
3
J. Scott McNally , Seong-Eun Kim1,3, John Roberts1,3, Gerald S. Treiman2,4,
Dennis L. Parker1,3
1
Utah Center for Advanced Imaging Research, 2VA Salt Lake City Health Care System, 3Radiology,
4
Surgery, University of Utah, Salt Lake City, Utah, 84108
Introduction
Reports suggest that atherosclerotic plaque at the carotid bifurcation correlates with areas
of flow reversal and low wall shear stress.
MRI can be used to delineate plaque
components including ulceration, erosion, and hemorrhage, which are thought to correlate
with symptoms. Using MRI, our goal is to determine if areas of flow reversal correlate with
these markers of plaque vulnerability and plaque location at the carotid bifurcation.
Methods
VA subjects scheduled for carotid endarterectomy are being scanned with a 3T MRI using
the following sequences: 2D T1w (0.5 x 0.5 x 2mm3) and 2D TSE to measure plaque
location, ulceration and erosion, 3D MPRAGE (0.5 X 0.5 x 1mm3) to measure
hemorrhage, 3D TOF (0.3 x 0.6 x 0.6mm3) and 2D Phase Contrast (0.8 x 0.8 x 3.0mm3)
with dual VENC to measure flow reversal. After tracing the regions of interest, areas of
these components are calculated.
Results and Discussion
Results obtained to date on subjects with atherosclerosis (16 diseased and 7 normal
carotid arteries) have shown: 1) Flow reversal in normal carotid arteries occurs along the
proximal internal carotid artery (ICA) and downstream of plaques.
2) Plaque area is
largest along the outer wall of the ICA. 3) Ulceration, erosion and hemorrhage occur
preferentially in areas exposed to flow reversal.
Conclusion
Initial results suggest that flow reversal correlates with plaque location and markers of
vulnerability.
This study may help determine the significance of flow reversal in the
progression of atherosclerosis.
MR-Angioclub East Lansing 2009
69
7.1 4D-MRA in combination with arterial spin labelling for selective
and functional information in patients with AVMs
W. A. Willinek1,G.M. Kukuk1, D.R. Hadizadeh1, J. Gieseke1, 2, J. Bergener1, G. Beck2, L. Geerts2, P.
Mürtz1, A. Boström3, H. Urbach1, J Schramm3, H. H. Schild1
1
Dpt. of Radiology, University of Bonn, Germany, 2 Philips Healthcare, Best, Netherlands, 3 Dpt. of
Neurosurgery, University of Bonn, Germany
Purpose: Arterial spin labelling with selective labelling pulses is a promising method
providing selective and functional information regarding brain perfusion territories, regional
cerebral vascular supply and functional collateral circulation. The purpose of this study
was to prospectively evaluate 4D-MRA in combination with selective arterial spin labelling
for perioperative assessment of cerebral AVMs.
Methods: In a prospective intraindividual comparative study 10 patients (6 female, 4 male;
mean age 35.8 years ± 12.2; range 20-58 years) diagnosed with symptomatic cerebral
AVMs underwent pre- and postoperative 4D-MRA, regional brain perfusion imaging using
selective arterial spin labelling and DSA. Institutional ethics committee approval and
written informed consent were obtained. 4D-MRA was performed using CENTRA keyhole
in combination with view sharing yielding a temporal resolution of 572 msec, whole brain
coverage and an isotropic voxel size of 1.1 x 1.1 x 1.1 mm³. Selective arterial spin
labelling was performed using the PULSAR labelling sequence for selective labelling of
both carotid arteries and the vertebrobasilar complex. All images were pre- and
postoperatively assessed by two radiologists in consensus regarding technical success
rate, preoperative assessment (Spetzler-Martin classification, identification of arterial
feeders, existence of anatomic variants / functional crossfilling) and completeness of
resection. In all cases DSA served as the standard of reference.
Results: 4D-MRA was successfully performed in 20/20 exams and enabled the same
Spetzler-Martin classification as DSA in all cases (100 %). 11/13 (85 %) feeding arteries
were identified by 4D-MRA. Selective arterial spin labelling was successful in 16/20 (80 %)
exams. Selective arterial spin labelling provided additional functional or anatomic
information in 2/16 exams and enabled the diagnosis of a cross-filling feeding artery that
was not identified by 4D-MRA but by DSA, thus improving the sensitivity of MRI in
identification of arterial feeders from 11/13 (85 %) to 12/13 (92 %). Postoperative
assessment confirmed complete resection of all AVMs in 100 % yielding a 100 %
concordance between 4D-MRA in combination with selective arterial spin labelling and
DSA.
Conclusion: 4D-MRA in combination with selective arterial spin labelling is a promising
tool for pre- and postoperative assessment of cerebral AVMs providing functional
information that so far has been gained only with selective DSA.
References:
[1] Golay X, Petersen ET, Hui F. Pulsed star labeling of arterial regions (PULSAR): a
robust regional perfusion technique for high field imaging. Magn Reson Med 2005;53:1521.
[2] Hendrikse J, van der Grond J, Hanzhang L et al. Flow territory mapping of the cerebral
arteries with regional perfusion MRI. Stroke 2004;35:882-887.
[3] Lim CC, Petersen ET, Ng I et al. MR regional perfusion imaging: visualizing functional
collateral circulation. AJNR 2007;28:447-448.
[4] Willinek WA, Hadizadeh DR, von Falkenhausen M. 4D time-resolved MR angiography
with keyhole (4D-TRAK): more then 60 times accelerated MRA using a combination of
CENTRA, keyhole and SENSE at 3.0T. JMRI 2008;27:1455-1460.
[5] Hadizadeh DR, von Falkenhausen M, Gieseke J et al. Cerebral arteriovenous
malformation: Spetzler-Martin classification at subsecond-temporal-resolution fourdimensional MR angiography compared with that at DSA. Radiology 2008;246:205-213.
70
MR-Angioclub East Lansing 2009
7.2 Intraindividual comparision between multislice CT and 4 D
TWIST MRA in the assessment of residual cerebral
arteriovenous malformations – a prospective study protocol
Authors: M. Essig, M. Voth, A. Zabel-Du-Bois, L. Schuster
Institution: Department of Radiology, German Cancer Research Center, Heidelberg, Germany
Rationale and Objectives
Small or residual cerebral arteriovenous malformations (AVMs) are hard to visualise with
MR angiographic techniques. Although MRA techniques like TOF or contrast enhanced
MRA allow high resolution they are not able to visualize small or slow flowing vessel
segments. However, these small vascular structures and their intra cranial course are
important for radiotherapy planning and follow-up assessments. Therefore MS-CTA is
used to assess complete obliteration if MRA proved negative. The aim of the presented
study protocol was now to assess the validity of a time resolved 3D contrast enhanced
MRA technique in direct comparision with CTA.
Methods
In an ongoing dual centric prospective study protocol we used a combination of high
temporal resolution (250ms) and high spatial resolution (1x1x1 mm) CE MRA using a
standard dose (0,1mmol) of the macrocyclic Gadobutrol (Gadovist®, Bayer, Berlin) for a
TWIST (Time-Resolved Imaging with Stochastic Trajectories) acquisitions. The new 4 D
MRA technique was intraindividually compared with MS CTA to assess the visibility of
residual small AVM compartments and the presence of arteriovenous shunting.
The imaging data were evaluated in a qualitative matter by two independent readers with
special attention to the morphologic features of the malformation and the influence of the
dynamic MRA studies.
Results
So far we were able to include 12 patients with no residual AVM components on
conventional TOF-MRA examinations (inclusion criteria). In10 out of these patients
residual AVM components could be visualized on the dynamic and high resolution contrast
enhanced MRA sequences. The MS CTA was able to visualize residual vessel
components in only 8 patients. In two patients massive artifacts from previous
embolisation hindered the visualisation. In two patients no vascular components were
seen on both CTA and contrast enhanced MRA. In the MRA studies the hemodynamic
information from the TWIST technique was found to be very helpful in the differentiation
between physiologic and pathologic vessels.
Conclusion
In conclusion these preliminary results could prove that high temporal and spatial
resolution 4D MRA is able to assess even small residual AVM compartments at a high
sensitivity.
Based on the initial patient studies, Gadobutrol with its unique high molar concentration
seems to be the ideal contrasting agent in this study concept. The high concentration
allowed for a small bolus which is ideal for the high temporal resolution of the MRA, the
higher relaxivity was found to be ideal for the vessel assessment in the high resolution
MRA.
MR-Angioclub East Lansing 2009
71
7.3 Our Strategy for the Surgical Planning with 3T MRA
in Detecting Unruptured Cerebral Aneurysms
1)
Keiji Igase , Ichiro Matsubara1), Masamori Arai1), Jyunji Goishi1), Hitoshi Miki2),
Kazuhiko Sadamoto1)
1)
Department of Neurosurgery, Washokai Sadamoto Hoapital
2)
Departments of Radiology, Ehime University School of Medicine
Purpose: Subarachnoid hemorrhage is the most disastrous and irrevocable disease out of
many cerebrovascular diseases except for a few fortunate cases, thus it has been
overwhelmingly emphasized to detect unruptured intracranial aneurysms (ANs) with less
invasive examination. Therefore, we made our original 3T MRA-centered strategy for the
surgical planning on detection of unruptured cerebral ANs and verified the detection
capability of them compared with flat-panel detector digital angiography (FD-DA).
Methods: In order to screen unruptured intracranial ANs 3T MRI (Signa Excite: GE) was
aggressively exploited in our hospital. First of all, out-patients with some cranial problems
can undergo 3T-MRI for the screening of cerebrovascular diseases including cerebral
ANs, and in the case of patients with ANs suspected on MIP (Minimum Intensity
Projection) image of MR Angiography, VR (volume rendering) images are briefly created,
with which if ANs are definitely diagnosed, there are two options to proceed treatments
depending on their size. For ANs over 5mm in size flat- panel detector digital angiography
(FD-DA) is planned for scrutinizing both size and shape of the aneurysms because of its
having an indication for the operation, on the other hand for ANs less than 5mm 3D-CT
Angiography precedes FD-DA. Objective was all out-patients initially underwent 3T MRI
during one year in 2006, where patients with ANs have been followed for 3 years.
Results: Out of 3318 of out-patients 286 patients were found to have unruptured ANs,
and 3 years after 157 patients were followed, in which 2 patients (1.3%) had the rupture of
ANs, 15 patients (5.2%) the operation for unruptured ANs, 19 (12.1%) patients the
enlargement of ANs, and 121 (77.1%) no change of the ANs. All of the operated 15
patients underwent both 3T-MRI and FD-DA, revealing that VR images of 3T MRI closely
coincided with 3D-images of FD-DA in size and shape of ANs.
Conclusion: In detecting unruptured cerebral ANs for surgical treatment 3T-MR
Angiography, especially VR images, are sufficiently useful and our strategy with 3T MRIcentered system should be reasonable and beneficial for less invasive examination.
72
MR-Angioclub East Lansing 2009
7.4 Hybrid of Opposite Contrast MR Angiography of the Brain
1
1
2
Faïza Admiraal-Behloul , Evert Blink , Tokunori Kimura ,
Toshiba Medical Systems Europe, Zoetermeer, the Netherlands, 2MRI Systems Development
Department, Toshiba Medical Systems Corp., Otawara-Shi, Tochigi-Ken, Japan
1
Purpose: To compare a new non-contrast enhanced MR angiography technique, to an
optimized Time of Flight technique (TOF) with MTC pulse, in the visualization of fast and
slow flow brain vessels using comparable scan time on a 1.5T MR system.
Methods: The Hybrid of Opposite
contrast (HOP) technique is a dualecho 3D gradient-echo sequence
where the first echo is used to
generate a TOF white blood image
and the second echo generates a
Flow Sensitive Black Blood image
using motion-probing gradients to
Figure 1: (a) HOP image (b) TOF image.
introduce intra-voxel flow dephasing
[1]. In this work, the 2-echo images are combined using a frequency weighted subtraction
that enhances slow-flow (small) vessels [1]. Fifteen volunteers (8 men, mean age 46.2 y,
range 31 to 77 y) were imaged on a 1,5T System (Vantage ZGV Atlas, Toshiba) using a
13-channel Atlas-head coil with a Parallel Imaging factor of 2. Both TOF and HOP images
were obtained using a matrix of 256x256, FOV of 22x22cm, in-plane resolution of
0.86x0.86mm, slice thickness of 1mm and a slab thickness of 7cm. For TOF, TR = 30ms,
TE = 8ms and acquisition time = 5min12s. For HOP, TR = 34ms, TE1 = 7.5ms, second
TE2 = 24.9ms, and acquisition time = 5min30s. Water excitation technique was used for fat
suppression.
A slice-selective off-resonance sync pulse was used in TOF or optimal
background signal suppression. No MTC pulse was used in HOP.
Results: HOP and TOF were visually comparable in the depiction of fast flow vessels in
all subjects. The low-flow vessels were better visualized by HOP in 13 subjects (see fig.1
for a representative case) and comparable in 2 subjects.
Conclusion: HOP-MRA is a promising technique for the visualization of small collateral
vessels which is technically difficult with a standard TOF technique especially at 1.5 T.
Reference: 1. T. Kimura et al, Magn Reson Med. 2009 Aug; 62(2):450-8.
MR-Angioclub East Lansing 2009
73
7.5 High Resolution Fast Inversion Recovery MRA (FIR-MRA)
E. T. Tan, N. G. Campeau, J. Huston III, S. J. Riederer
Department of Radiology, Mayo Clinic, Rochester, MN 55905 USA
Purpose – Compared to 3D time-of-flight (TOF), the FIR-MRA technique provides
superior vessel conspicuity in imaging of the intracranial arteries [1]. However, the intrinsic
signal modulation of FIR-MRA causes loss of vessel sharpness and small vessel signal.
The FIR angiogram is obtained by a difference between the acquired, un-subtracted
bright-blood and black-blood data. Interestingly, the bright-blood signal is larger than the
difference signal at k-space periphery, while the dark-blood signal is negligible at k-space
center. We hypothesize that these properties of the un-subtracted data may be harnessed
to improve vessel depiction of high-resolution FIR-MRA by re-synthesizing the FIR-MRA
data.
Methods – Two data re-synthesis steps are proposed. First, the use of the difference data
for central k-space and the un-subtracted bright-blood
4 cm
data for k-space periphery reduces signal modulation and
improves sharpness. Second, an additional subtraction of
a fraction λ of magnitude dark-blood data with a threshold
at zero reduces the level of residual tissue signal. The
optimum value for pass band frequency in step one was
determined to be 70% of maximum k-space, and that for
λ of step two was 10%. This technique was evaluated in
A
vivo at 3T.
Results – Fig. 1 shows improvements in the smoothness
and sharpness of large vessels, and in the conspicuity of
small vessel branches. The background noise and
residual tissue signal are also noticeably reduced.
Conclusion – Re-synthesis of FIR-MRA data results in
superior vessel depiction, as well as reduction in residual
tissue signal and background noise.
References – [1] Tan ET et al. ISMRM 2009, #91.
B
Fig. 1. Axial targeted MIPs
(35 mm thick) at the left
internal carotid artery before
(A) and after (B) resynthesis, with
improvements in vessel
depiction noted (arrows).
74
MR-Angioclub East Lansing 2009
7.6 3D Dual V E NC P C MR A us ing S piral P rojec tion Imaging
1
N. R. Zwart , and J. G. Pipe1
Keller Center for Imaging Innovation, Barrow Neurological Institute, Phoenix, Arizona, United States
1
P urpos e: The dual-VENC method of unaliasing a low VENC with a high VENC MRI data
set allows for a significant improvement in the velocity to noise ratio of PCMRA [1]. The
collection of additional data necessitates the use of a rapid imaging technique to maintain
feasible scan durations. The proposed method is a 3D k-space sampling trajectory called
Spiral Projection Imaging (SPI) [2]. This work demonstrates the effectiveness of SPI while
maintaining a short sample window and increased
undersampling. The synthesis of high VNR images is
explored with attention to problematic areas of dephasing.
Methods :
S P I:
Variable density spiral interleaves are
critically sampled up to 1/8 of the k-space radius (Fig. 1a).
At the edge of k-space, spirals are separated by a factor of 4
times the critical sample distance. Spiral planes are rotated
about kz to fill 3D k-space (Fig. 1b). The scan parameters
are: 24cm FOV, 0.8mm3 voxel, 300 dia. matrix, 20
interleaves, 120 planes, 19.5msec TR, and a 6min total scan
time for 7 volumes.
Dual VE NC :
Velocity encoded sets
were collected for a 100cm/s and 20cm/s VENC.
High
velocity gradients at vessel walls cause vessel narrowing in
unaliased low VENC images.
This is addressed by a
weighted combination of the high and low VENC data.
R es ults and Dis c us s ion:
SPI PCMRA scans were
collected with a GE 3T Signa Excite system (Fig. 1c).
Blurring near the sinuses becomes problematic with longer sampling windows than what is
used. Aliasing due to undersampling causes some smaller vessels to appear inconsistent.
Vessel diameter in high flow areas is regained by partial combination of high VENC data
with the unaliased low VENC data.
C onc lus ion: This method provides short scan times that make the added time required
by dual VENC techniques less prohibitive.
R eferenc es : [1] Lee, A. MRM, 33:122, 1995; [2] Irarrazabal, P. MRM, 33:656, 1995;
MR-Angioclub East Lansing 2009
75
7.7 High Resolution Simultaneous Angiography and Venography
(MRAV) with a Single Echo
1,2
Samuel Barnes, MS , E. Mark Haacke, PhD1,2
1. Wayne State University, Detroit 2. Loma Linda University, Loma Linda, CA
Purpose: In this work we develop novel acquisition and post processing techniques to
improve the quality of single echo MRAV acquisition using susceptibility weighted imaging
(SWI). Techniques recently presented have focused on double echo acquisitions which
will be harder to implement at very high fields.
Methods: All SWI images were acquired at 3T with TR/TE/FA/Resolution/BW =
30ms/20ms/15°/0.5x0.5x0.5mm3/50Hz/pixel or 160Hz/pixel. To visualize veins the data
was downsampled to a resolution of 0.5x0.5x2.0mm3 before the SWI processing.
Results: Using a higher bandwidth of 160 Hz/pixel as compared with 50 Hz/pixel
dramatically reduces flow dephasing, improving larger vessel visibility at the cost of a
reduced signal-to-noise ratio (Fig. 1). The high isotropic resolution also helps to reduce
dephasing across a voxel improving the MRA. The original isotropic resolution data
showed poor venous contrast due to the non-optimal aspect ratio of 1:1 which causes the
phase for certain orientations of veins to have the opposite sign (Figure 2). This lost
contrast is fully recovered by downsampling the high resolution data to a more optimal
aspect ratio of 1:4 (0.5x0.5x2 mm3). The downsampling takes advantage of the high
resolution data by reconstructing the slices in an overlapping pattern so the optimal partial
voluming of smaller structures is guaranteed thus improving image quality.
Figure 1. 50 Hz/pixel (left) shows almost
complete loss of the MCA and losses in
the middle of smaller vessels while
160Hz/pixel (right) shows minimal flow
losses.
Figure 2. Isotropic phase image (left)
shows veins switching from bright to
dark depending on orientation.
Downsampled image (right) shows
higher SNR and more homogenous
veins producing a better venography.
Figure 3. Maximum intensity
projection (left) over isotropic
data showing arteries and
minimum intensity projection
(right) over SWI processed
downsampled image.
Conclusion: By collecting data with high isotropic resolution and higher bandwidth, flow
related losses from higher order uncompensated effects can be reduced improving the
quality of the MRA extracted from the SWI data even at long echo times (20ms).
76
MR-Angioclub East Lansing 2009
7.8 Low Dose 3D Time-Resolved MR Angiography of the
Supraaortic Artery: Correlation to High Spatial
Resolution 3D Contrast-Enhanced MRA
Yoon-Joo Lee, So-Lyung Jung, Kook-Jin Ahn, Bum-soo Kim
Department of Radiology, Seoul St.Mary Hospital, The Catholic University of Korea
Purpose: To evaluate the effectiveness of low-dose, contrast-enhanced, time-resolved,
three dimensional (3D) magnetic resonance (MR) angiography (TR-MRA) in the
assessment of supraaortic vessel, and to compare the results with high-resolution contrast
enhanced MRA (HR-CEMRA).
Materials and Methods:
45 consecutive patients underwent contrast enhanced 3D TR-MRA and high spatial
resolution 3D CE-MRA for evaluation of neurovascular disease at 3T. Gadolinium-based
contrast medium was administered at a constant dose of 1ml for TR-MRA, and
0.1mmol/kg for HR-CEMRA. Two readers evaluated image quality using a four point scale
(from 0=excellent to 3=non-diagnostic), artifacts and findings on both datasets.
Results: The overall image quality for low dose TR-MRA was in the diagnostic range
(median 0, range 0-3). Two cases showed non-diagnostic image quality due to severe
motion in patients with acute ICA occlusion. Readers demonstrated additional
hemodynamic information on TR MRA in 3 patients with severe stenoocclusive lesions.
For the evaluation of arterial stenosis, TR-MRA well correlated with HR-CEMRA (r=0.668,
p<0.001). Of the 675 supraaortic arterial segments evaluated for stenosis or occlusion,
TR-MRA agreed with HR-CEMRA in 611 of 675 (90.5%), overestimated in 41 of 675
(6.1%), and underestimated 23 of 675 (3.4%).
Conclusion: TR-MRA can be achieved by administration of small contrast dose (1cc,
0.1mmol), and yields rapid and important functional and anatomical information in the
evaluation of supraaortic arteries. Due to limited spatial resolution, TR-MRA is has
tendency to overestimated the stenosis or occlusion of smaller intracranial arteries.
MR-Angioclub East Lansing 2009
77
7.9 Obstruction of IJV by Asymmetry of Lateral Mass of Atlas on
Head and Neck CEMRA and Contrast CT
1
Tae-Sub Chung, MD , Hye Mi Gweon, MD1, Sang Hyun Suh, MD1
Department of Diagnostic Radiology, Gangnam Severance Hospital 1
Yonsei University, Seoul 135-720, Republic of Korea
Purpose;
To evaluate that asymmetry of lateral mass of atlas could be the cause of high level
internal jugular vein (IJV) obstruction on head and neck contrast enhanced 3D MR
angiography (CE-MRA) and contrast enhanced computed tomography (CE-CT).
