22.01.2015 Views

6th International Workshop on Breast Densitometry and Breast ...

6th International Workshop on Breast Densitometry and Breast ...

6th International Workshop on Breast Densitometry and Breast ...

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

Create successful ePaper yourself

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

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong><br />

<strong>Breast</strong> <strong>Densitometry</strong> <strong>and</strong> <strong>Breast</strong><br />

Cancer Risk Assessment<br />

San Francisco 2013<br />

SPONSORED BY<br />

SUPPORTED BY<br />

Support is being provided by the American Cancer Society, through The<br />

L<strong>on</strong>gaberger Company, a direct selling company offering home products<br />

including h<strong>and</strong>crafted baskets made in Ohio, <strong>and</strong> the L<strong>on</strong>gaberger<br />

Horiz<strong>on</strong> of Hope Campaign, which provided a grant to the American<br />

Cancer Society for breast cancer research <strong>and</strong> educati<strong>on</strong>.<br />

Hologic, Inc.<br />

Matakina <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Limited<br />

U-Systems – A GE Healthcare Company<br />

VuCOMP, Inc.


TABLE OF CONTENTS<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Sp<strong>on</strong>sors .................................................................................................................................................. 2<br />

Page<br />

Program Thursday, June 6 th .................................................................................................................. 3<br />

Program Friday, June 7 th ....................................................................................................................... 4<br />

Speakers Abstracts................................................................................................................................. 6-25<br />

Poster Abstracts...................................................................................................................................... 26-83<br />

Participants ............................................................................................................................................ 84-89<br />

Notes ........................................................................................................................................................ 90-93<br />

Feedback Form....................................................................................................................................... 94<br />

TABLE OF CONTENTS<br />

1


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Sp<strong>on</strong>sored by the<br />

Daniel <strong>and</strong> Phyllis Da Costa Funds of the CPMC Foundati<strong>on</strong><br />

Sutter Health at California Pacific Medical Center<br />

UCSF Department of Epidemiology & Biostatistics<br />

UCSF Department of Medicine<br />

UCSF Department of Radiology <strong>and</strong> Bio Medical Imaging<br />

SPONSORS<br />

Supported by<br />

California <strong>Breast</strong> Cancer Research Program<br />

American Cancer Society<br />

Support is being provided by the American Cancer Society, through The L<strong>on</strong>gaberger Company, a direct selling<br />

company offering home products including h<strong>and</strong>crafted baskets made in Ohio, <strong>and</strong> the L<strong>on</strong>gaberger Horiz<strong>on</strong> of Hope<br />

Campaign, which provided a grant to the American Cancer Society for breast cancer research <strong>and</strong> educati<strong>on</strong>.<br />

Hologic, Inc.<br />

Matakina <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Limited<br />

U-Systems – A GE Healthcare Company<br />

VuCOMP, Inc.<br />

2


PROGRAM<br />

Thursday, June <str<strong>on</strong>g>6th</str<strong>on</strong>g><br />

8:00 - 9:00 Breakfast <strong>and</strong> Registrati<strong>on</strong><br />

3<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

9:00 - 9:10 Introducti<strong>on</strong><br />

9:10 - 9:20 <strong>Breast</strong> Density - What is a Woman to do NOW"<br />

Deborah Collyar<br />

The Politics <strong>and</strong> Practicality of Reporting <strong>Breast</strong> Density<br />

9:20 - 9:45 Cauti<strong>on</strong> – Kitty Litter, Silly Putty & Dense <strong>Breast</strong>s: Using State, Federal <strong>and</strong><br />

Regulatory Efforts to St<strong>and</strong>ardize the Communicati<strong>on</strong> of Dense <strong>Breast</strong> Tissue to<br />

Women<br />

Nancy M. Cappello, PhD - President & Founder, Are You Dense, Inc.<br />

9:45 - 10:10 Discussi<strong>on</strong> of the New Legislati<strong>on</strong> M<strong>and</strong>ating the Reporting of <strong>Breast</strong> Density,<br />

Perspectives from <strong>Breast</strong> Cancer Advocates Supporting the Legislati<strong>on</strong>, Discussi<strong>on</strong><br />

of the Benefits <strong>and</strong> Practical Limitati<strong>on</strong>s of the <strong>Breast</strong> Density Reporting in a<br />

Clinical Mammography Setting.<br />

Joe Simitian, Santa Clara County Supervisor <strong>and</strong> former California State Senator<br />

10:10 - 10:50 Applicati<strong>on</strong> of California SB 1538 <strong>Breast</strong> Density Notificati<strong>on</strong> Law in Clinical<br />

Practice (Panel)<br />

Meg Durbin, MD (Palo Alto Medical Foundati<strong>on</strong>); B<strong>on</strong>nie Joe, MD, PhD (UCSF);<br />

Debra Ikeda, MD (Stanford); Ed Sickles, MD (UCSF)<br />

10:50 - 11:05 Break (15 minutes)<br />

11:05 - 11:20 ABSTRACT: CIRRUS: A Fully-Automated Software Platform for <strong>Breast</strong> Cancer<br />

Risk Predicti<strong>on</strong> from Mammograms<br />

John Hopper, PhD - The University of Melbourne<br />

11:20 - 11:35 ABSTRACT: Several Mammographic Texture Measures, Including MTR, Add<br />

Simultaneously to Percentage Density Risk Segregati<strong>on</strong><br />

Mads Nielsen, PhD - University of Copenhagen<br />

11:35 - 12:30 Lunch (45 minutes)<br />

Comparative Measurement of <strong>Breast</strong> Density<br />

12:30 - 1:10 Comparis<strong>on</strong> of Alternative Methods for Estimating <strong>Breast</strong> Density<br />

Isabella dos Santos Silva, MD, MSc, PhD - L<strong>on</strong>d<strong>on</strong> School of Hygiene & Trophical<br />

Medicine<br />

1:10 - 1:50 <strong>Breast</strong> Cancer Risk: Is it Just About <strong>Breast</strong> Density<br />

Elizabeth Morris, MD, FACR - Memorial Sloan-Kettering Cancer Center<br />

1:50 - 2:30 Next Generati<strong>on</strong> Measurements of <strong>Breast</strong> Density - Tomosynthesis<br />

Despina K<strong>on</strong>tos, PhD - University of Pennsylvania<br />

2:30 - 2:45 Break (15 minutes)<br />

<strong>Breast</strong> Density <strong>and</strong> Biology<br />

2:45 - 3:25 Molecular Microenvir<strong>on</strong>ments of Mammographically Dense <strong>Breast</strong> Tissue<br />

Melissa Troester, PhD, MPH - UNC School of Public Health<br />

3:25 - 4:05 Collagen Alignment Surrounding <strong>Breast</strong> Tumors Facilitates Tumor Spread <strong>and</strong><br />

Metastasis<br />

Suzanne P<strong>on</strong>ik, PhD - University of Wisc<strong>on</strong>sin<br />

4:05 - 4:45 Summary of Day One Panel Discussi<strong>on</strong><br />

6:00 Speakers Dinner: Café Majestic in the Hotel Majestic<br />

PROGRAM


PROGRAM<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

PROGRAM<br />

Friday, June 7th<br />

8:30 - 9:00 Breakfast<br />

9:00 - 9:05 Introducti<strong>on</strong><br />

Genetics of <strong>Breast</strong> Density <strong>and</strong> <strong>Breast</strong> Cancer Risk<br />

9:05 - 9:45 Genetic Ancestry <strong>and</strong> Mammographic Density<br />

Elad Ziv, MD - UCSF<br />

9:45 - 10:25 The Relative C<strong>on</strong>tributi<strong>on</strong>s of BI-RADS Density <strong>and</strong> Genetics to <strong>Breast</strong> Cancer<br />

Risk<br />

Celine Vach<strong>on</strong>, PhD - Mayo Clinic<br />

10:25 -<br />

10:45<br />

10:45 -<br />

11:25<br />

11:25 -<br />

12:05<br />

Break (15 minutes)<br />

Risk Modeling: Putting It All Together<br />

<strong>Breast</strong> Cancer Risk Predicti<strong>on</strong> <strong>and</strong> Individualized Screening Based <strong>on</strong> Comm<strong>on</strong><br />

Genetic Variati<strong>on</strong>s <strong>and</strong> <strong>Breast</strong> Density Measurements<br />

Hatef Darabi, PhD - Karolinska Inst<br />

Benign <strong>Breast</strong> Disease <strong>and</strong> <strong>Breast</strong> Density<br />

Jeffrey A. Tice, MD - UCSF<br />

12:05 - 1:00 Lunch (55 minutes)<br />

1:00 - 1:15 ABSTRACT: Determinants of <strong>Breast</strong> Tissue Compositi<strong>on</strong> in 15-18 year Old<br />

Girls<br />

Norman Boyd, MD, DSc, FRCPC - Ontario Cancer Institute<br />

1:15 - 1:30 ABSTRACT: The Effect of Weight Change <strong>on</strong> Changes in <strong>Breast</strong> Measures Over<br />

Menopause<br />

Hanneke W<strong>and</strong>ers, MSc -Julius Ctr for Health Sciences & Primary Care, Epidemiology<br />

UMC Utrecht<br />

<strong>Breast</strong> Density <strong>and</strong> Risk Assessment in Clinical Practice<br />

1:30 - 2:10 Awareness of <strong>Breast</strong> Density <strong>and</strong> Its Relati<strong>on</strong>ship to Mammography Sensitivity<br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Am<strong>on</strong>g U.S. Women: Results of a Nati<strong>on</strong>al Survey<br />

Deb Rhodes, MD - Mayo Clinic<br />

2:10 - 2:50 <strong>Breast</strong>CARE: A primary Care Clinic-Based RCT to Increase <strong>Breast</strong> Cancer<br />

Knowledge <strong>and</strong> Discussi<strong>on</strong> of Risk <strong>and</strong> Lifestyle Behaviors<br />

Celia Kaplan, DrPH - UCSF<br />

2:50 - 3:05 Break (15 minutes)<br />

3:10 - 3:50 Impact of The Transiti<strong>on</strong> to Digital Mammography <strong>on</strong> the Benefits, Harms <strong>and</strong><br />

Costs of Screening for <strong>Breast</strong> Cancer in the US<br />

Natasha K. Stout, PhD - Harvard<br />

3:50 - 4:30 SUMMARY AND CLOSING: Panel Discussi<strong>on</strong><br />

4


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

ABSTRACTS<br />

FOR<br />

SPEAKERS<br />

5


BREAST DENSITY – WHAT IS A WOMAN TO DO NOW<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Deborah Collyar<br />

925.260.1006<br />

Deborah@tumortime.com<br />

UCSF <strong>and</strong> Patient Advocates in Research (PAIR)<br />

ABSTRACTS FOR SPEAKERS<br />

The good news… breast density seems to help explain more of the previously unknown risk factors for<br />

breast cancer. Unfortunately, all known risk factors <strong>on</strong>ly account for ~50% of breast cancers cases.<br />

The bad news… the cart has been placed before the horse, so to speak. Legislati<strong>on</strong> that requires doctors<br />

to inform women that they have high breast density sounds good, but without c<strong>on</strong>sistent measurements,<br />

st<strong>and</strong>ards, <strong>and</strong> plain-language principles, this may cause even more c<strong>on</strong>fusi<strong>on</strong> <strong>and</strong> fear for women.<br />

<strong>Breast</strong> density shares similar c<strong>on</strong>undrums that surfaced with BRCA1 <strong>and</strong> BRCA2 testing during its<br />

impetus. What opti<strong>on</strong>s (let al<strong>on</strong>e treatments) do doctors talk about with women who have high breast<br />

density What st<strong>and</strong>ard tool is being used to accurately assess breast density How do we explain the<br />

absolute risk of quadrant 3 <strong>and</strong> 4 (instead of relative risk), <strong>and</strong> how does that translate to the number of<br />

women in each category who actually get breast cancer each year<br />

Most importantly, who is going to put the informati<strong>on</strong> in c<strong>on</strong>text for each woman so that she can not <strong>on</strong>ly<br />

c<strong>on</strong>sider the opti<strong>on</strong>s, but can also determine which <strong>on</strong>es fit her unique lifestyle or decisi<strong>on</strong>-making<br />

process for important life decisi<strong>on</strong>s<br />

<strong>Breast</strong> density is also clouded by the c<strong>on</strong>fusing c<strong>on</strong>troversies surrounding the timing <strong>and</strong> accuracy of<br />

mammograms. Younger women a higher likelihood of dense breasts, which equates to less accurate<br />

mammograms, <strong>and</strong> yet they are told to have annual mammograms because of their higher risk. How can<br />

this be explained clearly so that it actually makes sense<br />

For instance, if a woman is at higher risk for breast cancer because of high breast density, what type of<br />

breast cancer might she get Is it more or less aggressive (e.g. what subtype is most comm<strong>on</strong>) Will<br />

additi<strong>on</strong>al mammograms, ultrasounds, MRIs or tomosynthesis (3-D mammograms) help find it at an<br />

earlier stage that will actually change its course, or just find more false positives that have their own<br />

physical, financial, <strong>and</strong> emoti<strong>on</strong>al c<strong>on</strong>sequences for women <strong>and</strong> their families<br />

St<strong>and</strong>ards <strong>and</strong> plain language are the most critical elements to ensure clear <strong>and</strong> useful informati<strong>on</strong> for<br />

women. Unfortunately, the genie is out of the bottle in California <strong>and</strong> other states without incorporating<br />

these elements.<br />

Women d<strong>on</strong>’t need more uncertainty <strong>and</strong> anxiety, or unnecessary biopsies with subsequent over-treatment.<br />

We’ve already created this situati<strong>on</strong> with Ductal Carcinoma In Situ (DCIS). Let’s not repeat this<br />

unfortunate reality with breast density.<br />

This situati<strong>on</strong> establishes a moral imperative to quickly work together to create the st<strong>and</strong>ards that should<br />

have been established before the legislati<strong>on</strong> became active. This call to acti<strong>on</strong> includes every<strong>on</strong>e who<br />

regulates, studies, screens, detects, explains, <strong>and</strong>/or treats breast cancer.<br />

6


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

CAUTION – KITTY LITTER, SILLY PUTTY & DENSE BREASTS: USING STATE, FEDERAL<br />

AND REGULATORY EFFORTS TO STANDARDIZE THE COMMUNICATION OF DENSE<br />

BREAST TISSUE TO WOMEN<br />

Nancy M. Cappello, Ph.D.<br />

Founder, Are You Dense Inc., Are You Dense Advocacy, Inc.<br />

203.232.9570<br />

nancy@areyoudense.org<br />

My radiologist knew I had dense breasts, my gynecologist knew I had dense breasts – the <strong>on</strong>ly <strong>on</strong>e who<br />

did not know was me - the woman with the dense breasts.<br />

When I was diagnosed in 2004 with advanced stage breast cancer which metastasized to 13 lymph nodes,<br />

I was baffled. After all, I received my “normal” mammography report, “the Happy Gram” six weeks<br />

prior, in additi<strong>on</strong> to a decade of normal mammography reports preceding this devastating news. How<br />

could my mammogram not find the “suspicious” 3 cm lesi<strong>on</strong> the same day that the ultrasound discovered<br />

it I questi<strong>on</strong>ed my doctors; after all, isn’t this why I go for yearly screenings For the first time, I was<br />

told that my “dense breast tissue” prevented my mammograms from finding my cancer.<br />

Searching for informati<strong>on</strong> in lay publicati<strong>on</strong>s, <strong>on</strong>line cancer <strong>and</strong> medical organizati<strong>on</strong>s, I found nothing<br />

about this “dense” c<strong>on</strong>diti<strong>on</strong> that harmed me greatly. I turned to the medical literature <strong>and</strong>, in 2004,<br />

during <strong>on</strong>e of the most vulnerable times in my life, I uncovered over a decade of scientific studies which<br />

c<strong>on</strong>cluded:<br />

<br />

<br />

<br />

<br />

<br />

<br />

40% of women have dense breast tissue;<br />

<strong>Breast</strong> Density is the str<strong>on</strong>gest predictor of the failure of mammography screening to detect cancer;<br />

There is a direct correlati<strong>on</strong> between tumor size at discovery <strong>and</strong> l<strong>on</strong>g term survivability;<br />

Women with the densest breasts are at a greater risk of having an interval cancer;<br />

There are additi<strong>on</strong>al tests, when added to mammography, that can significantly detect early-stage<br />

invasive cancers in dense breasts; <strong>and</strong><br />

Dense breast tissue is a well-established predictor of breast cancer risk<br />

Armed with knowledge, I shared the scientific evidence with my physicians. I asked them to c<strong>on</strong>sider informing<br />

patients about their dense tissue. Each of their resp<strong>on</strong>ses was “No, it is not the st<strong>and</strong>ard of care.”<br />

I then turned to the C<strong>on</strong>necticut legislature <strong>and</strong> in 2005, C<strong>on</strong>necticut became the first state in the nati<strong>on</strong> to<br />

legislate insurance coverage for ultrasound screening as a supplement to the mammogram for women<br />

with dense breast tissue. In 2009, C<strong>on</strong>necticut enacted another l<strong>and</strong>mark state law which st<strong>and</strong>ardized the<br />

communicati<strong>on</strong> of dense breast tissue through the mammography report.<br />

Using state, federal <strong>and</strong> regulatory efforts <strong>and</strong> the birth of two n<strong>on</strong>profit organizati<strong>on</strong>s, Are You Dense,<br />

Inc. <strong>and</strong> Are You Dense Advocacy, Inc., our missi<strong>on</strong> has fueled a grassroots movement with impactful<br />

accomplishments. This is a testament to the fact that there is no shortage of women, with recent “normal”<br />

mammography reports <strong>and</strong> subsequent advanced cancer diagnoses, because of their dense breast tissue.<br />

Legislators, medical professi<strong>on</strong>als <strong>and</strong> citizens have joined our missi<strong>on</strong> to ensure that women with dense<br />

breast tissue are informed participants in discussi<strong>on</strong>s with their health-care providers about their pers<strong>on</strong>al<br />

breast screening surveillance.<br />

ABSTRACTS FOR SPEAKERS<br />

7


ABSTRACTS FOR SPEAKERS<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

APPLICATION OF CALIFORNIA SB 1538 BREAST DENSITY NOTIFICATION LAW IN<br />

CLINICAL PRACTICE<br />

Meg Durbin (Palo Alto Medical Foundati<strong>on</strong>), Debra M. Ikeda (Stanford University), B<strong>on</strong>nie N. Joe<br />

(UCSF), Edward A Sickles (UCSF)<br />

On September 22, 2012 California State Senate Bill (SB 1538) (1) was signed into law m<strong>and</strong>ating that<br />

facilities who perform mammograms notify women identified with heterogeneously dense or dense breast<br />

tissue (2) <strong>on</strong> mammograms “based <strong>on</strong> the <strong>Breast</strong> Imaging Reporting <strong>and</strong> Data System established by the<br />

American College of Radiology”, “include in the summary of the written report that is sent to the patient,<br />

as required by federal law, the following notice”: "Your mammogram shows that your breast tissue is<br />

dense. Dense breast tissue is comm<strong>on</strong> <strong>and</strong> is not abnormal. However, dense breast tissue can make it<br />

harder to evaluate the results of your mammogram <strong>and</strong> may also be associated with an increased risk of<br />

breast cancer. This informati<strong>on</strong> about the results of your mammogram is given to you to raise your<br />

awareness <strong>and</strong> to inform your c<strong>on</strong>versati<strong>on</strong>s with your doctor. Together, you can decide which screening<br />

opti<strong>on</strong>s are right for you. A report of your results was sent to your physician." This statement was to be<br />

included within the lay letter informing women of their mammogram result under the federal<br />

Mammography Quality St<strong>and</strong>ards Act (42 U.S.C. Sec. 263b). The law further states “Nothing in this<br />

secti<strong>on</strong> shall be c<strong>on</strong>strued to create or impose liability <strong>on</strong> a health care facility for failing to comply with<br />

the requirements of this secti<strong>on</strong> prior to April 1, 2013. Nothing in this secti<strong>on</strong> shall be deemed to create a<br />

duty of care or other legal obligati<strong>on</strong> bey<strong>on</strong>d the duty to provide notice as set forth in this secti<strong>on</strong>.” In<br />

California, m<strong>and</strong>atory reporting requirements took effect <strong>on</strong> April 1, 2013 <strong>and</strong> is to remain in effect until<br />

January 1, 2019.<br />

<strong>Breast</strong> density notificati<strong>on</strong> laws are being c<strong>on</strong>sidered in Florida, North Carolina, Oreg<strong>on</strong>, <strong>and</strong> Utah<br />

(3), while California, C<strong>on</strong>necticut, New York, Texas, Virginia <strong>and</strong> Hawaii have passed laws. C<strong>on</strong>necticut<br />

screening breast ultrasound studies were reported recently (4-6), covered by insurance as directed by<br />

C<strong>on</strong>necticut law. However, the issue in California was how to frame policy to rec<strong>on</strong>cile the legislative<br />

intent of SB1538 notificati<strong>on</strong> with realistic practice patterns since SB1538 provided no guidance <strong>on</strong><br />

supplemental screening guidelines, <strong>and</strong>, unlike C<strong>on</strong>necticut, provided no funding for supplemental<br />

screening. In California, an academic <strong>and</strong> community-based working group of breast imagers <strong>and</strong> breast<br />

cancer risk specialists, the California <strong>Breast</strong> Density Informati<strong>on</strong> Group (CBDIG), formed in October<br />

2012 to investigate the literature related to issues raised by SB 1538 to provide an evidence-based<br />

resp<strong>on</strong>se to provide informati<strong>on</strong> for women <strong>on</strong> breast density <strong>and</strong> help facilities prepare policies. The<br />

result was a free, instituti<strong>on</strong>-neutral website breastdensity.info, instantly translatable into multiple<br />

languages with frequently asked questi<strong>on</strong>s <strong>and</strong> evidenced-based recommendati<strong>on</strong>s for supplemental<br />

screening of women identified with dense breast tissue, based <strong>on</strong> breast cancer risk assessment from<br />

nati<strong>on</strong>ally recognized guidelines. This presentati<strong>on</strong> will show data indicating that approximately 50% of<br />

women have heterogeneously dense or dense tissue <strong>on</strong> mammography (7) <strong>and</strong> report the results of the<br />

CBDIG website. However, we will also illustrate the impact of SB 1538 <strong>on</strong> facilities to prepare to notify<br />

<strong>and</strong> educate the approximately 2 milli<strong>on</strong> women with dense breast tissue, <strong>and</strong> their healthcare providers.<br />

Furthermore we will discuss gaps in informati<strong>on</strong> <strong>on</strong> how best to measure <strong>and</strong> report breast density,<br />

c<strong>on</strong>troversies <strong>on</strong> the risk of dense breast tissue, lack of validated models incorporating breast density into<br />

risk assessment, lack large trial data <strong>on</strong> supplemental screening in intermediate or average risk women,<br />

<strong>and</strong> preliminary data <strong>on</strong> tomosynthesis.<br />

1. http://www.leginfo.ca.gov/pub/11-12/bill/sen/sb_1501-1550/sb_1538_bill_20120922_chaptered.html (accessed May 3, 2013)<br />

2 D'Orsi C, Mendels<strong>on</strong> E, Ikeda D, al. e. <strong>Breast</strong> Imaging Reporting <strong>and</strong> Data System: ACR BI-RADS -- <strong>Breast</strong> Imaging Atlas. Rest<strong>on</strong>, VA:<br />