Materials and Methods;
Thirty cases among 1800cases which examined both head and neck CE-MRA and CECT were enrolled during last 5 years. The eleven cases had IJV obstruction and nineteen
cases had no IJV obstruction on CE-MRA. We defined obstruction group which had IJV
obstruction and control group which had no IJV obstruction on CE-MRA. The following
parameter was measured from axial images of CE-CT: 1) the diameter of IJV; 2) the
distance between styloid process and ipsilateral lateral mass of atlas; 3) maximum area of
lateral mass of atlas.
Results;
The incidence of IJV obstruction was 28% (504/1800 cases) of all reviewed head and
neck CE-MRA. The diameter of IJV and distance between styloid process and lateral
mass of atlas at IJV obstruction side in obstruction group were 1.6 ± 1.0mm and 4.1 ±
2.1mm. The diameter of IJV and distance between styloid process and lateral mass of
atlas were significantly narrower than those of contralateral normal side in obstruction
group (3.5 ± 1.7mm and 3.7 ± 3.2mm, p<0.001). The maximum area of lateral mass of
atlas at IJV obstruction side was significantly larger than that of contralateral normal side
in obstruction group (24.2 ± 10.5 mm2, p<0.001).
Conclusion: The cause of high level IJV obstruction on head and neck CE-MRA was
narrowing space between styloid process and lateral mass of atlas resulting from larger
area of lateral mass of atlas and induced IJV compression.
78
MR-Angioclub East Lansing 2009
®
8.1 Evaluation of Gd-DOTA (DOTAREM ) enhanced MRA compared
to time-of-flight MRA in the diagnosis of clinically significant
non-coronary arterial disease at 1.5 and 3.0 Tesla.
1
Dipan J. Shah , Lim Tae-Hwan2, Steven Wolff3.
Methodist DeBakey Heart & Vascular Center, The Methodist Hospital, Houston, TX.
2
Department of Radiology, Research Institute of Radiology, University of Ulsan, College of Medicine,
Asan Medical Center, Seoul, Korea.
3
Advanced Cardiovascular Imaging, New York, NY.
1
PURPOSE: To assess the diagnostic accuracy and safety of meglumine gadoterate (GdDOTA)-enhanced MRA over non-enhanced Time-of-Flight (TOF) MRA at 1.5 and 3.0
Tesla for clinically significant non-coronary arterial disease by comparing of each
technique with x-ray angiography.
MATERIAL AND METHODS: Multicenter, open-label, paired trial in 192 subjects (100 at
1.5 Tesla and 92 at 3.0 Tesla), age >18 years, (140 men, 52 women; mean [±SD] age,
63.7 ± 13.9 years) with suspected non-coronary arterial disease and scheduled to undergo
x-ray angiography were included and received an iv bolus of 0.1 mmol/kg Gd-DOTA.
Renal insufficiency was present in 15 (7.8%) of subjects. The percent agreement was
defined as the number of stenosis grades measured with MRA in agreement with x-ray at
segment level / total number of segments evaluated for the patient x 100. Sensitivity and
specificity were assessed at the segment level.
RESULTS: The arteries imaged were: aorto-iliac (39.6%); renal (18.2%); calf (13.0%);
femoral (12.5%); carotid (12.0%); and popliteal (4.7%).
There was a statistically
significantly greater mean (± SD) percent agreement of MRA to x-ray with Gd-DOTA MRA
vs TOF MRA (85.8% ± 19.8% vs 78.3% ± 24.9%, respectively; difference 7.4% ± 22.1%
[p<0.0001]). The sensitivity and the specificity of Gd-DOTA MRA were significantly greater
than TOF MRA (93.3% vs 85.7% [p=0.0004] and 89.7% vs 84.4%, [p=0.0019]
respectively). There were no serious drug-related adverse events and no spontaneously
reported cases of nephrogenic systemic fibrosis.
CONCLUSION:
Gd-DOTA-enhanced MRA provided significantly greater diagnostic
accuracy than TOF MRA at 1.5 and 3.0 Tesla in the diagnosis of non-coronary arterial
disease.
MR-Angioclub East Lansing 2009
79
8.2 Pulmonary MRA in 75 patients with dyspnea
ML Schiebler1, SK Nagle1, CJ Francois 1, RF Busse4, ACS Brau4,
JH Brittain4, TM Grist1, and SB Reeder 1, 2, 3
1. Department of Radiology, UW- Madison
2. Department of Medicine, UW-Madison
3. Department of Medical Physics, UW-Madison
4. Applied Science Lab, General Electric, Waukesha, WI
Purpose: Review of our clinical experience with a new contrast enhanced pulmonary
MRA (CE-MRA) sequence with 2D parallel imaging (ARC) 1
for the detection of
pulmonary embolism (PE) performed in 75 dyspneic patients.
Methods: A total of 75 CE-MRA (accelerated with 2D-ARC) pulmonary MRA exams in
patients with dyspnea were reviewed for the presence of emboli (PE), perfusion defects
within the lungs, and artifacts limiting diagnostic quality.
These artifacts fell into 4
categories: central vessel signal drop-out (CVSDO), central field-of-view (FOV) blurring,
bolus timing issues, and residual aliasing.
Results: 20 pulmonary emboli were detected in 11 patients. 27 lung perfusion defects
were demonstrated. Lobar (n=8), segmental (n=7) and sub-segmental (n=2) PE were
identified. Only 5 cases contained artifacts that required follow-up imaging with computed
tomography angiography (CTA) to exclude PE. In most cases of CVSDO, use of multiphasic imaging allowed differentiation from PE. Central FOV blurring and residual aliasing
were only a problem when the entire AP dimension of the patient was not included in the
FOV. Bolus-timing issues were not a significant problem.
Conclusion: CE-Pulmonary MRA with 2D-ARC allows near-isotropic high-resolution
whole chest coverage in a14s breath hold. This is now being used routinely to evaluate
young patients with suspected pulmonary embolism in order to limit radiation dose to this
vulnerable population at our institution.
Figure 1: Pulmonary embolus in patient with dyspnea. A). Pulmonary infarct (arrow) on
pre-contrast 2D ARC, B). Perfusion defect seen on dextro-phase of CE-MRA in left lower
lobe (arrow), C). Segmental embolus in LLL lateral segmental artery (arrow) causing the
perfusion defect seen in A and B.
A
B
C
References: 1. Schiebler et al, ISMRM 2008, Poster # 3928
80
MR-Angioclub East Lansing 2009
8.3 Origin and Frequency of artifacts in Contrast Enhanced
Pulmonary MRA in 80 patients with dyspnea
ML Schiebler1, SK Nagle1, CJ Francois 1, RF Busse4, ACS Brau4,
JH Brittain4, TM Grist1, and SB Reeder 1, 2, 3
1. Department of Radiology, UW- Madison
2. Department of Medicine, UW-Madison
3. Department of Medical Physics, UW-Madison
4. Applied Science Lab, General Electric, Waukesha, WI
Purpose: Clinical use of a new contrast enhanced pulmonary MRA sequence with 2D
parallel imaging (ARC) 1 for the detection of pulmonary embolism (PE) requires a careful
understanding of the normal range of artifacts to
help prevent a false positive
interpretation.
Methods: A total of 80 CE-MRA (accelerated with 2D-ARC) pulmonary MRA exams in
patients with dyspnea were reviewed for the presence of significant artifacts (Gibbs
ringing, cardiac motion induced blurring, aliasing, respiratory motion) necessitating further
imaging with CTA to rule out the presence of PE.
Results: Of the 80 exams, only 5 cases had artifacts on pulmonary MRA requiring further
imaging with CTA. These artifacts (4 cases) appeared as central hypo-intense foci that
resembled PE. The origin of these artifacts may be a Gibb’s ringing phenomenon or from
mixing of contrast (from the SVC) with un-opacified blood from the IVC. These occurred
most commonly in the right and left lower lobe (LLL) pulmonary arteries. In addition,
cardiac motion occasionally obscured the right middle lobe and lingular branches (1 case).
In this series, respiratory motion and aliasing were not problematic artifacts.
Conclusion: Understanding the artifacts is necessary when interpreting pulmonary MRA.
With understanding of these artifacts, highly diagnostic pulmonary MRA can be performed
in the vast majority of patients, greatly reducing the use of unnecessary radiation.
Figure 1: A. Gibbs ringing artifact simulating a central PE in the left lower lobe pulmonary
artery B. Cardiac motion induced blurring of right middle lobe arteries C (arrows).
Respiratory motion in a positive case of pulmonary embolism showing perfusion defect
(arrowhead) in left upper lobe and a LLL PE (proven at CTA).
References: 1. Schiebler et al, ISMRM 2008, Poster # 3928
MR-Angioclub East Lansing 2009
81
8.4 Gadolinium Enhanced Magnetic Resonance Angiography for
Pulmonary Embolism: Results of PIOPED III
Paul D, Stein, MD
Visiting Professor, Department of Medicine, School of Osteopathic Medicine, Michigan State University,
East Lansing, MIchigan
Purpose
PIOPED III is a multi-center prospective study of contrast-enhanced magnetic resonance
angiography (MRA) and venography (MRV) accuracy for diagnosis of acute pulmonary
embolism (PE). The study rationale was the assumed safety of MRA in the large number
of patients who have contraindications to CT angiography (CTA) related to iodine allergy,
impaired renal function, or ionizing radiation.
Materials and Methods
Patients were eligible for the study if they were suspected of having PE. Exclusions
included age under 18, inability to complete MRA within 72 hr of the reference test,
contraindications to MRA, critical illness, and other standard exclusions. All patients had
Wells’ score, MRA and one or more reference imaging tests (CTA, V/Q scan, digital
subtraction pulmonary angiography or lower extremity ultrasound). Basis for confirmation
of PE included CTA showing PE in main or lobar pulmonary arteries; positive DSA; and
either 1] positive CTA (not main or lobar) or positive CTV; or 2] high probability V/Q scan;
or 3] positive lower extremity US; in combination with high or intermediate Wells’ score.
Basis for exclusion of PE included the inverse of the confirmatory criteria, and patients
who did not meet the confirmatory or exclusionary criteria were classified as PE uncertain.
Results
During the course of the study, a novel disorder (nephrogenic systemic fibrosis) was
attributed to use of gadolinium in the presence of impaired renal function, and this
impacted recruitment negatively.
A total of 818 patients were enrolled.
The high
prevalence of relative contraindications to CTA noted in the PIOPED II study was
confirmed. The image quality, sensitivity and specificity of MRA were assessed. The
results will be presented in detail.
Conclusions
The use of MRA in patients with impaired renal function continues to evolve, while patients
with iodine allergy and radiation issues remain candidates for MRA. The role of MRA in
diagnosis of PE will be discussed.
82
MR-Angioclub East Lansing 2009
8.5 4D time-resolved MR angiography for non-invasive pulmonary
post-embolization AVM patency assessment
L Boussel, A Cernicanu, D Gamondes, C Khouatra, V Cottin, D Revel, P Douek. Lyon, France
Purpose: In Rendu-Osler disease, post-percutaneous embolization recurrence of
pulmonary arteriovenous malformation (AVM) patency is often difficult to assess noninvasively using CT because of its poor temporal resolution. We assess the capability of a
post IV Gd 4D time-resolved MR angiography (MRA) sequence to distinguish between
patent AVMs and healthy normal vessel by analyzing pulmonary arterial and venous
enhancement kinetic.
Methods: After IRB approval, 8 patients with 8 documented pulmonary AVMs (3
previously embolized (recurrence) and 5 untreated), prospectively underwent: a thoracic
MDCT scanner to localize the AVMs; a pulmonary digital substracted angiography (DSA)
to assess AVMs patency and a 4D time-resolved MRA with keyhole and view sharing
compression method at 3T (Philips Achieva). MRA was performed after IV injection of 15
ml of Gadovist (Bayer-Schering) at a 2cc/s rate with the following parameters: FOV:
500x350x240 mm, spatial resolution: 1.2x1.2x1.4 mm, keyhole factor: 20%, viewsharing
compression: 100%, dynamic scan time (temporal resolution): 1.2 s, total acqusition time:
22.7 s for 6 dynamic images. All images were consensually reviewed by two experienced
radiologists. Signal value of cross section AVMs afferent pulmonary arteries and efferent
veins were compared to reference arteries and veins located in an healthy pulmonary area
at the same distance from the hilum than the studied AVM vessels. The difference in Time
to Peak (dTTPav) for each couple artery/vein was calculated. A Mann Whitney test was
used to compare dTTPav for AVMs and reference vessels and recurrent and untreated
AVMs.
Results: A complete thoracic coverage with 6 dynamic time points was obtained for each
patient within a single breath hold and with sufficient spatial resolution to analyze distal
arteries and veins. dTTPav was significantly smaller in AVM (0.45 +/- 0.9 s) than in
reference vessels (3.75 +/- 1.35 s), p=0.0006. No significant difference was found
between recurrent and untreated AVMs, p=0.4.
Conclusion: 4D time-resolved MR angiography is a promising tool for non-invasive
thoracic AVM post-embolization patency assessment.
MR-Angioclub East Lansing 2009
83
8.6 Radial Sliding Window MRA in Pulmonary Hypertension
1,2
Timothy J. Carroll , Amir Davarpanah2, James C. Carr2, Michael Cuttica3,
2
4
1
John Sheehan , Sanjiv Shah , and Hyun Jeong
1
2
3
4
Biomedical Engineering, Radiology, Internal Medicine, and Cardiology,
Northwestern University, Chicago, IL
Purpose: To develop an MRI imaging protocol for the quantification of hemodynamic
changes resulting from pulmonary hypertension.
Methods: We have developed an approach to CE-MRA which is based on a previously
reported radially sampled MR fluoroscopic technique using sliding mask subtraction (1, 2,
3). This allows for better A/V separation
which
aids
in the identification
of pulmonary
Sliding
Window
Mask-Mode
Subtractions Improves
the Depiction of
The Distal Branches of the Pulmonary Veins.
branches (Figure 1). We tested
the hypothesis that bolus transit
times
are
pulmonary
patients.
(n=17,
indicators
of
hypertension
in
(a)
(b)
A series of patients
male=8,
female=9,
<age>=46 ± 9.7) were studied
with
elevated
mean
arterial
pressure (mPAP> 25 mm Hg)
measured
in
a
right
heart
catheterization
study
confirmed
diagnosis
the
which
of
pulmonary arterial hypertension.
Figure 1. Mask mode subtraction using (a) a precontrast mask image and (b) a “sliding mask” that is
a fixed time lag behind the angiogram. The
improved depiction of the pulmonary veins
(arrows).
ROIs were place to cover the proximal and distal
branches of the pulmonary arteries and veins. Arterial and venous transit times were
calculated individually as a test of the improved separation of the arterial and venous
phases.
Results/Discussion: We found that bolus transit times, as measured with our high frame
rate pulse sequence, were significantly (p<0.05) increased in pulmonary hypertension
patients.
Conclusions: CE-MRA of the pulmonary arteries may serve as an adjunct to cardiac
catheterization.
References: (1) Reiderer et all MRM 1998, (2) Cashen MRM 2007, (3) Jeong MRM 2009.
84
MR-Angioclub East Lansing 2009
8.7 Non-Contrast Enhanced Pulmonary Vein MRI with a Spatially
Selective Slab Inversion Preparation Sequence
Peng Hu1, Michael L. Chuang1, Kraig V. Kissinger1, Beth Goddu1, Lois A. Goepfert1, Neil M. Rofsky2,
Warren J. Manning1, Reza Nezafat1
1
Departments of Medicine and 2Radiology, Beth Israel Deaconess Medical, Boston, MA
Purpose: With recent reports of adverse effects of use of contrast agents, non-contrast
pulmonary vein (PV) imaging has been of interest [1,2]. We propose a non-contrast
enhanced free-breathing ECG-gated 3D thin-slab gradient echo sequence with a sagittal
slab-selective inversion for PV angiography.
Methods: A sagittal inversion slab was applied prior to
data acquisition to suppress cardiac structures adjacent to
the left atrium (LA) and PV (Fig. 1) thereby, improving
PV/LA conspicuity. The feasibility of the proposed method
was demonstrated in 5 healthy subjects. Contrast-to-noise
ratio (CNR) between LA and right atrium (RA), ascending
aorta and pulmonary artery was measured and compared
Fig. 1. Sequence diagram.
with conventional non-contrast imaging without inversion.
Results: Figure 2 shows improvement of PV and LA conspicuity using our method. Figure
3 shows an example volume view of PV image. Compared to the conventional GRE
without inversion, our technique increased the CNR between LA and RA and pulmonary
artery by 20 and 4 fold (p<0.01), respectively.
Conclusions: The proposed technique enhances the conspicuity of the PVs and LA with
minimal loss of SNR.
References:
1. Francois et al.,
Radiology 2009;
250:932-939
2. Krishnam et al.,
Invest. Radiology,
2009,June 25, ePub.
Fig 2: A comparison of images
acquired using a conventional GRE
(top row) and the proposed
technique (bottom row).
Fig 3: 3D volume view
of PV and LA acquired
using the proposed
method.
MR-Angioclub East Lansing 2009
85
8.8 MRA with the “No Phase Wrap”
Grace Choi, Martin R. Prince
New York, NY
PURPOSE: Aliasing artifact in the phase encoding direction, also known as wrap-around
artifact, occurs when patient anatomy extends beyond the field of view (FOV). Tissues
outside the FOV wrap around to the opposing edge in the phase-encode direction
superimposing unwanted signals on the area of interest which may interfere with
diagnosis.
Aliasing has become more problematic in abdominal and thoracic MRA
utilizing parallel imaging which may wrap the arms onto the middle of the torso
superimposing on major vessels.
Metallic, e.g. aluminum foil, sleeves have been
successful in eliminating wrap-around but heating of the metal and cumbersome
application of the foil has prevented this from being a viable clinical option. We have
developed sleeves that are convenient to apply and eliminates phase aliasing artifact with
negligible heating.
MATERIALS AND METHODS: Sleeves were constructed using silver and carbon fabrics
of varying mesh geometry and weights and treated with liquid silicone to absorb excess
heat. The opening diameter for the shoulder was minimized with an elastic liner to adjust
to patient size. Imaging was performed on 1.5T and 3.0T systems (GE HDx 14.0) using an
8-channel body array coil. Coronal and axial single shot fast spin echo (SSFSE), axial
steady state free precession and spoiled gradient echo sequences were performed with
and without sleeves to identify the optimal fabric.
A final design was evaluated on
abdominal, thoracic and unilateral upper extremity MR Angiography studies.
RESULTS: Phase wrap-around artifact was present on all images scanned without
sleeves. Corresponding images using sleeves showed no wrap-around artifact.
MRA was possible with smaller FOV without wrap around artifact from the arms on
all studies. MRA with parallel imaging was possible with smaller FOV.
CONCLUSION:
Silver and carbon sleeves effectively eliminate phase wrap-around
artifact from arms in patients who cannot tolerate elevating arms overhead for abdominal,
thoracic and unilateral upper extremity MRA.
86
MR-Angioclub East Lansing 2009
8.9 Unruptured Intracranial Aneurysms; Detection and Follow-up
on 3.0T MRA
1
Hitoshi Miki , K Igase3, I Matsubara3, I Kiriyama2, Kazuhiko Sadamaoto3
1
Department of Radiology, Ehime University School of Medicine,
Department of Radiology2 and Neurosurgery3, Sadamoto Hospital, JAPAN
Purpose: To evaluate the frequency, size, location and enlargement of unruptured
intracranial aneurysms (UIAs) found during screening with 3.0T MR angiography (MRA) at
our hospital.
Methods: 3D time-of-flight (TOF) MRA was performed for 3,414 cases (1,453 men, 1,961
women, a mean age of 65.7 years) without neurological sign during January to December
2006 at our hospital. All screening MRA was performed with a 3.0T MR system by using
an eight-element phased array head coil.
Results: The UIAs were found in 286 (8.4%) of the 3,414 cases, 93 men and 193 women,
with a mean age of 65.7 years. The locations of the UIAs of single aneurysm cases were
as follows: anterior cerebral artery (ACA) including anterior communicating artery (A-com)
in 43 lesions, middle cerebral artery (MCA) in 68 lesions, C1 portion of internal carotid
artery (ICA) in 44 lesions, C2 portion in 7 lesions, C3-5 portion in 79 lesions, and basilar
artery (BA) in two lesions and vertebral artery (VA) in 15 lesions. In 30 cases, multiple
UIAs were observed. Aneurysm size varied as follows: 143 lesions (50.5%) were less than
3 mm, 94 (33.2%) were ranged from 3 to 5 mm in size, 34 (12%) from 5 to 7 mm, 11
(3.8%) from 7 to 10 mm, and 4 (1.4%) were greater than 10 mm. The frequency of UIAs
on screening 3.0T MRA was higher than that of past screening reports with 1.5T MRA.
Especially, the frequency of small aneurysms less than 3mm markedly increased in our
study. One hundred forty of 286 cases were followed with serial MRA. Frequency of
enlargement was 13.6% (Nineteen cases).
Conclusion: 3.0T 3D TOF MRA should be excellent modalities for screening and
following up unruptured intracranial aneurysms.
MR-Angioclub East Lansing 2009
87
9.1 Comprehensive PC MR Imaging in Congenital Heart Disease
Oliver Wieben1,2, Kevin M. Johnson1, Elisabeth Nett1, Ben Landgraf1,2, Scott Reeder1,2,
2
3
3
2
2
Mark Schiebler , Sharda Srinivasan , Carter Ralphe , Darren Lum , and Chris Francois
Depts. of Medical Physics1, Radiology2 & Pediatric Cardiology3 - University of Wisconsin
Purpose: To further develop and validate highly accelerated radially sampled phase
contrast imaging (PC VIPR) for clinical use in congenital heart disease [1].
Methods: 24 consecutive CHD patients (range 9 weeks – 67 years) with a variety of
pathology including aortic coarctation (9), bicuspid aortic valve (6), tetralogy of Fallot (5),
atrial septal defects (3) among others were scanned at 1.5T and 3 T.
Typical scan
parameters: imaging volume = 320 x 320 x 180 mm3, readout = 256-320, (1.0-1.25 mm)3
acquired isotropic spatial resolution, VENC = 50-150 cm/s, TR/TE = 8.7/2.8 ms, flip = 10º ,
retrospective cardiac gating, scan time ~ 10 min (50% respiratory gating efficiency). CEMRA and 2D PC images were used for comparisons when available. Data processing in
customized analysis and visualization tools (Matlab & Ensight).