American College of Radiology; 2003.<br />

3. Lee CI, Bassett LW, Lehman CD. <strong>Breast</strong> density legislati<strong>on</strong>s <strong>and</strong> opportunities for patient-centered outcomes research. Radiology 2012<br />

Vol 264 No 3: 632-636<br />

4 Weigert J, Steenbergen S. The c<strong>on</strong>necticut experiment: the role of ultrasound in the screening of women with dense breasts. <strong>Breast</strong> J.<br />

2012;18(6):517-22.<br />

5. Parris T, Wakefield D, Frimmer H. Real world performance of screening breast ultrasound following enactment of C<strong>on</strong>necticut Bill 458.<br />

<strong>Breast</strong> J. 2013;19(1):64-70.<br />

6. Hooley RJ, Greenberg KL, Stackhouse RM, Geisel JL, Butler RS, Philpotts LE. Screening US in patients with mammographically dense<br />

breasts: initial experience with C<strong>on</strong>necticut Public Act 09-41. Radiology. 2012;265(1):59-69.<br />

7. D'orsi C, Mendels<strong>on</strong> E, Morris E, al e. ACR BI-RADS: <strong>Breast</strong> Imaging Reporting <strong>and</strong> Data System. 5th ed ed. Rest<strong>on</strong>, VA: American<br />

College of Radiology; 2013 (in press).<br />

8


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

CIRRUS: A FULLY-AUTOMATED SOFTWARE PLATFORM FOR BREAST CANCER RISK<br />

PREDICTION FROM MAMMOGRAMS<br />

Daniel F. Schmidt, Enes Makalic, Tu<strong>on</strong>g L. Nguyen, Laura Baglietto, Kavitha Krishnan, Jennifer St<strong>on</strong>e,<br />

Carmel Apicella, Melissa C. Southey, Dallas R. English, Graham G. Giles, John L. Hopper<br />

Centre for Molecular, Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytic Epidemiology, The University of Melbourne,<br />

Carlt<strong>on</strong> VIC 3053, Australia<br />

Background: There is informati<strong>on</strong> in mammograms, apart from the detecti<strong>on</strong> <strong>and</strong> masking of existing<br />

lesi<strong>on</strong>s, that predicts breast cancer risk. This c<strong>on</strong>cept is currently referred to as ‘mammographic density’,<br />

operati<strong>on</strong>ally defined by the total area of the mammographic image that appears light or white. The most<br />

comm<strong>on</strong>ly used software tool for measuring mammographic density is CUMULUS, a computer-assisted<br />

technique that derives the pixels in a digitised mammogram which corresp<strong>on</strong>d to user-determined<br />

‘mammographically dense regi<strong>on</strong>s’. CUMULUS measures of mammographic density, adjusted for age<br />

<strong>and</strong> body mass index, have been shown to predict breast cancer risk even after adjusting for other<br />

measured breast cancer risk factors.<br />

We have developed a fully-automated predictor of breast cancer risk from digitised mammographic<br />

images, which we call CIRRUS, that does not try to measure ‘mammographic density’ per se, but instead<br />

tries to identify agnostically the features in a mammographic image that best predict breast cancer risk.<br />

Methods: CIRRUS is based <strong>on</strong> image processing <strong>and</strong> machine learning techniques. It includes fully<br />

automated image cropping procedures which remove irrelevant features (e.g. pectoral muscle). In additi<strong>on</strong><br />

to the brightness of the pixels, CIRRUS uses informati<strong>on</strong> from other features in the mammogram, such as<br />

patterns <strong>and</strong> texture.<br />

CIRRUS was trained using 2,597 mammograms (608 cases <strong>and</strong> 1,989 c<strong>on</strong>trols) collected by the<br />

Melbourne Collaborative Cohort Study. Bayesian logistic regressi<strong>on</strong> with the ridge penalty was used to<br />

combine all the extracted image features into the final CIRRUS measure, which was compared with dense<br />

area <strong>and</strong> percent dense area measurements obtained using CUMULUS. The bootstrap technique was used<br />

to estimate the c<strong>on</strong>fidence intervals for the area under the receiver operator curve (AUC). We also<br />

measured CIRRUS for 544 m<strong>on</strong>ozygotic twin pairs (MZ), 339 dizygotic twin pairs (DZ) <strong>and</strong> 1,556 of<br />

their sisters from the Australian Mammographic Density Twins <strong>and</strong> Sisters Study, <strong>and</strong> analysed familial<br />

aspects under a multivariate normal model.<br />

Results: In terms of breast cancer risk predicti<strong>on</strong>, for both dense area <strong>and</strong> percent dense area measured by<br />

CUMULUS the mean AUC was 0.58 (CI 95%: 0.57 – 0.61). For the CIRRUS measure the AUC was 0.62<br />

(CI 95%: 0.58 - 0.64). CIRRUS measures were not markedly associated with age or any other measured<br />

breast cancer risk factor, which explained


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

SEVERAL MAMMOGRAPHIC TEXTURE MEASURES, INCLUDING MTR, ADD<br />

SIMULTANEOUSLY TO PERCENTAGE DENSITY RISK SEGREGATION<br />

J Marques 1 , M Lillholm 1 , DR Jørgensen 1 , P Diao 2 , K Petersen 2 , N Karssemeijer 3 , M Nielsen 1,2<br />

1 University of Copenhagen, Dept. of Comp. Science, 2 Biomediq A/S, 3 Radboud University, Nijmegen<br />

Medical Centre<br />

ABSTRACTS FOR SPEAKERS<br />

Purpose: Mammographic percentage density (PD) is established as a str<strong>on</strong>g risk factor for breast cancer.<br />

PD, however, relies <strong>on</strong> limited aspects of mammographic intensity appearance <strong>and</strong> variati<strong>on</strong> – several<br />

other measures have been suggested to cover more structural aspects of mammographic parenchymal<br />

texture. We evaluate a comprehensive list of such measures in terms of their breast cancer risk<br />

segregati<strong>on</strong>. Furthermore, we evaluate to which extent a machine-learning based texture measure,<br />

mammographic texture resemblance (MTR), <strong>and</strong> automated PD measurements add informati<strong>on</strong>.<br />

Materials <strong>and</strong> Methods: The study was based <strong>on</strong> a 495 women case-c<strong>on</strong>trol cohort from the Dutch<br />

screening program (age 58.0 ± 5.7 years). Cases <strong>and</strong> c<strong>on</strong>trols were screened as cancer free at baseline; the<br />

250 c<strong>on</strong>trols remained healthy 4 years later whereas the 245 cases were diagnosed 2-4 year after the<br />

initial mammography.<br />

All baseline mammograms were analyzed to derive the following measures of mammographic texture: i)<br />

PD scores were assessed by a trained radiologist using a Cumulus-like tool <strong>and</strong> automatically using a<br />

software tool, ii) Lacunarity, Markovian, Laws, Run Length, Fourier, Power Law, Wavelet, Variati<strong>on</strong>, <strong>and</strong><br />

Fractal Dimensi<strong>on</strong> as described in the papers by M<strong>and</strong>uca 1 , Heine 2 , <strong>and</strong> Giger 3 , <strong>and</strong> iii) the MTR measure<br />

using a software tool by Biomediq 4 .<br />

Left <strong>and</strong> right MLO views were processed separately <strong>and</strong> odds ratio (OR) <strong>and</strong> area under the ROC curve<br />

(AUC) for separati<strong>on</strong> between cases <strong>and</strong> c<strong>on</strong>trols were calculated for each measure.<br />

Two logistic regressi<strong>on</strong> combinati<strong>on</strong> models were c<strong>on</strong>structed: i) using all the measures described above<br />

<strong>and</strong> ii) the same except the radiologist PD were excluded. Both models iteratively excluded any measures<br />

that did not add significantly to the model.<br />

Results: The risk segregati<strong>on</strong>s for each texture measure (left/right MLO) are given in Table 1. The<br />

logistic regressi<strong>on</strong> model based gave an OR of 3.36 (p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Table 1 The risk segregati<strong>on</strong> of the included texture measures as odds ratio <strong>and</strong> AUC for left <strong>and</strong> right<br />

MLO views respectively. Stars indicate level of significance: * p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

COMPARISON OF ALTERNATIVE METHODS FOR ESTIMATING BREAST DENSITY<br />

Isabel dos Santos Silva 1 , Steve Allen 2 , Am<strong>and</strong>a Eng 1 , Zoe Garl<strong>and</strong> 1 , Sarah Vinnicombe 3 , Valerie<br />

McCormack 4 , JA Shepherd 5 , Mitch Dowsett 2<br />

1 L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical Medicine, L<strong>on</strong>d<strong>on</strong>, Engl<strong>and</strong>; 2 Royal Marsden Hospital, UK;<br />

3 University of Dundee, UK; 4 <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer, France; 5 University of<br />

California<br />

ABSTRACTS FOR SPEAKERS<br />

Mammographic density (MD), a trait which closely reflects the amount of radio-dense (n<strong>on</strong>-fatty) tissue of<br />

the breast, is <strong>on</strong>e of the str<strong>on</strong>gest known risk factors for breast cancer <strong>and</strong> a major determinant of<br />

sensitivity of mammographic screening. In many US states it is now m<strong>and</strong>atory for doctors to inform<br />

women if their breasts are dense (BI-RADS: 3 or 4), albeit it is unclear whether they will benefit from<br />

more intensive screening (e.g. shorter screening intervals, combinati<strong>on</strong> of mammography with ultrasound<br />

or MRI).<br />

Currently, there are several approaches to measuring MD in screen-film mammography. Qualitative<br />

assessments, such as BI-RADS, classify density into categories based <strong>on</strong> the subjective analysis of<br />

patterns observed <strong>on</strong> the mammogram while quantitative measurements determine the proporti<strong>on</strong> of the<br />

projected breast area that is composed of radio-dense tissue <strong>on</strong> a digitised screen-film. The quantitative<br />

semi-automatic threshold method, as implemented by the Cumulus software, is regarded as the “gold<br />

st<strong>and</strong>ard” for screen-film mammography. However, this method has limitati<strong>on</strong>s: it is subjective (i.e.<br />

observer-dependent); its area-based nature may be less efficient than a volume-based approach; <strong>and</strong> it is a<br />

rather time-c<strong>on</strong>suming <strong>and</strong> labour-intensive method. The latter, in particular, have precluded its<br />

incorporati<strong>on</strong> into routine clinical/screening practice. Fully automated methods have been developed<br />

which attempt to use volumetric approaches to quantify more precisely breast density <strong>on</strong> digitised screenfilms.<br />

But despite their alleged higher precisi<strong>on</strong>, such volumetric methods do not appear to predict breast<br />

cancer risk more accurately than the area-based approaches.<br />

As screen-film mammography is being replaced by full-field digital mammography (FFDM), new fullyautomated<br />

volumetric methods have recently been developed for assessment of density from digital<br />

images. Evaluati<strong>on</strong> of the performance of these methods have been mainly limited to establishing the<br />

correlati<strong>on</strong>s of their measurements with those from more established methods (e.g. BIRADS or Cumulus)<br />

<strong>and</strong>/or evaluating the extent to which their measurements are associated with known breast cancer risk<br />

factors. Ultimately, the best method for measuring breast density from digital images is the <strong>on</strong>e that yields<br />

the str<strong>on</strong>gest associati<strong>on</strong> with breast cancer risk.<br />

The development of a valid universal st<strong>and</strong>ard tool for measuring MD is vital for accurate risk predicti<strong>on</strong><br />

(<strong>on</strong> a populati<strong>on</strong> <strong>and</strong> individual level) <strong>and</strong> to tailor preventive measures, including breast screening,<br />

according to a woman’s risk. In this talk, I will present data from our <strong>on</strong>-going work <strong>on</strong> the evaluati<strong>on</strong> of<br />

alternative methods of measuring breast density <strong>on</strong> digital images. These include three area-based methods:<br />

(i) visual qualitative assessment using the BI-RADS classificati<strong>on</strong>; (ii) a semi-automated assessment using<br />

a thresholding technique (Cumulus); <strong>and</strong> (iii) an automated ImageJ-based approach which mimics<br />

Cumulus (the latter two optimised for FFDM). In additi<strong>on</strong>, we have evaluated three automated volumetric<br />

approaches: (iv) single X-ray absorptiometry (SXA) with calibrati<strong>on</strong> phantoms; (v) Volpara TM ; <strong>and</strong> (vi)<br />

Quantra TM . Overall, the findings showed that the density estimates yielded by the automated volumetric<br />

methods were at least as str<strong>on</strong>gly associated with breast cancer risk as those produced by the area-based<br />

approaches. Thus, these results support the use of volumetric-based approaches in screening/clinical<br />

settings <strong>and</strong> in large-scale studies.<br />

12


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BREAST CANCER RISK: IS IT JUST ABOUT BREAST DENSITY<br />

Elizabeth Morris MD FACR<br />

Chief, <strong>Breast</strong> Imaging Service, Department of Radiology, Memorial Sloan-Kettering Cancer Center<br />

<strong>Breast</strong> parenchyma depicted <strong>on</strong> mammography has been shown to provide valuable informati<strong>on</strong> about<br />

breast cancer risk. <strong>Breast</strong> tissue c<strong>on</strong>sists primarily of fibrogl<strong>and</strong>ular tissue <strong>and</strong> fat. Fibrogl<strong>and</strong>ular tissue<br />

is resp<strong>on</strong>sible for the density seen <strong>on</strong> mammography. The risk of breast cancer <strong>and</strong> women with<br />

mammographically dense breasts has been shown in multiple studies to be higher than in women with<br />

predominantly mammographically fatty breasts.<br />

<strong>Breast</strong> MRI is a st<strong>and</strong>ard part of the imaging workup in high risk women <strong>and</strong> in patients with known<br />

breast cancer. On breast MRI examinati<strong>on</strong>s, the amount of fibrogl<strong>and</strong>ular tissue can be seen <strong>and</strong> this<br />

likely corresp<strong>on</strong>ds to breast density seen <strong>on</strong> mammography. MRI however dem<strong>on</strong>strates additi<strong>on</strong>al<br />

informati<strong>on</strong> about the underlying fibrogl<strong>and</strong>ular tissue. As intravenous c<strong>on</strong>trast material is injected<br />

routinely for breast MRI examinati<strong>on</strong>s, it is been observed that native fibrogl<strong>and</strong>ular tissue will<br />

dem<strong>on</strong>strate variable enhancement patterns <strong>and</strong> levels of enhancement. The enhancement of the existing<br />

underlying fibrogl<strong>and</strong>ular tissue has been termed background parenchymal enhancement.<br />

As background parenchymal enhancement is related to vascular flow, it has been proposed that this may<br />

represent an imaging biomarker of the underlying proliferati<strong>on</strong> of fibrogl<strong>and</strong>ular tissue. Investigati<strong>on</strong>s<br />

have shown that there is an extremely str<strong>on</strong>g associati<strong>on</strong> between BPE <strong>and</strong> risk of breast cancer, at least a<br />

str<strong>on</strong>g as the associati<strong>on</strong> between mammographic density <strong>and</strong> breast cancer.<br />

There has been observed that background parenchymal enhancement is also affected by treatment<br />

changes <strong>and</strong> horm<strong>on</strong>al manipulati<strong>on</strong>. For example, tamoxifen therapy <strong>and</strong> aromatase inhibitors may<br />

decrease the intrinsic background parenchymal enhancement, dem<strong>on</strong>strating an imaging treatment<br />

resp<strong>on</strong>se. It has been shown that background parenchymal enhancement may fluctuate during the<br />

menstrual cycle <strong>and</strong> that horm<strong>on</strong>e replacement therapy can cause an increase in background parenchymal<br />

enhancement. Radiati<strong>on</strong> therapy to the breast causes marked reducti<strong>on</strong> in background parenchymal<br />

enhancement.<br />

As with breast density <strong>and</strong> distributi<strong>on</strong> of breast parenchyma <strong>on</strong> mammography, it appears that the<br />

background parenchymal enhancement of breast MRI is also extremely variable <strong>and</strong> women have<br />

different patterns <strong>and</strong> intensity of background parenchymal enhancement. In fact, it has been observed<br />

that not all mammographically dense breasts dem<strong>on</strong>strate increased background parenchymal<br />

enhancement. Therefore, it is possible that MRI can further stratify women at high risk for developing<br />

breast cancer <strong>on</strong> the basis of background parenchymal enhancement.<br />

1. King V, Brooks JD, Bernstein JL, Reiner AS, Pike MC, Morris EA. Background parenchymal<br />

enhancement at breast MR imaging <strong>and</strong> breast cancer risk. Radiology 2011; 260(1):50-60.<br />

PMID: 21493794<br />

2. King V, Goldfarb S, Brooks J, Sung JS, Pike MC, Nulsen B, Jozefara J, Dickler M, Morris EA.<br />

Effect of aromatase inhibitors <strong>on</strong> background parenchymal enhancement <strong>and</strong> amount of<br />

fibrogl<strong>and</strong>ular tissue <strong>on</strong> <strong>Breast</strong> MRI. Radiology. 2012 Sep; 264(3):670-8. PMID: 22771878.<br />

3. King V, Kaplan J, Pike MC, Liberman L, Dershaw DD, Lee CH, Brooks JD, Morris EA. Impact<br />

of tamoxifen <strong>on</strong> amount of fibrogl<strong>and</strong>ular tissue, background parenchymal enhancement, <strong>and</strong><br />

cysts <strong>on</strong> breast magnetic res<strong>on</strong>ance imaging. <strong>Breast</strong> J. 2012 Nov; 18(6):527-34. PMID:<br />

23002953.<br />

4. King V, Gu Y, Kaplan JB, Brooks JD, Pike MC, Morris EA. Impact of menopausal status <strong>on</strong><br />

background parenchymal enhancement <strong>and</strong> fibrogl<strong>and</strong>ular tissue <strong>on</strong> breast MRI. European<br />

Radiology. 2012 Dec; 22(12):2641-7. PMID: 22752463.<br />

ABSTRACTS FOR SPEAKERS<br />

13


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

NEXT GENERATION MEASUREMENTS OF BREAST DENSITY – TOMOSYNTHESIS<br />

Despina K<strong>on</strong>tos, PhD<br />

Department of Radiology, University of Pennsylvania<br />

ABSTRACTS FOR SPEAKERS<br />

Growing evidence suggests that breast density is an independent risk factor for breast cancer. Currently,<br />

the most widely used methods to estimate breast density rely <strong>on</strong> semi-automated thresholding of<br />

mammograms to estimate the percent of the dense tissue area versus the entire area of the breast.<br />

Although useful for breast cancer risk estimati<strong>on</strong>, these methods have limitati<strong>on</strong>s. Mammography is a<br />

projecti<strong>on</strong> imaging technique that visualizes the admixture of superimposed breast tissues.<br />

Therefore, such thresholding methods do not allow for estimating volumetric density, but instead provide<br />

an area-based estimate measured from the projecti<strong>on</strong> image of the breast. Digital breast tomosynthesis<br />

(DBT) is an emerging 3D x-ray imaging modality in which tomographic breast images are rec<strong>on</strong>structed<br />

from multiple low-dose x-ray source projecti<strong>on</strong>s. Measures of volumetric breast density from DBT<br />

images could provide more accurate measures of the dense breast tissue <strong>and</strong> ultimately result in more<br />

accurate measures of risk.<br />

This talk will provide an overview of the state-of-the art in the current research <strong>on</strong> developing volumetric<br />

breast density estimati<strong>on</strong> techniques for DBT, including both semi-automated <strong>and</strong> fully-automated<br />

approaches, <strong>and</strong> new approaches to characterize the volumetric complexity of the breast tissue using DBT<br />

parenchymal texture analysis. DBT breast density measures will be compared to other volumetric <strong>and</strong><br />

area-based multi-modality breast density estimati<strong>on</strong> approaches, including Digital Mammography <strong>and</strong><br />

<strong>Breast</strong> MRI, <strong>and</strong> implicati<strong>on</strong>s will be discussed for breast cancer risk estimati<strong>on</strong>.<br />

We envisi<strong>on</strong> a unique setting in which breast cancer risk assessment <strong>and</strong> patient educati<strong>on</strong> can be<br />

combined to empower women with knowledge about their pers<strong>on</strong>al risk <strong>and</strong> provide a fully automated<br />

risk assessment tool for physicians. The tomosynthesis FDA approval coupled with a rapidly evolving<br />

technology <strong>and</strong> a potential for superior clinical performance will so<strong>on</strong> determine the emerging role of<br />

DBT in clinical practice.<br />

Early trials suggest a significant benefit from DBT in reducing unnecessary recalls for women in the<br />

screening setting, including a potential for improving cancer detecti<strong>on</strong>. Fully-automated approaches for<br />

estimating volumetric breast density in DBT could provide a tool for incorporating quantitative measures<br />

of volumetric breast density in clinical breast cancer risk assessment.<br />

14


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MOLECULAR MICROENVIRONMENTS OF MAMMOGRAPHICALLY DENSE BREAST<br />

TISSUE<br />

Melissa A. Troester 1,2,3†<br />

1 Department of Epidemiology, 2 Department of Pathology <strong>and</strong> Laboratory Medicine, 3 Lineberger<br />

Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA<br />

Previous studies of breast tissue gene expressi<strong>on</strong> have dem<strong>on</strong>strated that the extratumoral<br />

microenvir<strong>on</strong>ment has substantial variability across individuals, some of which can be attributed to<br />

epidemiologic factors. To evaluate how mammographic density (MD) <strong>and</strong> breast tissue compositi<strong>on</strong><br />

relate to extratumoral microenvir<strong>on</strong>ment gene expressi<strong>on</strong>, we used data <strong>on</strong> 121 breast cancer patients<br />

from the populati<strong>on</strong>-based Polish Women’s <strong>Breast</strong> Cancer Study.<br />

<strong>Breast</strong> cancer cases were classified into two groups based <strong>on</strong> previously reported, biologically-defined<br />

extratumoral gene expressi<strong>on</strong>: the Active subtype, which is associated with high expressi<strong>on</strong> of genes<br />

related to fibrosis <strong>and</strong> wound resp<strong>on</strong>se, <strong>and</strong> the Inactive subtype, which has high expressi<strong>on</strong> of cellular<br />

adhesi<strong>on</strong> genes. MD was assessed using a quantitative, reliable computer-assisted thresholding method.<br />

<strong>Breast</strong> tissue compositi<strong>on</strong> was evaluated based <strong>on</strong> digital image analysis of tissue secti<strong>on</strong>s.<br />

The Inactive extratumoral subtype was associated with significantly higher percentage mammographic<br />

density (PD) <strong>and</strong> dense area (DA) in univariate analysis (PD: p=0.001; DA: p=0.049) <strong>and</strong> in multivariable<br />

analyses adjusted for age <strong>and</strong> body mass index (PD: p=0.004; DA: p=0.049). Inactive/higher-MD tissue<br />

was characterized by a significantly higher percentage of stroma <strong>and</strong> a significantly lower percentage of<br />

adipose tissue, with no significant change in epithelial c<strong>on</strong>tent.<br />

Analysis of published gene expressi<strong>on</strong> signatures suggested that Inactive/higher-MD tissue expressed<br />

increased estrogen resp<strong>on</strong>se <strong>and</strong> decreased TGF-β signaling. By linking novel molecular phenotypes with<br />

MD, our results indicate that MD reflects broad transcripti<strong>on</strong>al changes, including changes in both<br />

epithelia- <strong>and</strong> stroma-derived signaling.<br />

ABSTRACTS FOR SPEAKERS<br />

15


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

COLLAGEN ALIGNMENT SURROUNDING BREAST TUMORS FACILITATES TUMOR<br />

SPREAD AND METASTASIS<br />

Suzanne M. P<strong>on</strong>ik 1 , Kristin M. Riching 1 , Kevin W. Eliceiri 1 , Paolo P. Provenzano 2 , Patricia J. Keely 1<br />

ABSTRACTS FOR SPEAKERS<br />

1 Laboratory for Cellular <strong>and</strong> Molecular Biology <strong>and</strong> UW Carb<strong>on</strong>e Cancer Center, University of<br />

Wisc<strong>on</strong>sin, Madis<strong>on</strong>, 53706; 2 Department of Biomedical Engineering <strong>and</strong> UMCCC, University of<br />

Minnesota, Minneapolis, 55455<br />

While mammographic density has been correlated to increased risk of breast cancer in several studies,<br />

relatively little is known regarding the mechanisms by which mammographic density c<strong>on</strong>tributes to risk.<br />

Using mouse models of increased collagen in the c<strong>on</strong>nective tissue stroma, we find a causal link between<br />

increased stromal collagen <strong>and</strong> increased formati<strong>on</strong> of mammary tumors. Tumors that arise in dense<br />

collagen are more invasive <strong>and</strong> dem<strong>on</strong>strate dramatic changes in gene expressi<strong>on</strong> compared to tumors that<br />

arise in n<strong>on</strong>-dense mammary tissue. At the structural level, we find that mammary tissue has n<strong>on</strong>-aligned,<br />

curly collagen fibers that are altered during tumor progressi<strong>on</strong>.<br />

These changes, termed Tumor Associated Collagen Signatures (TACS) manifest as a c<strong>on</strong>sistent<br />

progressi<strong>on</strong> from increased unaligned collagen (TACS-1), to straight collagen fibers (TACS-2) to straight<br />

collagen fibers aligned perpendicular to the tumor/stromal boundary (TACS-3). Mechanistically, TACS-3<br />

collagen provides a “highway” <strong>on</strong> which tumor cells are able to migrate out from the tumor <strong>and</strong> invade<br />

into distal sites. TACS-3 signatures correlate to increased metastases in mouse models. In mouse models,<br />

similar rearrangements of collagen are also observed at the sites of lung metastasis, suggesting the<br />

collagen rearrangements facilitate not <strong>on</strong>ly spread, but also survival in metastatic sites. Importantly,<br />

collagen structural changes are also found in human breast cancer: analysis of 203 archived samples from<br />

patients with invasive breast cancer dem<strong>on</strong>strates that the presence of TACS-3 collagen significantly<br />

predicts increased relapse.<br />

Thus, TACS-3 collagen represents a potential biomarker. Current work is investigating ways that<br />

collagen changes can be targeted to better treat breast cancer in women with dense breast tissue.<br />

16


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

GENETIC MARKERS OF BREAST DENSITY-ETHNIC ASSOCIATION TO BREAST DENSITY<br />

Elad Ziv, MD<br />

Department of Medicine, University of California San Francisco<br />

Introducti<strong>on</strong>: Percent mammographic density (PMD) adjusted for age <strong>and</strong> BMI is <strong>on</strong>e of the str<strong>on</strong>gest<br />

risk factors for breast cancer <strong>and</strong> is known to be approximately 60 percent heritable. Here we report a<br />

finding of an associati<strong>on</strong> between genetic ancestry <strong>and</strong> adjusted PMD.<br />

Methods: We selected self-identified Caucasian women in the California Pacific Medical Center<br />

Research Institute Cohort whose screening mammograms placed them in the top or bottom quintiles of<br />

age- <strong>and</strong> body mass index-adjusted PMD. Our final data set included 474 women with the highest<br />

adjusted PMD <strong>and</strong> 469 with the lowest genotyped <strong>on</strong> the Illumina 1M platform. Principal comp<strong>on</strong>ent<br />

analysis (PCA) <strong>and</strong> identity-by-descent (IBD) analyses allowed us to infer the women's genetic<br />

ancestry <strong>and</strong> correlate it with adjusted PMD.<br />

Results: Women of Ashkenazi Jewish ancestry, as defined by the first principal comp<strong>on</strong>ent (PC1) of<br />

PCA <strong>and</strong> identity-by-descent analyses, represented approximately 15 percent of the sample. Ashkenazi<br />

Jewish ancestry, defined by PC1, was associated with higher adjusted PMD (p = 0.004). Using<br />

multivariate regressi<strong>on</strong> to adjust for epidemiologic factors associated with PMD, including age at parity<br />

<strong>and</strong> use of postmenopausal horm<strong>on</strong>e therapy, did not attenuate the associati<strong>on</strong>.<br />

C<strong>on</strong>clusi<strong>on</strong>: Women of Ashkenazi Jewish ancestry based <strong>on</strong> genetic analysis are more likely to have<br />

high age- <strong>and</strong> BMI-adjusted PMD. Ashkenazi Jews may have a unique set of genetic variants or<br />

envir<strong>on</strong>mental risk factors that increase mammographic density.<br />

ABSTRACTS FOR SPEAKERS<br />

17


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

THE RELATIVE CONTRIBUTIONS OF BI-RADS DENSITY AND GENETICS TO BREAST<br />

CANCER RISK<br />

Celine M. Vach<strong>on</strong> 1 , Peter Fasching 2 , Fergus Couch 1,3 , Christopher Scott 1 , Matthew Jensen 1 , Dana<br />

Whaley 4 , Kathleen R. Br<strong>and</strong>t 4 , Elad Ziv 5 , Karla Kerlikowske 5 , V. Shane Pankratz 1<br />

ABSTRACTS FOR SPEAKERS<br />

1 Department of Health Sciences Research, Divisi<strong>on</strong> of Epidemiology, Mayo Clinic College of Medicine,<br />

Rochester, MN; 2 Department of Gynecology <strong>and</strong> Obstetrics, University Hospital Erlangen Friedrich-<br />

Alex<strong>and</strong>er-University Erlangen-Nuremberg Erlangen, Germany; 3 Divisi<strong>on</strong> of Experimental Pathology,<br />

Department of Laboratory Medicine <strong>and</strong> Pathology , Mayo Clinic College of Medicine, Rochester, MN;<br />

4 Divis<strong>on</strong> of <strong>Breast</strong> Imaging, Department of Radiology, Mayo Clinic College of Medicine, Rochester, MN;<br />

5 Departments of Epidemiology <strong>and</strong> Biostatistics <strong>and</strong> General Internal Medicine Secti<strong>on</strong>, Department of<br />

Veterans Affairs, University of California, San Francisco, CA<br />

Background: <strong>Breast</strong> density is <strong>on</strong>e of the str<strong>on</strong>gest risk factors for breast cancer (BC), with a high<br />

populati<strong>on</strong> attributable risk. Recent genetic studies have identified 77 comm<strong>on</strong> susceptibility loci for BC<br />

which together explain 14% of familial risk. Only <strong>on</strong>e study to date has examined the c<strong>on</strong>tributi<strong>on</strong> of a<br />

subset of these variants (18 SNPs) to risk models that include breast density. We examine whether the 77<br />

genetic variants provide additi<strong>on</strong>al discriminati<strong>on</strong> of BC risk relative to age- <strong>and</strong> BMI- adjusted BI-<br />

RADS density. Further, we extend the BCSC risk model that incorporates <strong>Breast</strong> Imaging Reporting <strong>and</strong><br />

Data System (BI-RADS) density <strong>and</strong> clinical factors to examine the additi<strong>on</strong>al c<strong>on</strong>tributi<strong>on</strong> of the 77<br />

SNPs to BC risk.<br />

Methods: The sample c<strong>on</strong>sisted of 1643 cases <strong>and</strong> 2497 c<strong>on</strong>trols from three clinic-based studies with<br />

clinical risk factors, including BI-RADS density, <strong>and</strong> genotype informati<strong>on</strong> <strong>on</strong> the 77 loci. We formulated<br />

a polygenic SNP score (77-SNP) from the reported per-SNP odds ratios, <strong>and</strong> used quartiles of this score in<br />

analyses. We evaluated whether BI-RADS density <strong>and</strong> the 77-SNP score were independent risk factors for<br />

BC, adjusted for age, BMI <strong>and</strong> study, <strong>and</strong> assessed whether the BIRADS-BC associati<strong>on</strong> differed by SNP<br />

quartiles. AUC values were compared between models with <strong>and</strong> without the 77-SNP score, using<br />

DeL<strong>on</strong>g’s method, to evaluate whether inclusi<strong>on</strong> of the 77-SNP score significantly improved the<br />

discriminatory ability of the model. We extended the BCSC risk model by incorporating into it the odds<br />

ratios (OR) for the 77-SNP groups, <strong>and</strong> estimated 5-year risks for the subjects in <strong>on</strong>e of the three studies<br />

with 456 cases, 1166 c<strong>on</strong>trols. Five-year risks from the BCSC risk model were compared to those from<br />

the BCSC+77-SNP model by computing net reclassificati<strong>on</strong> improvement (NRI) for cases <strong>and</strong> c<strong>on</strong>trols<br />

using published 5-year risk groupings (4.0).<br />

Results: BI-RADS density, adjusted for age, BMI <strong>and</strong> study, was a significant risk factor for BC (OR<br />

(95% CI): 0.6 (0.4-0.7), 1.3 (1.1-1.5) <strong>and</strong> 1.8 (1.4-2.3) for BI-RADS 1, 3 <strong>and</strong> 4 vs. 2; AUC=0.66). The<br />

77-SNP score (adjusted for age, BMI <strong>and</strong> study) was similarly associated with BC (OR (95% CI): 0.6<br />

(0.5-0.7); 1.5 (1.3-1.8); 1.7 (1.4-2.0) for Quartiles 1, 3 <strong>and</strong> 4 vs. 2; AUC=0.675). BI-RADS density <strong>and</strong><br />

77-SNP score were independent risk factors for BC, <strong>and</strong> together showed greater discriminati<strong>on</strong> of risk<br />

compared to the model with BI-RADS al<strong>on</strong>e (adjusted for age <strong>and</strong> BMI) (AUC=0.69; UC=0.028,<br />

p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BREAST CANCER RISK PREDICTION AND INDIVIDUALIZED SCREENING BASED ON<br />

COMMON GENETIC VARIATIONS AND BREAST DENSITY MEASUREMENTS<br />

Hatef Darabi<br />

Karolinska Institutet, Stockholm, Sweden<br />

Over the last decade several breast cancer risk alleles have been identified which has led to an increased<br />

interest in individualised risk predicti<strong>on</strong> for clinical purposes. In the present work we examine several<br />

models for predicting absolute risk, in particular we examine the performance of 18 breast cancer risk<br />

single-nucleotide polymorphisms (SNPs), together with mammographic percentage density (PD), body<br />

mass index (BMI) <strong>and</strong> clinical risk factors in predicting absolute risk of breast cancer.<br />

Adding mammographic PD, BMI <strong>and</strong> all 18 SNPs to a baseline Swedish Gail model improved the<br />

discriminatory accuracy (the AUC statistic) from 55% to 62%. The net reclassificati<strong>on</strong> improvement<br />

(NRI) was used to assess improvement in classificati<strong>on</strong> of women into low, intermediate, <strong>and</strong> high<br />

categories of 5-year risk, where significant positive reclassificati<strong>on</strong> was observed (NRI= 0.170).<br />

Researchers participating in the Collaborative Oncological Gene-envir<strong>on</strong>ment Study (COGS) have now<br />

identified more than 40 new SNPs associated with breast cancer. In additi<strong>on</strong> to the discovery of new<br />

susceptibility loci, the findings reported can be applied to derive estimates for disease risk based <strong>on</strong> an<br />

individual’s genetic profile, <strong>and</strong> it is expected to improve the ability to predict which women are at<br />

greater risk of developing breast cancer.<br />

ABSTRACTS FOR SPEAKERS<br />

19


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BENIGN BREAST DISEASE, MAMMOGRAPHIC BREAST DENSITY AND THE RISK OF<br />

BREAST CANCER<br />

Jeffrey A. Tice, MD; Ellen S. O’Meara, PhD; D<strong>on</strong>ald L. Weaver, MD; Celine Vach<strong>on</strong>, PhD; Rachel<br />

Ballard-Barbash, MD, MPH; Karla Kerlikowske, MD<br />

ABSTRACTS FOR SPEAKERS<br />

Divisi<strong>on</strong> of General Internal Medicine, Department of Medicine; General Internal Medicine Secti<strong>on</strong>,<br />

Department of Veteran Affairs <strong>and</strong> Departments of Medicine <strong>and</strong> Epidemiology <strong>and</strong> Biostatistics (Drs.<br />

Kerlikowske); University of California, San Francisco, San Francisco, CA. Group Health Research<br />

Institute (Dr. O’Meara); Department of Pathology, University of Verm<strong>on</strong>t College of Medicine <strong>and</strong><br />

Verm<strong>on</strong>t Cancer Center (Dr. Weaver). Applied Research Program, Divisi<strong>on</strong> of Cancer C<strong>on</strong>trol <strong>and</strong><br />

Populati<strong>on</strong> Sciences, Nati<strong>on</strong>al Cancer Institute (Dr. Ballard-Barbash).<br />

Background: Benign breast disease <strong>and</strong> high breast density are prevalent, str<strong>on</strong>g risk factors for breast<br />

cancer. Women with both risk factors may be at very high risk.<br />

Methods: We included 42,818 women participating in the <strong>Breast</strong> Cancer Surveillance C<strong>on</strong>sortium<br />

(BCSC) from 1994 through 2009 who had no prior diagnosis of breast cancer <strong>and</strong> had underg<strong>on</strong>e at least<br />

<strong>on</strong>e benign breast biopsy <strong>and</strong> mammogram; 1,359 women developed incident breast cancer in 6.1 years of<br />

follow-up (78% invasive, 22% DCIS). Community radiologists rated breast density using <strong>Breast</strong> Imaging<br />

Reporting <strong>and</strong> Data System categories: almost entirely fat (low density); scattered fibrogl<strong>and</strong>ular densities<br />

(average density); heterogeneously dense (high density); extremely dense (very high density).<br />

Results: In Cox regressi<strong>on</strong> models, benign breast disease <strong>and</strong> breast density were independently<br />

associated with breast cancer. Atypical hyperplasia <strong>and</strong> very high density was uncomm<strong>on</strong> (0.6% of<br />

biopsies), but associated with the highest risk [hazard ratio (HR); 5.3, 95% c<strong>on</strong>fidence interval (CI) 3.5-<br />

8.1] compared to n<strong>on</strong>-proliferative changes <strong>and</strong> average density. Proliferative disease without atypia (26%<br />

of biopsies) was associated with elevated risk that varied little across levels of density: average (HR 1.4,<br />

95% CI 1.1-1.7), high (HR 2.0, 95% CI 1.7-2.4), or very high (HR 2.0, 95% CI 1.5-2.7). Low breast<br />

density (4.5% of biopsies) was associated with low risk (HRs


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

DETERMINANTS OF BREAST TISSUE COMPOSITION IN 15-18 YEAR OLD GIRLS<br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Norman F. Boyd*, Ella Huszti, Greg Stanisz, Sofia Chavez, Qing Li, Linda Lint<strong>on</strong>, M<strong>on</strong>ica Taylor, Sara<br />

Hennessy, Lisa J. Martin, Salom<strong>on</strong> Minkin<br />

Campbell Institute for <strong>Breast</strong> Cancer Research Ontario Cancer Institute<br />

Background. <strong>Breast</strong> tissue is especially susceptible to carcinogenic influences in early life <strong>and</strong> several<br />

variables in early life, including birth weight, growth in adolescence <strong>and</strong> attained height, influence risk of<br />

breast cancer in later life. We examine here the associati<strong>on</strong> of these risk factors in early life with<br />

variati<strong>on</strong>s in breast tissue compositi<strong>on</strong> in 15-18 year old girls.<br />

Methods. We recruited 437 girls aged 15-18, <strong>and</strong> used magnetic res<strong>on</strong>ance scans to measure the water<br />

<strong>and</strong> fat c<strong>on</strong>tent of the breast. All scans were obtained in the follicular phase of the menstrual cycle. We<br />

also made anthropometric measurements. We obtained from mothers by questi<strong>on</strong>naire a history of their<br />

daughter’s birth weight, breast-feeding <strong>and</strong> growth during childhood, <strong>and</strong> of maternal health during<br />

pregnancy with the daughter.<br />

Results. At ages 15-18 years, percent water in the breast was positively associated with: greater birth<br />

weight (adjusted for age <strong>and</strong> durati<strong>on</strong> of breast feeding; p=0.06); l<strong>on</strong>ger durati<strong>on</strong> of breast-feeding<br />

(adjusted for age <strong>and</strong> birth weight; p=0.02); <strong>and</strong> greater increase in height between ages 9 <strong>and</strong> 12 years<br />

(adjusted for age, weight <strong>and</strong> relative height at age 9; p=0.0007). Growth between ages 12 <strong>and</strong> 15<br />

(adjusted for age, weight <strong>and</strong> relative height at age 12; p=0.6) was not associated with percent breast<br />

water at ages 15-18.<br />

Greater birth weight (p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

THE EFFECT OF WEIGHT CHANGE ON CHANGES IN BREAST MEASURES OVER<br />

MENOPAUSE<br />

J.O.P. W<strong>and</strong>ers 1 , M.F. Bakker 1 , P.H.M. Peeters 1,2 , C.H. van Gils 1<br />

ABSTRACTS FOR SPEAKERS<br />

1 Julius Center for Health Sciences <strong>and</strong> Primary Care, University Medical Center Utrecht, POBox 85500,<br />

3508 GA Utrecht, The Netherl<strong>and</strong>s; 2 Department of Epidemiology, Public Health <strong>and</strong> Primary Care,<br />

Faculty of Medicine, Imperial College, Norfolk Place, L<strong>on</strong>d<strong>on</strong> W2 1PG, UK<br />

Introducti<strong>on</strong>: Higher BMI <strong>and</strong> weight have been related to lower percent density in cross-secti<strong>on</strong>al<br />

studies. However, little is known <strong>on</strong> how weight change over time is l<strong>on</strong>gitudinally related to changes in<br />

mammographic density. Here we investigate, in a populati<strong>on</strong> going through menopause, how dense area,<br />

n<strong>on</strong>-dense area <strong>and</strong> percent density change <strong>and</strong> to what extent this is influenced by weight change over<br />

menopause.<br />

Methods: 652 women, aged 49 to 61 years at recruitment, who participated in the Prospect-EPIC cohort<br />

<strong>and</strong> went through menopause <strong>on</strong> average within 5 years after recruitment, were included in this study to<br />

examine the effect of weight change <strong>on</strong> mammographic measures over menopause. All women had a pre<strong>and</strong><br />

post-menopausal mammogram <strong>and</strong> informati<strong>on</strong> about their weight at the time of these mammograms<br />

was available. A quantitative computer-assisted method (Cumulus) was used for breast measure<br />

assessment. Women were divided into 3 groups according to their percentage weight change over<br />

menopause: 1) weight loss (more than 1.5% weight loss), 2) stable weight (not more than 1.5% weight<br />

loss or weight gain) <strong>and</strong> 3) weight gain (more than 1.5% weight gain).<br />

Results: Percentage weight change ranged from -20.3% to +125.2% . 190 (29%) women lost more than<br />

1.5% of their body weight at recruitment, 138 (21%) women remained weight stable <strong>and</strong> 324 (50%)<br />

women gained more than 1.5% of their body weight at recruitment. The mean absolute change in n<strong>on</strong>dense<br />

area in the weight loss, stable weight <strong>and</strong> weight gain group was -3.7 cm 2 (95% CI: -7.8 ; 0.5), -4.6<br />

cm 2 (95% CI: -8.9 ; -0.3) <strong>and</strong> 4.5 cm 2 (95% CI: 1.8 ; 7.2), respectively (p-value for a linear trend, p-trend<br />

< 0.001). The mean absolute change in dense area was -16.2 cm 2 (95% CI: -19.6 ; -12.8), -15.0 cm 2 (95%<br />

CI: -18.0 ; -12.0) <strong>and</strong> -16.3 cm 2 (95% CI: -18.1 ; -14.4), respectively (p-trend=0.87). Finally, the mean<br />

absolute change in percentage breast density was -5.7% (95% CI: -7.7 ; -3.7), -5.0% (95% CI: -7.2 ; -<br />

2.8) <strong>and</strong> -9.3% (95% CI: -10.7 ; -7.8), respectively (p-trend=0.001).<br />

C<strong>on</strong>clusi<strong>on</strong>: Women who go through menopause <strong>on</strong> average show a decrease in both dense area <strong>and</strong><br />

percent density. The decrease in dense area is independent of weight change. The decrease in percent<br />

density is larger in women who gain weight than in women with stable weight or weight loss. This is due<br />

to an increase in the n<strong>on</strong>-dense (fatty) tissue in this group. The role of the fatty breast tissue in breast<br />

cancer risk is subject of further investigati<strong>on</strong>.<br />

22


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

AWARENESS OF BREAST DENSITY AND ITS RELATIONSHIP TO MAMMOGRAPHY<br />

SENSITIVITY AND BREAST CANCER RISK AMONG U.S. WOMEN: RESULTS OF A<br />

NATIONAL SURVEY<br />

Rhodes DJ, Radecki-Breitkopf C, Ziegenfuss J*, Jenkins SM, Vach<strong>on</strong> CM.<br />

Mayo Clinic, Rochester MN (*HealthPartners, Minneapolis MN)<br />

Background: Recent legislati<strong>on</strong> m<strong>and</strong>ating breast density (BD) notificati<strong>on</strong> assumes a knowledge deficit<br />

am<strong>on</strong>g screening-aged women, but little is known about women’s awareness of BD or its impact <strong>on</strong><br />

mammography sensitivity <strong>and</strong> breast cancer risk.<br />

Methods: Using a probability-based web panel (KnowledgePanel) representative of the United States<br />

populati<strong>on</strong>, we surveyed women aged 40 -74 years regarding their awareness of BD <strong>and</strong> knowledge of the<br />

impact of BD <strong>on</strong> mammography sensitivity <strong>and</strong> breast cancer risk.<br />

Results: Of 2,311 women surveyed, 1,506 completed questi<strong>on</strong>naires. Overall, 58% of women had heard<br />

of BD. Factors significantly correlated with a higher likelihood of having heard of BD include: age older<br />

than 50 years (p < 0.001); White race/ethnicity (p < 0.0001); higher income <strong>and</strong> educati<strong>on</strong>al attainment (p<br />

< 0.0001); self-described health status as “very good” or “excellent”(p < 0.01); use of horm<strong>on</strong>e therapy (p<br />

< 0.01); prior mammography (p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BREASTCARE: A PRIMARY CARE CLINIC-BASED RCT TO INCREASE BREAST CANCER<br />

KNOWLEDGE AND DISCUSSION OF RISK AND LIFESTYLE BEHAVIORS<br />

Celia P. Kaplan, DrPH, MA, Jennifer Livaudais-Toman, PhD, Steven Gregorich, PhD, Jeffrey Tice, MD,<br />