Results: PC VIPR data sets were successfully acquired in all patients. All anatomical
structures visualized on CE MRA images were identified on the angiograms derived from
the PC VIPR images. On a case-by-case basis, additional hemodynamic information was
obtained including visualization of flow patterns, flow quantification, and transstenotic
pressure gradients. PC VIPR flow measurements were more accurate than the clinical
routine targeted 2D PC measurements.
Conclusion: The radially undersampled 10 min PC MRI acquisition coupled with the
developed post-processing tool has proven to be a versatile tool for imaging in CHD.
Coupled with an efficient motion correction it can possibly replace exams in the young
currently performed under anesthesia with more patient friendly sedation.
e
SVC
f
g
RPA
L
P
A posterior view, (b) atrial defect: 1.34 L/min, (c)
Fig 1: (a-d) Pulmonary venoblar syndrome – (a)
analomous pulmonary venous return “Scimitar Vein”: 0.42 L/min, (d) abnormal systemic artery: flow to
the right lung., (e-f) Double inlet left ventricle status post bidirectional Glenn – particle traces and flow
waveforms, (g) Aortic coarctation - pressure map.
88
MR-Angioclub East Lansing 2009
9.2 Ultrasound-guided Cardiac Gating for Coronary MRA
G. Liu1, R. Walcarius2, X.L. Qi2, A. Dick2, G. A. Wright1
1Medical Biophysics, University of Toronto, Toronto, Ontario, Canada,
2Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
Purpose: Mistakes on the order of tens of milliseconds in the timing of imaging windows
can incur significant motion artifacts in coronary MRA. We present a new, ultrasoundbased method for identifying quiescent periods by monitoring the velocity of the interventricular septum (IVS), a correlate of flow into and out of the left ventricle [1]. We
demonstrate that, compared with using a cine SSFP pre-scan, our determination of gating
parameters produces sharper coronary MRA images.
Methods: Right coronary artery (RCA) imaging was performed on four healthy volunteers
using a GE Signa 1.5T system. Two sequences were used: (1) T2-prep spiral GRE during
6 to 15s breath-holds with 0.77 x 0.77 x 3.6 mm resolution; and (2) respiratory navigated
3D fat-sat SSFP, with 1.4 x 1.4 x 2.0 mm resolution. ECG gating parameters were
determined by, first, ultrasound imaging of the IVS, and second, a SSFP cine of the
4-chamber view [2]. Differences in gating parameters produced different results in RCA
visualization. Two experienced observers chose the better images of the two data sets in
a blinded, head-to-head comparison.
Results: One observer always favoured the MRAs guided by the ultrasound pre-scan.
The second observer found image quality to be better for the ultrasound method in 7 out of
8 comparisons, and equal between the two methods in the remaining case. Figures 1 and
2 show some sample comparisons.
Figure 1: 2D RCA MRA (74 bpm)
ECG gating parameters obtained by (left) an
ultrasound pre-scan [onset: 600ms; duration:
53ms], and (right) a cine MR pre-scan [onset:
432ms;duration: 158ms].
Figure 2: 3D RCA MRA (62b pm)
ECG gating parameters obtained by (left) an
ultrasound pre-scan [onset: 650ms;duration:
158ms], and (right) a cine MR pre-scan
[onset:561ms;duration:165ms].
Conclusion: In this study, RCA MRA produced sharper images under the guidance of an
ultrasound pre-scan versus the guidance of a MR cine pre-scan.
References: [1] Mundigler G et al., JClinBasicCardio 2002 (5).
[2] Jahnke et al., Radiology 2006 (239).
MR-Angioclub East Lansing 2009
89
9.3 Contrast-Enhanced Whole-Heart Coronary MRA at 3T Using
Gradient Echo Interleaved EPI (GRE-EPI)
1
2
1
Himanshu Bhat , Sven Zuehlsdorff , Debiao Li .
Northwestern University, Chicago, IL, 2Siemens Medical Solutions, Chicago, IL
1
Purpose: Whole-heart coronary MRA is a promising technique for detecting coronary
artery disease; however its major drawback is the long scan time on the order of 10-15
minutes. The goal of this work was to reduce the
scan time of whole-heart coronary MRA by using
a GRE-EPI [1] sequence at 3T.
Methods: 6 echoes (TR = 10.6 ms) were
acquired after each RF pulse. To minimize kspace
modulations,
scan
parameters
were
selected using simulations of the Bloch equation.
A
dual
reference
scan
phase
correction
technique was used for accurate echo alignment
in the presence of increased off-resonance
effects at 3T. The GRE-EPI readout was
combined with GRAPPA [2] for a further
reduction in scan time.
Results: Whole-heart coronary artery images
were acquired in 7 volunteers with a spatial
resolution of 1.0 x 1.0 x 2.0 mm3 in an average
scan time of 2.6 ± 0.6 minutes with an average
navigator efficiency of 41.7 ± 9.7%. Fig. 1 shows
coronary artery images from 2 volunteers using
the contrast-enhanced GRE-EPI (a-c) and GRE
(d-f) techniques acquired in separate sessions
for comparison purposes. The imaging times with
GRE-EPI were 1.6 and 2.6 minutes and those
with GRE were 4.2 and 8.7 minutes. Both
sequences
show
similar
depiction
of
the
Fig 1. Coronary artery images using a
contrast-enhanced GRE-EPI sequence
(a-c) and a contrast-enhanced GRE
sequence (5d-f), acquired in separate
scan sessions.
coronary arteries.
Conclusion: Compared with current techniques, the proposed GRE-EPI method
represents a factor of 3 reduction in scan time and a factor of 2 reduction in contrast dose.
References: [1] MRM 30(5):609-16, 1993. [2] MRM 47(6):1202-10, 2002.
90
MR-Angioclub East Lansing 2009
9.4 Feasibility of Whole-Heart Coronary MRA on 3 Tesla Using
Ultrashort-TR SSFP VIPR
1
1
1
1
J. Xie , P. Lai , H. Bhat , and D. Li
Departments of Radiology and Biomedical Engineering, Northwestern University, Chicago, IL, United
States
1
Purpose: The purpose of the work was to evaluate the feasibility of whole-heart coronary
MRA on 3.0T using SSFP and verify that ultrashort TR with VIPR allows good coronary
MRA image quality with SSFP.
Methods: Eight healthy volunteers were studied on a 3.0 Tesla Siemens whole-body
scanner during free breathing. An ECG-triggered, navigator-gated SSFP VIPR sequence
was used for data acquisition. The imaging parameters were: TR/TE = 3.0 ms/1.5 ms,
bandwidth/pixel was 868 Hz, resolution = 1.3*1.3*1.3 mm3.15360~16720 projections, 512
readout points per projection were collected. With the SAR limitation, a flip angle of 50~60
degree was used. Adiabatic T2 preparation scheme was used with a T2-prep time of 40
ms [3]. SPIR (Spectral Presaturation Inversion Recovery) was used to suppress the fat
signal.
Results: The acquisition time of whole-heart MRA ranged between 9 to 13 min. Both left
and right coronary arteries
from
four volunteers
were
successfully visualized. Figure
1 is a multiplanar reformatted
(MPR)
LAD,
image
RCA,
Excellent
blood
and
contrast
and
observed
delineating
LCX.
between
myocardium
and
no
is
banding
artifacts are present. Average
Figure 1. MPR images acquired with ultrashort-TR SSFP
VIPR from two volunteers. Note the good delineation of
coronary arteries.
image quality scores were
3.10 with a SD of 0.41.
Conclusion: With non-slab-selective excitation for VIPR, TR could be decreased to 3.0
ms as compared to 4.0 ms usually required for slab-selective SSFP. As a result, no
apparent image artifacts were observed in the region of interest and excellent delineation
of coronary arteries were obtained in our volunteer studies.
References: [1] Bi X, et al. JMRI 2005;22:206-212
[2] Shea SM, et al. JMRI 2002;15:597-602
[3] Nezafat R, et al. MRM 2006;55:858-864
MR-Angioclub East Lansing 2009
91
9.5 Cardiac Imaging: Methods for the Detection of
Intramyocardial Fat
James W Goldfarb PhD
Saint Francis Hospital, Roslyn, NY and Stony Brook University, Stony Brook, NY
Purpose: Detection and characterization of myocardial infarction has been shown to be
critical for both the right and left ventricles. While the primary technique for detection of
fibrosis and necrosis is late gadolinium-enhanced (LGE) imaging, chronic myocardial
infarction is often associated with fatty replacement. In this presentation, we will discuss
the available methods for detection of intramyocardial fat along with their advantages and
disadvantages. Examples from each technique will be presented.
Methods and Results: Techniques considered include T1 weighted imaging (native T1
weighting or use of inversion or saturation pulses)(1), bSSFP CINE, opposed phased
imaging, water-fat separation (2), chemical fat saturation preparation pulses, and late
gadolinium-enhanced water-fat separated imaging(3). Accurate sizing of myocardial fat
deposition, may only be achievable using “in-phase” MR images due to the loss of signal
at water-fat tissue boundaries in opposed phase images. Opposed phase images may
offer increased sensitivity for small fat deposits due to this “artifact” at tissue boundaries.
Reliability, ease of use, accuracy, sensitivity, specificity, availability and speed are all
important issues for widespread clinical usage.
Due to its simplicity, precontrast CT
imaging (4) is a quick, high resolution, accurate technique, but does not have the
specificity for fat detection as does MR imaging.
Though CT techniques are under
development, MR imaging currently has better methods for contrast-enhanced infarct
detection and the quantitative assessment of ventricular function.
Conclusions: MR imaging has many techniques for the detection of intramyocardial fat.
Though water-fat separation and CT imaging provide the best images, other techniques
may be relevant if detection rather than accurate sizing is needed.
1.
2.
3.
4.
92
Goldfarb J, Arnold S, Roth M, McLaughlin J, Reichek N. Magnetic Resonance
Shows Fatty Replacement of Left Ventricular Myocardium after Myocardial
Infarction. Circulation 2005; 112:II-470.
Reeder SB, Markl M, Yu H, Hellinger JC, Herfkens RJ, Pelc NJ. Cardiac CINE
imaging with IDEAL water-fat separation and steady-state free precession. J
Magn Reson Imaging 2005; 22:44-52.
Goldfarb JW. Fat-water separated delayed hyperenhanced myocardial infarct
imaging. Magn Reson Med 2008; 60:503-509.
Zafar HM, Litt HI, Torigian DA. CT imaging features and frequency of left
ventricular myocardial fat in patients with CT findings of chronic left ventricular
myocardial infarction. Clin Radiol 2008; 63:256-262.
MR-Angioclub East Lansing 2009
9.6 3D spiral high-resolution late gadolinium enhancement
Dana C. Peters, Peng Hu, Reza Nezafat, Warren J. Manning
Beth Israel Deaconess Medical Center, Dept of Medicine, Harvard Medical School
Purpose: Late gadolinium enhancement (LGE) is peerless among imaging modalities for
visualizing scar/fibrosis in the heart. High resolution LGE imaging is an important goal to
improve visualization of small regions of scar, such as papillary muscle scar, complex scar
related to ventricular tachycardia, and RF ablation scar. Spiral imaging with spectral
spatial pulses is attractive for high-resolution LGE because it provides excellent fatsuppression, high SNR efficiency, and good motion ghosting properties. We compared a
high resolution 3D spiral LGE sequence with our current 3D Cartesian protocol.
Methods: Four subjects were imaged 20-30 minutes after injection of 0.2mmol/kg GdDTPA. Spiral scan parameters include 3D, ecg-gating, and NAV-gating, water-selective
RF pulses, 1.3 x 1.3 x 5 mm3 spatial resolution, imaging time = 60 heart-beats (neglecting
dead time), 14 interleaves, 14 ms acquisition window, and 38 slice-encodings.
The
sequence acquires 5 TRs per heart-beat, using a kz-centric acquisition, TRTEq=
23ms/3.6ms/40°.
Results: Scans were diagnostic in all subjects.
Figure 1 compares the spiral LGE
sequence with the Cartesian sequences. The image quality and sharpness is similar to
the 3D LGE sequence which took 3x longer. Conclusions: The high resolution 3D LGE
spiral approach is promising for visualizing scar in the heart.
A
B
C
Figure 1: A) Low resolution 3D LGE (2 x 2 x 5 mm) acquired in 2 minutes. B) 3D
Cartesian LGE with 1.2 x 1.2 x 5 mm resolution, in 6 minutes. C) Spiral 3D LGE with 1.3 x
1.3 x 5 mm spatial resolution, acquired in 2 minute. All times assume 50% NAVefficiency.
MR-Angioclub East Lansing 2009
93
9.7 Temporal Filtering for Sliding Window Time-resolved
Angiography: Beyond Density Compensation Solutions
Grabow B, Wu H, Block WF, Samsonov AA
University of Wisconsin–Madison, Madison, WI USA
Introduction: Methods such as “tornado filtering” [1-2] and KWIC [3] exploit the variable
sampling density in k-space trajectories such as radial acquisitions to produce a temporal
or parametric series of images from a single acquisition. A gridding reconstruction utilizes
a density compensation function (DCF) to limit the aperture of oversampled spatial
frequencies to a specific time point or contrast-weighting. The sampling aperture widens
with the k-space radius to limit the effects of undersampling artefacts as the sampling
density decreases. The filters are poorly compatible with iterative reconstruction because
of: 1) suboptimal SNR; 2) mixing of temporal information, especially in smaller objects.
We present a method to design such filters for iterative methods that is not based on using
a DCF.
Methods: The DCF in [1-2] is designed to minimize errors in the spatial point spread
function (psf). As the k-space radius increases and data become undersampled, these
approaches weight data across the widening temporal footprint [4] evenly, degrading
temporal fidelity. We instead use an iterative reconstruction that derives an image
estimate
fi
that
fWEf-s
i = argmin
fi
(
designed function
fits
1/2
ii
Wi
the
(
acquired
) 2)
data
s
and
the
encoding
matrix
E
:
. The minimization is weighted with a specially
that emphasizes the current time frame over all spatial frequencies
while minimizing the influence of adjacent temporal data and simultaneously providing
optimized image SNR relative to conventional DCF. The implementation provide benefits
from parallel imaging by including coil sensitivity information in E .
Results and Discussion: The figure
to the right shows four simulated
contrast-enhancing vessels over a
background of linearly enhancing
peripheral tissue at a midpoint in scan
where the peak vessel enhancement
is at the distal (inferior) point of the
image.
There is no horizontal
variation
between
the
vessel
enhancement patterns. Notice that
that with the traditional filter, the
larger
vessels
show
variation
vertically across the vessel but the
smallest vessel shows no change
across the image.
The iterative
methods show a much closer
depiction of the actual enhancement
for all widths of blood vessel. The
iterative method gives more flexibility in the design of the temporal filter, as the
minimization process essentially corrects errors due to variations in sampling density.
There is a cost in noise amplification, which ultimately limits increases in temporal fidelity.
1) Barger et al MRM 2002 2) Liu et al IEEE-TMI 2006 3) Song et al, MRM 2000 4) Mostardi and
Riederer et al, MRM, 2009.
94
MR-Angioclub East Lansing 2009
10.1 Angiographic and Hemodynamic Assessment of the Hepatic
Vasculature in Portal Venous Hypertension
using High Resolution PC VIPR
Kevin M. Johnson, Oliver Wieben, Chris Francois, Ben Landgraf, Scott B. Reeder
Departments of Medical Physics and Radiology, University of Wisconsin, Madison, WI, USA
Purpose: Several investigators have proposed the use of non-invasive flow measurement
techniques such as PC MR [1,2] for the evaluation of hepatic flow patterns and rates in
patients with portal venous hypertension; however, with existing techniques evaluation of
the entire hepatic vasculature can be challenging due to long scan times and limited
coverage. In this work, we present initial results using an accelerated 3D radial sequence
for whole abdomen CINE, 3D PC (PC VIPR).
Methods: Both normal volunteers and patients with liver disease were scanned on a 3T
MR scanner (MR750, GE Healthcare) using a 32-channel torso array coil. PC VIPR was
performed
using
adaptive
bellows
respiratory gating with 50% efficiency,
1.25mm isotropic resolution, 32 x 32 x
24cm3 FOV, Venc = 25cm/s, retrospective
cardiac gating, for a total scan time of
approximately 10minutes.
Results:
Figure
1,
shows
example
anatomical images derived using PC
VIPR. Both arteries and veins are well
visualized with high resolution. In Figure 2,
vortical flow can be observed in a
representative visualization of the portal
vein.
Figure 1. Example PC VIPR anatomical
images of the hepatic artery (left) and portal
vein (right). Top images show axial limited
MIPs while bottom images show coronal
limited MIPs.
Conclusion: Whole abdomen PC VIPR allows
anatomical and hemodynamic visualization of
the entire liver vasculature, the splenic and
renal vasculature, making it a very promising for
non-contrast enhanced evaluation of the hepatic
vasculature in portal venous hypertension.
References: 1. Yzet et al. EJR 08 In Press.
2.Stankovic et al. ISMRM 09 #3856
Figure 2. Example visualization on the
flow in the portal vein, demonstrating
laminar mixing from the splenic vein
(blue) and SMV (red)
MR-Angioclub East Lansing 2009
95
10.2 Renal MR angiography: multicenter intraindividual
comparison of gadobenate dimeglumine and gadofosveset
trisodium
G Schneider1, M Pasowicz2, J Vymazal3, Z Seidl4, M Aschauer5,
6
7
8
9
M Konopka , D Bilecen , R Iezzi , C Ballarati
1. Homburg University Hospital, Homburg/Saar, Germany; 2. John Paul II Hospital, Krakow, Poland; 3.
Na Homolce Hospital, Prague, Czech Republic; 4. Neurologicka Klinika, Prague, Czech Republic; 5.
University of Graz, Graz, Austria; 6. NZOZ Slaskie Centrum Diagnostyki Obrazowej, Katowice, Poland;
7. University of Basel, Basel, Switzerland; 8. Università G. D'Annunzio, Chieti, Italy; 9. Hospital
Valduce, Como, Italy
Purpose: To prospectively compare gadobenate dimeglumine and gadofosveset trisodium
for contrast-enhanced MR angiography (CE-MRA) of the renal arteries.
Methods: 38 subjects with renal vascular disease underwent a first CE-MRA exam with
0.1 mmol/kg gadobenate dimeglumine followed 3-12 days later by a second exam with
0.03 mmol/kg gadofosveset. For both agents, identical T1w SPGR sequences were used
to acquire first-pass (FP) coronal images during breath-hold. In 16/38 patients additional
steady-state (SS) sagittal and axial images were acquired with gadofosveset. DSA was
performed in 34 patients. Images were evaluated by 3 blinded readers in terms of
sensitivity, specificity, accuracy, and positive and negative predictive values (PPV and
NPV) for detection of significant (≥51%) renal artery stenosis compared to DSA. Findings
were compared using McNemar and Wald tests; assessments of FP diagnostic preference
were evaluated using the Wilcoxon Signed Rank test; and reader agreement (kappa [])
was determined. A full safety evaluation was performed.
Results: Gadobenate dimeglumine was consistently superior to gadofosveset (sensitivity:
76-87% vs 68-76%; specificity: 92-99% vs 91-94%; accuracy: 89-96% vs 86-90%; PPV:
70-94% vs 65-76%; NPV: 94-97% vs 92-94%). Significant superiority for gadobenate
dimeglumine was noted by 2 readers for specificity (P≤0.02), accuracy (P≤0.005), and
PPV (P≤0.018). SS images provided no additional benefit for gadofosveset. Three-reader
agreement was excellent (=0.776-0.855). Readers 1, 2, and 3 preferred gadobenate
dimeglumine in 11, 17, and 13 patients and gadofosveset in 5, 4, and 5 patients; no
preference was expressed for the remaining subjects. Adverse events were reported for
2/38 [5.3%] with gadofosveset but 0/39 [0%] with gadobenate dimeglumine.
Conclusion: Better reader preference and diagnostic performance was obtained with 0.1
mmol/kg gadobenate dimeglumine vs 0.03 mmol/kg of the intravascular blood pool agent
gadofosveset for CE-MRA of the renal arteries.
96
MR-Angioclub East Lansing 2009
10.3 FINESS (Flow Inversion-prepared Non-contrast Enhancement
in the Steady State): a novel technique for non-contrast renal MRA
Manojkumar Saranathan1, Ersin Bayram1, and James Glockner2
GE Healthcare, Rochester MN & 2Dept. of Radiology, Mayo Clinic, Rochester MN
1
Purpose: To evaluate a novel balanced SSFP-Dixon technique for non-contrast MRA of
the renal vasculature in a single breath-hold
Methods: A 3D dual-echo bipolar readout balanced SSFP pulse sequence with a robust
two-point Dixon algorithm1 for fat-water separation was developed. This enabled use of
radial fan-beam segmentation in ky-kz. Each fan-beam was acquired after a slab selective
180° pulse that effected venous and background suppression2. The radial fan-beam
scheme enabled us to acquire the 3D volume in a single 20-22s breath-hold, eliminating
the need for respiratory triggering, which is sub-optimal in some patients. Parameters: 70°
flip, TR/TE1/TE2 6.2/1.4/2.8 ms, 256x224 matrix, 35 cm FOV, 2 mm thick, 32-40 slices,
TI=900ms. All subjects were imaged on a GE Excite system with a 8-channel torso array
coil under an IRB-approved protocol.
Results and Discussion: Fig. 1 Right renal artery stenosis depicted using a 22s BH
FINESS volume rendering (top) and confirmed on conventional x-ray angiography
(bottom). Fig. 2. Right renal artery stenosis depicted using a 23s BH FINESS sequence
(top) compared to MR contrast enhanced angiography (bottom).
FINESS afforded
excellent visualization of stenoses in a short 20-22s breathhold with marked insensitivity to
Bo inhomogeneities. Preliminary results are encouraging, and suggest that this technique
may have clinical utility for rapid, breath-held non-contrast MRA.
Figure 1
Figure 2
Refs: [1] Ma et al. MRM. 52:415-419 (2004) [2] Takei et al. Proc ISMRM, p3420 (2008)
MR-Angioclub East Lansing 2009
97
10.4 Magnetic Resonance Angiography of the skin for perforatorbased autologous breast reconstruction
I
Tiffany Newman, MD , Julie Vasile MD II, Joshua Levine, MD II, David Greenspun, MD II, MSc, Robert J.
Allen M.D., A.P.M.C, F.A.C.S II, Minh-Tam Chao B.S.R.T(R) MR I, Martin R. Prince, MD, PhD I
I
Radiology, Weill Cornell Imaging at New York Presbyterian, New York, NY, United States, II The
Center for Microsurgical Breast Reconstruction, New York, NY, United States.
Purpose: Autologous breast
reconstruction after mastectomy using
abdominal and gluteal perforator artery
flaps has gained popularity due to
preservation of the donor site muscle and
function. We evaluate skin MRA accuracy
for the preoperative mapping of perforating
arteries and flap volume estimation.
Methods – Pre-operative MRA on 25
consecutive patients undergoing perforator
artery based autologous breast
reconstruction was performed at 1.5 Tesla
using axial 3D LAVA of the skin overlying
abdominal and/or gluteal regions with 20ml
gadobenate. Perforator artery size and
coordinates relative to umbilicus or top of
gluteal crease on 3D MRA were
compared to findings at surgery.
Reconstructed breast volume estimates
from volume rendered MRA images were
also compared to weights at harvesting.
Results – One hundred twenty-five
perforator arteries were found at surgery to
be located within 1cm of the coordinates
measured on MRA and were surgically
verified to be suitable for flap perfusion.
Surgery verified the arterial course and
caliber through the rectus and gluteal
muscles visualized on MRA in 47 of 48
arteries. Volume rendering of 3D MRA
accurately predicted breast reconstruction
volumes.
Figures: Preoperative MRA of the
skin of the abdomen (top 2 images)
and gluteal (bottom 2 images)
regions in different patients
undergoing perforator based
breast reconstruction
Conclusion - This study of 25 patients undergoing breast reconstruction shows that 1.5T
MRA safely and accurately identifies precise location measurements and vascular
anatomy of abdominal wall and gluteal perforator arteries for guiding autologous flap
harvesting.
98
MR-Angioclub East Lansing 2009
10.5 Time-SLIP versus DSA in Patients with Renal Artery Stenosis
1
Isabelle Parienty , Faïza Admiraal-Behloul2 , Francis Jouniaux1 , Guy Rostoker3
2
Centre d’Imagerie du bois de verrière, Antony, France. Toshiba Medical systems,
Zoetermeer, the Netherlands. 3Service de Néphrologie, Centre Hospitalier Claude Galien,
Paris, France.
1
Purpose: To compare the findings in non-contrast enhanced MRA using the Time Spatial
Labeling Inversion Pulse (Time-SLIP) technique [1]
to those of Digital Subtraction Angiography (DSA) in
patient with significant renal artery stenosis (>60%).
Methods: Thirty Patients (12 man, mean age 72 ± 11
y) with renal insufficiency and suspected renal artery
stenosis were explored. Time-SLIP images were
obtained on a 1.5T MRI system (Vantage, TOSHIBA,
Tokyo), using an SSFP sequence with respiratorygating and the following parameters: TR=5.2 ms,
TI=1200 to 1800 ms, TE=2.6 ms FA 120, FOV 35x35
cm, Matrix 256X256, Speeder Factor 2 , 35 slices,
Fat Sat on, and time= 4.30 min. The image quality
was visually assessed by an experienced radiologist
and scored as: poor: no contrast in the distal
branches but interpretable, moderate: moderate
contrast in the distal branches and good: strong
contrast from the ostium to the segmental arteries.
the
degree
of
stenosis
was
estimated
using
measurement tools on a post-processing workstation
(GPW, Toshiba Medical Systems). A degree of
stenosis of 60% or higher was considered as
significant. In all patients with a significant stenosis, a
DSA was performed.
Figure 1 :. (a) Time-SLIP image
(scored as moderate) revealed an
ostial stenosis on the right main
renal artery estimated at 60%,
(see arrow). (b) DSA confirming
the findings.
Results: The Time SLIP images were scored as good in 24 patients, moderate in 5
patients and poor in 1 patient. We detected 18 significant stenosis in 17 patients. In all 17
patients the DSA confirmed our findings . Figure 1 shows a representative case.
Conclusion: Time-SLIP is a reliable technique for renal artery stenosis screening and
diagnostic in patients with moderate to severe renal dysfunction. A quantitative
comparative study where different degrees of stenosis, from 20% to total occlusion, is ongoing.
Reference: 1. D. Utsunomiya et al. Circ J 2008; 72: 1627–1630 .
MR-Angioclub East Lansing 2009
99
10.6 Simultaneous Renal Angiography and Perfusion Measurement
Using Time-Resolved MRA
1
Katherine L. Wright , Raymond F. Muzic1,2, Nicole Seiberlich2, Yu-Hua Fang1, Stephen R. Yutzy1,
Mark A. Griswold1,2, Vikas Gulani2
1
Dept. of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
2
Dept. of Radiology, University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH
Purpose Time resolved angiography with stochastic trajectories (TWIST), a 3D view
sharing technique, with GRAPPA is used to achieve sufficient spatial and temporal
resolution to simultaneously acquire an angiography exam and perfusion measurement
with a single contrast dose.
Methods Two normal subjects underwent a TWIST renal MRA
exam at 3T (Magnetom Verio, Siemens, Erlangen, Germany)
according to local IRB protocol. Imaging parameters: FLASH; 0.05
mmol/kg of Gd-BOPTA (Multihance; Bracco Diagnostics Inc.,
Princeton, NJ), pA=0.2, pB=0.4 [1], TR/TE/FA=2.5ms/0.95ms/21°,
TA=3.7s/volume, Res=1.4x1.4x1.5mm3, FOV=350x284x108mm3,
GRAPPA R=3. Contrast dynamics were evaluated for manually
segmented tissues using a two-compartment model [2].
Results A representative MIP from a single angiographic frame for
Figure 1. MIP from
a single time frame.
an asymptomatic volunteer is shown in Fig 1. This confirms the
feasibility of using this method for renal angiography. Fig 2 depicts time course data and
model fits for the same volunteer. The rate constant Ktrans was measured as 4.24 (cortex)
and 0.14 (medulla), yielding perfusion measurements of 6.63 and 0.22 ml g-1 min-1,
respectively.
Conclusions The initial feasibility of this method has been demonstrated. This could have
significant clinical benefit, as it could potentially assess perfusion deficits created by renal
artery stenosis with a gadolinium dose several times smaller than previously needed
forperfusion and angiography measurements
[3].
References [1] Song, et al. MRM 2009; 61:12428.
Figure 2. Time course data from ROIs
and model fits.
[2] Tofts, et. al. JMRI 1999; 10:223-32.
[3]. Michaely et al. Radiol 2006; 238:586-96.
Figure 2. Time course data from ROIs and model fits
100
MR-Angioclub East Lansing 2009
10.7 Assessing Kidney Perfusion using Arterial Spin Labeling and
Radial Acquisition for Rapid Characterization of Inflow Dynamics
1
N. Artz , K. Johnson1, Y. Huang1, E. Sadowski2, S. Fain1,2
Medical Physics and 2Radiology, University of Wisconsin, Madison, WI, United States
1
Purpose: Quantifying arterial spin labeling (ASL) perfusion measurements, especially in
diseased subjects who may demonstrate a wide range of blood flows, benefits from data
at multiple delay times1. This research aims to efficiently acquire data at multiple delay
times using a radial approach.
Methods: FAIR ASL was performed on a healthy volunteer in a 1.5 T MR scanner. From
0.1 to 2.1 seconds following inversion, a 2D radial balanced SSFP readout acquired
unique projections with the following parameters: slice orientation = oblique-sagittal, slice
thickness = 8 mm, TR/TE/flip = 5.4/2.7ms/30°, BW = 250.33 kHz, FOV = 34 cm, and
matrix = 128 x 128. Control (non-selective inversion) and tag (selective inversion) were
alternated until 55 pairs were acquired in 11 minutes. The unique radial lines from all
related inversions were combined and partitioned into twenty time frames, each with a
temporal window of 55 ms. A cortical ROI was
Tag
Control
Difference
used for signal analysis.
Results:
The perfusion-weighted difference
image (at a delay of 1.4s) suffers from poor SNR
[Fig 1]. However, the cortical ROI signal vs. time Figure 1 Sagittal control, tag, and
difference image at delay time of 1.4
frame curve demonstrates the correct trend for
sec.
perfusion with the tag showing higher signal than
the control [Fig 2]. Further pulse sequence development could reduce the TR, BW and
off-resonance effects.
HYPR related reconstruction
Cortical ROI Signal vs. Time during Blood Inflow
techniques may improve SNR and further reduce scan
Conclusion: Preliminary results suggest that a radial
approach may efficiently acquire perfusion data at
Tag
Signal (a.u.)
time.
Time Frames During Inflow
Control
Null
multiple delay times in a clinically feasible scan time.
0.6
1.1
1.6
Delay Time (sec)
1
References: Parkes et al. Magn Reson Med. 2002; 48(1):
27-41.
Figure 2 Tag and control
inversion recovery signal
vs time for a cortical ROI
during the blood inflow
period. Scan time: 11
minutes
MR-Angioclub East Lansing 2009
101
10.8 Imaging Capabilities for Real-time Guidance and Verification
of Transcatheter Arterial Chemoembolization (TACE) Procedures
WF Block, EK Brodsky, E Bultman, H Wu, A Samsonov, SB Reeder, O Unal
University of Wisconsin–Madison, Madison, WI USA
Introduction: Transcatheter arterial chemoembolization (TACE) provides targeted
delivery of chemotherapy and embolizing agents to liver lesions using X-ray fluoroscopic
guidance. The inability of X-ray to adequately visualize the tumor and the tumor’s vascular
supply can cause mistreatment, and thus potentially unnecessary damage to healthy liver
tissue or incomplete tumor treatment. Our long term aim is to develop an integrated MR
real-time imaging system which can visualize liver tumors and adjacent vasculature
through dynamic 3D imaging while also verifying the treatment area. Here we describe
milestones reached as we move towards system integration.
Methods: MR-guided TACE will require catheter tracking, tumor localization, vascular
visualization, and treatment validation. Tumor localization has previously been described
at the Angio Club. Examining the vascular territory distal to the catheter position is
performed via an intra-arterial contrast injection with a 3D stack of stars sequence which
can be oriented obliquely in relation to the hepatic vasculature. Verification of the
treatment region in TACE is normally performed with a followup CT that is sensitive to the
iodine in the ethiodized oil that is preferentially taken up in the treated region.
We
demonstrate how treatment can be verified immediately by detecting the oil in ethiodized
oil in the fat images generated by a single breath-hold MR IDEAL scan.
The imaging tasks for this procedure require both real-time, near real-time, and
non-real-time capabilities with multiple contrast mechanisms. We are porting catheter
tracking (described by Brodsky and Unal at this meeting),
vascular imaging, and
treatment verification using the virtual scanner capabilities provided by the
RtHawk
imaging platform (HeartVista, Palo Alto, CA) onto a GE 1.5T HDx system. In the RtHawk
paradigm, users exert imaging control through a stand-alone processor which modifies a
very simple and flexible imaging sequence.
Results and Discussion: Oblique time-resolved CE-MRA of the hepatic vein is shown at
2 s intervals using a stack of stars sequence in Figure a, with time frames advancing left to
right. Using an MR IDEAL fat image to validate
the treatment region after TACE is shown in Fig.
c, where the hyperintense region corresponds to
the hyperintense region shown in the CT
followup (Fig b).
102
MR-Angioclub East Lansing 2009
11.1 Non-Contrast-Enhanced MR Identification of DVT
M Louis Lauzon, Houman Mahallati, Linda Andersen, and Richard Frayne
Seaman Family MR Research Centre, University of Calgary, Calgary, AB
Purpose: Pulmonary embolism is the 3rd most common cause of death in US hospitals.
Common sources of emboli come from pelvic or lower extremity deep vein thrombosis
(DVT). We hypothesize that identifying thigh-to-calf DVT is clinically important, so we are
investigating high-resolution non-contrast-enhanced (NCE) thrombus MR imaging.
Methods: We adapted a 3D direct thrombus imaging sequence1,2 by adding a flowsuppressing bipolar gradient. All images were acquired on a 3.0T scanner (Signa VH/i; GE
Healthcare) using a 4-channel torso phased-array coil, coronal orientation, 15º flip angle,
40 cm FOV, 9.2 ms TR, 320×320 in-plane matrix, 90-120 slices for the thigh and calf/knee
regions, 2.0 mm slice thickness, velocity suppression of 20 cm/s and above, TE of 2.0/5.4
ms without/with flow suppression, and one signal average, leading to scan times of 4.5 to
6.0 minutes per region. The internal review board approved the study protocol, and each
subject gave written informed consent before imaging. Certified body radiologists
interpreted the MR thrombus images to determine the presence of clots.
Results: The figure below is from a patient with known DVT in the superficial femoral vein.
The conspicuity of clot in a given slice is similar without (A) or with (B) flow suppression,
but the maximum intensity projection (MIP) image provides significantly better thrombus
identification with (D) than without (C) flow suppression.
Conclusion: The visualization of lower extremity DVT using high resolution NCE MR
imaging is feasible, and shows great clinical promise and potential.
A
B
C
D
References: 1Moody. Lancet 1997,350:1073. 2Moody. J Thromb Haemost 2003,1:1403.
MR-Angioclub East Lansing 2009
103
11.2 Susceptibility mapping as a means to image veins
1,2, 3
2
E. Mark Haacke, PhD
and Jin Tang
1. Wayne State University, Detroit, MI, USA
2. McMaster University, Hamilton, Ontario, Canada
3. The MRI Institute for Biomedical Research, Detroit, MI, USA
Introduction: The ability to image oxygen saturation is tantamount to being able to follow
tissue function in the brain. This is important for monitoring patients with stroke, multiple
sclerosis and even tumors. Recently, a new approach to susceptibility weighted imaging
(SWI) called susceptibility mapping (1,2) has been proposed. We refer to this new
approach as SWIM. This method offers the ability to monitor susceptibility and correlate it
with oxygen saturation, the focus of this work.
Materials and methods: In order to extract the susceptibility, the phase from a gradient
echo sequence is required. In this study, we use the high pass filtered phase image from
an SWI scan. This phase image is then Fourier transformed back to k-space, filtered with
an inverse regularized filter, and then forward transformed back to the image domain. The
resulting image is now a susceptibility map of the veins and the tissue in the brain. High
resolution SWI data with isotropic resolution of 0.5mm at 4T were collected. Three echo
times were used: 11.6ms, 15ms and 19.2ms.
Results: The susceptibility maps for all three echoes give similar results. Although the
agreement is not perfect, the susceptibility values appear to be independent of echo time
as they should be. They also predict that the oxygen saturation is roughly 0.5ppm in SI
units which is consistent with expected in vivo values.
Discussion and Conclusion: The ability to map veins throughout the entire brain and to
extract venous oxygen saturation is an important adjunct to MRI methodology in the study
of diseases that affect the brain’s hemodynamics. We have presented here an approach
that promises the ability to provide this information for veins much larger than a voxel. The
method is fast and simple and can be applied to any SWI data with phase.
References: 1. Deville G, Bernier M, Delrieux J. NMR multiple echoes observed in solid
3He. Physical Review B 1979;19:5666-5688. 2. Salomir R, Senneville BD, Moonen CT. A
Fast Calculation Method for Magnetic Field In homogeneity due to an Arbitrary Distribution
of Bulk Susceptibility. Concepts in MR Part B (MR Eng) 2003;198:26-34.
104
MR-Angioclub East Lansing 2009
11.3 Modified CAPR MRA: Improved Imaging of the Arterial and
Venous Phases
P. M. Mostardi, C. R. Haider, N. G. Campeau, S. J. Riederer
Department of Radiology, Mayo Clinic, Rochester, MN 55905 USA
Purpose – Acquisition parameters for time-resolved MRA are typically constant
throughout a scan. We hypothesize that the image quality of the arterial and venous
phases of an intracranial MRA can be substantially improved by dynamically changing
acquisition parameters, optimizing spatial resolutions and frame rates as dictated by the
relevant physiology. Tailoring acquisition parameters to match the specific demands of
temporal and spatial resolution of the vascular region of interest is essential in the
development of a Comprehensive Neurovascular Exam (CNVE).
Methods – The CAPR pulse sequence [1] was modified to allow dynamic change of
matrix size, SENSE acceleration, k-space center size, and view sharing factor at a
specified time during the acquisition. Volunteer studies were performed in which a viewshared time-resolved view order (2.25 sec/frame, 0.86 x
1.38 x 2.00 mm3) was executed to capture the arterial
A
phase, and then by seamlessly switching to a high spatial
A
resolution single-phase view order (25 sec, 0.86 x 0.86 x
1.00 mm3) a venogram was acquired. The reconstruction
was modified to automatically account for the change of
CAPR acquisition parameters.
Results – Fig. 1 shows images from a single timeresolved MRA scan in which several distinct arterial
frames were captured as well as a venogram. The arterial
B
images are optimized for high temporal resolution,
whereas the venous phase accentuates spatial resolution
and SNR.
Conclusion – By dynamically changing view order
parameters, the intracranial arterial and venous systems
can be imaged with high quality in a single scan. Further
enhancements to the CNVE will allow modification of the
FOV from a large aorta-based FOV to one limited to the
brain.
References – [1] Haider CR, MRM 60:3(2008).
Fig. 1. Sagittal MIPs of one
arterial time frame (A) and
the subsequent venogram
(B) both taken from the
modified acquisition.
MR-Angioclub East Lansing 2009
105
11.4 CAMERA: Contrast-enhanced Angiography with Multi-Echo
and RAdial k-space
1
Hyun Jeong , Christopher Eddleman2, Saurabh Shah3, Guilherme Dabus4, Timothy J. Carroll1,4
1
Biomed. Engineering, 2Neurosurgery, and 4Radiology, Northwestern University, Chicago, IL
3
Siemens Medical Solutions, Chicago, IL
Purpose: A new fast acquisition technique for 4D contrast-enhanced MRA is introduced.
It allows a shorter temporal footprint, which samples the contrast bolus more frequently
than previous techniques for more accurate dynamic information.
Methods: 3D radial “stack-of-stars” k-space is acquired with multiple echoes in partition
direction (kz), similar to in a centric segmented EPI (1), which shortens the temporal
footprint by up to 60%. Intracranial MRA images of healthy volunteers and AVM patients
were acquired on a Siemens 3T Trio, using CAMERA and the previously developed radial
sequence. Sliding window reconstruction (2, 3) was used to increase apparent frame rate.
AVM patient images were correlated with X-Ray angiography.
Results and Discussion: The images acquired with CAMERA (Nechoes=4) resulted in
significantly higher CNR values than single-echo acquisition. Flip angle optimization is a
known problem with high field CE-MRA, due to SAR. With our multi-echo approach, we
are able to increase the TR while reducing temporal footprint. The longer TR used in
multi-echo acquisitions allows more flexibility in the choice of flip angles for optimal
contrast weighting of the FLASH sequence. Figure 1 shows arterial, nidal, and venous
phases of the time-resolved MRA of an AVM, and a corresponding X-Ray angiogram.
Figure 1: 4D MRA of an AVM using
CAMERA. NRO=192, Nproj=192,
Npartition=30 (10% OS), Nechoes=4,
FA=45°, TR=6ms, TE=1.5, 2.5, 3.5,
4.5 ms, FOV=220x220x3 mm.
Conclusion: A novel fast 4D MRA technique based
on radial k-space and multi-echo has been developed.
Temporal footprint=9s. Arrows
indicate early drainage.
References:
(1)Beck G et al. MRM, 2001 (2)Riederer et al. MRM, 1988 (3)Cashen et al. MRM, 2007
106
MR-Angioclub East Lansing 2009
11.5 Time-Resolved, Vessel-Selective, Cerebral Angiography Using
Arterial Spin Labelling
Philip M. Robson1, Weiying Dai1, Ajit Shankaranarayanan2, Neil M. Rofsky1, David C. Alsop1
Beth Israel Deaconess Medical Center, Boston, MA, 2Global Applied Science Laboratory, GE
Healthcare, Menlo Park, CA
1
PURPOSE: X-ray Digital Subtraction Angiography (DSA) is the conventional, yet highly
invasive method for morphologic and haemodynamic assessment of the cerebral
vasculature [1-2]. The purpose of this work is to assess an Arterial Spin Labelling (ASL)
MRI method capable of time-resolved inflow visualisation and vessel-selective labelling of
feeding vessels, similarly to X-ray DSA, without the use of contrast material.
METHODS: Temporally resolved image frames were obtained by varying the duration of
labelling before commencing imaging.
A modification of pulsed-continuous labelling
(pCASL) incorporating additional gradient pulses, allowed labelling to be targeted to the
ICA [3].
Labelled signal was acquired whilst in the lumen using a bSSFP read-out.
Background suppression was used to provide robust subtraction images. Imaging time
was 1.5 min. Quantitative measurements included arterial transit time to vessel segments,
residual contralateral signal, and labelling efficiency for vessel-selective labelling.
RESULTS: MIP images with temporal resolution of 200 ms (Figure), and high selectivity
and efficiency were obtained (5% contralateral signal, 73% relative efficiency). Normal
variations of the vasculature were identifiable by ASL-MRA in our cohort of 6 healthy
volunteers.
CONCLUSION:
This non-invasive ASL technique provides high quality angiographic
images of the vasculature, able to show haemo-dynamic function; it may be of particular
importance for assessing conditions exhibiting altered or differen-tial arterial transit and
collateral flow pathways.
Left to right: labelling durations of 600, 800, 1000, and 2000 ms
REFERENCES:
1) Borisch I et al., AJNR 2003;24(6):1117-1122; 2) Grzyska U et al.,
Neuroradiology1990;32(4):296-299; 3) Dai W et al., Proc. ISMRM 2008:184.
MR-Angioclub East Lansing 2009
107
11.6 Quantifying L umen G eometry from R outine C arotid C E MR A
1
2
3
Payam B. Bijari , Luca Antiga, PhD , Bruce Wasserman, MD , David A. Steinman, PhD1
1
Biomedical Simulation Laboratory, University of Toronto, Toronto, ON, Canada
2
Bioengineering Department, Mario Negri Institute for Pharmacological Research, Ranica, Italy
3
Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
Purpose: Recent work has demonstrated a strong relationship between the exposure of
the normal carotid bifurcation to disturbed flow and its three-dimensional (3D) geometry
[1]. Insofar as 3D contrast-enhanced MRA (CEMRA) is now almost routine, it becomes
possible to consider large-scale studies of local risk factors for atherosclerosis. Before this
happens, however, it is important to assess the repeatability of these geometric
measurements from routine CEMRA.