Karla Kerlikowske, MD, Rena Pasick, PhD, Alice Chen, MD, Eliseo J. Perez-Stable, MD, Leah S.<br />

Karliner, MD<br />

ABSTRACTS FOR SPEAKERS<br />

Background. Despite the availability of breast cancer risk assessment tools <strong>and</strong> interventi<strong>on</strong>s for risk<br />

reducti<strong>on</strong>, these tools are not well integrated into clinical practice. As a result, many women do not<br />

engage in a discussi<strong>on</strong> of their breast cancer risk with their physician, which may lead to underuse of<br />

effective risk reducti<strong>on</strong> interventi<strong>on</strong>s.<br />

Methods. We c<strong>on</strong>ducted a r<strong>and</strong>omized c<strong>on</strong>trolled trial that compared usual care to a tablet-pers<strong>on</strong>al<br />

computer (PC) based breast cancer risk assessment <strong>and</strong> educati<strong>on</strong> interventi<strong>on</strong> (<strong>Breast</strong>CARE) delivered in<br />

a primary care setting. We enrolled women aged 40-74 years with no pers<strong>on</strong>al breast cancer history prior<br />

to their scheduled primary care visits at two clinics (<strong>on</strong>e academic medical center, <strong>on</strong>e safety-net)<br />

between June 2011 <strong>and</strong> August 2012, <strong>and</strong> r<strong>and</strong>omized them to Interventi<strong>on</strong> or Usual Care (UC) arms,<br />

stratified by race/ethnicity. The Interventi<strong>on</strong> group completed the <strong>Breast</strong>CARE interventi<strong>on</strong> at the clinic<br />

just prior to their clinic visit <strong>and</strong> women <strong>and</strong> physicians received a tailored risk report. The UC group<br />

completed a teleph<strong>on</strong>e-based risk assessment after their clinic visit. We categorized women as high or<br />

average risk based <strong>on</strong> family history using the Referral Screening Tool (RST) or breast cancer risk factors<br />

using the Gail/<strong>Breast</strong> Cancer Surveillance C<strong>on</strong>sortium (BCSC) risk models. We c<strong>on</strong>tacted all women for<br />

a follow-up teleph<strong>on</strong>e survey <strong>on</strong>e week after risk assessment. We used generalized estimating equati<strong>on</strong>s to<br />

account for clustering by physician, <strong>and</strong> to estimate differences at follow-up between Interventi<strong>on</strong> <strong>and</strong> UC<br />

groups in above average knowledge of breast cancer risk factors (cut-point based <strong>on</strong> mean knowledge<br />

score from sample), <strong>and</strong> discussi<strong>on</strong> of breast cancer risk <strong>and</strong> lifestyle behaviors (exercise <strong>and</strong> weight)<br />

with physician.<br />

Results. A total of 1,278 women completed risk assessments <strong>and</strong> signed c<strong>on</strong>sent forms (596 Interventi<strong>on</strong><br />

<strong>and</strong> 665 UC) <strong>and</strong> 1,235 (97%) completed follow-up interviews (580 Interventi<strong>on</strong> <strong>and</strong> 655 UC). The mean<br />

sample age was 56 years (SD=9) with 35% n<strong>on</strong>-Latina White, 23% Latina, 22% African American, 18%<br />

Asian/Pacific Isl<strong>and</strong>er <strong>and</strong> 2% Native American/other. Demographic characteristics <strong>and</strong> breast cancer risk<br />

distributi<strong>on</strong>s were well-balanced between Interventi<strong>on</strong> <strong>and</strong> UC groups. Nine percent of women qualified<br />

for high-risk referral based <strong>on</strong> RST score <strong>and</strong> 16% qualified based <strong>on</strong> Gail/BCSC models. Compared to<br />

women in the UC group, those in the Interventi<strong>on</strong> group reported greater knowledge of breast cancer risk<br />

factors (69% vs. 55%), <strong>and</strong> more discussi<strong>on</strong> of breast cancer risk (41% vs. 14%), exercise (75% vs. 61%)<br />

<strong>and</strong> weight (67% vs. 57%) with physician. Discussi<strong>on</strong> of risk was greatest am<strong>on</strong>g women at the highest<br />

risk for breast cancer who received the interventi<strong>on</strong> (51% Interventi<strong>on</strong> vs. 18% UC). Multivariable<br />

analysis results are presented in Table 1.<br />

Table 1. Multivariable analysis at follow-up*<br />

Above average<br />

knowledge of breast<br />

cancer risk factors<br />

24<br />

Discussed breast<br />

cancer risk with<br />

physician<br />

Discussed regular<br />

exercise with<br />

physician<br />

Discussed weight<br />

with physician<br />

OR (95% CI) OR (95% CI) OR (95% CI) OR (95% CI)<br />

(ref=UC) Interventi<strong>on</strong> group 1.78 (1.37-2.31)° 3.94 (2.90-5.37)° 1.95 (1.51-2.51)° 1.60 (1.26-2.02)°<br />

*Analyses account for clustering of observati<strong>on</strong>s by physician <strong>and</strong> are adjusted for clinic site <strong>and</strong> objective breast cancer risk<br />

°p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

IMPACT OF THE TRANSITION TO DIGITAL MAMMOGRAPHY ON THE BENEFITS,<br />

HARMS AND COSTS OF SCREENING FOR BREAST CANCER IN THE US<br />

Natasha K. Stout, PhD 1 ; S<strong>and</strong>ra J. Lee, DSc 2 ; Clyde B. Schechter, MD MA 3 ; Oguzhan Alagoz, PhD 4,5 ;<br />

D<strong>on</strong>ald Berry, PhD 6 ; Diana S.M. Buist, PhD 7 ; Mucahit Cevik 4 ; Gary Chisholm 6 ; Harry J. de K<strong>on</strong>ing, MD<br />

PhD 8 ; Hui Huang, MS 2 ; Rebecca Hubbard PhD 7 ; Karla Kerlikowske, MD 9 ; Mark Munsell, MS 6 ; Anna N.<br />

A. Tostes<strong>on</strong>, ScD 10 ; Amy Trentham-Dietz, PhD 5 ; Nicolien T. van Ravesteyn, MSc 8 ; Diana L. Miglioretti,<br />

PhD 11 ; Jeanne S. M<strong>and</strong>elblatt, MD MPH 12<br />

1 Department of Populati<strong>on</strong> Medicine, Harvard Medical School <strong>and</strong> Harvard Pilgrim Health Care<br />

Institute, Bost<strong>on</strong>, MA; 2 Dana-Farber Cancer Institute, Bost<strong>on</strong>, MA; 3 Departments of Family & Social<br />

Medicine <strong>and</strong> Epidemiology & Populati<strong>on</strong> Health, Albert Einstein College of Medicine, Br<strong>on</strong>x, New York;<br />

4 Department of Industrial <strong>and</strong> Systems Engineering, University of Wisc<strong>on</strong>sin, Madis<strong>on</strong>, WI; 5 Department<br />

of Populati<strong>on</strong> Health Sciences <strong>and</strong> Carb<strong>on</strong>e Cancer Center, University of Wisc<strong>on</strong>sin, Madis<strong>on</strong>, WI; 6 The<br />

University of Texas M.D. Anders<strong>on</strong> Cancer Center, Houst<strong>on</strong>, TX; 7 Group Health Research Institute,<br />

Seattle, WA; 8 Department of Public Health, Erasmus MC, Rotterdam, The Netherl<strong>and</strong>s; 9 Departments of<br />

Epidemiology <strong>and</strong> Biostatistics, <strong>and</strong> General Internal Medicine Secti<strong>on</strong>, Department of Veterans Affairs,<br />

University of California, San Francisco, CA; 10 The Dartmouth Institute for Health Policy <strong>and</strong> Clinical<br />

Practice, Geisel School of Medicine at Dartmouth, Leban<strong>on</strong>, NH; 11 Department of Public Health<br />

Sciences, School of Medicine, University of California, Davis; 12 Department of Oncology, Georgetown<br />

University Medical Center <strong>and</strong> Cancer Preventi<strong>on</strong> <strong>and</strong> C<strong>on</strong>trol Program, Lombardi Comprehensive<br />

Cancer Center<br />

Fueled in part by the 2005 Digital Mammographic Imaging Trial results showing improved sensitivity for<br />

selected subgroups of women, digital mammography has quickly replaced plain film mammography for<br />

routine breast cancer screening. Use in community settings supports the modest improvements in<br />

sensitivity for younger women <strong>and</strong> women with radiographically dense breasts seen in the trial, however,<br />

specificity is reduced. Further, digital has higher cost compared with plain film mammography. The<br />

impact of the switch in technology from film to digital mammography <strong>on</strong> women’s health <strong>and</strong> ec<strong>on</strong>omic<br />

costs is unclear.<br />

Therefore, we will 1) assess the health effects <strong>and</strong> m<strong>on</strong>etary costs of screening mammography using<br />

digital mammography instead of film according the 2009 US Preventive Services Task Force breast<br />

cancer screening guidelines, <strong>and</strong> 2) examine alternative screening schedules using digital mammography<br />

that extend screening to younger women or women with dense breast tissue, two subgroups where digital<br />

has been shown to perform better than film mammography. This analysis represents a collaborati<strong>on</strong><br />

between researchers from the Nati<strong>on</strong>al Cancer Institute’s Cancer Interventi<strong>on</strong> <strong>and</strong> Surveillance Modeling<br />

Network (CISNET) <strong>and</strong> <strong>Breast</strong> Cancer Surveillance C<strong>on</strong>sortium (BCSC). We will use a comparative<br />

modeling approach with five independent CISNET computer models of U.S. breast cancer epidemiology.<br />

The models account for differential risk breast cancer risk <strong>and</strong> mammography performance by breast<br />

density using inputs based <strong>on</strong> data from the BCSC.<br />

Model outcomes will include life years, quality-adjusted life years, costs, breast cancer mortality <strong>and</strong> false<br />

positives per screening scenario. We will compute incremental cost-effectiveness ratios for the digital<br />

screening strategies both within each model <strong>and</strong> across the five models to underst<strong>and</strong> the value of<br />

alternative screening scenarios. The comparative modeling approach using five models will allow us to<br />

better account for uncertainty in breast cancer natural history <strong>on</strong> analysis c<strong>on</strong>clusi<strong>on</strong>s about the benefits<br />

<strong>and</strong> harms of screening.<br />

ABSTRACTS FOR SPEAKERS<br />

25


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

POSTER<br />

ABSTRACTS<br />

26


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Poster<br />

#<br />

P1<br />

P2<br />

P3<br />

P4<br />

P5<br />

P6<br />

P7<br />

P8<br />

P9<br />

P10<br />

P11<br />

P12<br />

P13<br />

P14<br />

P15<br />

P16<br />

P17<br />

P18<br />

P19<br />

P20<br />

P21<br />

P22<br />

Poster Title<br />

EXPLORING A RELATIONSHIP BETWEEN EXTRACELLULAR MATRIX<br />

STIFFNESS AND MAMMOGRAPHIC DENSITY IN THE HUMAN BREAST<br />

COMPARISON OF BREAST DENSITY MEASUREMENTS WITH A<br />

MAMMOGRAPHIC VOLUMETRIC AND AREA ALGORITHM AND<br />

MAGNETIC RESONANCE IMAGING<br />

COMPARISON OF CUMULUS AREA DENSITY WITH TWO AUTOMATED<br />

VOLUMETRIC BREAST DENSITY ALGORITHMS<br />

COMPARISON OF THREE METHODS FOR DENSITY ASSESSMENT AND<br />

THEIR ASSOCIATION TO BREAST CANCER RISK<br />

MEASUREMENT OF COMPRESSED BREAST THICKNESS FOR BREAST<br />

DENSITY ASSESSMENT USING A GAMES CONSOLE INPUT DEVICE<br />

ASSOCIATIONS OF MAMMOGRAPHIC DENSE AND NON-DENSE AREA<br />

AND BODY MASS INDEX WITH RISK OF BREAST CANCER<br />

PREDICTION OF MAMMOGRAPHIC DENSITY USING REPRODUCTIVE<br />

AND LIFESTYLE CHARACTERISTICS<br />

MAMMOGRAPHIC DESNITY AND RISK OF BREAST CANCER BY AGE<br />

AND TUMOR CHARACTERISTICS<br />

MAMMOGRAPHIC PIXEL VARIANCE AND RISK OF BREAST CANCER IN<br />

A NESTED CASE-CONTROL STUDY<br />

VALIDATION OF A FULLY AUTOMATED, HIGH THROUGHPUT<br />

MAMMOGRAPHIC DENSITY MEASUREMENT TOOL FOR USE WITH<br />

PROCESSED DIGITAL MAMMOGRAMS<br />

RESULTS OF SUPPLEMENTAL ULTRASOUND SCREENING IN WOMEN<br />

WITH AND WITHOUT DENSE BREAST TISSUE<br />

COMPARISON OF CUMULUS MAMMOGRAPHIC DENSITY READINGS<br />

BETWEEN FUJIFILM COMPUTED RADIOGRAPHY DIGITAL<br />

MAMMOGRAPHIC IMAGES IN RAW AND DISPLAY FORMATS<br />

RISK ASSOCIATION OF SUBREGIONAL MEASUREMENTS OF BREAST<br />

DENSITY<br />

THE ASSOCIATION BETWEEN VITAMIN D INTAKE, SEASON AND<br />

MAMMOGRAPHIC DENSITY IN NORWEGIAN POSTMENOPAUSAL<br />

WOMEN<br />

THE ASSOCIATION BETWEEN GEOGRAPHIC STATUS AND<br />

MAMMOGRAPHIC DENSITY<br />

INTER-SCAN REPRODUCIBILITY OF MAMMOGRAPHIC DENSITY<br />

MEASUREMENTS<br />

MULTIPLE METABOLIC RISK FACTORS AND MAMMOGRAPHIC BREAST<br />

DENSITY IN THE NEW YORK CITY MULTIETHNIC BREAST CANCER<br />

PROJECT<br />

GENETIC TESTING FOR BREAST CANCER RISK ESTIMATION – A COST -<br />

EFFECTIVENESS ANALYSIS<br />

ADIPOSITY AND MAMMOGRAPHIC BREAST DENSITY IN<br />

PREMENOPAUSAL CHILEAN WOMEN<br />

RELATIONSHIP OF SERUM ESTROGEN LEVELS WITH AREA AND VOLUME<br />

MEASURES OF MAMMOGRAPHIC DENSITY AMONG WOMEN<br />

UNDERGOING BREAST BIOPSY FOR CLINICAL INDICATIONS<br />

MAMMOGRAPHIC DENSITY PHENOTYPES AND RISK OF BREAST CANCER:<br />

A META-ANALYSIS<br />

MAMMOGRAPHIC DENSITY AND EARLY MAMMOGRAPHIC SCREENING<br />

PERFORMANCE MEASURES IN THE NORWEGIAN BREAST CANCER<br />

SCREENING PROGRAM<br />

Author<br />

Acerbi<br />

Al<strong>on</strong>zo<br />

Al<strong>on</strong>zo<br />

Assi<br />

Astley<br />

Baglietto (Hopper)<br />

Bakker<br />

Bertr<strong>and</strong><br />

Brentnall<br />

Couwenberg<br />

Dean<br />

Dickens<br />

Duewer<br />

Ellingjord-Dale<br />

Emaus<br />

Emaus<br />

Engmann<br />

Folse<br />

Garmendia<br />

Gierach<br />

Graff<br />

Hofvind<br />

27


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P23<br />

P24<br />

P25<br />

P26<br />

P27<br />

P28<br />

P29<br />

P30<br />

P31<br />

P32<br />

P33<br />

CHILDHOOD AND ADOLESCENT GROWTH PREDICTS MAMMOGRAPHIC<br />

DENSITY MEASURES ASSOCIATED WITH BREAST CANCER RISK IN MID-<br />

LIFE: A PROSPECTIVE STUDY<br />

BACKGROUND PARENCHYMAL UPTAKE OF Tc-99m SESTAMIBI ON<br />

MOLECULAR BREAST IMAGING IN WOMEN WITH DENSE BREASTS<br />

IMPACT OF CALIFORNIA BREAST DENSITY NOTIFICATION LAW SB 1538<br />

ON CALIFORNIA WOMEN AND THEIR HEALTH CARE PROVIDERS<br />

CHALLENGES IN DE-IDENTIFYING FILM-SCREEN MAMMOGRAMS PRIOR<br />

TO BREAST DENSITY ASSESSMENT FOR THE NOVICE PRACTITIONER<br />

ASSOCIATION BETWEEN AUTOMATED, VOLUMETRIC BREAST DENSITY<br />

MEASURES AND BREAST CANCER IN A LARGE SCREENING POPULATION<br />

LONGITUDINAL TRACKING WITH TIME OF MAMMOGRAPHIC DENSITY<br />

MEASURES THAT PREDICT BREAST CANCER<br />

MULTI-VENDOR VALIDATION OF A FULL-AUTOMATED BREAST DENSITY<br />

ESTIMATION METHOD APPLICATION TO BOTH RAW AND PROCESSED<br />

DIGITAL MAMMOGRAMS<br />

GENETIC DETERMINANTS OF FIBROGLANDULAR BREAST TISSUE:<br />

IMPLICATIONS ON USING MAMMOGRAPHIC DENSITY MEASURES IN<br />

BREAST CANCER RISK ASSESSMENT<br />

FINE-MAPPING OF THREE MAMMOGRAPHIC DENSITY LOCI USING NEXT-<br />

GENERATION SEQUENCING<br />

APPLICATION OF SHAPE AND APPEARANCE MODEL APPROACH TO<br />

MAMMOGRAMS<br />

METHODS TO ASSESS AND REPRESENT MAMMOGRAPHIC DENSITY: AN<br />

ANALYSIS OF FOUR CASE-CONTROL STUDIES<br />

28<br />

Hopper<br />

Hruska<br />

Ikeda<br />

(Hargreaves/Fellow)<br />

Jobling (Forbes)<br />

Kallenberg<br />

Kavitha (Hopper)<br />

K<strong>on</strong>tos<br />

K<strong>on</strong>tos<br />

Lindstrom<br />

Malkov<br />

Maskarinec<br />

P34 INTERNATIONAL POOLING PROJECT OF MAMMOGRAPHIC DENSITY McCormack<br />

AN APPROACH TO THE STANDARDIZATION OF PERCENT<br />

P35 MAMMOGRAPHIC DENSITY TO ENABLE JOINT ANALYSIS OF DATA<br />

ACROSS MULTIPLE CENTERS<br />

Pankratz<br />

P36<br />

BREAST DEVELOPMENT AT DIFFERENT BREAST TANNER STAGES:<br />

EVIDENCE FROM THE GROWTH OBESITY CHILEAN COHORT STUDY<br />

Pereira<br />

P37<br />

DUAL X-RAY ABSORPTIOMETRY AN ALTERNATIVE METHOD TO MEASURE<br />

VOLUMETRIC BREAST DENSITY<br />

Pereira<br />

P38<br />

AUTOMATED DENSITY RISK SCORING WITH LEARNED HIERARCHICAL<br />

FEATURES<br />

Petersen<br />

P39<br />

ASSURE - ADAPTING BREAST CANCER SCREENING STRATEGY USING<br />

PERSONALISED RISK ESTIMATION<br />

Platel<br />

P40 BREAST DENSITY INFORM: NEW YORK STATE AND FEDERAL INITIATIVES Pushkin<br />

CIRCULATING INSULIN-LIKE GROWTH FACTOR-1, INSULIN-LIKE GROWTH<br />

P41<br />

FACTOR BINDING PROTEIN-3, GENETIC POLYMORPHISMS, AND<br />

Rice<br />

MAMMOGRAPHIC DENSITY IN PREMENOPAUSAL MEXICAN WOMEN:<br />

RESULTS FROM THE ESMAESTRAS COHORT<br />

P42<br />

P43<br />

P44<br />

P45<br />

P46<br />

P47<br />

ADJUSTING FOR INTER-OBSERVER VARIATION IN VISUAL ASSESSMENT<br />

OF PERCENTAGE BREAST DENSITY ON A CONTINUOUS SCALE AND<br />

DETERMINING WOMEN AT HIGH RISK OF DEVELOPING BREAST CANCER<br />

RISK<br />

VISUAL ANALOGUE SCALE ASSESSMENT OF PERCENTAGE BREAST<br />

DENSITY: A STUDY OF AGREEMENT BETWEEN OBSERVERS AND WITH<br />

INTERACTIVE THRESHOLDING<br />

LARGE INTERNATIONAL STUDY REVEALS NOVEL ASSOCIATIONS<br />

BETWEEN COMMON BREAST CANCER SUSCEPTIBILITY VARIANTS AND<br />

RISK-PREDICTING MAMMOGRAPHIC DENSITY MEASURES<br />

META-ANALYSIS OF TWELVE GENOME-WIDE ASSOCIATION STUDIES<br />

(GWAS) IDENTIFIES NOVEL GENETIC LOCI ASSOCIATED WITH<br />

MAMMOGRAPHIC DENSITY PHENOTYPES<br />

NON-INVASIVE OPTICAL ASSESSMENT OF BREAST DENSITY AND<br />

IDENTIFICATION OF HIGH-RISK SUBJECTS<br />

RETROSPECTIVE ANALYSIS OF BREAST DENSITY IN TRIPLE NEGATIVE<br />

BREAST CANCERS<br />

Sergeant (Astley)<br />

Sergeant (Astley)<br />

St<strong>on</strong>e<br />

Tamimi<br />

Tar<strong>on</strong>i<br />

Vijayaraghavan


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P48<br />

A FULLY-AUTOMATED METHOD FOR QUANTIFYING FIBROGLANDULAR<br />

TISSUE AND BACKGROUND PARENCHYMAL ENHANCEMENT IN BREAST<br />

MRI<br />

Wu<br />

29


P1<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

EXPLORING A RELATIONSHIP BETWEEN EXTRACELLULAR MATRIX STIFFNESS<br />

AND MAMMOGRAPHIC DENSITY IN THE HUMAN BREAST<br />

Irene Acerbi 1,2 , Alfred Au 2 , Ori Maller 1,2 , Quanming Shi 2,3 , Jan Liphardt 2,3 , Yunn-yi Chen 4 , Shelley<br />

Hwang 5 , Valerie M. Weaver 1,2,6,7,8<br />

1 Department of Surgery <strong>and</strong> Center for Bioengineering <strong>and</strong> Tissue Regenerati<strong>on</strong>, University of California,<br />

San Francisco, CA, USA; 2 Bay Area Physical Sciences in Oncology Center, Berkeley, CA, USA<br />

3 Department of Physics, University of California, Berkeley, CA, USA; 4 UCSF Carol Buck <strong>Breast</strong> Cancer<br />

Center, San Francisco, CA, USA; 5 Department of Pathology, University of California, San Francisco, CA,<br />

USA; 6 Department of Surgery, Duke University, Durham, NC, USA; 7 Institute for Regenerati<strong>on</strong> Medicine,<br />

University of California San Francisco, CA, USA; 8 Department of Bioengineering <strong>and</strong> Therapeutic<br />

Sciences, University of California, San Francisco, CA, USA; 9 Department of Anatomy, University of<br />

California, San Francisco, CA, USA<br />

Mammographic density (MD) is associated with high collagen c<strong>on</strong>tent in the extracellular matrix<br />