Methods: As part of the ARIC Carotid MRI study’s repeatability protocol, the left or right
carotid bifurcations of 60 participants were scanned twice at a mean(±SD) interval of
65±36 days. 3D contrast-enhanced MR angiograms were acquired at 1.5T using bilateral
phased-array RF surface coils at the following spatial resolutions: a coronal slab
partitioned into 56 2-mm thick slices, zero-padded to 1 mm; and 200-mm2 field-of-view
acquired at 256x160, zero-padded to 512x512. Our Vascular Modeling ToolKit
(www.vmtk.org) was used to segment the lumen surface rapidly (typically < 5 minutes) and
with minimal operator interaction, from which a number of geometric parameters were
extracted automatically as described by Lee et al. [1].
Results: Of the 60 pairs, 9 were excluded because one or both carotid bifurcations
resisted segmentation by the rapid protocol. Intra-class
Parameter
ICC
correlation coefficients (ICC), tabulated to the right and
Angle
0.80
based on analysis of the remaining pairs, revealed
Planarity
0.54
excellent repeatability for three of the four geometric
Area Ratio
0.93
factors despite the relatively coarse CEMRA spatial
Tortuosity
0.89
resolutions, possible repositioning effects, and
purposefully rapid segmentation protocol. These results
were found to be insensitive to operator’s grading of the
segmented lumen surface as excellent (37%), adequate (41%) or poor (22%).
Interestingly, area ratio and tortuosity, the geometric parameters that together best predict
the burden of disturbed flow [1], were found to have the best repeatability.
Conclusion: In most cases 3D level set segmentation and geometric factor extraction can
be performed reproducibility, with minimal operator intervention, from non-ideal CEMRA
data. This opens up the possibility for cost-effective retrospective or prospective studies
on the role of local risk factors in vascular disease.
[1] Lee SW, Antiga L, Spence JD, Steinman DA. Geometry of the carotid bifurcation
anticipates its exposure to disturbed flow. Stroke 2008 Aug; 39(8): 2341-7.
108
MR-Angioclub East Lansing 2009
11.7 Magnetic source MRI for quantitative brain iron mapping
L. de Rochefort, T. Liu, I. Khalidov, J. Liu, B. Kressler, J. Wu, M.R. Prince, Yi Wang
Departments of Radiology and Biomedical Engineering, Cornell University, New York
Purpose: Iron deposition in the brain can result from cerebral microbleeds (CMB), which
may be associated with an increased risk of devastating intracerebral hemorrhage (ICH),
especially in patients anticoagulated with medicines such as warfarin. Studies have
indicated that the severity of CMB is strongly related to the occurrence of ICH (1).
Quantification of iron deposition in CMB may help quantitatively manage the risk of
warfarin associated ICH. Currently, dark regions in T2* weighted MRI have been used to
identify the presence of iron (2). This hypointensity caused by intravoxel variation of local
magnetic fields depends on imaging parameters and source-voxel geometry, may be
confused with other signal voids such as those caused by calcium deposits.
Method/Results/Conclusions: We propose a novel magnetic source MRI (msMRI)
approach to generate quantitative susceptibility maps for quantitative assessment of CMB
iron deposits. The average local magnetic field in a voxel is a convolution of a dipole field
kernel with the iron magnetization (mass density X B0 X iron susceptibility). So the
susceptibility as a tissue material property can be determined by solving the inverse
problem from magnetic field to susceptibility source. Unfortunately, straightforward
inversion generates no meaningful susceptibility mapping because of severe noise
propagation near the zero points of the dipole kernel. We propose to develop a novel
robust inversion method by making full use of all information in the T2* gradient echo
image data. The phase image (typically neglected in MRI) contains the intravoxel average
field information and is used to generate a local magnetic field map. The magnitude image
contains intravoxel field variation information and is used to guide the inverse algorithm
through a regularization term. We have obtained very encouraging preliminary data
indicating that our inverse approach is highly viable for mapping CMB susceptibilities.
References: 1. Lee SH, et al. Neurology 2009; 72:171-176. 2. Haacke EM, et al. Magn
Reson Imaging 2005; 23:1-25.
MR-Angioclub East Lansing 2009
109
11.8 Targeted Glyco-Magnetic Fe3O4 Nanoprobes for Detection and
Molecular Imaging of Atherosclerosis
Kheireddine El-Boubbou,a,d Medha N. Kamat,a,d David C. Zhu,b Ruiping Huang,c George Abela,c
Xuefei Huang*a
Department of Chemistry,a Departments of Radiology and Psychology,b Department of Medicine,c
Michigan State University, East Lansing MI 48824
Cardiovascular diseases, often associated with atherosclerosis, are the leading cause of
morbidity and mortality in the world. Despite the significant progress in cardiology, there
remain large unmet needs to early detect atherosclerotic plaques, especially those which
are prone to ruptures causing heart attacks and strokes. One of the major causes of such
dramatic event is “inflammation” which occurs during early onset of the disease leading to
over-expression of adhesion molecules. This initiates an immune response that eventually
leads to the formation of the plaque. Such adhesion cell-surface glycoprotein receptors
including the cluster of differentiation (CD44) expressed on leukocytes presents a unique
opportunity for the detection of the disease in its preliminary state. Our proposed work is
based on the surveillance that hyaluronic acid (HA) is upregulated in atherosclerotic
lesions and CD44, its principal receptor, is involved in several atherogenic processes.
Polyvalent HA is expected to displace native HA from cell surface CD44 to reduce the
development of atherosclerosis. Thus, we engineered novel highly dispersed hyaluronic
functionalized superparamagnetic iron oxide nanoparticles (HA-DESPIONs) as proficient
probes to non-invasively target atherosclerotic plaques using magnetic resonance imaging
(MRI). The targeting agents on the external surface of the magnetic nanoparticles will
allow the selective labeling of the plaques. Herein, imaging of atherosclerotic plaques in
rabbits was successfully examined. Due to the paramagnetic nature of the nanoparticles,
their binding with the plaques greatly enhances the contrast from the surrounding tissue,
allowing ready plaque detection by MRI. We anticipate that such novel nanoprobes HADESPIONs will not only deepen our fundamental understanding of the molecular and
cellular events characterizing unstable atherosclerotic plaques, but also be potentially
developed into a highly innovative therapy for atherosclerosis.
110
MR-Angioclub East Lansing 2009
12.1 4D DSA and Fluoroscopy: A New Challenge for MRA?
C A. Mistretta, E Oberstar, B Davis, E Brodsky and CM Strother
When DSA was first introduced, it was hoped that X-ray angiography could be
accomplished with intravenous injections. Ultimately, the overlap occurring in the 2D
projections led to the need for repeated injections and image quality was often marginal
due to poor SNR. In recent years 3D rotational DSA techniques, based on rotating C-arm
gantrys and large area flat panel detectors have been developed. 3D angiographic nontime-resolved volumes are reconstructed following IV or IA injection of Iodine. Image
quality is excellent due to the combination of information over the course of a 5-20 second
injection and rotation.
In recent years accelerated MRA methods have been developed to permit temporal
resolution of about 0.75 seconds over isotropic 320 x 320 x320 voxel volumes. These
methods have been modified to permit the acquisition of time resolved 4D DSA data sets
with matrices up to 512 x 512 x 512 with frame rates of 20 per second using either the
intrinsic projection information in the 3D rotational DSA acquisition or using a combination
of these data and an auxiliary conventional single projection DSA run using an IV or IA
injection. Real-time fluoroscopic catheter information can be embedded within a roadmap
formed from any of the 4D DSA time frames and can be viewed from any angle without
gantry rotation.
Methods
Rotational DSA data are acquired using either intra-arterial or intravenous injections of
iodine using typical rotation time of 5-20 seconds. Temporal information is embedded in
the reconstructed 3D rotational vascular system using multiplicative projection processing
(MPP) using time resolved information from either a separate conventional DSA
examination or using the intrinsic projections used to form the rotational reconstruction.
Whereas conventional reconstruction from projections typically requires a number of
projections dictated by the Nyquist theorem, one or two projections suffice to embed the
temporal information in the rotational DSA vessels due to the sparsity of angiographic data
sets. Shadowing artifacts occur when signals from the time resolved data set are
projected through. However the use of biplane acquisition or the use of separated
projections from the intrinsic data set are effective in resolving these. In the latter case
temporal resolution is reduced in proportion to the required angular separation. Artifacts
can also be reduced based on analysis of contrast time curves where shadowing artifacts
will appear as anomalies. .4D fluoroscopy is implemented using MPP using a subtracted
catheter-only data set that is multiplied into the vascular tree.
Results are shown below.
Figure 1 shows the AP and Lateral MIP images of the acquired 3D rotational study formed
with a 5 second rotation following an intra-arterial contrast injection. The first four frames
of the time resolved AP and lateral MIPS through the 4D DSA data set are also shown.
The spatial resolution of the images shown has been reduced by a factor of eight relative
to what is possible due to memory limitations of the MATLAB software used to reconstruct
these images. This study was performed using a single AP conventional DSA acquisition.
MR-Angioclub East Lansing 2009
111
Figure 1 Rotational DSA MIPS and MIPS through the 4D DSA time frames. All time
frames can be rotated in arbitrary directions.
Figure 2 illustrates two orthogonal views of a single fluoroscopic time frame obtained using
single plane fluoroscopy. The catheter can be viewed from arbitrary directions without
gantry rotation. When biplane fluoroscopy is used the catheter representation is accurate
from all views. When a single projection is used the catheter position is constrained to be
in the center of the vessel in the orthogonal view since the projection from the acquired
view produces a sheet of intensity through the vessel. This may provide no disadvantage
since the position of the catheter in a normal fluoroscopic view is also unknown in one
direction. In spite of this, the advancement of the catheter tip is well represented.
Figure2 Orthogonal
views of a single
fluoroscopic time frame
from single plane
fluoroscopic exposure.
The catheter is shown
in white and can be
view from arbitrary
directions without
gantry rotation.
Figure 3 shows a color display of the time to ½ maximum intensity for the vessels
contained in one of the 4D DSA time frames.
Figure 3 Color display of time to ½ peak. The color scale is in units of 0.25 seconds.
4D DSA is the result of an approximate reconstruction method and typically suppresses
parenchymal information, so semi-quantitative impressions of perfusion deficits will have
to be inferred based on localized models involving the differences in calculated vascular
transit times combined with already available cerebral blood volume maps.
112
MR-Angioclub East Lansing 2009
12.2 MR Angiography of muscular and collateral arteries in
peripheral arterial disease: reproducibility of morphological and
functional vascular status
Bas Versluis1,4, MD; MD; Patty J. Nelemans3, MD, PhD; Joachim E. Wildberger1,4, MD, PhD; Walter H.
1,4
1,4
Backes , PhD; Tim Leiner , MD, PhD
Maastricht University Medical Center, Departments of Radiology1 and Epidemiology2 and
3
Cardiovascular Research Institute Maastricht (CARIM), 5Atrium Medical Center Heerlen, 6University
Medical Center Utrecht
Purpose: Vascular adaptations contribute to the recovery of peripheral arterial disease
(PAD). The aim of this study was to determine the reproducibility of the number of arteries
in the upper leg as well as arterial flow.
Methods: 10 patients with proven PAD (Fontaine stage II) and 10 healthy volunteers were
included. All subjects underwent CE-MRA covering the entire muscular volume of the
upper legs twice, with a time interval of at least 72 hours. Reproducibility was evaluated in
terms of the smallest noticable difference by the repeatability coefficient (RC), the
coefficient of variation and the intraclass correlation coefficient between the two scans and
the two readers.
Results: Interscan RC for the number of vessels was 1.1 for both patients and volunteers,
meaning an increase of 1 vessel per measurement plane between two scans would
already indicate a significant effect. Interscan RC for flow was 1.5 mL/s for patients and
1.6 mL/s in volunteers. Interreader repeatability coefficient was approximately four times
higher, indicating that the same reader is recommended for follow-up studies.
Conclusions: Quantification of the morphologic vascular status using artery count and
flow measurements proved reproducible in both patients and healthy volunteers. Because
of the high reproducibility, CE-MRA might be helpful in quantifying the development of
arterial collateral formation in patients with PAD and can help to understand to complexity
of this process.
MR-Angioclub East Lansing 2009
113
12.3 Multicenter Studies: Lessons Learned from ADNI
Matt A. Bernstein, Jeffrey L. Gunter, and Clifford R. Jack Jr
Mayo Clinic, Rochester, MN, USA
The Alzheimer’s Disease Neuroimaging Initiative (ADNI) [1, 2] is a six-year, 800-subject
observational study to assess how well the combined information obtained from MRI, PET,
other biological markers, as well as from clinical and neuropsychological assessment can
measure the progression of mild cognitive impairment (MCI) and early Alzheimer’s
disease (AD). All of the subjects are imaged with 1.5T MRI, and a subset (25%) with 3T
MRI. Half of the subjects also receive FDG PET, and a smaller subset of 120 subjects
receives PIB PET.
A total of approximately 5500 MRI exams are planned over the
Execution Phase of the study, which is scheduled to be completed in 2010. All of the
image data are readily available via the Internet to any researcher.
Details about the ADNI MR imaging protocol and its development process are
documented in [2]. A total of 89 scanners with 38 discrete combinations of vendor/field
strength/software revision/hardware configuration are supported. Detailed lists of imaging
parameters for those configurations are posted and are publicly available at
http://www.loni.ucla.edu/ADNI/Research/Cores/ . The wide variety of supported platforms
greatly increases the complexity of the management of the study.
In this talk, with benefit of hindsight, a few lessons learned about managing the MR
portion of a large, multicenter study will be discussed. In particular, the following points will
be covered:
- Quantitative and automated QC and standardization methods [3,4].
- Advantages of a phased approach, i.e. a “prep phase” or mini-dry run for a large study.
- Collaboration with MRI equipment vendors and how it can be mutually beneficial.
Preliminary results from ADNI will also be presented, indicating that the sensitivity of
MRI methods compares very favorably with PET and neuropsychological testing. The
particular experiences reported here relate to an Alzheimer’s disease study, but most of
the lessons learned are quite general and apply to large MR angiography multicenter
studies as well.
1. Mueller SG, Weiner MW, Thal LJ, et al. Alzheimers Dement. 2005 ;1(1):55-66.
2. Jack CR, Bernstein MA, Fox NC et al., J Magn Reson Imaging 2008 ;27(4):685-91.
3. Gunter JL, Bernstein MA, Borowski BJ et al, Med Phys. 2009 Jun;36(6):2193-205.
4. Mortamet B, Bernstein MA, Jack CR et al, Magn Reson Med. 2009 Jun 12;62(2):365372.
114
MR-Angioclub East Lansing 2009
12.4 Imaging Considerations in Serial Studies of Vascular Disease
M Sakamoto, H Kroll, V Rayz, L Boussel, A Martin, and D Saloner
Department of Radiology and Biomedical Imaging, VA Medical Center/UCSF
Purpose
To investigate strategies for extracting quantitative estimates of important
descriptors of vascular disease that would permit a determination of compositional and
geometric changes in patients with atherosclerotic and/or aneurysmal disease.
Methods We selected 18 patients from a large cohort of subjects who underwent two
serial MRA studies of the extracranial carotid arteries and 32 subjects with intracranial
aneurysms who underwent at least two serial studies generally acquired one-year apart.
Quantitative measures of the lumenal volume was measured manually by three
independent readers and using a semi-automated algorithm on TOF- MRA, CE-MRA, and
T1-black blood MRI. Lumen boundaries were determined either using a visual threshhold
or from histogram and/or profile analyses. In order to account for threshholding variation
from factors such as coil sensitivity profiles, a reference segment of normal vessel was
selected for calibration.
Results
Excellent inter-reader and intra-reader reproducibility was demonstrated.
In
carotid disease it was found that appropriate selection of histogram parameters provided
essentially equivalent volume measures for all sequences. In aneurysmal disease, it was
found that volume values were reproducible to within 3% for diameters larger than 5 mms,
to within 10% for diameters greater than 3 mms, and that smaller aneurysms were
associated with substantial uncertainty.
Conclusions The combination of MRI and MRA provides information on the temporal
evolution of both the lumenal geometry and disease of the vessel wall such as atheroma
or juxtalumenal thrombus.
However, the absence of normalized values for signal
intensities compromises the ability to unambiguously define vessel wall composition and
the precise location of vessel edges.
With careful attention to signal intensity
characteristics and using appropriate reference standards it was found that reliable
estimates of serial changes could be extracted from MRI/A. These approaches have
significant advantages in longitudinal studies. One specific example is the ability to use
MRA studies to quantitate the vascular lumen thereby reducing the imperative to obtain
high quality black-blood lumenal definition in studies of atherosclerosis.
MR-Angioclub East Lansing 2009
115
12.5 Towards Abdominal MRA at 7 Tesla
Ladd ME, Umutlu L, Maderwald S, Kinner S, Orzada S, Antoch G, Kraff O,
Ladd SC, Brote I, Bitz AK, Schaefer L, Quick HH, Lauenstein TC
1
Erwin L. Hahn Institute for MRI, University Duisburg-Essen, Essen, Germany
Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital,
Essen, Germany
2
Purpose: Imaging in anatomic regions with large cross-section is challenging at 7T
because of RF wavelength effects in the tissue. Thus far, 7T imaging has therefore been
primarily limited to the head or extremities. Only few reports of 7T for cardiac or abdominal
imaging are available [1-3]. The purpose of this study was to perform an exploratory study
of the potential of 7T for performing MRI and MRA in the abdomen.
Methods: All examinations were performed on a 7T whole-body MRI system (Magnetom
7T, Siemens) in a total of 20 subjects. A custom-built flexible 8-channel RF
transmit/receive body coil consisting of stripline elements and suitable for static RF
shimming was used [4]. Non-contrast-enhanced MRI was performed including T1w 3D
FLASH, T1w fat-saturated 2D FLASH, 2D T1w in- and opposed-phase FLASH, 2D TOF,
quasi T2w 2D TrueFISP, and T2w TSE.
Results: T1w imaging at 7T in general revealed excellent conspicuity of small anatomical
structures and the hepatic and renal vasculature (Fig. 1). The arterial system was bright
and the venous system primarily dark regardless of slice orientation. TrueFISP provided
unexpectedly good image quality (Fig. 2); however, both it and TSE remained challenging
due to SAR restrictions.
Conclusions: These results are promising for the future of performing abdominal MRA at
7 Tesla. The inherently high signal of the arterial system in T1w imaging shows the
potential for MRA without contrast agent. Subsequent studies in healthy volunteers and
patients will further assess these sequences and the 7T appearance of pathologies.
Fig. 1: Non-enhanced T1w imaging. Left and middle:
FLASH 2D of the upper abdomen. Note the inherently
high vasculature signal. TR/TE = 130/3.6 ms, flip 70°, BW
2
405 Hz/pixel, FOV 400x400 mm , matrix 512x512,
13 slices, thickness 2 mm, Grappa R=2, TA 31 s. Right:
TOF MIP in a further subject.
Fig. 2: TrueFISP provides good
overview of the liver vasculature. TR/TE
= 3.5/1.5 ms, flip 50°, BW 975 Hz/pixel,
2
FOV 400x400 mm , matrix 256x320, 21
slices, thickness 4 mm, TA 18 s.
References: [1] Vaughan JT, et al. MRM. 2009;61:244-8. [3] Maderwald S,et al. ISMRM 2009, p. 821.
[2] Snyder CJ, et al. MRM. 2009;61:517-2 [4] Bitz AK, et al. ISMRM 2009, p. 4767.
116
MR-Angioclub East Lansing 2009
1,2
12.6 7 Tesla Cardiac MRI in Humans
Harald H. Quick , S.Maderwald1, S.Orzada1, A.K.Bitz1, I.Brote1, O.Kraff1, L.C.Schaefer1, M.E.Ladd1,2
1
Erwin L. Hahn Institute for MRI, UNESCO World Cultural Heritage Zollverein, Essen, Germany
2
University Hospital Essen, Department of Diagnostic and Interventional Radiology, Essen, Germany
Purpose: Human cardiac MRI at 7 Tesla is a potentially challenging endeavor due to
inhomogeneous RF signal transmission [1] caused by the heart’s position deep within the
body and due to the reduced Larmor wavelength at 7 T of approximately 12 cm, which is
shorter than the dimensions of the human body and may thus lead to destructive B1
interference (signal voids). Additionally, the specific absorption rate (SAR) at this field
strength often constrains the choice of imaging sequence parameters [2]. The purpose of
this study was to transfer initial experiences in animal cardiac MRI at 7T [3, 4] to human in
vivo cardiac imaging using a custom-built flexible 2x4-channel RF transmit/receive body
coil.
Methods: All examinations were performed on a 7T whole-body MRI system (Magnetom
7T, Siemens, Erlangen). A custom-built flexible 2x4-channel RF transmit/receive body coil
for 7T human imaging was used for RF signal transmission and reception. Seven healthy
volunteers (4 male, 3 female) were placed head-first supine with the chest at the isocenter
of the magnet and within the sensitive region of the coil. The imaging protocol
encompassed cardiac function along standard views using peripheral pulse-triggered Cine
FLASH sequences with 20 phases per RR-interval. The image quality was visually
assessed for signal homogeneity and myocardium-to-blood contrast.
Results: All seven subjects could be successfully examined. The coil, driven in CP mode,
qualitatively provided relatively homogeneous B1 signal over the sensitive volume. Some
regions in the images, however, showed destructive interference with associated signal
voids. The Cine FLASH sequence provided good imaging quality and signal homogeneity
over almost the entire heart and with good myocardium-to-blood contrast; the achieved
spatial resolution was 1.4 x 1.4 x 4 mm3. For perfectly timed and triggered cardiac images,
however, ECG triggering seems mandatory. Peripheral pulse gating, as used in this study,
in part was associated with imprecise triggering leading to mild motion artifacts.
Fig. 1: Cine FLASH images of the human heart in vivo at 7T. a) short axis, b) 2-chamber, c) 4
chamber, d) LVOT, and e) LVOT 2nd. 1 out of 25 cine phases is shown for each orientation.
Conclusion: These initial results can be considered as a first step towards human in vivo
cardiac imaging at 7 Tesla high-field MRI. Subsequent studies in patients will further
assess these sequences in high-field cardiac imaging and additional cardiac protocols
including late enhancement.
References: [1] DelaBarre et al.; ISMRM 2007, p. 3867; [2] Maderwald et al.; ISMRM 2008, p. 2716.[3]
Quick et al.; MRA-Club 2008, Graz, p. 84.;[4] Quick et al.; ISMRM 2008, p. 1023.
MR-Angioclub East Lansing 2009
117
12.7 The Role of Cholesterol Crystals in Acute Cardiovascular
Events: Identifying the Cause for Gender Differences in Clinical
Presentation
George S. Abela MD, Ameeth Vedre MD, Fadi Shamoun MD, Majid Moughal MD, Department of
Medicine, Division of Cardiology, Michigan State University, East Lansing, MI
Purpose: Plaque rupture has been seen more frequently in men and erosion in women.
To explain this we evaluated the physical factors related to cholesterol crystallization.
Methods: The amount of cholesterol, temperature, hydration and pH were varied and rate
and amount of volume expansion measured in vitro. Cholesterol powder (1,2,3 g) was
dissolved in corn oil at varying temperatures (22-44°C) and allowed to crystallize. Effect of
pH and hydration of cholesterol were also evaluated. Fibrous membranes (rabbit
pericardium) with similar thickness and composition to the plaque cap were placed in the
path of growing crystals to assess potential for damage. Tissue preparation for scanning
electron microscopy (SEM) was performed without ethanol to avoid dissolving the
cholesterol crystals.
Results: The volume and rate of cholesterol expansion was directly related to the amount
of cholesterol present (r=0.98; p<0.01, r=0.99; p<0.01 respectively). Low temperature,
hydration and higher pH all significantly increased volume expansion. By gross
examination, the expanding crystals tore the fibrous membrane while in others it just
perforated the membrane. By SEM cholesterol crystals were seen perforating the fibrous
membranes.
Conclusions: Greater amounts of cholesterol result in more crystallization and volume
displacement that can tear fibrous membranes. Thus, the larger necrotic cores seen in
men that probably have higher cholesterol content would result in more rupture. This may
help explain in part the gender difference in clinical symptoms of more ruptures in men
compared to more erosion in women.
118
MR-Angioclub East Lansing 2009
P.1 CE-MRA with tailored 3D random sampling patterns and
nonlinear parallel imaging reconstruction
1
2
3
F. Knoll , C. Clason , F. Ebner , M. Aschauer3, R. Stollberger1
Institute of Medical Engineering, TU Graz, Austria, 2Institute for Mathematics and Scientific Computing,
University of Graz, Austria, 3Department of Radiology, Medical University Graz
1
Introduction: Variable density 3D random sampling trajectories which were introduced in
the context of compressed sensing [1], have great potential for subsampled MR
angiography techniques. The goal of this work was to present a parameter-free method to
construct tailored variable density sampling patterns, which can be used together with a
nonlinear parallel imaging method [2, 3] to allow the use of very high acceleration factors.
Methods: A CE-MRA dataset (3D Gradient Echo Sequence, TR/TE=3.74/1.48ms,
FA=30°, matrix: 448x352x40, resolution: 0.55x0.55x0.70mm3) of the carotid arteries was
acquired on a clinical 3T system and subsampled retrospectively, to simulate an
accelerated acquisition. Parallel imaging with 8 receiver coils, comparable to the spatial
positions of the individual elements of the receiver head coil that was used in our
experiments, was simulated by use of Biot Savart’s law. The proposed method to generate
the variable density 3D random sampling pattern is to use the scan of the same anatomic
region of a different patient or a healthy volunteer as a template. The power density
spectrum of this template can be used as a reference to generate a probability density
function which is then used to construct the sampling pattern. Patterns that are generated
in this way can be pre-computed for different types of scans or anatomical regions. Image
reconstruction was performed with an iteratively regularized Gauss-Newton method
(IRGN) [2].
Results and Discussion: Our results show that excellent image quality can be achieved
even for very high acceleration factors like R=30 (Fig.1.) without any application of
temporal view sharing. Only a slight decrease of SNR and a minimally reduced contrast
for the smallest vessels result for an acceleration factor of 30. The mean RMS difference
to the original fully sampled data set over all 40 slices was 0.055 with a standard deviation
of 0.036. The proposed approach to generate the variable density sampling pattern
ensures the sampled ratio of low to high
frequency sample points is reasonable for
angiography scans. One major advantage is
that the method is completely free of any
user defined parameters. Our experiments
showed that the method is robust regarding
the choice of the reference image, as the
only information that has to be obtained is an
estimate of the ratio of high to low frequency
components. The exact anatomical details
are not important in this context.
Fig. 1: CE-MRA Dataset of the carotid arteries:
(a) Fully sampled data set
(b) IRGN reconstruction, R=30
References: [1] Lustig et al., MRM 58: 11821195 (2007), [2] Uecker et al., MRM 60: 674-682, [3] Knoll et al., ISMRM 2009: 2721
MR-Angioclub East Lansing 2009
119
P.2 Handling Motion in Sparse MRI with Whiskers
Jason Mendes, Dennis L. Parker, University of Utah, UCAIR, Salt Lake City, Utah
PURPOSE: For dynamic images, pixel intensities that vary smoothly or periodically can
be sparsely represented in a Wavelet-Fourier space and recovered from undersampled kspace data using Compressed Sensing (1,2).
Patient motion that is not periodic or
smooth can therefore be a problem in the reconstruction. We investigate the application
of Compressed Sensing to a novel segmented data sampling technique called Whiskers
(for the whiskers looking pattern in k-space and lack of a clever acronym) to reduce the
effects of patient motion.
METHODS: In general, the minimum number of k-space samples required to produce
good results in sparse reconstruction is approximately four times the number of sparse
coefficients (3). It is therefore beneficial to detect and correct as much patient motion as
possible to maximize temporal sparsity and thus reduce the total number of k-space
samples required.
This is accomplished using a hybrid Radial-Cartesian sampling
technique called Whiskers. The Whiskers k-space trajectories of a single segment are
shown in Figure 1. Readout and phase encoding directions are alternated each TR.
Figure 1: Whiskers segment (a)
combined with segment (b)
yields a radial like coverage
while sampling the central part
of k-space rectilinearly.
(a)
(c)
(b)
(c)
RESULTS AND CONCLUSION: The new Whiskers design has been implemented with a
Turbo Spin Echo sequence and shown to provide good detection of both translational and
rotational motion. Aliasing artifacts due to Whiskers undersampling are similar to that of
Radial undersampling, but more incoherent, making Whiskers well suited to be used in
conjunction with Compressed Sensing.
Additionally, the most distinct artifact of
Compressed Sensing is the loss of low-contrast features in the image (1).
Because
Whiskers fully samples the central portion of k-space it should be able to recover much of
that loss. The simulated results look promising and the authors intend next to apply the
technique to vascular imaging.
ACKNOWLEDGMENTS:
1. Lustig M, et. al. Magn Reson Med 2007;58:1182–1195.
2. Lustig M, et. al. ISMRM, Seattle, 2006. p 2420.
3. Tsaig Y, et. al. Signal Process 2006;86:533–548
120
MR-Angioclub East Lansing 2009
P.3 Improved Carotid imaging with HASTE using a reduced FOV
and increased gradient performance
Jordan P. Hulet5, Seong-Eun Kim1,3, Gerald S. Treiman2,4, Dennis L. Parker1,3,5
Utah Center for Advanced Imaging Research, 2VA Salt Lake City Health Care System, 3Radiology,
4
Surgery, 5Biomedical Informatics, University of Utah
1
Purpose
MR imaging of the carotid artery often suffers from motion artifacts. Single shot
sequences, such as HASTE, can reduce the effect of motion artifacts but often produce
blurry images due a long echo train. We have explored the use of both a reduced FOV
technique (shown below) and increased gradient performance in order to shorten the echo
train length, decrease blurring, and improve image quality.
Methods
The standard Siemens HASTE sequence was modified to image a reduced field of view
(rFOV) in the phase-encoding direction while preventing aliasing artifacts by moving the
gradient waveforms for the 180° refocusing pulses from the slice select to the phase
encode axis and adjusting gradient amplitude to accommodate the reduced field of view. A
volunteer was scanned using both regular HASTE and rFOV-HASTE.
Results
Images obtained using both sequences are shown in Figure 1. rFOV-HASTE resulted in
images with significantly less blurring which more clearly visualize the carotid arteries.
Figure 1: Carotid scans using regular (a) HASTE and (b) rFOV-HASTE. The HASTE
images were cropped to match the reduced FOV (25%) of the rFOV-HASTE images.
Discussion
Using rFOV with HASTE shortens the echo train resulting in improved carotid images.
Additionally, a custom gradient insert designed to increase gradient performance is
currently under construction. The rFOV-HASTE sequence has been modified to utilize this
increased gradient performance and further shorten the echo train.
MR-Angioclub East Lansing 2009
121
P.4 Towards Continuously Moving Table NCE Peripheral MRA
Randall B. Stafford1,5, Mohammad Sabati4, Houman Mahallati2,5, Richard Frayne1-3,5
1
2
3
Departments of Physics and Astronomy, Radiology, Clinical Neurosciences, University of Calgary,
Calgary, AB, Canada; 4Department of Radiology, University of Miami, Miami, FL, USA; 5Seaman
Family MR Research Centre, Foothills Medical Centre, Calgary, AB, Canada
Purpose: Peripheral arterial disease (PAD) requires large field-of-view (LFOV)
angiography. PAD patients with renal artery stenosis may be at risk of NSF [1,2]. The
purpose of this project is to combine the balanced steady-state free precession (bSSFP)
Dixon method [3,4] with the continuously moving table (CMT) technique [5,6] for LFOV
NCE peripheral MRA. Here, we assess the feasibility through computer simulation.
Methods: A peripheral anthropomorphic
phantom consisting of fat, marrow, arterial
and venous blood, and muscle was
constructed in MATLAB (The MathWorks,
Inc.). Arterial segments were modified with
varying levels of stenosis. LFOV bSSFP
Dixon method [3,4] image data sets were
generated using bSSFP magnetization
evolution with a table velocity of 0.98 cm/s.
Stationary images were generated for
difference comparison. Maximum intensity
Fig. 1: Simulated bSSFP Dixon method MIP
images of anthropomorphic phantom (left)
stationary data set, (centre) CMT data set, and
(right) difference. Arrows indicate stenoses.
projection (MIP) images were produced for
all data sets.
Results: The water-only MIP images [3,4] are shown in Fig. 1. The simulated table
velocity (0.98 cm/s) allowed the tissue to achieve steady-state magnetization, and allowed
for 100% hybrid-space coverage [5]. The blurring artifacts in the CMT images are due to
partial-volume effects associated with the coarse phantom resolution and the table motion.
This blurring effect would be reduced in a real CMT acquisition [5,6].
Conclusion: The simulations presented here have successfully demonstrated the
compatibility of the bSSFP Dixon method with the CMT technique. The next step in this
project is to collect LFOV images in vivo, and assess vessel conspicuity.
References: [1] Albers, Am J Kidney Dis 1994;24:636, [2] Thomsen, Eur Radiol
2006;16:2619, [3] Huang, MRM 2004;51:243, [4] Stafford, MRM 2008;59:430, [5] Kruger,
MRM 2002;47:224, [6] Sabati, PMB 2003;48:2739.
122
MR-Angioclub East Lansing 2009
P.5 R2* Calibration Phantoms for Cardiovascular Studies
Matthew T. Latourette and James E. Siebert, Michigan State University, East Lansing
Purpose:
R2* quantitation has been applied to assess iron overload and, recently, to
characterize hemorrhage in atherosclerotic plaque.1-3 Calibration reference phantoms exhibiting
long-term chemical stability and temperature-insensitive R2*/T2* signal, needed for longitudinal
studies, have not been adequately addressed in the literature. This study aims to improve R2*
quantitation reproducibility via development of a calibration reference. The availability of a
stable reference standard is important for R2*-based research studies for monitoring data
quality, decreasing the variance of pooled intra-site study data, and detecting and correcting
bias in multi-site studies.
Methods: A gel mixture matching the T1 and T2 of pediatric brain was doped with SPIO
nanoparticles (Feridex) to produce phantoms with varying R2*. NiCl2 was used to modify T1
because its relaxation behavior is largely independent of temperature, unlike other popular
doping agents.4,5 Agarose was used to modify T2 and promote gel formation. Carrageenan
aided formation of strong, tissue-like gels.6 A methylisothiazolinone-based (MIT) preservative
provided long-term antimicrobial protection. Heat and stirring was applied to the mixture of
NiCl2, carrageenan, agarose, and MIT to dissolve it in water. The solution was poured into
polysulfone bottles, chilled, and sealed. Imaging was performed at 0.7T to verify the T1 and T2
of these phantoms. T1 was computed by fitting the mean ROI signal intensity for each of 5
axial SPGR series to S(TR)=S0(1-e-TR/T1). Mean signal intensity in 6 axial spin echo series ROIs
was fit to S(TE)=S0e-TE/T2 to compute T2. R2* calibration phantoms were prepared with the
same underlying composition, varying only the SPIO concentration.
Results and Discussion: Average T1 and T2 for the SPIO-free phantoms at 0.7T was 838 ms
and 89.5 ms respectively. R2* as a function of SPIO concentration of the doped phantoms was
38.5±1.3 s-1 at 63.6 µM, 44.4±1.7 s-1 at 72.7 µM, 55.5±1.9 s-1 at 90.9 µM, and 68.6±1.9 s-1 at
127 µM. Follow-up imaging results describing measurement reproducibility, R2* measurements
for other field strengths, and an evaluation of the thermal stability of phantom R2* will be
presented.
References: 1Siebert JE et al, MR Angio Club, 2006. 2Zhu DC et al, ISMRM, 16:2841, 2008.
3
Zhu DC et al, ISMRM, 17:606, 2009. 4Kraft KA et al, MRM, 5:555-562, 1987.
5
Waiter GD,
6
Foster MA, Magn Reson Imaging, 15:929-938, 1997. Yoshimura K et al, MRM, 50:1011-1017,
2003.
MR-Angioclub East Lansing 2009
123
P.6 Pictorial Review of Supra-Aortic Artery Pathologies as
Visualised with MRA using Blood Pool Contrast Agent
Bethapudi S & Roditi G
Glasgow Royal Infirmary, Alexandra Parade, Glasgow G31 2ER
Purpose: Review the appearances of carotid, vertebral & subclavian artery disease as
demonstrated by Contrast-Enhanced Magnetic Resonance Angiography (CE-MRA) with
blood pool contrast agent. To understand the utility of high resolution 3D steady state
phase imaging in adding value to conventional first pass CE-MRA.
Background: The blood pool contrast agent Gadofosveset trisodium allows first pass CEMRA at high spatial resolution with low gadolinium dose due to its high specific relaxivity in
human serum. The prolonged blood pool residency further allows very high spatial
resolution imaging during the steady state phase with acquisition of isotropic voxels that
allow reformat in any plane without loss of spatial resolution along with depiction of
adjacent structures not seen on usual CE-MRA.
Procedure Details: Our experience of MRA in patients imaged with blood pool contrast
agent for suspected carotid, vertebral or subclavian disease is presented. Patients were
imaged in usual first pass and subsequently in steady state phase with high spatial
resolution. Results are presented as a pictorial review of the spectrum of pathologies
encountered
including
atherosclerotic
steno-occlusive
disease,
subclavian
steal
syndrome, vertebral dissection, carotid body tumour, surgical endarterectomy sites, large
vessel vasculitis, thoracic outlet syndromes and depiction of extra-anatomical bypass
grafts. Particular emphasis is placed on the utility of high spatial resolution steady state
imaging to extend coverage, delineate the vascular wall and evaluate associated
structures.
Conclusion: Blood pool contrast-enhanced MRA of the supra-aortic vasculature is
effective in allowing comprehensive evaluation of various arterial pathologies, particularly
in demonstrating extraluminal pathology.
124
MR-Angioclub East Lansing 2009
P.7 Robust Clinical Application of Time-Resolved MRA
K. A. Blackham, G.S Sandhu, R.C. Gilkeson, M.A. Griswold, V. Gulani1
Department of Radiology, University Hospitals of Cleveland, Cleveland, Ohio, United States
Purpose:
To present a review of the clinical applications of time resolved MR
angiography (trMRA) throughout the body.
Methods:
A series of patients were imaged with dynamic
contrast enhanced TWIST-MRA (Siemens Verio, 3.0 T,
TR/TE=minimal ; representative 3.28ms/1.51 ms; matrix 256320 x 176-256 x 64-80, GRAPPA 2-3, flip angle 21-25˚, TA 1.54 s/ frame, # frames varied according to application).
Results: The TWIST sequence provides a robust clinical tool
for trMRA that can be employed to solve clinical problems noninvasively. In children, applications included assessment of
arterial
inflow
and
venous
outflow
in
arteriovenous
malformations (AVMs), mapping of thoracic and abdominal
anomalous vessels, and assessment for renal artery stenosis.
The patients were free-breathing, and resolution as high as
Fig.1: Sub-volume
MIP of a contrast
enhanced trMRA in a
child showing a
thoracic kidney in the
right hemithorax and
an aneurysm in the
renal artery.
0.9x0.7x0.9 cm3 was routinely achieved with a single dose of
Magnevist contrast. Applications in adults included AVMs in the brain and throughout the
body with mapping of feeder/draining vessels, chest pulmonary angiography and
venography, assessment of renal artery stenosis in patients unable to provide breathholds, assessment of AV fistulas, dynamic imaging of popliteal artery entrapment
syndrome,
pelvic congestion syndrome, assessment of arterial anatomy and disease
when contrast dynamics in two legs were drastically different, and assessment of
perforator anatomy for fibular free-flap transfer operations, often with half the standard
dose of contrast (by weight). Near isotropic datasets allowed multiplanar reconstruction of
images at multiple timeframes, and dynamic MIP images allowed visualization resembling
traditional angiograms.
Conclusion: TWIST-MRA is a rapid, robust technique that is non-invasive and can be
used routinely and for problem solving in a variety of settings throughout the body.
References: 1. Petkova M, et al. J Magn Reson Imaging 2009; 29:7-122. 2. Sandhu G et
al. Proc ISMRM 2009; 589
MR-Angioclub East Lansing 2009
125
P.8 Reproducibility of Aortic Pulse Wave Velocity Measurements
Obtained with Phase Contrast Magnetic Resonance (PCMR) and
Applanation Tonometry
Jonathan Suever, BS,† David Huneycutt, MD,* Enrique Rojas-Campos, MD,* Francesca Cardarelli,
MD,* Sam Fielden, BS,* Arthur Stillman, MD, PhD,* Paolo Raggi, MD,* John N. Oshinski, PhD,*†
†Emory/Georgia Tech, Department of Biomedical Engineering, Atlanta, GA
*Emory University School of Medicine, Atlanta, GA
Purpose: Increased aortic pulse wave velocity (PWV) results from decreased vessel
compliance and can be due to increased age, atherosclerosis and/or hypertension (Farrar
et al. Circ. 1991). The goal of this study was to compare reproducibility of PWV
measurements obtained via applanation tonometry (AT), a clinically accepted method, to a
new PCMR method using cross-correlation analysis.
Methods: PWV was measured in seven asymptomatic patients with an elevated CT
coronary calcium score, and ten healthy volunteers. Oblique sagittal images in the plane
of the aorta (“Candy Cane” view) were acquired with a velocity encoding in the foot-head
direction. Cross-correlation was used to determine time shifts between velocity curves at
adjacent points along the aorta. The slope of a regression line fit to the transit time vs.
location data was used to estimate PWV
(Fielden et al. JMRI 2008). For AT, a
Sphygmocor device (AtCor Medical) was
used to measure pressure waveforms at
carotid and femoral locations and the PWV
was determined using a standard transfer
formula.
Inter-scan
variability
for
each
method was assessed with the coefficient of
variation (CoV).
Results: PCMR had a significantly lower
inter-scan CoV than AT (Figure 1). The better
reproducibility may allow for the enrollment of
fewer patients for trials of interventions
targeted at vessel wall compliance.
Conclusion: PWV estimates using PCMR
and
cross-correlation
processing
have
Figure 1: Inter-scan coefficient of
variation for AT and PCMR
superior reproducibility as compared to AT.
126
MR-Angioclub East Lansing 2009
P.9 Motion-compensated, flow-independent, non-contrastenhanced renal MR angiography
Gregory J. Wilson1,2, George R. Oliveira2, Liesbeth Geerts1, Jeffrey H. Maki2
1 – Philips Healthcare, Cleveland, USA and Best, Netherlands;
2 -- University of Washington Dept of Radiology, Seattle, WA, USA
Purpose Evaluate a new MR technique for high resolution imaging of renal arteries.
While breath-hold contrast-enhanced MRA (CE-MRA) of renal arteries provides useful
images, the achievable spatial resolution can be limited by breath-hold duration, cardiac
motion [1], respiratory motion [2], and the contrast agent bolus profile [3]. The flowindependent, non-contrast-enhanced method implemented here reduces or eliminates
these resolution-limiting effects.
Methods
The technique has been evaluated in 3 healthy volunteers. The cardiac-
triggered, balanced TFE acquisition employed respiratory navigator gating, fat saturation,
and magnetization preparation (T2 Prep). Data was acquired only during diastole, the
most quiescent period of flow and cardiac-related motion, and without inversion pulses. As
a result, the sequence was flow-independent. Axial volumes were acquired with resolution
of 1.68x1.72x2.4mm3 (reconstructed to 0.73x0.73x1.2mm3) in 4:26 with 70% navigator
efficiency and 2 averages.
Results The acquisition was successful in each volunteer. High resolution images were
produced without the limitations of motion or bolus profile blurring. Flow independence
was demonstrated by lack of in-flow signal modulation in coronal acquisitions, and lack of
venous signal suppression by application of in-flow saturation bands. As a consequence,
veins were visible in the images.
Conclusion The new technique provided high resolution images for evaluation of renal
artery lumen morphology. Further study is planned to evaluate the diagnostic value of this
technique.
Figure 1: High resolution, axial source image
displaying left renal artery.
Figure2: Curved
reformat showing
bilateral renal
arteries.