(ECM) <strong>and</strong> greater risk to malignancy. Data from our group <strong>and</strong> others have highlighted the importance<br />

of mechanical cues from the ECM in breast tissue homeostasis <strong>and</strong> tumor progressi<strong>on</strong> [Paszek et al.,<br />

Cancer Cell 2005; Levental et al., Cell, 2009; reviewed in Yu et al., Trends Cell Biol, 2010].<br />

Nevertheless, a role for ECM stiffness in tumor cell initiati<strong>on</strong> is unknown. In this study, we hypothesize<br />

that high MD correlates with stiffer breast ECM <strong>and</strong> enhances women’s risk for breast cancer compared<br />

to low MD.<br />

To test this hypothesis, we studied breast tissues obtained through prophylactic mastectomy from<br />

women with high MD (BIRADS 4) versus low (BIRADS 1). From each surgically excised breast,<br />

samples of 0.5cm x 0.5cm x 1cm dimensi<strong>on</strong> were removed from the retroareolar regi<strong>on</strong> <strong>and</strong> from 4<br />

peripheral quadrants. Sample secti<strong>on</strong>s were subjected to biophysical, morphological <strong>and</strong> biochemical<br />

analysis. Biophysical analysis included the applicati<strong>on</strong> of Atomic Force Microscopy (AFM) to obtain an<br />

extensive force map of distinct anatomical regi<strong>on</strong>s of the ECM associated with the intra-lobular <strong>and</strong> interlobular<br />

ECM. ECM architecture has been analyzed using two phot<strong>on</strong>s <strong>and</strong> SIM-POL imaging coupled<br />

with picrosirius staining, polarized light imaging <strong>and</strong> image quantificati<strong>on</strong>. Biochemical <strong>and</strong><br />

morphological analysis c<strong>on</strong>sisted of immunohistochemistry for markers that detect mechano-signaling in<br />

the epithelium <strong>and</strong> stromal fibroblasts, <strong>and</strong> H&E to visualize cellular <strong>and</strong> ECM organizati<strong>on</strong>.<br />

We studied samples from the different 5 quadrants of a same breast. We found that the ECM<br />

associated with the upper outer regi<strong>on</strong>, was in tendency stiffer (AFM) <strong>and</strong> had higher tensi<strong>on</strong> (SIMPOL<br />

birefringence) than the other peripheral quadrants. We show heterogeneity within multiple regi<strong>on</strong>s of the<br />

same breast, with the upper outer regi<strong>on</strong>, where most tumors occur, c<strong>on</strong>tained linearized collagen fibrils.<br />

Most invasive breast cancers arise from the ductal epithelium. Therefore, we studied the ECM<br />

associated with the Terminal Ductal Lobular Units (TDLU): inter-lobular ECM (am<strong>on</strong>g the TLDU), <strong>and</strong><br />

intra-lobular ECM (within the TDLU). The inter-lobular ECM of the breast c<strong>on</strong>tained linearized collagen<br />

fibrils <strong>and</strong> was relatively stiffer. In c<strong>on</strong>trast, we found that the intra-lobular ECM associated with the<br />

TDLU in the breast c<strong>on</strong>tained less linearized collagen fibrils <strong>and</strong> was compliant.<br />

In a small cohort, breast tissues from of the upper outer regi<strong>on</strong> were examine to directly explore<br />

the relati<strong>on</strong>ship between extracellular matrix stiffness <strong>and</strong> high MD across a patient populati<strong>on</strong>. Our<br />

preliminary data suggest an increase in collagen abundance <strong>and</strong> higher c<strong>on</strong>tent of linearized collagen<br />

fibers adjacent to TLDUs were associated with high MD. Our future efforts will focus <strong>on</strong> increasing the<br />

size of the cohort to further establish a correlative relati<strong>on</strong>ship between MD <strong>and</strong> ECM stiffness. We will<br />

also examine the significance of this relati<strong>on</strong>ship in tumor cell initiati<strong>on</strong>.<br />

ABSTRACTS<br />

Acknowledgements: supported by W81XWH-05-1-0330 <strong>and</strong> R01 CA138818-01A1 to VMW, 1U01 ES019458-01<br />

to VMW <strong>and</strong> ZW, <strong>and</strong> P50 CA 58207 to JG, VW, SH <strong>and</strong> LC, U54CA143836-01 to JL <strong>and</strong> VW, <strong>and</strong> Susan G.<br />

Komen for the Cure PSF12230246 to IA.<br />

30


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P2 COMPARISON OF BREAST DENSITY MEASUREMENTS WITH A<br />

MAMMOGRAPHIC VOLUMETRIC AND AREA ALGORITHM AND MAGNETIC<br />

RESONANCE IMAGING<br />

O Al<strong>on</strong>zo-Proulx 1 , J Mainprize 1 , E. Warner 1 , J. Harvey 2 , M. Yaffe 1<br />

1 Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Tor<strong>on</strong>to, Ontario Canada, M4N 3M5 ;<br />

2 University of Virginia Health System, Charlottesville, Virginia<br />

Introducti<strong>on</strong>: Area <strong>and</strong> volumetric breast density (ABD <strong>and</strong> VBD) is typically determined from twodimensi<strong>on</strong>al<br />

x-ray projecti<strong>on</strong> mammograms. In this study we compare the values of ABD, VBD, dense<br />

volume <strong>and</strong> total breast volume estimated in this way, with measurements obtained directly from a threedimensi<strong>on</strong>al<br />

breast MRI image dataset.<br />

ABSTRACTS<br />

Materials <strong>and</strong> Methods: Our VBD algorithm, CUMULUS V, has been described earlier [1,2]. A<br />

calibrati<strong>on</strong> surface relating the imaging signal to the compositi<strong>on</strong> <strong>and</strong> thickness of breast phantoms is<br />

obtained experimentally, <strong>and</strong> corrected using simulati<strong>on</strong> to compensate for the attenuati<strong>on</strong> difference<br />

between the phantom materials <strong>and</strong> breast tissue. The resp<strong>on</strong>se of the thickness readout of the<br />

mammography machine is characterized to allow for an accurate estimate of the breast thickness under<br />

compressi<strong>on</strong>. The ABD measurements were d<strong>on</strong>e using the CUMULUS area algorithm. The MR images<br />

were obtained from a study of high-risk women [3]. The images were segmented in four steps. 1) the<br />

inhomogeneities arising from the coil sensitivity were corrected semi-automatically in each slice; 2) a<br />

user delineated the chest wall <strong>and</strong> air boundaries; 3) a user manually selected the signal peaks due to fatty<br />

<strong>and</strong> fibrogl<strong>and</strong>ular tissue; 4) a soft-threshold functi<strong>on</strong> was applied between the peaks to transform the<br />

image into fracti<strong>on</strong>al density c<strong>on</strong>tent. Images from 80 patients (left <strong>and</strong> right breast) were analyzed in this<br />

study.<br />

Results: The digital mammograms were obtained, <strong>on</strong> average, 3 years after the MR exam, when the mean<br />

age of the women was 45 years. Figure 1 <strong>and</strong> Table 1 compare the mammography <strong>and</strong> MR measurements.<br />

The mean VBD <strong>and</strong> volumes were 32.3 % <strong>and</strong> 677 cm 3 , respectively, for the mammography measurement<br />

versus 32.3 % <strong>and</strong> 645 cm 3 for the MR measurement. The comparis<strong>on</strong> with ABD has not been completed<br />

at this point.<br />

Figure 1: comparis<strong>on</strong> between the mammographic density algorithm <strong>and</strong> MRI for the VBD (left) <strong>and</strong> breast<br />

volume (right).<br />

31


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

RMS diff. Pears<strong>on</strong> Slope Intercept<br />

correlati<strong>on</strong><br />

VBD 10.1 % 0.801 1.05 -1.6 %<br />

Volume 157 cm 3 0.934 1.14 -55 cm 3<br />

Dense volume 66 cm 3 0.733 0.81 33 cm 3<br />

Table 1: Root mean square (RMS) difference, correlati<strong>on</strong>, <strong>and</strong> linear least square fits between the MR <strong>and</strong><br />

mammography measurements.<br />

Discussi<strong>on</strong> <strong>and</strong> c<strong>on</strong>clusi<strong>on</strong>: There was good correlati<strong>on</strong> <strong>and</strong> agreement between the MR <strong>and</strong><br />

mammography measurements. The accuracies of the VBD algorithm <strong>and</strong> MR segmentati<strong>on</strong> are 3-5 <strong>and</strong> 2-<br />

4 percentage points, respectively, thus we can expect an error of 4-7 % between the two. Further errors<br />

are expected to occur due to the time difference between the exams, <strong>and</strong> from differences in the imaged<br />

field-of-view. To the best of our knowledge, <strong>on</strong>ly <strong>on</strong>e other study has investigated the relati<strong>on</strong> between<br />

MR <strong>and</strong> mammography density measurements [4].<br />

References:<br />

1. Al<strong>on</strong>zo-Proulx O et al. Phys. Med. Biol. 2010 55 3027-44<br />

2. Al<strong>on</strong>zo-Proulx O et al. Phys. Med. Biol. 2012 57 7443-57.<br />

3. Warner E, et al. J Clin Oncol. 2011 29(13) 1664-9.<br />

4. Van Engel<strong>and</strong> S et al. IEEE Trans. Med. Imaging 2006 25 273-82<br />

ABSTRACTS<br />

32


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P3 COMPARISON OF CUMULUS AREA DENSITY WITH TWO AUTOMATED<br />

VOLUMETRIC BREAST DENSITY ALGORITHMS<br />

Olivier Al<strong>on</strong>zo-Proulx 1 , Martin Yaffe 1 , Jennifer Harvey 2 .<br />

1 Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Tor<strong>on</strong>to, Ontario Canada, M4N 3M5;<br />

2 University of Virginia Health System, Charlottesville, Virginia<br />

Purpose: Measurements of breast density made with the Cumulus interactive threshold algorithm have<br />

been shown to be moderately associated with breast cancer risk. In this work we compare two volumetric<br />

density (VBD) measures (the volume proporti<strong>on</strong> of fibrogl<strong>and</strong>ular tissue), with Cumulus area percent<br />

density (PD) to validate the volumetric algorithms.<br />

ABSTRACTS<br />

Methods <strong>and</strong> materials: 310 women who had both film-screen (FS) <strong>and</strong> digital mammograms as part of<br />

the ACRIN DMIST were analyzed for PD (left CC, FS) <strong>and</strong> VBD (left <strong>and</strong> right CC, digital). One VBD<br />

algorithm, ‘Cumulus Volume’ is based <strong>on</strong> a prior calibrati<strong>on</strong> of the imaging signal versus tissue thickness<br />

<strong>and</strong> compositi<strong>on</strong>. The sec<strong>on</strong>d algorithm, ‘Volpara’, finds a pixel where <strong>on</strong>ly fatty tissue is present, whose<br />

signal intensity is then used as a reference to determine the density <strong>on</strong> the image. PD <strong>and</strong> both VBD<br />

values were compared, <strong>and</strong> density color maps were visually reviewed. The relati<strong>on</strong> between the VBD of<br />

the left-right pairs was investigated, as well as the relati<strong>on</strong> between the two volumetric algorithms.<br />

Results: The correlati<strong>on</strong> between Cumulus PD with Cumulus VBD <strong>and</strong> Volpara VBD yielded good<br />

results (R 2 correlati<strong>on</strong> of 0.74 <strong>and</strong> 0.76 respectively, using a 2 nd order polynomial fit). Outliers between<br />

Cumulus PD <strong>and</strong> Cumulus VBD were due to thin breasts, which have a higher volume density because of<br />

the skin c<strong>on</strong>tributi<strong>on</strong>, to thick breasts where the thickness estimati<strong>on</strong> was err<strong>on</strong>eous, or from poorly<br />

exposed images. Outliers between Cumulus PD <strong>and</strong> Volpara VBD were due to the algorithm being unable<br />

to find an adequate reference fatty pixel <strong>on</strong> dense breasts. The linear correlati<strong>on</strong> between Volpara <strong>and</strong><br />

Cumulus VBD was very good (R 2 =0.82), <strong>and</strong> Volpara VBD overall had lower values since it excludes the<br />

effect of skin in the volume density. The linear correlati<strong>on</strong> between the left <strong>and</strong> right breast using<br />

Cumulus VBD was excellent (R 2 =0.94).<br />

C<strong>on</strong>clusi<strong>on</strong>: Values obtained using either volumetric algorithms showed good correlati<strong>on</strong> to the area<br />

method, with <strong>on</strong>ly a few outliers, indicating that VBD can potentially be used in breast cancer risk<br />

models. The algorithms were str<strong>on</strong>gly correlated with each other. The left <strong>and</strong> right Cumulus VBD were<br />

very str<strong>on</strong>gly correlated, an indicati<strong>on</strong> of the algorithm’s reliability.<br />

Clinical relevance: Volumetric breast density measurements are automated, perfectly reproducible,<br />

correlate well with st<strong>and</strong>ard area density methods, <strong>and</strong> can thus be used to develop breast cancer risk<br />

models.<br />

Figure:<br />

33


P4<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

COMPARISON OF THREE METHODS FOR DENSITY ASSESSMENT AND THEIR<br />

ASSOCIATION TO BREAST CANCER RISK<br />

V. Assi 1 , J. Warwick 2 , N. Perry 3 , K. Pinker-Domenig 4 , S. Duffy 1<br />

1 Centre for Cancer Preventi<strong>on</strong>, Wolfs<strong>on</strong> Institute of Preventive Medicine, Queen Mary University of<br />

L<strong>on</strong>d<strong>on</strong>, L<strong>on</strong>d<strong>on</strong>, UK; 2 Imperial Clinical Trials Unit, School of Public Health, Faculty of Medicine,<br />

Imperial College L<strong>on</strong>d<strong>on</strong>, L<strong>on</strong>d<strong>on</strong>, UK; 3 The Princess Grace Hospital, The L<strong>on</strong>d<strong>on</strong> <strong>Breast</strong> Institute,<br />

L<strong>on</strong>d<strong>on</strong>, UK; 4 Medical University Vienna, Department of Radiology, Vienna, Austria<br />

Introducti<strong>on</strong> Despite mammographic density being recognised as a str<strong>on</strong>g risk factor for breast cancer,<br />

its applicati<strong>on</strong> in risk-predicti<strong>on</strong> c<strong>on</strong>texts is currently limited. This is partly due to difficulties in obtaining<br />

reliable density readings. New volumetric <strong>and</strong> fully-automated approaches, e.g. Quantra <strong>and</strong> Volpara, are<br />

expected to ensure a better performance: eliminating inter- <strong>and</strong> intra- reader variability, allowing the<br />

assessment of density in large numbers of women, <strong>and</strong> improving the risk-predicti<strong>on</strong> by estimating more<br />

directly the amount of dense tissue.<br />

Purpose Evaluati<strong>on</strong> of the estimates of mammographic density provided by Quantra <strong>and</strong> Volpara,<br />

compared to BIRADS, a well-established visual assessment method, as an indicator of breast cancer risk.<br />

Methods Quantra <strong>and</strong> Volpara are two software applicati<strong>on</strong>s that, from full-field digital mammograms<br />

(FFDM), estimate the total volume (cm 3 ) of the breast <strong>and</strong> of the dense tissue (absolute density). From<br />

these, percent density <strong>and</strong> n<strong>on</strong>-dense volume can be computed. The performance of BIRADS, Quantra<br />

<strong>and</strong> Volpara, as an indicator of breast cancer risk, was assessed using st<strong>and</strong>ard logistic regressi<strong>on</strong> adjusted<br />

for age <strong>on</strong> data from a retrospective case-c<strong>on</strong>trol study, comprising 200 cases <strong>and</strong> 200 c<strong>on</strong>trols.<br />

Relati<strong>on</strong>ship between the volumetric methods <strong>and</strong> Birads was also investigated.<br />

Results Of the Quantra measures <strong>on</strong>ly absolute density had a significant associati<strong>on</strong> with risk, OR 1.04<br />

per 10 cm 3 [1.00 – 1.07], after age adjustment. Likewise, using Volpara, density in absolute terms was the<br />

most predictive, OR 1.07 per 10 cm 3 [1.00 – 1.13]. St<strong>and</strong>ardising the odds ratios dem<strong>on</strong>strates that the<br />

two volumetric methods give very similar results: an increase of <strong>on</strong>e st<strong>and</strong>ard deviati<strong>on</strong> in absolute<br />

density was associated with age-adjusted ORs of 1.25 [1.02 - 1.53] <strong>and</strong> 1.26 [1.01 - 1.56], respectively for<br />

Quantra <strong>and</strong> Volpara. Absolute densities from these two methods were str<strong>on</strong>gly correlated (82%),<br />

although neither Quantra nor Volpara were associated with BIRADS classificati<strong>on</strong>. Women in the highest<br />

Birads dense category were twice as likely to develop cancer as women in the least dense BIRADS<br />

category, which is somewhat weaker than expected <strong>and</strong> may indicate poorer predictive ability of this<br />

assessment method when performed <strong>on</strong> digital images. This could also partially explain why Birads <strong>and</strong><br />

the two volumetric methods appear unrelated.<br />

ABSTRACTS<br />

C<strong>on</strong>clusi<strong>on</strong>s Both volumetric methods had a significant propensity in discriminating cases from c<strong>on</strong>trols.<br />

Absolute density was the most predictive. For digital images, absolute density measured with Quantra <strong>and</strong><br />

Volpara may be a better indicator of breast cancer risk than BIRADS.<br />

34


P5<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MEASUREMENT OF COMPRESSED BREAST THICKNESS FOR BREAST DENSITY<br />

ASSESSMENT USING A GAMES CONSOLE INPUT DEVICE<br />

Jeremy Hewes *1 , Andrew Williams<strong>on</strong> *1 , Philip No<strong>on</strong>an 2 , Jamie C. Sergeant 2 , Tina Dunn 3 ,<br />

Claire Mercer 3 , Miriam Griffiths 3 , Sam Haste 2 , Alan Huft<strong>on</strong> 2 , Sue Astley 2<br />

1 School of Physics <strong>and</strong> Astr<strong>on</strong>omy, University of Manchester, Manchester, UK ; 2 Centre for Imaging<br />

Sciences, Institute for Populati<strong>on</strong> Health, University of Manchester, Manchester, UK; 3 Nightingale<br />

Centre <strong>and</strong> Genesis Preventi<strong>on</strong> Centre, University Hospital of South Manchester, Manchester, UK<br />

* joint first authors<br />

Introducti<strong>on</strong>: Knowledge of compressed breast thickness is essential for accurate density assessment,<br />

but it has been shown that mammography system thickness outputs are inaccurate <strong>and</strong> compressi<strong>on</strong> plates<br />

tilt <strong>and</strong> bend for patient comfort. Furthermore, at the breast periphery the breast loses c<strong>on</strong>tact with the<br />

compressi<strong>on</strong> plate, <strong>and</strong> models or estimates of thickness must be used. Our aim is to design <strong>and</strong> evaluate a<br />

measurement method to overcome these limitati<strong>on</strong>s.<br />

ABSTRACTS<br />

The Microsoft Kinect games c<strong>on</strong>sole input device provides a low-cost range finder that is suitable for use<br />

in medical applicati<strong>on</strong>s. It projects an infra-red speckle pattern <strong>on</strong>to the object; this is observed by an<br />

infra-red detector. Depth is measured by analysing distorti<strong>on</strong>s in the received image. Here we assess the<br />

suitability of the Kinect for measuring the thickness of the compressed breast including the peripheral<br />

regi<strong>on</strong>. We have investigated the following properties: ability to image skin through the compressi<strong>on</strong><br />

plate; impact of ambient lighting; accuracy; angular <strong>and</strong> distance dependence.<br />

Method: We used a Kinect with open-source Point Cloud Library <strong>and</strong> Microsoft’s Kinect for Windows<br />

SDK including Kinect Fusi<strong>on</strong>; this can compare <strong>and</strong> combine successive frames streamed from the<br />

Kinect, enabling the creati<strong>on</strong> of a 360 o degree model of an object <strong>and</strong> improving the accuracy of<br />

measurement.<br />

Experiments to investigate Kinect imaging of different materials, the impact of ambient lighting,<br />

dependence <strong>on</strong> distance <strong>and</strong> the angular resp<strong>on</strong>se of the device were c<strong>on</strong>ducted in laboratory c<strong>on</strong>diti<strong>on</strong>s.<br />

Further imaging was carried out using clinical mammography systems <strong>and</strong> a breast phantom.<br />

Distance dependence was investigated using a flat surface at distances from 50-150cm, in 10cm<br />

increments (50-100cm) <strong>and</strong> 25cm increments (>100cm). At each distance 20 frames were acquired <strong>and</strong><br />

averaged. A regi<strong>on</strong> of interest was selected in the resulting image, <strong>and</strong> corrected for tilt using plane fitting.<br />

The st<strong>and</strong>ard deviati<strong>on</strong> of depth informati<strong>on</strong> of all points described al<strong>on</strong>g the surface yielded a<br />

measurement of the Kinect’s precisi<strong>on</strong>. The degree of averaging necessary was investigated by capturing<br />

150 frames at distances from 60-130cm <strong>and</strong> analysing the precisi<strong>on</strong> as the number of frames averaged<br />

was reduced. Angular resp<strong>on</strong>se was investigated by moving the Kinect to angles from 0 to 45 degrees <strong>and</strong><br />

analysing the Kinect output. Investigati<strong>on</strong>s of dependence <strong>on</strong> material <strong>and</strong> ambient lighting were also<br />

undertaken.<br />

Results: Precisi<strong>on</strong> was 0.8mm at 50cm, 1.7mm at 100cm, <strong>and</strong> 11.1mm at 150cm; 20-frame averaging<br />

was found to be optimal. The Kinect’s resp<strong>on</strong>se is highly material-dependent, <strong>and</strong> was reduced in<br />

envir<strong>on</strong>ments with a high intensity of infra-red light. For reflective materials viewed head-<strong>on</strong>, such as the<br />

breast support platform, the reflecti<strong>on</strong> of the Kinect’s laser projector can interfere with depth-sensing<br />

capabilities.<br />

C<strong>on</strong>clusi<strong>on</strong>s: The Kinect provides a fast, cheap <strong>and</strong> effective method for measuring depth informati<strong>on</strong> to<br />

millimetre accuracy at distances typical of digital mammography units. Furthermore, Kinect Fusi<strong>on</strong><br />

promises to provide a good model for the compressed breast thickness in 3-D.<br />

35


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Figure: A Kinect structured light image of a breast phantom compressed in a mammography system.<br />

ABSTRACTS<br />

36


P6<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

ASSOCIATIONS OF MAMMOGRAPHIC DENSE AND NON-DENSE AREA AND BODY<br />

MASS INDEX WITH RISK OF BREAST CANCER<br />

Laura Baglietto 1,2 ; Kavitha Krishnan 1,2 ; Jennifer St<strong>on</strong>e 2 ; Carmel Apicella 2 ; Melissa C. Southey 1,3 ; Dallas<br />

R. English 1,2 ; John L. Hopper 2 ; Graham G. Giles 1,2,4 .<br />

1 Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia (2) Centre for Molecular,<br />

Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytical Epidemiology, The University of Melbourne, Australia (3)<br />

Department of Pathology, The University of Melbourne, Australia; (4) Department of Epidemiology <strong>and</strong><br />