References
1-Kaandorp DW, JMRI2000;12:924-8; 2-Vasbinder GB, JMRI2002;16:685-96; 3-Fain SB,
MRM1999;42:1106-16.
MR-Angioclub East Lansing 2009
127
P.10 Vascular response during visual stimulation at 3T MRI:
functional phase contrast angiography (fPCA) study
1
1
Sang-Hoon Kim , Chang-Ki Kang , Young-Bo Kim1, Zang-Hee Cho1, 2
1. Neuroscience Research Institute, Gachon University of Medicine and Science, Korea
2. Department of Radiological Sciences, University of California, USA
Purpose: The purpose of present study is to investigate direct arterial response with
quantitative velocity analysis using phase contrast angiography (PCA) sequence [1,2].
Methods: PCA sequence was used with the following parameters: 0.67x0.67x1.0 mm3 of
imaging resolution, 36 slices, and 20 cm/s of velocity encoding (VENC) for all directions.
Total acquisition time was 10 m 18 s for 3 sessions, including 2 rests and 1 visual
stimulation (a flashing checker board). A custom-built surface coil was designed for
angiographic purpose and tuned to 3T. Twelve subjects participated in this study. Circular
region of interests (ROIs) consisting of 21 pixels was used to measure the velocity
difference on the different diameters. We found the ROI showing a maximum change and
defined as peak ROI and distal ROIs for vessels distal to the peak ROI. Velocity changes
of the selected ROIs were measured.
Results: Fig. 1 showed the vascular change acquired from PCA and calculated the
velocity and its percentage change during stimulation. Velocity change during stimulation
was clearly delineated, which were indicated with arrows (a). Mean velocity change
between rest and stimulation periods were 0.64 and 0.42 cm/s from a peak ROI and distal
ROIs (b), respectively, which are corresponding to 42.3 and 29.6 of percentage change as
shown in Fig. 1(c).
Conclusion: PCA for functional angiography (fPCA) at 3T MRI could provide vascular
response during brain function, including the vasculature as well as the velocity changes
of related vessels.
References:
1. Iadecola C, et al., J Neurophysiol 1997.
2. Cho ZH, et al., Neuroimage 2008.
Fig. 1. Quantitative velocity analysis of fPCA
128
MR-Angioclub East Lansing 2009
P.11 Optimization of Phase-contrast MR-based Flow Velocimetry
and Shear Stress Measurement
Taeho Kim1, Jihyea Seo1, Seongsik Bang1, Hyeonwoo Choi1, Yongmin Chang2, Tae-Seob Chung3,
Jongmin Lee4
1
2
4
Department of Biomedical Engineering, Molecular Medicine, and Radiology, Kyungpook National
3
University; Department of Radiology, Yonsei University
Purpose: To measure the pixel-by-pixel flow velocity and shear stress from PC MR
images, an optimized method was suggested and verified.
Material and Methods: A self-developed straight steady flow model was scanned by 3T
MRI with a velocity-encoded PC sequence. The pixel-by-pixel flow velocity and shear
stress was measured using self-made program and were compared with the physically
measured reference data. A comparison between the program and a commercial
velocimetry system was also performed. Subsequently velocity and shear stress were
measured in curved steady flow model to confirm shifted peak velocity and shear stress
toward the outer side of lumen.
Results: The velocity measured with self-made program showed a significant correlation
with the physically measured velocity and was superior to the commercial system (R2 =
0.85 vs. 0.75, respectively). The calculated mean shear stress had a significant correlation
with the physically measured mean shear stress (R2 = 0.95). The curved flow model
showed a significantly shifted peak velocity and shear stress zones toward the outside of
the flow.
Conclusion: The technique to measure pixel-by-pixel velocity and shear stress of steady
flow from velocity-encoded phase-contrast MRI was optimized and verified to be superior
to the use of a commercial system.
MR-Angioclub East Lansing 2009
129
P.12 Blood Flow Patterns in the Abdominal Aorta of Mice:
Implications for AAA Localization
1
1
3
2,3
1,2
Smbat Amirbekian , Robert Long , Michelle Consolini , W. Robert Taylor , John Oshinski
Department of Radiology1, Biomedical Engineering2, and Medicine (Cardiology)3, Emory University
School of Medicine, Atlanta, GA
Introduction: Abdominal aortic aneurysms (AAA) localize almost exclusively in the infrarenal aorta in man. Humans experience a period of reverse flow during early diastole in
the infra-renal aorta during each cardiac cycle. This reverse flow causes oscillatory wall
shear stress (OWSS) to be present in the infra-renal aorta of humans and has been linked
to AAA formation. In contrast to humans, all mouse models of AAA form aneurysms in the
supra-renal aorta. The presence of reverse flow in the mouse aorta is unknown.
Purpose: The goal of this study was to examine flow patterns in the mouse aorta and
evaluate whether reverse flow exist in the infra-renal or supra-renal aorta.
Methods: Blood flow was measured with a phase contrast magnetic resonance (PCMR)
sequence the supra-renal and infra-renal abdominal aorta of 18 wild type C57BL/6 mice
and 15 ApoE-/- mice (most prevalent mouse model of AAA). Measurements were made
using a 4.7T Varian system.
Results: Results indicate that unlike humans, there is no reversal of flow in the infra-renal
or supra-renal aorta of wild type or ApoE-/- mice. Distensibility of the mouse aortic wall as
a percent of diameter (63 + 18%), is higher than reported values for the human aorta.
Conclusion. Wild type and ApoE-/- mice do not experience the reverse flow associated
with OWSS and AAA in the infra-renal aorta that is observed in humans.
130
MR-Angioclub East Lansing 2009
P.13 3D Vessel Wall imaging of multiple vascular beds
1
Niranjan Balu , Jinnan Wang2, Chun Yuan1
1.
University of Washington, Seattle, WA
2.
Philips Medical Systems, BriarCliff Manor, Ny
Introduction: Black-blood MRI of atherosclerotic plaque allows identification of plaque
morphological and compositional features and thereby helps in assessing disease burden
and risk of thromboembolism due to plaque
Representative
rupture. Recently, a fast isotropic 3D black-blood
images
(a)
b
sequence for carotids has been shown to provide
Sagittal carotid
with
calificate
comprehensive plaque information in all spatial
(b)
Montage
directions [1]. Since atherosclerosis is a systemic
from slices to
show
entire
disease, plaque is coexistent in multiple vascular
femoral artery
beds. 3D MRI of multiple vascular beds is an ideal
covered
(c)
tool
to
study
the
pathophysiology
of
Cross-section
of
abdominal
atherosclerosis as a systemic disease.
aorta
Aim: To develop a time-efficient 3D isotropic
imaging protocol for carotid vessel wall imaging of
a
the carotids, aorta and femoral arteries
Methods: A 3D FLASH sequence with MSDE [2]
preparation was optimized (table) for carotid, aorta
and femoral imaging.
Results: Vessel wall images of diagnostic image
quality were obtained in the carotids, aorta and
femoral arteries (figure). Scan time was
approximately 2 min, 3.5 min and 10 min for
coverage of approximately 15 cm, 16 cm and 50
cm in the carotids, femoral and aorta respectively.
Conculsions: Time-efficient vessel wall imaging
c
of large arteries can be achieved within a one-hour
scan time by use of the new sequence.
References:
[1] Balu N et al, ISMRM 09 [2] Wang et al, MRM
07;58:973-981
Vascular Bed
Acquisition plane
Resolution, mm2
Field-Of-View, mm2
Slice thickness , mm
# of slices
TR/TE, ms
Flip angle, °
Turbo factor
Bandwidth, Hz/pixel
Number of averages
Number of stations
Scan time, min:s
Carotid
Coronal
0.7
250×160
0.7
100
10/4.8
6
90
134.3
1
1
2:03
Aorta
Coronal
1.4
350×300
1.4
86
7.7/3.7
6
35
269.1
1
1
3.23
Femoral
Coronal
1.0
300×350
1.0
150
7.6/3.5
6
100
191.6
1
3
10.24
MR-Angioclub East Lansing 2009
131
P.14 Feasibility Study of Combining 3D SSFP with T2prep
Inversion Recovery (T2IR) for Black Blood Vessel Wall Imaging
Keigo Kawaji1,2, Thanh Nguyen2, Pascal Spincemaille2, Martin R. Prince2, Yi Wang1,2
1
Department of Biomedical Engineering, Cornell University, Ithaca, NY.
2
Department of Radiology, Weill Cornell Medical College, New York, NY.
Background:
T2IR preparation vessel wall imaging provides flow-insensitive global
black-blood (BB) suppression at the expense of SNR [1-3].
A recent study [3] has
demonstrated the feasibility of T2IR preparation in 2D Fast-Spin-Echo for BB vessel wall
imaging. In this report, we examine the feasibility of imaging vessel walls using a 3D
Steady-State Free Precession (SSFP) sequence with T2IR preparation.
Methods: 4 healthy volunteers were scanned using a 4-channel cardiac coil for signal
reception. Axial views of the popliteal artery were acquired using gated 3D SSFP with
T2IR preparation on a 1.5T scanner (GE Signa HDx) using the following scan parameters:
imaging matrix 256x256x50, slice thickness=2mm, flip angle=60o, FOV=16cm, NEX=2,
VPS=100,
TE=1.6ms,
TR=4.0ms,
TEeff=120ms,
TI=225-375ms, with a scan time of approximately 4
mins for a nominal heart rate of 60bpm.
Fat
saturation was also used.
Results:
a
)
c)
Figures 1 a,b) show axial views of the
popliteal artery, and 1 c) shows a reformatted view
from the 3D T2IR SSFP images. The vessel walls
were clearly depicted in these images.
Discussion:
b
)
Preliminary data demonstrate the
feasibility of BB imaging of the vessel wall using a 3D
T2IR prepared SSFP sequence.
References: 1. Brittain et al. MRM 1997; 2. Liu et al.
ISMRM 08 pp3079; 3. Nguyen et al. ISMRM 09 pp607.
132
Figure 1: a) Axial view of 3D T2IR
SSFP image of knee. b) Popliteal
artery c) Reformatted view.
MR-Angioclub East Lansing 2009
P.15 Identification of Optimal First-Order Gradient Moment for
Flow-Sensitive Dephasing (FSD) Preparation
Z. Fan1,2, X. Bi3, and D. Li1,2
Radiology, 2Biomedical Engineering, Northwestern University, Chicago, IL, USA;
3
Siemens Medical Solutions USA, Inc., Chicago, IL, USA
1
Purpose: The first-order gradient moment, m1, is a measure of the blood signalsuppression capability of the FSD preparative module. This work aimed to develop an m1scout approach to rapidly identify the optimal m1, which may help improve vessel wall
imaging and FSD-based MRA quality.
Methods: FSD-induced signal suppression is voxel size-dependent as its underline
mechanism is the intravoxel velocity variation [1]. For a FSD-prepared 3D isotropic
resolution imaging with major flows in the readout direction, we proposed to run the same
imaging seqence in 2D mode to rapidly identify the optimal m1. This requires the 2D
imaging plane perpendicular to the major vessel of interest, FSD gradient pulses applied
in the slice-select direction, and the in-plane resolution identical to that of 3D imaging. We
hypothesized that the flow-void effects to occur in the 3D case could be reflected by the
2D case with the same m1 used. This approach was tested on flow phantom (Gd-doped
water 0.25mM, a silicone tube of 4.8-mm ID connected to a pump) at various flow rate (15,
20, 30, 40, 50, 60 cm/s). The 2D m1-scout sequence was implemented based on FSDprepared balanced SSFP which was also used in 3D imaging for verification. Spatial
resolution: isotropic 0.9-mm for 3D, 0.9-mm in-plane and 5-mm thickness for 2D. The 2D
scan continuously acquired 11 measurements (images) (Fig. 1), whereas the 3D scan
acquired only 6 images due to long scan time.
Results: The lumen signal intensity measurements from the 2D and 3D images were
significantly correlated at all the velocities tested (Mean Pearson correlation coefficient =
0.988, p < 0.001). Fig. 2 shows the case of flow rate = 20 cm/s.
Conclusion: The optimal m1 value may be identified using the fast 2D scout approach. In
vivo experiments are underway to further verify its applicability.
References: [1] Nguyen TD et al. JMRI 2008; 28:1092-100.
Fig 1. Schematic of the 2D m1-scout scan. A total 11 images were
collected. The first uses m1 = 0, while the latter uses incremental
m1 values (user specified.)
Fig 2. Corresponding 2D and 3D images (a) and
signal intensity measurements (b).
2D
3D
a
60
80
100
120
140
1.20
Signal Intensity
m1= 0
2D Scout
1.00
0.80
V = 20 cm/m
0.60
3D Scan
0.40
0.20
0.00
b
0
60
70
80
90
100 110 120 130 140 150
First-order gradient moment (mT.ms2/m)
MR-Angioclub East Lansing 2009
133
P.16 Combined Segmentation of Lumen and Intraplaque
Hemorrhage in Black-blood T1-Weighted Carotid Imaging
Rahul Sarkar1, General Leung1,2 and Alan R. Moody1,2
University of Toronto and 2Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
1
Purpose: We investigated the use of a novel automatic segmentation approach to allow
localization of intraplaque hemorrhage (IPH) with respect to the lumen in black-blood T1
weighted carotid images.
Methods: From the original image, a fuzzy connectedness map is produced using a
generalized fuzzy spel affinity [1] for lumen detection.
The fuzzy thresholded lumen
contour is downsampled to form control points for a b-spline curve to smooth image noise
at the boundary. A radial distance metric from the segmented lumen center of mass is
combined with intensity thresholding for segmentation of IPH. Images used for training
the fuzzy spel affinity and testing were obtained as previously described [2].
Results: Figure 1 shows the segmentation results produced from a representative image
showing intraplaque hemorrhage. The fuzzy segmentation step is highly efficient at gross
lumen detection, but demonstrates sensitivity to noise along border pixels. The derived bspline curve is effective in smoothing the border noise without significantly distorting the
lumen shape. IPH is accurately detected by thresholding within a defined radial distance
metric (5mm) from the segmented lumen.
Conclusion: The described approach is effective in automatically segmenting the lumen
and IPH to allow their co-localization in black blood T1weighted images.
Figure 1. From left: 1) Original image; 2) Fuzzy detected lumen (red) showing border noise
(blue arrow); 3) b-spline corrected fuzzy lumen (red) showing reduced border noise (green
arrow); 4) Corrected lumen (red) and automatically segmented IPH (yellow).
References: [1] Carvalho PAA 1999. [2] Leung ISMRM 2009.
134
MR-Angioclub East Lansing 2009
P.17 Automatic Registration of Multiparametric T1 Weighted
Images Using FOV-Selective Mutual Information
Rahul Sarkar1, General Leung1,2 and Alan R. Moody1,2
University of Toronto and 2Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
1
Purpose:
Multiparametric T1weighted imaging may be useful in identifying several
features of atherosclerotic plaque [1]. Here we investigate a technique to automatically
and reliably register the parametric images using normalized mutual information [2].
Methods:
All algorithm and user interface development was done in MATLAB (The
Mathworks). Mutiparametric datasets were obtained as previously described [1]. For
each dataset, a normalized mutual information algorithm was applied to register blackblood post-contrast and bright-blood delayed enhancement images to the reference blackblood pre-contrast image. Algorithm performance was observed under different field of
view (FOV) selections for reference and non-reference images.
Results: The algorithm was effective in accurately registering images of various nonreference image FOVs (Figure 1). Reference image FOV was an important factor in
algorithm efficiency. Small reference FOVs (40mm x 40 mm) including the vessel wall and
few surrounding features proved optimal in terms of accuracy and reduced computational
time significantly. Deformable motion effects in large reference FOVs sometimes led to
misregistration that was corrected by reducing the FOV size.
Conclusion: Multiparametric T1 weighted images may be accurately co-registered with a
normalized mutual information algorithm using small reference image FOVs.
Figure 1. Multiparametric T1
images with pre-contrast black
blood (left), post-contrast black
blood (middle), and bright blood
delayed enhancement (right).
Top panel shows unregistered
images of different FOVs.
Bottom panel shows the same
images registered to the
reference pre-contrast image.
References: [1] Leung ISMRM 2009. [2] Collignon IPMI 1995.
MR-Angioclub East Lansing 2009
135
P.18 MR fluid dynamics using 4D-Flow for intracranial aneurysms
with growing blebs and a ruptured intracranial aneurysm
Haruo Isoda1, Hisaya Hiramatsu2, Yasuhide Ohkura3, Takashi Kosugi3, Masaya Hirano4, Shuhei
Yamashita1, Yasuo Takehara1, Marcus T. Alley5, Harumi Sakahara1; 1Department of Radiology and
2
Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan;
3
4
Renaissance of Technology Corporation, Hamamatsu, Japan; GE Yokogawa Medical Systems, Hino,
Japan; 5 Department of Radiology, Stanford University School of Medicine, Stanford, USA.
PURPOSE: The purpose of our study was to investigate the relationship between
hemodynamics and growing blebs or intracranial aneurysm ruptures with the use of MR
fluid dynamics (MRFD) using 4D-Flow (1).
METHODS: This study included an unruptured 6mm right IC-Ach artery aneurysm with
growing bleb, an unruptured 4mm left IC aneurysm (C2 and C3 segment) with growing
bleb and a ruptured 8mm right IC-Ach artery aneurysm with hematoma in the medial
aspect of the right temporal lobe. 4D-Flow was performed by a 1.5T MR scanner. We
calculated and evaluated streamlines, wall shear stress (WSS) and the oscillatory shear
index (OSI) of these intracranial aneurysms based on 4D-Flow with our in-house software.
RESULTS: The top of the spiral flow was near the bleb in the unruptured IC-Ach artery
aneurysm. The WSS was lower and the OSI was higher near the bleb. The top of the
spiral flow was present at the growing bleb of the unruptured IC artery aneurysm and the
WSS was lower and the OSI was higher at this point. A disturbed spiral flow was noted in
the ruptured IC-Ach artery aneurysm. Lower WSS and higher OSI were observed in an
area containing the rupture point.
CONCLUSION: Bleb formation or rupture points were observed near the apex of the spiral
flow with low WSS and high OSI. Based on previously published papers reporting that low
WSS or high OSI causes degeneration and apoptosis of endothelial cells, we
hypothesized that endothelial cells at the apex of the spiral flow in the aneurysms were
much more vulnerable to degeneration or apoptosis.
REFERENCES: 1. Markl M, et al. J Magn Reson Imaging. 2003;17:499-506.
136
MR-Angioclub East Lansing 2009
P.19 Phase-Field Dithering for Active Catheter Tracking
1
Charles L. Dumoulin , Richard P. Mallozzi2, Robert D. Darrow3, and Ehud J. Schmidt4
1
2
3
Cincinnati Children’s Hospital, ONI Medical Systems, Inc., General Electric Global Research, and
4
Brigham and Women’s Hospital
Purpose: Active MR tracking can become difficult in low Signal-to-Noise (SNR)
conditions, especially when magnetic susceptibility of the device is not well matched to its
surroundings and when unwanted MR signal is coupled into the receive channel (e.g. from
poorly shielded cables).
A strategy to increase the robustness of active MR tracking of
micro-coils in these conditions was developed, and tested.
Methods: The method employs dephasing magnetic field gradient pulses that are applied
orthogonal to the frequency encoding gradient pulse used in conventional point-source
MR tracking.
In subsequent acquisitions the orthogonal dephasing gradient pulse is
rotated, while maintaining a perpendicular orientation with respect to the frequency
encoding gradient. The dephasing gradient creates a spatially-dependent phase shift in
directions perpendicular to the frequency encoding gradient. Since the desired MR signal
for robust MR tracking comes from the small volume of nuclear spins near the small
detection coil, the desired signal is not dramatically altered by the dephasing gradient.
Undesired MR signals arising from larger volumes (e.g. due to coupling with the body
and/or surface coils), on the other hand, are dephased and reduced in signal intensity.
Since the approach requires no a priori knowledge of the micro-coil orientation with
respect to the main magnetic field, data from several orthogonal dephasing gradients is
acquired and analyzed in real-time. One of several selection algorithms is employed to
identify the “best” data for use in coil localization.
Results and Conclusions:
This approach was tested in flow phantoms and animal
models, with several multiplexing schemes, including the Hadamard and zero-phase
reference approaches. It was found to provide improved MR tracking of un-tuned microcoils. It also dramatically improved MR tracking robustness in low SNR conditions and
permitted tracking of micro-coils that were inductively coupled to the body coil.
MR-Angioclub East Lansing 2009
137
P.20 Interventional Device Tracking and Imaging Using an
Extensible Real-Time System
Ethan Brodsky and Orhan Unal
Radiology and Medical Physics, University of Wisconsin, Madison, WI, USA
PURPOSE: To demonstrate tracking of interventional devices using RTHawk, extensible
software architecture for real-time MR imaging.
METHODS: RTHawk is a software architecture that permits a variety of pulse sequences,
acquisition trajectories, and reconstruction techniques to be easily developed and
interleaved at run-time on GE scanners [1]. We have previously demonstrated
simultaneously tracking a catheter and imaging an endovascular device using a customdesigned software solution that was tied to a particular pulse sequence implementation
and was very difficult to modify or extend [2,3]. We have therefore implemented two
catheter tracking techniques in RTHawk – the previously described 3DPR technique that
is amenable to simultaneous tracking and imaging, as well as a simple three-orthogonalview tracking acquisition that can easily be interleaved with Cartesian imaging.
RESULTS AND DISCUSSION: Initial testing shows that the previously developed tracking
and imaging techniques can be easily adapted to run in this environment, without requiring
any pulse sequence programming. A real-time display shows one-dimensional profiles for
each axis from which the signal peak is detected and tracked.
CONCLUSIONS: Initial results suggest that interventional sequences can easily be
adapted to work within this framework. It abstracts away much of the complexity of pulse
sequence and reconstruction programming, allowing simple intuitive representations of the
acquisition trajectory and reconstruction pipeline. Visualization of final and intermediate
results is also greatly simplified.
Figure 1: These one-dimensional profiles show the signal picked up by the endovascular
device’s tip tracking coil for projections along each of the orthogonal gradient axes. Pulse
sequences was Fast GRE, 4° flip, 12.5 ms TR, 256 sample readout over a 40 cm FOV for
a 1.6 mm resolution.
Figure 2: This plot of peak position over time shows the tip position as the device is
moved in a circle in the x-z plane.