Preventive Medicine, M<strong>on</strong>ash University, Melbourne, Australia<br />

Mammographic density measures are associated with risk of breast cancer. Traditi<strong>on</strong>ally mammographic<br />

density has been quantified as percent mammographic density (PMD), <strong>and</strong> few studies have investigated<br />

the c<strong>on</strong>current roles of absolute dense <strong>and</strong> n<strong>on</strong>-dense mammographic areas, body mass index (BMI), age<br />

at mammogram <strong>and</strong> diagnosis <strong>on</strong> breast cancer risk.<br />

ABSTRACTS<br />

We c<strong>on</strong>ducted a matched case-c<strong>on</strong>trol study nested within the Melbourne Collaborative Cohort Study<br />

(MCCS) to estimate the associati<strong>on</strong>s of dense area, n<strong>on</strong>-dense area, <strong>and</strong> BMI with breast cancer risk under<br />

alternative causal models.<br />

Dense area was positively associated with risk <strong>and</strong> the strength of this associati<strong>on</strong> was <strong>on</strong>ly slightly<br />

influenced by the choice of the causal model. The associati<strong>on</strong> between n<strong>on</strong>-dense area <strong>and</strong> breast cancer<br />

risk was negative under a direct causal relati<strong>on</strong> <strong>and</strong> null otherwise. The choice of causal model affected<br />

the individual risk predicti<strong>on</strong>s, especially for women with extreme values of n<strong>on</strong>-dense area. It also<br />

impacted <strong>on</strong> the estimates of the associati<strong>on</strong>s, interpretati<strong>on</strong>s, <strong>and</strong> the risk predicti<strong>on</strong>s which might vary<br />

c<strong>on</strong>siderably whether it is assumed that the risk factor is PMD or a linear combinati<strong>on</strong> of dense <strong>and</strong> n<strong>on</strong>dense<br />

area.<br />

An alternative explanati<strong>on</strong> for the negative associati<strong>on</strong> observed between n<strong>on</strong>-dense area <strong>and</strong> breast<br />

cancer risk could be the intrinsic collinearity between dense <strong>and</strong> n<strong>on</strong>-dense area, whose sum is<br />

c<strong>on</strong>strained to be equal to the total breast area. The presence of two negatively correlated predictors in the<br />

same model could lead to biased estimates of their associati<strong>on</strong>s, resulting in a spurious negative<br />

associati<strong>on</strong> of n<strong>on</strong>-dense area <strong>and</strong> an under-estimate of the associati<strong>on</strong> of dense-area.<br />

With our data we have not been able to choose whether a model based <strong>on</strong> PMD or <strong>on</strong> a linear<br />

combinati<strong>on</strong> of dense <strong>and</strong> n<strong>on</strong>-dense areas provides a better fit. We have shown that, for n<strong>on</strong>-dense area<br />

close to the populati<strong>on</strong> median, the two models are similar in their predicti<strong>on</strong>s, whereas for more extreme<br />

values of n<strong>on</strong>-dense area they provide quite different predicti<strong>on</strong>s. A validati<strong>on</strong> study c<strong>on</strong>ducted <strong>on</strong> a<br />

sample of women with more extreme values of n<strong>on</strong>-dense area might help to determine which of these<br />

two models provide a better fit to the data.<br />

37


P7<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

PREDICTION OF MAMMOGRAPHIC DENSITY USING REPRODUCTIVE AND<br />

LIFESTYLE CHARACTERISTICS<br />

Mariëtte Lokate 1, Linda M. Peelen 1, Marije F. Bakker 1, Petra H.M. Peeters 1, Carla H. van Gils 1<br />

1 Julius Center for Health Sciences <strong>and</strong> Primary Care, University Medical Center, PO Box 85500, 3508<br />

GA Utrecht, the Netherl<strong>and</strong>s<br />

Introducti<strong>on</strong>: <strong>Breast</strong> density is <strong>on</strong>e of the str<strong>on</strong>gest breast cancer risk factors, <strong>and</strong> is most comm<strong>on</strong>ly<br />

assessed using mammography. Since most mammographic breast cancer screening programs start at the<br />

age of 50 years, knowledge of this risk factor becomes available relatively late in life. Earlier informati<strong>on</strong><br />

<strong>on</strong> this risk factor could help in planning preventive or early detecti<strong>on</strong> strategies in these high-risk<br />

women. In this study, we investigated if we can predict mammographic density using characteristics that<br />

are easy to obtain earlier in a woman’s life.<br />

Methods: Within the EPIC-NL cohort, we assessed mammographic density of 871 women aged 50 or 51<br />

years at mammographic examinati<strong>on</strong>. We c<strong>on</strong>structed predicti<strong>on</strong> models with mammographic density as<br />

c<strong>on</strong>tinuous, categorical (


P8<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MAMMOGRAPHIC DENSITY AND RISK OF BREAST CANCER BY AGE AND<br />

TUMOR CHARACTERISTICS<br />

Kimberly A. Bertr<strong>and</strong> 1,2 , Rulla M. Tamimi 1,2 , Christopher G. Scott 3 , Matthew R. Jensen 3 , V. Shane<br />

Pankratz 3 , Daniel Visscher 4 , Aar<strong>on</strong> Norman 5 , Fergus Couch 5,6, , John Shepherd 7 , Bo Fan 7 , Yunn-Yi Chen 8 ,<br />

Lin Ma 9 , Andrew H. Beck, 10 Walter C. Willett, 1,2,11 Steven R. Cummings, 12 Karla Kerlikowske 13 , Celine<br />

M. Vach<strong>on</strong> 5<br />

ABSTRACTS<br />

1 Channing Divisi<strong>on</strong> of Network Medicine, Department of Medicine, Brigham <strong>and</strong> Women’s Hospital <strong>and</strong><br />

Harvard Medical School, Bost<strong>on</strong>, MA; 2 Department of Epidemiology, Harvard School of Public Health,<br />

Bost<strong>on</strong>, MA; 3 Divisi<strong>on</strong> of Biomedical Statistics <strong>and</strong> Informatics, Mayo Clinic College of Medicine,<br />

Rochester, MN; 4 Department of Anatomic Pathology, Mayo Clinic College of Medicine, Rochester, MN;<br />

5 Department of Health Sciences Research, Divisi<strong>on</strong> of Epidemiology, Mayo Clinic College of Medicine,<br />

Rochester, MN; 6 Department of Experimental Pathology <strong>and</strong> Laboratory Medicine, Mayo Clinic College<br />

of Medicine, Rochester, MN; 7 Department of Radiology, University of California, San Francisco, CA;<br />

8 Department of Pathology, University of California, San Francisco, CA; 9 Department of Medicine,<br />

University of California, San Francisco, CA; 10 Department of Pathology, Beth Israel Deac<strong>on</strong>ess Medical<br />

Center <strong>and</strong> Harvard Medical School, Bost<strong>on</strong>, MA; 11 Department of Nutriti<strong>on</strong>, Harvard School of Public<br />

Health, Bost<strong>on</strong>, MA; 12 San Francisco Coordinating Center, California Pacific Medical Center Research<br />

Institute; 13 Departments of Epidemiology <strong>and</strong> Biostatistics <strong>and</strong> General Internal Medicine Secti<strong>on</strong>,<br />

Department of Veterans Affairs, University of California, San Francisco, CA<br />

Background: Evidence <strong>on</strong> the associati<strong>on</strong> between mammographic density (MD) <strong>and</strong> breast cancer<br />

according to age <strong>and</strong> morphologic/histologic characteristics of tumors is mixed. Underst<strong>and</strong>ing whether<br />

MD is associated with all breast tumor subtypes <strong>and</strong> whether the strength of associati<strong>on</strong> varies by age is<br />

important prior to utilizing MD in risk models.<br />

Methods: Data were pooled from six cohort or case-c<strong>on</strong>trol studies including 3414 women with breast<br />

cancer <strong>and</strong> 7199 without who underwent screening mammography. Percent MD was assessed from<br />

digitized film-screen mammograms using a computer-assisted threshold technique. We used polytomous<br />

logistic regressi<strong>on</strong> to calculate the odds of breast cancer according to tumor type, histopathological<br />

characteristics <strong>and</strong> receptor (ER, PR, HER2) status by age (51%) vs. average density (11-25%). Women ages 2.1 cm) vs. smaller (0.1-1 cm <strong>and</strong> 1.1-2 cm) tumor size<br />

<strong>and</strong> positive vs. negative lymph node status (P trend


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Table. Pooled associati<strong>on</strong>s of categorical percent MD with breast cancer by ER status <strong>and</strong><br />

age.<br />

Ages


P9<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MAMMOGRAPHIC PIXEL VARIANCE AND RISK OF BREAST CANCER IN A<br />

NESTED CASE-CONTROL STUDY<br />

Adam R. Brentnall 1 , Jane Warwick 2 , Elizabeth Pinney 1 , Jennifer St<strong>on</strong>e 3 , Ruth M.L. Warren 4 , Anth<strong>on</strong>y<br />

Howell 5 , Jack Cuzick 1<br />

1 Centre for Cancer Preventi<strong>on</strong>, Wolfs<strong>on</strong> Institute of Preventive Medicine, Queen Mary University of<br />

L<strong>on</strong>d<strong>on</strong>, L<strong>on</strong>d<strong>on</strong>, UK; 2 Imperial Clinical Trials Unit, School of Public Health, Faculty of Medicine,<br />

Imperial College L<strong>on</strong>d<strong>on</strong>, L<strong>on</strong>d<strong>on</strong>, UK; 3 Centre for Molecular, Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytic<br />

(MEGA) Epidemiology, The University of Melbourne, Melbourne, Australia; 4 Cambridge <strong>Breast</strong> Unit,<br />

Addenbrooke's Hospital, Cambridge, UK; 5 Genesis <strong>Breast</strong> Cancer Preventi<strong>on</strong> Centre, University<br />

Hospital of South Manchester, Manchester, UK<br />

A nested case-c<strong>on</strong>trol study within the <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <strong>Breast</strong> Cancer Interventi<strong>on</strong> Study (IBIS-I) was used<br />

to assess a recent proposal from Heine et al (2012) to measure breast density automatically as the st<strong>and</strong>ard<br />

deviati<strong>on</strong> of pixel intensity within the breast. The performance of our implementati<strong>on</strong> of the method was<br />

compared with two percent density measures.<br />

ABSTRACTS<br />

Women were enrolled to the placebo arm, <strong>and</strong> had around twice the populati<strong>on</strong> risk of developing breast<br />

cancer. Case subjects were 49 women diagnosed with breast cancer at or after their first follow-up<br />

mammogram, <strong>and</strong> c<strong>on</strong>trol subjects were 468 women from the same eight UK centres without breast<br />

cancer. The original mammograms were taken using a variety of different machines <strong>and</strong> scanned at an 8-<br />

bit greyscale resoluti<strong>on</strong>, but no further informati<strong>on</strong> is known about them, so c<strong>on</strong>trols were also matched<br />

within centre by year of mammogram. The mean time between baseline mammogram <strong>and</strong> diagnosis was<br />

6.1 years (inter-quartile range, IQR, 4.0 - 8.6). Mean age at baseline was 50.6 years (IQR 46.4 - 54.5),<br />

mean body mass index (BMI) was 26.7 kg/m2 (IQR 23.4 - 29.0) <strong>and</strong> 52% were pre-menopausal.<br />

Mammographic density was assessed at baseline by three methods: visually <strong>on</strong> an x-ray film illuminator<br />

by a single reader (RW) in 5% intervals, semi-automatically by a single reader (JS) as a c<strong>on</strong>tinuous<br />

percent measure using computer software (cumulus 3.0), <strong>and</strong> a fully automatic estimate of the st<strong>and</strong>ard<br />

deviati<strong>on</strong> of pixels within the breast (V measure). The V measure was calculated by an algorithm to (1)<br />

remove the background <strong>and</strong> pectoral muscle, (2) trim 25% of pixels from the breast edge, <strong>and</strong> (3)<br />

calculate the sample st<strong>and</strong>ard deviati<strong>on</strong> of the remaining pixels. It used MLO views, taking the mean<br />

when both views were available (530 women).<br />

Linear regressi<strong>on</strong> was used to adjust percentage density for age <strong>and</strong> BMI. The different density measures<br />

were st<strong>and</strong>ardised by dividing by their sample st<strong>and</strong>ard deviati<strong>on</strong> (SD) to facilitate comparis<strong>on</strong> between<br />

measures. Pears<strong>on</strong>'s correlati<strong>on</strong> coefficient quantified associati<strong>on</strong> between the density measures. Odds<br />

ratios were obtained by c<strong>on</strong>diti<strong>on</strong>al logistic regressi<strong>on</strong>.<br />

Pears<strong>on</strong> correlati<strong>on</strong> of the V measure between left <strong>and</strong> right MLO views was 0.88 (95% CI 0.86 - 0.90).<br />

The V measure was moderately correlated with percentage density when assessed visually (Pears<strong>on</strong><br />

correlati<strong>on</strong> 0.25, 95% CI 0.17 - 0.32) <strong>and</strong> semi-automatically (Pears<strong>on</strong> correlati<strong>on</strong> 0.34, 95% CI 0.27 -<br />

0.42), but the percentage measures were more str<strong>on</strong>gly correlated with each other (Pears<strong>on</strong> correlati<strong>on</strong><br />

0.85, 95% CI 0.83 - 0.88). The V measure was at least as predictive (st<strong>and</strong>ardised measure odds ratio<br />

(SOR) 1.63 per SD, 95% CI 1.13 - 2.36) as visual percent density (SOR 1.39, 95% CI 1.00 - 1.92) <strong>and</strong><br />

cumulus (SOR 1.28, 95% CI 0.93 - 1.77). This was also found when visual percent density was adjusted<br />

for age <strong>and</strong> BMI (SOR 1.53, 95% CI 1.15 - 2.04).<br />

C<strong>on</strong>clusi<strong>on</strong>: The automated V measure appeared to be as predictive of future breast cancer as visuallyassessed<br />

<strong>and</strong> semi-automatic percentage density in this case-c<strong>on</strong>trol study.<br />

41


P10<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

VALIDATION OF A FULLY AUTOMATED, HIGH THROUGHPUT MAMMOGRAPHIC<br />

DENSITY MEASUREMENT TOOL FOR USE WITH PROCESSED DIGITAL<br />

MAMMOGRAMS<br />

AM Couwenberg, CH van Gils, J Li, R Pijnappel, RK Charaghv<strong>and</strong>i, M Hartman, HM Verkooijen<br />

Introducti<strong>on</strong>: Recently, a public domain image-processing software (ImageJ) tool was developed to<br />

mimic Cumulus mammographic density readings. This fully-automated method has been validated for<br />

digitized film screen mammograms (JM Li et al. 2012). i Here we validate the Image J density reading tool<br />

in processed full field digital mammograms (FFDM), which are currently the most comm<strong>on</strong>ly available<br />

image types. We also correlate Image J readings with the clinically widely-used BIRADS density<br />

classificati<strong>on</strong>.<br />

Methods: This study includes Dutch women who underwent a FFDM mammography between 2001 <strong>and</strong><br />

2011 at the University Medical Center Utrecht, the Netherl<strong>and</strong>s. Only processed images were available.<br />

We composed a training set for model building (n=170 women, 331 images MLO <strong>and</strong> CC view of the<br />

c<strong>on</strong>tralateral breast) <strong>and</strong> a test set for model assessment (530 women, 4,429 images MLO <strong>and</strong> CC view of<br />

the c<strong>on</strong>tralateral breast). The training set was read with Cumulus by <strong>on</strong>e trained reader <strong>and</strong> used to build<br />

the automated Image J mammographic density model. This model was used for the density assessment in<br />

the test set. These automated density readings were compared with BIRADS density readings by a<br />

dedicated breast radiologist. We used Pears<strong>on</strong> product-moment correlati<strong>on</strong> coefficient (r) to compare<br />

Cumulus measurements with the automated measurements in the training set. Correlati<strong>on</strong> <strong>and</strong> of the<br />

automated method with BIRADS density readings in the test set was estimated using Spearman<br />

correlati<strong>on</strong> coefficient. As a further validati<strong>on</strong>, we investigated the associati<strong>on</strong> between breast cancer risk<br />

factors <strong>and</strong> the automated density measurements for the patients in our test set, using generalized linear<br />

models.<br />

Results: We observed a high correlati<strong>on</strong> between the automated method <strong>and</strong> Cumulus (r=0.90 95% Cl:<br />

0.86 – 0.93). We also found a high correlati<strong>on</strong> between the automated measure <strong>and</strong> the BIRADS density<br />

classificati<strong>on</strong> in the test set (spearman r= 0.862). Women with a higher density as measured with the<br />

automated method, were significantly younger, more often premenopausal, had a lower number of<br />

children, <strong>and</strong> more often a benign breast lesi<strong>on</strong>s or a family history of breast cancer.<br />

C<strong>on</strong>clusi<strong>on</strong>: We dem<strong>on</strong>strated that the high-throughput automated density assessment tool, based <strong>on</strong> the<br />

analysis of the established method of Cumulus <strong>and</strong> previously validated <strong>on</strong> digitized film-screen<br />

mammograms, can be used <strong>on</strong> processed digital mammograms. Its results also str<strong>on</strong>gly correlate with the<br />

clinically widely-used BIRADS density readings <strong>and</strong> with established breast cancer risk factors.<br />

ABSTRACTS<br />

•<br />

1 Li et al. High-throughput mammographic-density measurement: a tool for risk predicti<strong>on</strong> of breast cancer.<br />

<strong>Breast</strong> Cancer Research 2012, 14:R114.<br />

42


P11<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

RESULTS OF SUPPLEMENTAL ULTRASOUND SCREENING IN WOMEN WITH AND<br />

WITHOUT DENSE BREAST TISSUE<br />

Judy C Dean MD<br />

Recent awareness of the increased risk of breast cancer, <strong>and</strong> higher rate of interval cancer that escapes<br />

detecti<strong>on</strong> with mammography have led to recommendati<strong>on</strong>s for supplemental screening with ultrasound,<br />

yet little is known about the diagnostic yield of these examinati<strong>on</strong>s.<br />

Since November, 2005 I have performed almost 10,000 automated whole breast ultrasound studies with<br />

digital mammograms, primarily in women with either increased breast cancer risk or increased breast<br />

density. Stratificati<strong>on</strong> by breast density (as analyzed visually using the Bi-Rads classificati<strong>on</strong> system)<br />

shows significantly more cancers detected in women with increased breast density.<br />

Cancer Detecti<strong>on</strong>s by Imaging Method:<br />

ABSTRACTS<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Mam <strong>on</strong>ly Both ABUS<br />

<strong>on</strong>ly<br />

Either<br />

The table above shows that half of all cancers detected were identified with automated breast ultrasound <strong>on</strong>ly, <strong>and</strong><br />

not imaged by mammography. Only <strong>on</strong>e of the additi<strong>on</strong>al cancers detected by ultrasound was DCIS, <strong>and</strong> all others<br />

were invasive cancers.<br />

43


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Cancers detected per 1000 women screened, by Bi-Rads Density Score:<br />

20<br />

15<br />

10<br />

5<br />

0<br />

In women with Bi-Rads density 4, per 1000 women screened 17 cancers were detected. For density 3, 10.5 per<br />

1000, <strong>and</strong> for density 1 <strong>and</strong> 2, 4 per 1000.<br />

Cancers detected by ultrasound <strong>on</strong>ly per 1000 women screened by Bi-Rads density:<br />

18<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

1 & 2 3 4<br />

1 & 2 3 4<br />

ABSTRACTS<br />

Additi<strong>on</strong>al cancers detected with ultrasound <strong>on</strong>ly <strong>and</strong> not visualized with mammography were approximately 12 per<br />

1000 for density 4, almost 4 per 1000 for density 3, <strong>and</strong> <strong>on</strong>ly 1 per 1000 for density 1 <strong>and</strong> 2.<br />

This data c<strong>on</strong>firms that ultrasound will detect significantly more cancers than mammography al<strong>on</strong>e in women with<br />

dense breast tissue. Since nearly all the additi<strong>on</strong>al cancers detected are invasive tumors, these are likely to be more<br />

clinically relevant than cancers detected with mammography al<strong>on</strong>e, which are 30 to 50% n<strong>on</strong>-invasive.<br />

44


P12<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

COMPARISON OF CUMULUS MAMMOGRAPHIC DENSITY READINGS BETWEEN<br />

FUJIFILM COMPUTED RADIOGRAPHY DIGITAL MAMMOGRAPHIC IMAGES IN<br />

RAW AND DISPLAY FORMATS<br />

Caroline Dickens, Valerie McCormack<br />

Secti<strong>on</strong> of Envir<strong>on</strong>ment <strong>and</strong> Radiati<strong>on</strong>, <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer, Ly<strong>on</strong> 69008,<br />

FRANCE.<br />

Background: The processing of digital mammograms from their raw to processed or display formats in<br />

order to improve tumor detecti<strong>on</strong> involves manipulati<strong>on</strong> of the image c<strong>on</strong>trast by means of proprietary<br />

image processing algorithms, which differ between vendors. It has been recommended that raw images<br />

be used for estimati<strong>on</strong> of mammographic density as they maintain image c<strong>on</strong>trast, but in some instances<br />

they are not available. Recent studies have shown a good correlati<strong>on</strong> in percent breast density (PD)<br />

between raw <strong>and</strong> processed images acquired using the General Electric Senographe FFDM<br />

mammography units. We wished to see if similar comparability exists between PD readings performed<br />

<strong>on</strong> raw <strong>and</strong> processed images obtained using the Fujifilm Computed Radiography (CR) imaging system.<br />

ABSTRACTS<br />

Methods: Bilateral raw <strong>and</strong> processed craniocaudal (CC) Fujifilm CR images were obtained for 100<br />

women. Images were processed using the Fujifilm Image Intelligence algorithm. 400 images (100<br />

women x 2 breasts x 2 formats) were an<strong>on</strong>ymized <strong>and</strong> r<strong>and</strong>omly sorted into 5 batches of 80 prior to<br />

reading. PD was estimated by a trained observer (VM) using an interactive-threshold method (Cumulus3).<br />

Comparis<strong>on</strong>s of PD <strong>and</strong> absolute dense area between the raw <strong>and</strong> processed images were c<strong>on</strong>ducted using<br />

Spearman’s rank correlati<strong>on</strong> coefficient (r), t-tests for mean differences <strong>and</strong> kappa statistics for the<br />

agreement of categories. Differences according to level of density were also examined.<br />

Results: Minimum-maximum ranges (0-55% for raw <strong>and</strong> 0-58% for processed images) were similar in<br />

the two image output formats, but processed images had a larger st<strong>and</strong>ard deviati<strong>on</strong> (14.8 vs 12.8). Mean<br />

PD was 22.8% (95% c<strong>on</strong>fidence interval (CI): 21.0, 24.6) for raw images <strong>and</strong> 25.9 (23.8, 27.9) for<br />

processed images. Processed images had a mean PD 3.1 (2.0, 4.1) absolute percentage points higher than<br />

in the corresp<strong>on</strong>ding raw image. For raw images with PD in the range


P13<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

RISK ASSOCIATION OF SUBREGIONAL MEASUREMENTS OF BREAST<br />

DENSITY<br />

Duewer, Fred 1 , Fan, Bo 1 , Karla, Kerlikowske 2,3 , Ma, Lin 2,3 , Malkov, Serghei 1 , <strong>and</strong> Shepherd, John 1<br />

University of California, San Francisco, Department of Radiology 1 , Department of Epidemiology <strong>and</strong><br />

Biostatistics 2 , <strong>and</strong> Department of Medicine 3<br />

Although breast density is, excepting age, the str<strong>on</strong>gest known risk factor for breast cancer, little is known<br />

about the spatial variati<strong>on</strong> in breast density. Our hypothesis is that the associati<strong>on</strong> of local fibrogl<strong>and</strong>ular<br />

volume with breast cancer in a given subregi<strong>on</strong> of the breast varies based <strong>on</strong> the positi<strong>on</strong> of that subregi<strong>on</strong><br />

relative to the nipple <strong>and</strong> breast outline.<br />

We used single x-ray absorptiometry to obtain pixel-by-pixel maps of breast density <strong>and</strong> spatially<br />

normalized each breast density map using estimated midplane positi<strong>on</strong> <strong>and</strong> the breast outline <strong>on</strong> casec<strong>on</strong>trol<br />

data set c<strong>on</strong>sisting of 278 cases <strong>and</strong> 834 c<strong>on</strong>trols matched by age <strong>and</strong> body mass index. We<br />

measured the absolute fibrogl<strong>and</strong>ular volume in 100 subregi<strong>on</strong>s for each image. We compared average<br />

fibrogl<strong>and</strong>ular volume in subregi<strong>on</strong>s <strong>and</strong> the absolute left-right difference in each subregi<strong>on</strong> (asymmetry)<br />

depending <strong>on</strong> case-c<strong>on</strong>trol status. We found that average fibrogl<strong>and</strong>ular volume was higher in cases than<br />

c<strong>on</strong>trols for all subregi<strong>on</strong>s <strong>and</strong> that left-right fibrogl<strong>and</strong>ular volume asymmetry was also higher for cases<br />

than for c<strong>on</strong>trols.<br />

We tested for regi<strong>on</strong>al differences in breast cancer risk associati<strong>on</strong> between cases <strong>and</strong> c<strong>on</strong>trols <strong>and</strong><br />

according to age <strong>and</strong> body mass index by measuring the P-value of the t-test between cases <strong>and</strong> c<strong>on</strong>trols<br />

for each subregi<strong>on</strong>. We found that subregi<strong>on</strong>s closer to the nipple differed more significantly between<br />

cases <strong>and</strong> c<strong>on</strong>trols. In additi<strong>on</strong>, we used principal comp<strong>on</strong>ent analysis <strong>on</strong> the average fibrogl<strong>and</strong>ular<br />

volume in the 100 subregi<strong>on</strong>s <strong>and</strong> identified three principal comp<strong>on</strong>ents that c<strong>on</strong>tributed significantly to<br />

breast cancer risk (P


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P14 THE ASSOCIATION BETWEEN VITAMIN D INTAKE, SEASON AND<br />

MAMMOGRAPHIC DENSITY IN NORWEGIAN POSTMENOPAUSAL WOMEN<br />

Merete Ellingjord-Dale¹, Isabel dos Santos Silva ² , Tom Grotmol³, Solveig Hofvind³, Lene Frost<br />

Andersen¹, Samera Azeem Qureshi¹, Marianne Skov Markussen¹, Elisabeth Couto⁵, Giske Ursin 1,3,4<br />

1 University of Oslo, Norway, 2 L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical Medicine, UK , 3 Cancer Registry of<br />

Norway, Norway, 4 University of Southern California, USA, ⁵Norwegian Knowledge Centre for the Health<br />

Services, Norway<br />

ABSTRACTS<br />

Background: It is unclear whether vitamin D intake is associated with breast cancer risk. There is some,<br />

albeit inc<strong>on</strong>sistent, evidence that mammographic density (MD) varies by seas<strong>on</strong> of the year or by vitamin<br />

D intake. We evaluated the associati<strong>on</strong> of MD with seas<strong>on</strong>ality <strong>and</strong> vitamin D intake am<strong>on</strong>g a subset of<br />

women who participated in the Norwegian <strong>Breast</strong> Cancer Screening Program (NBCSP) in 2004 or in<br />

2008.<br />

Methods: We c<strong>on</strong>ducted a cross-secti<strong>on</strong>al study to examine the associati<strong>on</strong> of MD with vitamin D intake<br />

<strong>and</strong> seas<strong>on</strong>ality am<strong>on</strong>g 2662 postmenopausal women aged 50-69 years. Vitamin D intake (estimated with<br />

<strong>and</strong> without taking into account supplement use) was ascertained through a self-administered<br />

questi<strong>on</strong>naire. Density <strong>on</strong> digitized analogue films was measured using a computer assisted method<br />

(Madena, University of Southern California). Linear regressi<strong>on</strong> methods were used to determine the<br />

associati<strong>on</strong> of MD with vitamin D intake <strong>and</strong> the seas<strong>on</strong> in which the mammogram was obtained.<br />

Specifically we estimated least square means of MD across categories of the exposure variables, while<br />

adjusting for potential c<strong>on</strong>founders.<br />

Results: There was no associati<strong>on</strong> between intake of vitamin D <strong>and</strong> MD, across quartiles of vitamin D<br />

intake (0-5, 6-8, 9-12 <strong>and</strong> 13-49 mcg per day, estimated with supplements). Mean MD was 18.5%<br />

(SD=15.3%) in women with the lowest, <strong>and</strong> 18.9% (SD=15.6%) in women with the highest intake of<br />

vitamin D (p for trend=0.73). There was no associati<strong>on</strong> between vitamin D intake <strong>and</strong> MD in women who<br />

had their mammogram during January-May, i.e. at the time when vitamin D levels are presumed to be at<br />

the lowest (p for trend= 0.10).<br />

Similarly, there was no statistical significant associati<strong>on</strong> between m<strong>on</strong>th of mammography <strong>and</strong> MD in<br />

either the 2004 or the 2008 data.<br />

C<strong>on</strong>clusi<strong>on</strong>: Our cross-secti<strong>on</strong>al study found no evidence of an associati<strong>on</strong> of MD with vitamin D intake<br />

or m<strong>on</strong>th/seas<strong>on</strong> of mammography.<br />

47


P15<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

THE ASSOCIATION BETWEEN GEOGRAPHIC STATUS AND MAMMOGRAPHIC<br />

DENSITY<br />

Marleen J. Emaus 1 , Marije F. Bakker 1 , Petra H.M. Peeters 1, 2 , Carla H. van Gils 1<br />

1 Julius Center for Health Sciences <strong>and</strong> Primary Care, University Medical Center, PO Box 85500, 3508<br />

GA Utrecht, the Netherl<strong>and</strong>s; 2 Department of Epidemiology, Public Health <strong>and</strong> Primary Care, Faculty of<br />

Medicine, Imperial College, Norfolk Place, L<strong>on</strong>d<strong>on</strong> W2 1PG, UK<br />

Introducti<strong>on</strong>: It is frequently observed that mammographic density varies depending up<strong>on</strong> geographic<br />

area. It is unclear whether these differences are related to the degree of urbanizati<strong>on</strong> (as proxy variable for<br />

envir<strong>on</strong>mental factors such as exposure to traffic emissi<strong>on</strong>s) or to differences in social ec<strong>on</strong>omic status<br />

(SES) (as proxy variable for lifestyle <strong>and</strong> reproductive characteristics). In the present study we try to<br />

unravel this issue within the Dutch screening populati<strong>on</strong>.<br />

Methods: The study was performed within the Prospect-EPIC cohort. Prospect-EPIC participants were<br />

recruited through the breast cancer screening program in Utrecht <strong>and</strong> vicinity between 1993 <strong>and</strong> 1997.<br />

Urbanizati<strong>on</strong> was categorized according to the number of addresses per kilometer squared using the 5-<br />

group classificati<strong>on</strong> established by Statistics Netherl<strong>and</strong>s. SES was measured at neighborhood level<br />

(Statusscore) using data from Statistics Netherl<strong>and</strong>s as well as at individual level (educati<strong>on</strong>al level of the<br />

participant). General linear models were used to study the associati<strong>on</strong> between the degree of urbanizati<strong>on</strong><br />

<strong>and</strong> SES <strong>on</strong> the <strong>on</strong>e h<strong>and</strong> <strong>and</strong> mammographic density <strong>on</strong> the other h<strong>and</strong> (i.e. percent density <strong>and</strong> dense<br />

area). The models were adjusted for age <strong>and</strong> BMI. Analyses with additi<strong>on</strong>al adjustment for lifestyle <strong>and</strong><br />

horm<strong>on</strong>al risk factors for breast density will be presented at the c<strong>on</strong>ference.<br />

Results: We included 2,805 women with a mean age of 57.3 years (SD: 5.9 years). Women living in<br />

urban areas showed higher percent density than women in rural areas (adjusted mean percent density urban =<br />

21.9%, 95%CI 21.1-22.8 versus adjusted mean percent density rural = 15.1, 95% CI 12.8-17.7, p-<br />

trend


P16<br />

INTER-SCAN REPRODUCIBILITY OF MAMMOGRAPHIC DENSITY<br />

MEASUREMENTS<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Mariëtte Lokate 1 , Marleen J. Emaus 1 , Michiel Kallenberg 2,3 , Nico Karssemeijer 2 , Wouter B. Veldhuis 4 ,<br />

Petra H.M. Peeters 1 , Carla H. van Gils 1<br />

1 Julius Center for Health Sciences <strong>and</strong> Primary Care, University Medical Center, PO Box 85500, 3508<br />

GA Utrecht, the Netherl<strong>and</strong>s; 2 Department of Radiology, Radboud University Nijmegen Medical Center,<br />

Geert Grooteplein Zuid 18, 6525 GA Nijmegen, the Netherl<strong>and</strong>s; 3 Matakina Technology Ltd, PO Box<br />

24404, Manners Street Central, Wellingt<strong>on</strong>, 6142, New Zeal<strong>and</strong>; 4 Department of Radiology, University<br />

Medical Center, PO Box 85500, 3508 GA Utrecht, the Netherl<strong>and</strong>s<br />

Introducti<strong>on</strong>: Changes in mammographic density are frequently used as an intermediate marker to<br />

evaluate the effect of interventi<strong>on</strong>s for reducing breast cancer risk. We investigated the robustness of<br />

measurement of changes in mammographic density. We assessed c<strong>on</strong>sistency of mammographic density<br />

measurements between pairs of mammograms. They were made within a short time interval, in which no<br />

actual change in mammographic density can be expected.<br />

ABSTRACTS<br />

Methods: We included 170 women who underwent two mammographic examinati<strong>on</strong>s within a median<br />

follow-up period of 12 days (maximum 6 m<strong>on</strong>ths) : 106 had two Full-Field Digital Mammograms<br />

(FFDM), 12 had two Film-Screen Mammograms (FSM) <strong>and</strong> 52 had <strong>on</strong>e FSM <strong>and</strong> <strong>on</strong>e FFDM.<br />

Mammographic density was assessed by a single reader using a computer assisted threshold method<br />

(Cumulus). The correlati<strong>on</strong> between percent mammographic density measurements was estimated using<br />

intraclass correlati<strong>on</strong> coefficients <strong>and</strong> we also calculated mean absolute difference in density measures<br />

between both mammograms. Linear regressi<strong>on</strong> was performed to adjust for differences in acquisiti<strong>on</strong><br />

parameters between mammograms.<br />

Results: The intraclass correlati<strong>on</strong> between percent density measurements <strong>on</strong> two FFDMs was 0.93<br />

(95%CI 0.91–0.95), between two FSMs 0.94 (95% CI 0.84-0.98), <strong>and</strong> between FFDM <strong>and</strong> FSM 0.72<br />

(0.61-0.80). The mean absolute difference in percent density between two FFDMs of the same woman<br />

was 4.3% (95% CI 3.8% to 4.9%) <strong>and</strong> between two FSMs 6.5% (95% CI 3.4% to 9.6%). On average,<br />

FSM showed a higher percent density compared to FFDM in the same women (difference 12.9% (95% CI<br />

11% to 14.8%). Adjustment for differences in exposure, peak kilo voltage, compressi<strong>on</strong> force <strong>and</strong> breast<br />

thickness between mammograms did not alter the results.<br />

C<strong>on</strong>clusi<strong>on</strong>: When studying changes in mammographic density, it is important to compare mammograms<br />

of the same type (i.e. either all FSM or all FFDM). Although intraclass correlati<strong>on</strong> between density<br />

measured <strong>on</strong> mammograms of the same type (either FSM or FFDM) is high, the absolute differences in<br />

percent density are of a magnitude that may influence detectability of real changes in mammographic<br />

density.<br />

49


P17<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MULTIPLE METABOLIC RISK FACTORS AND MAMMOGRAPHIC BREAST<br />

DENSITY IN THE NEW YORK CITY MULTIETHNIC BREAST CANCER PROJECT<br />

Parisa Tehranifar, 1,2,3 Diane Reynolds, 4 Xiaozhou Fan, 1 Bernadette Boden-Albala, 5 Natalie Engmann, 1<br />

Julie D. Flom, 1 Mary Beth Terry 1,2<br />

1 Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY ;<br />

2 Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY ;<br />

3 The Center for the Study of Social Inequalities <strong>and</strong> Health, Columbia University Mailman School of<br />

Public Health, New York,, NY; 4 School of Nursing, L<strong>on</strong>g Isl<strong>and</strong> University, Brooklyn Campus, Brooklyn,<br />

NY; 5 Divisi<strong>on</strong> of Social Epidemiology, Department of Health Evidence <strong>and</strong> Policy, Department of<br />

Neurology, ICAHN School of Medicine at Mount Sinai, New York, NY<br />

Acknowledgement: We would like to thank the study participants for c<strong>on</strong>tributing data to this analysis <strong>and</strong><br />

the following research staff for assisting with data collecti<strong>on</strong> <strong>and</strong> recruitment activities: Diane Levy,<br />

Wendy Lewis, Gladys Rivera, Zoe Qu<strong>and</strong>t, Joy White, Jessica Cabildo <strong>and</strong> Renata Khanis. This research<br />

was funded by grants from the Nati<strong>on</strong>al Cancer Institute (grant numbers: U54 CA101598; K07<br />

CA151777), Susan B. Komen Foundati<strong>on</strong> Career Catalyst award (grant number KG110331) <strong>and</strong> the<br />

Nati<strong>on</strong>al Institute of Envir<strong>on</strong>mental Health Sciences Center Support (grant number ES009089).<br />

Background: Metabolic abnormalities have been individually associated with breast cancer risk. More<br />

recently, studies have also shown modest positive associati<strong>on</strong>s between the presence of multiple<br />

metabolic c<strong>on</strong>diti<strong>on</strong>s, as frequently defined by the metabolic syndrome (MetS), <strong>and</strong> breast cancer risk in<br />

postmenopausal women. Few studies have examined whether the presence of multiple metabolic risk<br />

factors are similarly associated with breast density, an intermediate marker of breast cancer risk. We<br />

examined whether obesity, diabetes, hypertensi<strong>on</strong>, <strong>and</strong> elevated cholesterol, individually <strong>and</strong> in<br />

combinati<strong>on</strong>, are associated with breast density.<br />

Methods: We digitized film mammograms <strong>and</strong> measured percent density <strong>and</strong> dense area using a<br />

computer-assisted method in a sample of 191 women, recruited from a single radiology facility during<br />

their screening mammography appointment (age range=40-61 years; 42% African American, 22% African<br />

Caribbean, 22% White, 11% Hispanic, 3% other ethnicities). We used in-pers<strong>on</strong> computer assisted<br />

interviews to collect detailed epidemiologic data at the time of recruitment. We used linear regressi<strong>on</strong><br />

models to examine the associati<strong>on</strong>s of each metabolic c<strong>on</strong>diti<strong>on</strong> <strong>and</strong> the number of co-occurring<br />

metabolic c<strong>on</strong>diti<strong>on</strong>s (0, 1, 2, <strong>and</strong> 3 or 4 c<strong>on</strong>diti<strong>on</strong>s) with percent density <strong>and</strong> dense breast area.<br />

ABSTRACTS<br />

Results: In age adjusted models of individual metabolic c<strong>on</strong>diti<strong>on</strong>s, hypertensi<strong>on</strong> <strong>and</strong> obese body mass<br />

index (≥30 kg/m 2 ) were inversely associated with percent density, <strong>and</strong> high blood cholesterol was<br />

inversely associated with dense area. The presence of multiple metabolic c<strong>on</strong>diti<strong>on</strong>s was associated with<br />

lower percent density <strong>and</strong> dense area in multivariable models (2 c<strong>on</strong>diti<strong>on</strong>s <strong>and</strong> 3 or 4 c<strong>on</strong>diti<strong>on</strong>s vs. 0<br />

c<strong>on</strong>diti<strong>on</strong>s were associated with 9.4% (95% CI:-13.2, -5.5) <strong>and</strong> 10.7% (95% CI:-15.2, -6.2) reducti<strong>on</strong> in<br />

percent density <strong>and</strong> with 5.5 cm 2 (95% CI: -10.8, -0.3) <strong>and</strong> 8.4 cm 2 (95% CI: -14.5, -2.4) smaller dense<br />

area).<br />

C<strong>on</strong>clusi<strong>on</strong>s: The presence of multiple metabolic c<strong>on</strong>diti<strong>on</strong>s was associated with lower levels of relative<br />

<strong>and</strong> absolute measures of breast density in a predominantly racial minority sample of women. The<br />

reducti<strong>on</strong> in breast density associated with multiple metabolic c<strong>on</strong>diti<strong>on</strong>s was greater than the influence of<br />

any single c<strong>on</strong>diti<strong>on</strong> <strong>on</strong> breast density. Our results suggest that the positive associati<strong>on</strong> between<br />

metabolic abnormalities <strong>and</strong> breast cancer may not involve pathways related to breast density, <strong>and</strong> may be<br />

str<strong>on</strong>ger after accounting for the influence of metabolic c<strong>on</strong>diti<strong>on</strong>s <strong>on</strong> breast density.<br />

50


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P18 GENETIC TESTING FOR BREAST CANCER RISK ESTIMATION – A COST -<br />

EFFECTIVENESS ANALYSIS<br />

Henri Folse 1 (corresp<strong>on</strong>ding <strong>and</strong> presenting author), Tuan Dinh 1 , Richard Allman 2<br />

1 Archimedes, Inc., 2 Genetic Technologies, Ltd.<br />

Genetic testing based <strong>on</strong> seven single-nucleotide polymorphisms (7SNP) can improve individualized<br />

estimates of lifetime risk of breast cancer relative to the Gail risk test al<strong>on</strong>e, for the purpose of<br />

recommending MRI screening for women at high risk.<br />

ABSTRACTS<br />

An individual-based, c<strong>on</strong>tinuous-time simulati<strong>on</strong> model of breast cancer <strong>and</strong> health care processes was<br />

used to simulate women in a virtual trial comparing the use of the 7SNP test to the Gail risk test al<strong>on</strong>e to<br />

categorize patients as either low risk or high risk. Low risk patients received annual mammogram, while<br />

high risk patients received annual MRI. Cancer incidence was based <strong>on</strong> Surveillance, Epidemiology, <strong>and</strong><br />

End Results (SEER) data <strong>and</strong> validated to the Cancer Preventi<strong>on</strong> Study II (CPS-II) Nutriti<strong>on</strong> Cohort data<br />

set. Risk factors are drawn from the Nati<strong>on</strong>al Health <strong>and</strong> Nutriti<strong>on</strong> Examinati<strong>on</strong> Survey (NHANES-4) <strong>and</strong><br />

Prostate, Lung, Colorectal, <strong>and</strong> Ovarian Cancer Screening Trial (PLCO) data sets. Mammogram<br />

characteristics were derived from the <strong>Breast</strong> Cancer Surveillance C<strong>on</strong>sortium (BCSC) dataset.<br />

Other parameters were derived from published literature. The 7SNP test (vs Gail al<strong>on</strong>e) saved 0.00734<br />

quality-adjusted life-years (QALYs) per pers<strong>on</strong> at a cost of $1,971 per pers<strong>on</strong> ($268,386 per QALY).<br />

Limiting the 7SNP test to <strong>on</strong>ly those patients with a lifetime Gail risk of 16 – 28% resulted in a cost of<br />

$163,264 per QALY. These results were sensitive to the age at which the test is given, the discount rate,<br />

<strong>and</strong> the costs of the genetic test <strong>and</strong> MRI. The cost-effectiveness of using the 7SNP test for patients with<br />

intermediate Gail risk is similar to that of other recommended strategies, including annual MRI for<br />

patients with a lifetime risk greater than 20% or BRCA1/2 mutati<strong>on</strong>s.<br />

51


P19<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

ADIPOSITY AND MAMMOGRAPHIC BREAST DENSITY IN PREMENOPAUSAL<br />

CHILEAN WOMEN<br />

Maria Luisa Garmendia 1 , Ana Pereira 1 , John Shepherd 2 , Ralph Highnam 3 , Camila Corvalán 1 ,<br />

1 Institute of Nutriti<strong>on</strong> <strong>and</strong> Food Technology, University of Chile, Chile; 2 Department of Radiology <strong>and</strong><br />

Biomedical Imaging University of California at San Francisco, United States; 3 Mātakina Technology<br />

Limited, Wellingt<strong>on</strong>, New Zeal<strong>and</strong><br />

Background: <strong>Breast</strong> density is <strong>on</strong>e of the str<strong>on</strong>gest risk factors for breast cancer. In premenopausal<br />

women, obesity decreases the risk of breast cancer, but the relati<strong>on</strong>ship between breast dense tissue <strong>and</strong><br />

obesity remains unclear.<br />

Objectives: To assess the relati<strong>on</strong>ship between adiposity indicators <strong>and</strong> absolute breast dense volume<br />

(fibrogl<strong>and</strong>ular tissue volume) <strong>and</strong> to evaluate metabolic <strong>and</strong> horm<strong>on</strong>al factors as potential mediators in<br />

these relati<strong>on</strong>ships.<br />

Methods:<br />

Design: This is a cross-secti<strong>on</strong>al study of the mothers of the girls participating in the Growth <strong>and</strong> Obesity<br />

Chilean cohort Study (GOCS) participants. GOCS is a populati<strong>on</strong>-based ambispective cohort of 1196<br />

children born in 2002 in six low <strong>and</strong> middle-income counties from the South-area of Santiago, Chile. The<br />

study sample is representative of the populati<strong>on</strong> that receives care at the Nati<strong>on</strong>al Health Services System<br />

(70% of the entire populati<strong>on</strong>). In 2011-2012, 400 premenopausal mothers of the GOCS girls (mean age =<br />

36.9 years, SD= 6.4) were included in a l<strong>on</strong>gitudinal study of breast cancer risk factors (DERCAM<br />

cohort).<br />

Procedures: Weight, height, body mass index (BMI= weight (kg)/ height(m) 2 ), waist circumference<br />

(WC), <strong>and</strong> waist-to-height ratio (WHR) were measured by trained nutriti<strong>on</strong>ists. Percentage total fat was<br />

estimate by bioimpedance (Tanita). All women underwent digital mammography during the follicular<br />

phase of their menstrual cycle. Volumetric measures of breast density (absolute fibrogl<strong>and</strong>ular volume,<br />

adipose volume, <strong>and</strong> percentage fibrogl<strong>and</strong>ular volume) were determined using an automated method<br />