REFERENCES: [1] Santos et al. Proc IEEE Eng Med Biol Soc. 2:1048,’04
[2] Brodsky et al. MRM 56:247, ‘06
[3] Unal et al. Proc MRA Club 18:4.2, ’06
138
MR-Angioclub East Lansing 2009
P.21 Simplified Catheter-based Multimode Coil for Active MR
Tracking and Intravascular Imaging
Mahdi Salmani Rahimi, Krishna Kurpad, Orhan Unal
Departments of Biomedical Engineering, Radiology and Medical Physics, University of WisconsinMadison
PURPOSE: To simplify the design of a multi-mode intravascular MR catheter-based coil
that combines the functionalities of an active tip-tracking coil and an imaging coil and to
resolve the ambiguity of having two tip-tracking signal peaks.
METHODS: Unlike earlier designs [1] that had a separate solenoid at the catheter tip to
provide the localized tracking signal, in this design the inductor in the required matching
network of the tuned imaging coil has been exploited for tip tracking. Quarter-wavelength
π-matching network was placed
a
at the distal tip with a wirewound solenoid as the inductor.
1a
shows
the
coil
schematic. Cable lengths were
c
Tracking Coil
Decoupling Box
and GE 1.5T
Scanner
Amp.
Figure
b
Imaging Loop
(λ/4 long)
adjusted so that the solenoid in
the
matching
network
is
in
resonance during the receive
cycle of the scan. To increase
the number of turns in the
inductor to improve tip-tracking
signal
and
avoid
λ/4 Transformer
Freq.
Figure 1: (a) Schematic of the catheter coil design. (b) 313
micron resolution axial image of a grapefruit obtained with the
imaging loop using a 2D GRE technique. (c) Signal profile from
manual prescan, TG=100. Tracking coil peak is visible and
easily detectable from broader imaging signal in the coil with
one counter turn.
unwanted
increase in the competing imaging signal, some turns in the imaging coil have been wound
in the opposite direction.
RESULTS: Figure 1b shows a high resolution axial image of a grapefruit obtained with the
imaging loop of the multimode probe using a 2D GRE sequence (FOV=80x80mm,
256X256, TR/TE=200/3.1 ms, FA=90o, and NEX=5). Since there is only one solenoid in
this design, the ambiguity in determining tip position caused by having more than one
localized peak in earlier designs can also be avoided as seen in Figure 1c.
CONCLUSIONS: Our preliminary results suggest that multimode intravascular probes can
be
simplified
while
improving
active
tip-tracking
and
imaging
functionalities.
REFERENCES:
[1] Unal O, et al. ISMRM 1398, 2006.
MR-Angioclub East Lansing 2009
139
P.22 Transmit Power Optimization for Tracking, Wireless Marker
and Imaging applications of a Multi-mode Endovascular coil.
Krishna N. Kurpad and Orhan Unal
Departments of Radiology and Medical Physics, University of Wisconsin-Madison
PURPOSE: To optimize body coil transmit power settings to manipulate transverse
magnetization in various regions in the vicinity of a MR multi-mode endovascular coil for
optimal operation in tip tracking, wireless marker, and endovascular imaging modes.
METHODS: The multimode coil 1,2 (Fig. 1a), being inductively coupled to the transmit coil,
behaves as a local B1 field magnifier. The seriesconnected tip tracking and imaging components of
the multimode coil are constructed with different
conductor densities, resulting in different transverse
magnetization in their vicinity. A series of 21 coronal
images of a tube phantom containing saline solution
doped
with
copper
sulphate
(T1=150ms)
was
obtained using a FGRE pulse sequence (TR/TE= 200/4 ms, FA=90o, FOV=6cm, 256x256)
on a GE Signa 1.5T clinical scanner. The images were obtained by incrementing the
transmit gain setting in steps of 1 dB between the limits of 0 dB and 20 dB. Graphs of the
mean signal intensity in three small regions (Fig. 1b) located close to 1) the tracking coil,
2) the conductors of the imaging coil, and 3) one cm away from the imaging coil were
plotted against the transmit gain setting for the corresponding images.
RESULTS: The graphs in Fig. 2 show that transmit power settings may be suitably
adjusted to generate 1) distinct tracking
peak, 2) broad region of sensitivity for
imaging, or 3) visualize the conductors of
the multimode coil for wireless marker
applications.
CONCLUSIONS:
Preliminary
results
show that the current induced in the
multimode coil by the transmitted B1 field
results
in
magnetization
varying
due
to
transverse
the
varying
conductor density of its various components. The transmit power may therefore be
optimized for optimal operation of the multimode coil in all three functional modes.
REFERENCES:1Kurpad KN,et al, ISMRM 292, 2007, 2Unal O, et al. ISMRM 1398, 2006
140
MR-Angioclub East Lansing 2009
Sponsors
Lantheus Medical Imaging, a worldwide leader in diagnostic
imaging for 50 years, is committed to developing innovative
solutions for the diagnosis and management of disease. For more
information, please visit www.lantheus.com.
"Bayer Schering Pharma AG has a worldwide leading position in
developing and manufacturing contrast media for use in x-rays,
computed tomography (CT), magnetic resonance imaging (MRI)
and in contrast media application systems.
www.bayerhealthcare.com
Bracco Imaging S.p.A, is one of the World’s leading companies in
the diagnostic imaging business. Bracco Imaging S.p.A products
are distributed in USA by Bracco Diagnostic Inc., Princeton.
www.braccoimaging.com
GE Healthcare's expertise in medical imaging and information
technologies, medical diagnostics, patient monitoring systems,
disease research, drug discovery and biopharmaceuticals is
dedicated to detecting disease earlier and tailoring treatment for
individual patients. For more information visit
www.gehealthcare.com
syngo TimCT offers continuous table move for CT-like scanning.
Setting the trend in MRI with MAGNETOM ESSENZA, Avanto,
Espree, and Verio. Siemens. Answering the questions of life.
www.siemens.com/mr
Guerbet is the only pharmaceutical group fully dedicated to
medical imaging, offering a comprehensive range of contrast
agents for X-ray and MRI worldwide. Guerbet’s innovation focuses
on molecular imaging for MRI or nuclear medicine applications,
enabling diseases to be targeted earlier and more precisely.
www.guerbet.com
MR-Angioclub East Lansing 2009
141
Sponsors
Philips Healthcare is a premier supplier of healthcare solutions.
The MR portfolio provides unique, advanced features enabling our
users to extend the realms of vascular MR imaging.
www.medical.philips.com
TopSpins Inc. is dedicated to helping MR imaging centers
implement safe, accurate, state-of-the-art contrast enhanced MR
Angiography and dynamic contrast-enhanced MR imaging studies
for the benefit of their patients. www.topspins.com
Robarts Research Institute harnesses the power of cell biology,
genomics and advanced imaging technologies to investigate
disorders of the neurological, cardiovascular and immune
systems. It is a research Institute within the Schulich School of
Medicine & Dentistry at The University of Western Ontario.
www.robarts.ca
Founded in 1878, The University of Western Ontario is one of
Canada's oldest universities and today is committed to providing
the best student experience among Canada's leading researchintensive universities. A vibrant centre of learning located in
London, Ontario, Western is home to more than 30,000 students
and attracts external support of over $200 million annually for
research projects. www.uwo.ca
As an integral part of the Michigan State University HealthTeam,
the Department of Radiology has served the Mid-Michigan
community since 1975. Our dedicated team of health care
professionals has pioneered research and clinical applications in
radiology. www.rad.msu.edu
142
MR-Angioclub East Lansing 2009
Insert Lantheus file
MR-Angioclub East Lansing 2009
143
Notes
144
MR-Angioclub East Lansing 2009
Notes
MR-Angioclub East Lansing 2009
145
Notes
146
MR-Angioclub East Lansing 2009
Notes
MR-Angioclub East Lansing 2009
147
Presenter Index
Page
A
Abela, George
The Role of Cholesterol Crystals in Acute Cardiovascular Events:
Indentifying the Cause for Gender Differences in Clinical Presentation
Admirral - Behloul, Faiza
Hybrid of Opposite Contrast MR Angiography of the Brain
Artz, Nathan
Assessing Kidney Perfusion using Arterial Spin Labeling and Radial
Acquisition for Rapid Characterization of Inflow Dynamics
Aschauer, Manuela
Gadofoveset Excretion into Human Breast Milk
CE-MRA with tailored 3D random sampling patterns and nonlinear parallel
imaging reconstruction (P1)
118
73
101
54
119
B
Balu, Niranjan
3D Vessel Wall imaging of multiple vascular beds (P13)
Barnes, Samuel
High Resolution Simultaneous Angiography and Venography (MRAV) with
a Single Echo
Bernstein, Matt
Multicenter Studies: Lessons Learned from ADNI
Bhat, Himanshu
Contrast-Enhanced Whole-Heart Coronary MRA at 3T Using Gradient
Echo Interleaved EPI (GRE-EPI)
Blackham, Kristine
Robust Clinical Application of Time-Resolved MRA(P7)
Bley, Thorsten
Non-invasive Trans-Stenotic Pressure Measurements with 3D Phase
Contrast MRA: Validation against Endovascular Pressure Measurements
in Swine
Block, Walter
Imaging Capabilities for Real-time Guidance and Verification of
Transcatheter Arterial Chemoembolization (TACE) Procedures
Bousell, Loic
4D time-resolved MR angiography for non-invasive pulmonary postembolization AVM patency assessment
Brodsky, Ethan
Interventional Device Tracking and Imaging Using an Extensible Real-time
System (P20)
131
76
114
90
125
61
102
83
138
C
Carr, James
Non Contrast MRA of the Hand in Patients with Raynauds disease using
Flow Sensitized Dephasing Prepared SSFP
Carroll, Timothy
Radial Sliding Window MRA in Pulmonary Hypertension
Choi, Grace
MRA with the “No Phase Wrap”
Chung, Tae-Sub
Obstruction of IJV by Asymmetry of Lateral Mass of Atlas on Head and
Neck CEMRA and Contrast CT
148
MR-Angioclub East Lansing 2009
50
84
86
78
Author Index
Page
D
Dumoulin, Charles
Phase-Field Dithering for Active Catheter Tracking (P19)
137
E
El-Boubbou, Kheireddine
Targeted Glyco-Magnetic Fe304 Nanoprobes for Detection and Molecular
Imaging of Atherosclerosis
Essig, Marco
Intraindividual comparison between multislice CT and 4D TWIST MRA in
the assessment of residual cerebral ateriovenous malformations – a
prospective study protocol
110
71
F
Fan, Zhaoyang
3D Noncontrast MRA using FSD – Prepared Balanced SSFP
Identification of Optimal First-Order Gradient Moment for Slow-Sensitive
Dephasing (FSD) Preparation (P15)
Fielden, Samuel
Balanced-gradient TSE for Non-Contrast Peripheral MRA
Francois, Christopher
Flow assessment of arterial dissections using 3D radial phase contrast MR
Angiography
Frydrychowicz, Alex
Analysis of aortic hemodynamics after treatments for coarctation using
flow-sensitive 4D MRA at 3T
49
133
27
63
62
G
Ge, Lan
Myocardial Perfusion MRI in Canines with Improved Spatial Coverage,
Resolution and SNR
Geerts, Liesbeth
Non-CE Imaging of the Pulmonary Arteries
Goldfarb, James
Cardiac Imaging: Methods for the Detection of Intramyocardial Fat
Grabow, Ben
Temporal Filtering for Sliding Window Time-resolved Angiography; Beyond
Density Compensation Solutions
Grist, Thomas
Overview of Gd-BOPTA Phase III Trial for CEMRA: What are the results,
and how do we move forward?
Griswold, Mark
A Simple View of Compressed Sensing and How it Could Change
Everything We Do in MRI and MRA
32
24
92
94
55
29
H
Haacke, Mark
High Resolution Perfusion Weighted Imaging
66
Susceptibility mapping as a means to image veins
104
MR-Angioclub East Lansing 2009
149
Author Index
H
Hadley, Rock
A 16 Channel Anterior Neck RF Coil for Cervical Carotid MRA
Haider, Clifton
A Comparison of Time-Resolved 3D CE-MRA with Peripheral Run-off CTA
in the Calves
Hibberd, Mark
An Update on the Clinical Experience with Gadofosveset
Hu, Peng
Non-Contrast Enhanced Pulmonary Vein MRI with a Spatially Selective
Slab Inversion Preparation
Hulet, Jordan
Improved Carotid Imaging with HASTE using a reduced FOV and
increased gradient performance (P3)
Page
42
47
52
85
121
I
Igase, Keiji
Our Strategy for the Surgical Planning with 3T MRA in Detecting
Unruptured Cerebral Aneurysms
Isodo, Haruo
MR fluid dynamics using 4D-Flow for intracranial aneurysms with growing
blebs and a rupture intracranial aneurysm (P18)
72
136
J
Jeong, Hyun
CAMERA: Contrast-enhanced Angiography with Multi-Echo and Radial kspace
Johnson, Casey
Two-Station Time-Resolved CE-MRA of the Lower Legs
Johnson, Kevin
Accelerated Time Resolved Inflow with 3D Radial bSSFP
Angiographic and Hemodynamic Assessment of the Hepatic Vasculature
in Portal Venous Hypertension using High Resolution PC VIPR
106
51
25
95
K
Kang, Chang-Ki
Vascular response during visual stimulation at 3T MRI: functional phase
contrast angiography (fPCA) study (P10)
Kawaji, Keigo
Feasibility Study of Combining 3D SSFP with T2prep inversion Recovery
(T2IR) for Black Blood Vessel Wall Imaging (P14)
Kecskemeti, Steven
Stack of Stars 4D Phase Contrast Velocimetry of the Circle of Willis
Kerwin, William
Fibrous Cap Thickness Assessment: Fact or Fiction?
Kim, Bum-soo
Low Dose 3D Time-Resolved MR Angiography of the Supraaortic Artery:
Correlation to High Spatial Resolution 3D Contrast-Enhanced MRA
Kim, Seong-Eun
Improved Black Blood Multi-Contrast Protocol for In-vivo Atherosclerotic
Imaging
150
MR-Angioclub East Lansing 2009
128
132
65
38
77
40
Author Index
K
Page
Kurpad, Krishna
Transmit Power Optimization for Tracking, Wireless Marker and Imaging
applications of a Multi-mode Endovascular coil (P22)
140
Page
L
Ladd, Mark
Toward Abdominal MRA at 7 Tesla
Latourette, Matthews
R*2 Calibration Phantoms for Cardiovascular Studies (P5)
Laub, Gerhard
Low-Dose 4D MR Angiography
Lauzon, Louis
Non Contrast-Enhanced MR identification of DVT
Lee, Jongmin
Optimization of Phase-Contrast MR-based Flow Velocimetry and Shear
Stress Measurement (P11)
Leiner, Tim
Gadobenate dimeglumine vs. gadopentetate dimeglumine for peripheral
MR angiography: Comparison with DSA
Li, Rui
Gradient Echo Based Sequence Provides More Information from Ex Vivo
Carotid Plaque Specimens
Liu, Garry
Ultrasound guided cardiac gating for coronary MRA
Liu, Jing
Self-gated Free Breathing 3D Cardiac Cine Imaging with Data Acquisition
During Slice Encoding
Lu, Zheng-Rong
Manganese Based Biodegradable Macromolecular MRI Contrast Agents
for Cardiovascular Imaging
116
123
26
103
129
43
39
89
28
60
M
Maki, Jeffrey
Dose Comparison between Conventional and High Relaxivity Contrast
Agents in Peripheral MRA
Marinelli, Luca
Accelerated velocity imaging using compressed sensing
Markl, Michael
Wall Shear Stress in Normal and Atherosclerotic Carotid Arteries
McNally, Scott
MR imaging and significance of flow reversal and carotid atherosclerosis:
Initial results
Mendes, Jason
Handling Motion in Sparse MRA with Whiskers (P2)
Miki, Hitoshi
Unruptured Intracranial Aneurysms; Detection and Follow-up on 3.0T MRA
Mistretta, Chuck
4D DSA and Fluoroscopy: A New Challenge for MRA?
MR-Angioclub East Lansing 2009
44
67
68
69
120
87
111112
151
Author Index
Page
M
Mostardi, Petrice
Modified CAPR MRA: Improved Imaging of the Arterial and Venous
Phases
105
Page
N
Newman, Tiffany
Magnetic Resonance Angiography of the skin for perforator-based
autologous breast reconstruction
98
O
Oshinski, John
Blood Flow Patterns in the Abdominal Aorta of Mice: Implications for AAA
Localization (P12)
Ota, Hideki
Carotid Intraplaque Hemorrhage is Associated with Enlargement of Lipidrich Necrotic Core and Plaque Volume Over TimeL In Vivo 3T MRI
Prospective Study
130
35
P
Parienty, Isabelle
Time-SLIP versus DSA in Patients with Renal Artery Stenosis
Parsons, Edward
A Re-analysis of MS-325 (gadofoveset trisodium) Clinical Trial Date in
Support of US-FDA Approval
Peters, Dana
3D spiral high- resolution late gadolinium enhancement
Prince,Martin
Risk Factors for NSF: A Meta-analysis
99
53
93
58
Q
Quick, Harald
7 Tesla Cardiac MRA in Humans
117
R
Rahimi, Mahdi Salmani
Simplified Catheter-based Multimode Coil for Active MR Tracking and
Intravascular Imaging (P21)
Reeder, Scott
High Temporal and High Spatial Resolution Perfusion Imaging of
Hepatocellular Carcinoma of the Liver
Robson, Philip
Time-Resolved, Vessel-selective, Cerebral Angiography Using Arterial
Spin Labeling
Roditi, Giles
Retrospective 7 year study of the Incidence of Nephrogenic System
Fibrosis in Patients Investigated with Gadolinium Contrast-Enhanced
Renal Magnetic Resonance Angiography
Pictorial Review of Supra-Aortic Artery Pathologies as Visualised with
MRA using Blood Pool Contrast Agent (P6)
152
MR-Angioclub East Lansing 2009
139
64
107
57
124
Author Index
S
Saloner, David
Imaging Considerations in Serial Studies of Vascular Disease
Saranathan, Manojkumar
FINESS (Flow Inversion-prepared Non-contrast Enchancement in the
Steady State): A novel technique for non-contrast renal MRA
Sarkar, Rahul
Combined Segmentation of Lumen and Intraplaque Hemorrhage in Blackblood T1-weighted Carotid Imaging (P16)
115
97
134
Automatic Registration of Multiparametic T1 Weighted Images using FOVSelective Mutual Information (P17)
Schiebler, Mark
Pulmonary MRA in 75 patients with dyspnea
135
Origin and Frequency of artifacts in Contrast Enhanced Pulmonary MRA in
80 patients with dyspnea
Schneider, Guenther
Safety of Gadobenate dimeglumine (Gd-BOPTA) in Cardiovascular
Imaging of Pediatric Patients
81
Renal MR angiography: multicenter intraindividual comparison of
gadobenate dimeglumine and gadofosveset trisodium
Seiberlich, Nicole
Reconstruction of MR Angiography Images Using Gradient Descent with
Sparsification
Shah, Dipan
Evaluation of Gd-DOTA (DOTAREM) enhanced MRA compared to time-offlight MRA in the diagnosis of clinically significant non-coronary arterial
disease at 1.5 and 3.0 Tesla
Sheehan, John
Flow and Motion-Insensitive Unenhanced MR Angiography of the
Peripheral Vascular System – A pilot study in the lower extremity
Stafford, Randall
Towards Continuously Moving Table MCE Peripheral MRA (P4)
Stein, Paul
Gadolinium Enhanced Magnetic Resonance Angiography for Pulmonary
Embolism: Results of PIOPED III
Steinman, David
Quantifying Lumen Geometry from Routine Carotid CEMRA
Suever, Jonathan
Reproducibility of Aortic Pulse Wave Velocity Measurements Obtained
with Phase Contrast Magnetic Resonance (PCMR) and Applanation
Tonometry (P8)
96
80
56
33
79
46
122
82
108
126
T
Tan, Ek Tsoon
High Resolution Fast Inversion Recovery MRA (FIR-MRA)
MR-Angioclub East Lansing 2009
74
153
Author Index
V
Varani, James
Extracellular matrix metabolism in organ-cultured skin from patients with
end-stage renal disease: Response to gadolinium based MRI contrast
agents
Velikina, Julia
Design of Compressed Sensing Reconstruction for Highly Accelerated
Time-Resolved MR Angiography
Versluis, Bas
MR Angiography of muscular and collateral arteries in peripheral and
arterial disease: reproducibility of morphological and functional vascular
status
Voth, M
Peripheral MRA with Continuous Table (CTM) Movements in Combination
with High Temporal and Spatial Resolution TWIST – MRA with 0.1 mmol/lg
Gadobutrol at 3.0 T
59
30
113
45
W
Wang, Jinnan
Improved Intraplaque Hemorrhage Detections by a Phase Sensitive IRTFE
(SPI) Sequence
Wang, Kang
3D Time-Resolved MR Angiography of Lower Extremities using Cartesian
Interleaved Variable Density Sampling and HYPR Reconstruction
Wang, Yi
3D peripheral vessel wall MRI with flow-insensitive blood suppression and
isotropic resolution at 3 Tesla
Magnetic source MRI for quantitative brain iron mapping
Wieben, Oliver
Comprehensive PC MR Imaging in Congenital Heart Disease
Willinek, Winfried
4D-MRA in combination with arterial spin labeling for selective and
functional information in patients with AVMs
Wilson, Gregory
Motion-compensated, flow-independent, non-contrast enhanced renal MR
angiography (P9)
Wright, Katherine
Simultaneous Renal Angiography and Perfusion measurement Using
Time-Resolved MRA
Wu, Yijing
Low Dose HYPR FLOW
37
48
41
109
88
70
127
100
31
X
Xie, Jingsi
Feasibility of Whole-Heart Coronary MRA on 3 Tesla Using Ultrashort-TR
SSFP VIPR
91
Y
Yuan, Chun
Carotid Plaque Imaging and Clinical Risk Assessment
154
MR-Angioclub East Lansing 2009
34
Author Index
Z
Zhu, David
The 3D SHINE Sequence Optimizes the Quantification of Carotid
Intraplaque Hemorrhage
Zwart, Nick
3D Dual VENC PCMRA using Spiral Projection Imaging
MR-Angioclub East Lansing 2009
36
75
155