(Volpara®, Matakina Technology, Wellingt<strong>on</strong>, New Zeal<strong>and</strong>). Age, educati<strong>on</strong> <strong>and</strong> parity were selfreported<br />

by questi<strong>on</strong>naire. Fasting insulin, glycaemia, insulin growth factor-1 (IGF-1), estr<strong>on</strong>e, estradiol,<br />

progester<strong>on</strong>e, <strong>and</strong>rostenedi<strong>on</strong>e, follicle-stimulating horm<strong>on</strong>e (FSH) <strong>and</strong> sex horm<strong>on</strong>e binding globulin<br />

(SHBG) were measured in the first week of the follicular phase of the menstrual cycle.<br />

ABSTRACTS<br />

Statistical analyses: We used crude <strong>and</strong> adjusted linear regressi<strong>on</strong> models to quantify associati<strong>on</strong>s<br />

between adiposity indicators <strong>and</strong> breast dense volume (log-transformed).<br />

Results: 39% of the women were overweight <strong>and</strong> 31% obese. All adiposity indicators were positively <strong>and</strong><br />

similarly associated with log-dense volume [ie, BMI, β st<strong>and</strong>ardized regressi<strong>on</strong> coefficient: 0.02 (95% CI:<br />

0.01, 0.03); WC, β st<strong>and</strong>ardized regressi<strong>on</strong> coefficient: 0.01 (95% CI: 0.01, 0.13)]; these associati<strong>on</strong>s<br />

remained significant after adjusting by sociodemographic <strong>and</strong> metabolic <strong>and</strong> horm<strong>on</strong>al factors.<br />

C<strong>on</strong>clusi<strong>on</strong>: In premenopausal Chilean women adiposity markers, either measured by BMI, WC, WHR<br />

<strong>and</strong> percentage of total fat, are positively associated with breast dense volume. This associati<strong>on</strong> is <strong>on</strong>ly<br />

partially explained by metabolic <strong>and</strong> breast cell proliferative horm<strong>on</strong>al factors, suggesting that other<br />

pathways may also be involved. L<strong>on</strong>gitudinal studies are needed to c<strong>on</strong>firm our findings <strong>and</strong> to better<br />

underst<strong>and</strong> the links between obesity, breast dense tissue <strong>and</strong> breast cancer risk, in premenopausal<br />

women.<br />

52


P20<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

RELATIONSHIP OF SERUM ESTROGEN LEVELS WITH AREA AND VOLUME<br />

MEASURES OF MAMMOGRAPHIC DENSITY AMONG WOMEN UNDERGOING<br />

BREAST BIOPSY FOR CLINICAL INDICATIONS<br />

Gretchen Gierach 1 , Deesha Patel 1 , R<strong>on</strong>i Falk 1 , Ruth Pfeiffer 1 , Berta Geller 2 , Pamela Vacek 2 , D<strong>on</strong>ald<br />

Weaver 2 , Rachael Chicoine 2 , John Shepherd 3 , Jeff Wang 3 , Bo Fan 3 , Xia Xu 5 , Timothy Veenstra 5 , Barbara<br />

Fuhrman 4 , Louise Brint<strong>on</strong> 1 , <strong>and</strong> Mark Sherman 1<br />

1<br />

Nati<strong>on</strong>al Cancer Institute, Nati<strong>on</strong>al Institutes of Health, Rockville, MD, USA; 2 University of Verm<strong>on</strong>t,<br />

Burlingt<strong>on</strong>, VT, USA; 3 University of California, San Francisco, San Francisco, CA, USA; 4 University of<br />

Arkansas for Medical Sciences, Little Rock, AR, USA; 5 Frederick Nati<strong>on</strong>al Laboratory for Cancer<br />

Research, SAIC-Frederick, Inc., Frederick, MD, USA<br />

ABSTRACTS<br />

Background: Mammographic density (MD), assessed as the area of n<strong>on</strong>-fatty appearing tissue divided by<br />

the total breast area (MD-A), is an established breast cancer risk factor am<strong>on</strong>g pre- <strong>and</strong> postmenopausal<br />

women. Although increased exposure to estrogens is hypothesized to increase both MD <strong>and</strong> breast cancer<br />

risk, data relating serum estrogen levels to MD are inc<strong>on</strong>sistent. These inc<strong>on</strong>sistencies may reflect<br />

differences in methods of measuring estrogens, the specific estrogens assessed, <strong>and</strong> variable techniques<br />

for measuring MD. To elucidate these relati<strong>on</strong>ships, we examined the associati<strong>on</strong>s of a panel of serum<br />

estrogens <strong>and</strong> estrogen metabolites (EM), as measured by a newly developed assay, with both area (MD-<br />

A) <strong>and</strong> volume (MD-V) density measures in a cross-secti<strong>on</strong>al study of women undergoing breast biopsy.<br />

Methods: Pre- <strong>and</strong> postmenopausal women (n=194), ages 40-65, undergoing a diagnostic image-guided<br />

breast biopsy at a facility of the Verm<strong>on</strong>t <strong>Breast</strong> Cancer Surveillance System were enrolled (2007-2010).<br />

Serum parent estrogens, estr<strong>on</strong>e (E 1 ) <strong>and</strong> estradiol (E 2 ), <strong>and</strong> their 2-, 4-, <strong>and</strong> 16-hydroxylated metabolites<br />

(c<strong>on</strong>jugated <strong>and</strong> unc<strong>on</strong>jugated forms, the latter of which are thought to be more biologically available)<br />

were measured using a validated, sensitive liquid chromatography t<strong>and</strong>em mass spectrometry assay.<br />

Laboratory coefficients of variati<strong>on</strong> were less than 3% for all analytes. MD-A <strong>and</strong> MD-V were assessed<br />

in craniocaudal views of the breast c<strong>on</strong>tralateral to the biopsy target in digital mammograms using<br />

computerized thresholding software (cm 2 ) <strong>and</strong> single X-ray absorptiometry (mL). Linear regressi<strong>on</strong><br />

models were used to estimate associati<strong>on</strong>s of estrogens with square-root transformed MD-A <strong>and</strong> MD-V,<br />

expressed as absolute values <strong>and</strong> as percentages of breast area or volume, adjusted for the critical<br />

c<strong>on</strong>founders of age <strong>and</strong> body mass index. Analyses were stratified by menopausal status <strong>and</strong>, in<br />

premenopausal women, by menstrual cycle phase.<br />

Results: Am<strong>on</strong>g premenopausal women (n=106), circulating levels of most individual EM were not<br />

associated with MD-A or MD-V, although positive associati<strong>on</strong>s between percent MD-A <strong>and</strong> levels of<br />

unc<strong>on</strong>jugated E 1 <strong>and</strong> unc<strong>on</strong>jugated E 2 were suggested for luteal phase women (E 1 : p=0.08; E 2 : p=0.07).<br />

Am<strong>on</strong>g postmenopausal women (n=88), higher circulating levels of unc<strong>on</strong>jugated E 1 showed an<br />

associati<strong>on</strong> with increased percent MD-V (p=0.049), but not with MD-A. Higher unc<strong>on</strong>jugated E 2 levels<br />

showed n<strong>on</strong>-significant positive relati<strong>on</strong>ships with absolute <strong>and</strong> percent MD-A <strong>and</strong> MD-V measures (pvalues<br />

ranged from 0.07-0.11). Levels of the 2-, 4- <strong>and</strong> 16-hydroxylati<strong>on</strong> pathway EM were positively<br />

associated with MD-A measures (percent MD-A: p=0.009, p=0.02, p=0.02; absolute MD-A: p=0.05,<br />

p=0.09, p=0.03, respectively), but not with MD-V.<br />

C<strong>on</strong>clusi<strong>on</strong>s: Positive associati<strong>on</strong>s between serum estrogen profiles <strong>and</strong> MD were apparent am<strong>on</strong>g<br />

women undergoing breast biopsy for clinical indicati<strong>on</strong>s. Further, our findings suggest that different<br />

measures of MD may provide different informati<strong>on</strong> about the biologic underpinnings of breast tissue<br />

compositi<strong>on</strong>. Future studies assessing relati<strong>on</strong>ships of estrogen metabolites <strong>and</strong> MD may provide clues<br />

about the etiology of breast cancer.<br />

53


P21<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MAMMOGRAPHIC DENSITY PHENOTYPES AND RISK OF BREAST CANCER: A<br />

META-ANALYSIS<br />

Andreas Petterss<strong>on</strong> 1 , Rebecca E. Graff 1 , Laura Baglietto 2, 3 , Marije F. Bakker 4 , Norman Boyd 5 , KeeSeng<br />

Chia 6 , Kamila Czene 7 , Isabel dos Santos Silva 8 , Louise Erikss<strong>on</strong> 7 , Graham G. Giles 2, 3, 9 , Per Hall 7 , Mikael<br />

Hartman 6, 7, 9 , John L. Hopper 3 , Kavitha Krishnan 2, 3 , Jingmei Li 11 , Qing Li 5 , Gertraud Maskarinec 12 ,<br />

Valerie McCormack 13 , Ian Pagano 12 , Bernard A. Rosner 14, 15 , Christopher Scott 16 , Jennifer St<strong>on</strong>e 3 , Giske<br />

Ursin 17, 18 , Celine Vach<strong>on</strong> 16 , Carla H. van Gils 4 , Chia Si<strong>on</strong>g W<strong>on</strong>g 6 1, 14<br />

, Rulla M. Tamimi<br />

1) Department of Epidemiology, Harvard School of Public Health, Bost<strong>on</strong>, MA, USA; 2) Cancer<br />

Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia; 3) Centre for Molecular,<br />

Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytical Epidemiology, The University of Melbourne, Australia; 4) Julius<br />

Center for Health Sciences <strong>and</strong> Primary Care, University Medical Center, Utrecht, The Netherl<strong>and</strong>s;<br />

5) Campbell Family Institute for <strong>Breast</strong> Cancer Research, Ontario Cancer Institute, Tor<strong>on</strong>to, Ontario,<br />

Canada; 6) Saw Swee Hock School of Public Health, Nati<strong>on</strong>al University of Singapore, Nati<strong>on</strong>al<br />

University Health System, Singapore; 7) Department of Medical Epidemiology <strong>and</strong> Biostatistics,<br />

Karolinska Institutet, Stockholm, Sweden; 8) Department of N<strong>on</strong>-Communicable Disease Epidemiology,<br />

L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical Medicine, L<strong>on</strong>d<strong>on</strong>, United Kingdom; 9) Department of<br />

Epidemiology <strong>and</strong> Preventive Medicine, M<strong>on</strong>ash University, Melbourne, Australia; 10) Department of<br />

Surgery, Y<strong>on</strong>g Loo Lin School of Medicine, Nati<strong>on</strong>al University of Singapore, Singapore; 11) Human<br />

Genetics Divisi<strong>on</strong>, Genome Institute of Singapore, Singapore; 12) Department of Epidemiology, University<br />

of Hawaii Cancer Center, H<strong>on</strong>olulu, HI, USA; 13) Secti<strong>on</strong> of Envir<strong>on</strong>ment <strong>and</strong> Radiati<strong>on</strong>, <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g><br />

Agency for Research <strong>on</strong> Cancer, Ly<strong>on</strong>, France; 14) Channing Divisi<strong>on</strong> of Network Medicine, Department<br />

of Medicine, Brigham <strong>and</strong> Women’s Hospital <strong>and</strong> Harvard Medical School, Bost<strong>on</strong>, MA, USA;<br />

15) Department of Biostatistics, Harvard School of Public Health, Bost<strong>on</strong>, MA, USA; 16) Department of<br />

Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA; 17) Department of<br />

Nutriti<strong>on</strong>, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; 18) Department of<br />

Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA<br />

Background: Fibrogl<strong>and</strong>ular breast tissue appears dense <strong>on</strong> mammogram; fat appears n<strong>on</strong>dense. We<br />

investigated whether absolute or percent dense area more str<strong>on</strong>gly predicts breast cancer risk, <strong>and</strong> whether<br />

absolute n<strong>on</strong>dense area is independently associated with risk.<br />

Methods: We c<strong>on</strong>ducted a meta-analysis of 13 case-c<strong>on</strong>trol studies, each having provided parameter<br />

estimates <strong>and</strong> st<strong>and</strong>ard errors from logistic regressi<strong>on</strong>s for associati<strong>on</strong>s between <strong>on</strong>e st<strong>and</strong>ard deviati<strong>on</strong><br />

(SD) increments in mammographic density phenotypes <strong>and</strong> breast cancer risk. We used r<strong>and</strong>om-effects<br />

models to calculate pooled odds ratios (ORs) <strong>and</strong> 95% c<strong>on</strong>fidence intervals (CIs).<br />

ABSTRACTS<br />

Results: Am<strong>on</strong>g premenopausal women (1,776 cases; 2,834 c<strong>on</strong>trols), summary ORs for <strong>on</strong>e SD<br />

increments were 1.37 (95% CI: 1.29-1.47) for absolute dense area, 0.78 (95% CI: 0.71-0.86) for absolute<br />

n<strong>on</strong>dense area, <strong>and</strong> 1.52 (95% CI: 1.39-1.66) for percent dense area when pooling estimates adjusted for<br />

age, BMI <strong>and</strong> parity. Corresp<strong>on</strong>ding ORs am<strong>on</strong>g postmenopausal women (6,643 cases; 11,187 c<strong>on</strong>trols)<br />

were 1.38 (95% CI: 1.31-1.44), 0.79 (95% CI: 0.73-0.85), <strong>and</strong> 1.53 (95% CI: 1.44-1.64). After additi<strong>on</strong>al<br />

adjustment for absolute dense area, associati<strong>on</strong>s between absolute n<strong>on</strong>dense area <strong>and</strong> breast cancer<br />

became attenuated or null in several studies <strong>and</strong> summary ORs became 0.82 (95% CI: 0.71-0.94; P-<br />

heterogeneity: 0.02) for premenopausal <strong>and</strong> 0.85 (95% CI: 0.75-0.96; P-heterogeneity:


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P22 MAMMOGRAPHIC DENSITY AND EARLY MAMMOGRAPHIC SCREENING<br />

PERFORMANCE MEASURES IN THE NORWEGIAN BREAST CANCER SCREENING<br />

PROGRAM<br />

Solveig Hofvind 1,2* Isabel dos-Santos-Silva 3 , Merete Ellingjord-Dale 4 , Sofie Sebuødegård 1 , Giske Ursin 1,5<br />

1 Cancer Registry of Norway, Oslo, Norway; 3 Department of N<strong>on</strong>-Communicable Disease Epidemiology,<br />

L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical Medicine, L<strong>on</strong>d<strong>on</strong>, UK ; 4 Institute of Basic Medical Sciences,<br />

University of Oslo, Oslo, Norway ; 2 Oslo <strong>and</strong> Akershus University College of Applied Sciences, Oslo,<br />

Norway; 5 University of Southern California, Los Angeles, CA<br />

ABSTRACTS<br />

Background: It is well known that mammographic density (MD) increases the risk of breast cancer <strong>and</strong><br />

reduces sensitivity of mammographic screening. However, less is known <strong>on</strong> how MD may affect early<br />

performance measures in mammographic screening am<strong>on</strong>g those recalled for further assessments. We<br />

used data collected by the populati<strong>on</strong>-based Norwegian <strong>Breast</strong> Cancer Screening Program (NBCSP) to<br />

investigate associati<strong>on</strong>s of MD <strong>and</strong> the percentage of cancers detected am<strong>on</strong>g women recalled for further<br />

assessment (positive predictive value <strong>on</strong>e, PPV-1), as well as percentage of cancers detected am<strong>on</strong>g the<br />

women who underwent a biopsy (fine needle aspirati<strong>on</strong> cytology or core needle biopsy)(PPV-2) <strong>and</strong> MD<br />

associati<strong>on</strong>s with prognostic histopathological tumor characteristics.<br />

Methods: We analyzed data from women screened in the NBCSP during 1996-2010 who were<br />

subsequently recalled for further assessments. Visual assessment of MD was routinely performed <strong>on</strong><br />

analogue (80%) or digital mammograms by radiologists with expertise in mammography using an inhouse<br />

visual 3-category scale (low: MD


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P23 CHILDHOOD AND ADOLESCENT GROWTH PREDICTS MAMMOGRAPHIC<br />

DENSITY MEASURES ASSOCIATED WITH BREAST CANCER RISK IN MID-LIFE: A<br />

PROSPECTIVE STUDY<br />

John L. Hopper 1 , Jennifer St<strong>on</strong>e 1 , Carmel Apicella 1 , Linh T. Nguyen 1 , Quang M. Bui 1 , Gillian S. Dite 1 ,<br />

Daniel F. Schmidt 1 , Enes Makalic 1 , Melanie C Mathes<strong>on</strong> 1 , Shyamali Dharmage 1 , Graham G. Giles 2<br />

1 Centre for Molecular, Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytic Epidemiology, The University of Melbourne,<br />

Carlt<strong>on</strong> VIC 3053, Australia; 2 Cancer Epidemiology Centre, Cancer Council Victoria, Carlt<strong>on</strong> VIC 3053,<br />

Australia<br />

Background: Mammographic density (MD) adjusted for age <strong>and</strong> body mass index (BMI) is associated<br />

with breast cancer risk. We c<strong>on</strong>ducted a prospective study to determine if growth in childhood <strong>and</strong><br />

adolescence predicts risk-associated MD.<br />

Methods: In 1968, the Tasmanian Asthma Study measured height <strong>and</strong> weight for almost all school<br />

children born in 1961. We obtained measured heights <strong>and</strong> weights for this cohort from annual school<br />

records for 1969 to 1977. Between 2009 <strong>and</strong> 2010, for 215 of these women we administered a<br />

questi<strong>on</strong>naire, including height <strong>and</strong> weight, <strong>and</strong> digitised at least <strong>on</strong>e mammogram. Absolute <strong>and</strong> percent<br />

MD were measured using the computer-assisted method Cumulus. Preece-Baines growth curves were<br />

fitted <strong>and</strong> missing data imputed under a multivariate normal model. We used linear regressi<strong>on</strong> to assess<br />

the associati<strong>on</strong> between square root MD <strong>and</strong> covariates.<br />

Results: Both absolute <strong>and</strong> percent MD adjusted for age <strong>and</strong> current BMI were negatively associated with<br />

log BMI at each of ages 7 to 15, <strong>and</strong> positively associated with age at menarche. When fitted together, log<br />

BMI at age 15 remained negatively associated (p < 0.00001), explaining about 15% of the variance (cf.<br />


P24<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BACKGROUND PARENCHYMAL UPTAKE OF Tc-99m SESTAMIBI ON MOLECULAR<br />

BREAST IMAGING IN WOMEN WITH DENSE BREASTS<br />

Hruska CB 1 , C<strong>on</strong>ners AL 1 , J<strong>on</strong>es KN 1 , Lingineni R 2 , Carter R 2 , Shah S 3 , Visscher D 3 , Rhodes DJ 4 ,<br />

Vach<strong>on</strong> C 2<br />

1 Departments of Radiology, 2 Health Sciences Research, 3 Laboratory Medicine <strong>and</strong> 4 Pathology, <strong>and</strong><br />

Medicine, Mayo Clinic, Rochester, MN<br />

Background: While evaluating molecular breast imaging (MBI) for adjunct screening in women with<br />

dense breasts, various presentati<strong>on</strong>s of background parenchymal uptake (BPU), or uptake of Tc-99m<br />

sestamibi in normal fibrogl<strong>and</strong>ular tissue (FT), were observed. We describe the prevalence of BPU in<br />

dense breasts <strong>and</strong> examine clinical <strong>and</strong> histologic risk factors associated with BPU.<br />

ABSTRACTS<br />

Methods: Screening MBI exams performed between April 2010 <strong>and</strong> March 2012 at Mayo Clinic in<br />

women with dense breasts (BI-RADS D2 - scattered densities; D3 - heterogeneously dense, or D4 -<br />

extremely dense) were reviewed. Participants with breast implants or cancer diagnosed at screening were<br />

excluded. BPU intensity was subjectively categorized by two radiologists as photopenic (uptake in FT <<br />

subcutaneous fat), mild (uptake in FT = fat), moderate (uptake in FT up to 2x fat), or marked (uptake in<br />

FT > 2x fat). Associati<strong>on</strong> of BPU with age, BI-RADS mammographic density, percent density measured<br />

by Cumulus, menopausal status, <strong>and</strong> use of horm<strong>on</strong>al medicati<strong>on</strong>s was examined using Kruskal-Wallis<br />

<strong>and</strong> Chi-Square analyses. To examine associati<strong>on</strong> of histological factors with BPU, prospective coreneedle<br />

breast biopsy of 23 postmenopausal women was performed in regi<strong>on</strong>s of mammographically dense<br />

tissue dem<strong>on</strong>strating either photopenic (18 women) or marked (5 women) BPU.<br />

Results: In 1274 MBI exams, BPU was photopenic in 273 (21%), mild in 826 (65%), <strong>and</strong> moderate or<br />

marked in 175 (14%). Significant differences in distributi<strong>on</strong> of BPU were observed with age, BI-RADS<br />

density, percent density, menopausal status, <strong>and</strong> postmenopausal horm<strong>on</strong>e use (all p75% involuted).<br />

C<strong>on</strong>clusi<strong>on</strong>: Moderate/marked BPU occurred more often in denser breasts <strong>and</strong> in women who were<br />

younger, pre- or perimenopausal, or using exogenous horm<strong>on</strong>es. In each density category, substantial<br />

proporti<strong>on</strong>s of both moderate/marked <strong>and</strong> photopenic BPU were observed, establishing that similarappearing<br />

FT <strong>on</strong> mammography can dem<strong>on</strong>strate c<strong>on</strong>siderable differences in BPU <strong>on</strong> MBI.<br />

57


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Photopenic<br />

Marked<br />

Figure 1. In these 3 patients, who were all postmenopausal<br />

<strong>and</strong> not using any exogenous horm<strong>on</strong>es, the anatomic<br />

appearance of fibrogl<strong>and</strong>ular tissue is similar <strong>on</strong> digital<br />

mammography (top row). However, the background<br />

parenchymal uptake of Tc-99m sestamibi <strong>on</strong> MBI (bottom<br />

row) dem<strong>on</strong>strates differing behavior of the dense tissue,<br />

which may indicate differences in individual risk.<br />

Figure 2. Core biopsy specimens obtained from two<br />

postmenopausal patients – a 58 year-old woman with<br />

photopenic background uptake <strong>and</strong> a 62 year-old<br />

woman with marked background uptake <strong>on</strong> MBI. The<br />

tissue with marked uptake showed higher Ki-67 index,<br />

less lobular involuti<strong>on</strong>, <strong>and</strong> a greater proporti<strong>on</strong> of<br />

epithelium versus stroma compared to the tissue with<br />

photopenic uptake.<br />

ABSTRACTS<br />

58


P25<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Impact of California <strong>Breast</strong> Density Notificati<strong>on</strong> Law SB 1538 <strong>on</strong> California Women <strong>and</strong><br />

Their Health Care Providers<br />

Debra M. Ikeda, J<strong>on</strong>athan Hargreaves, Elissa Price, Jafi Lips<strong>on</strong>, B<strong>on</strong>nie N. Joe, Karen Lindfors, R. James<br />

Brenner, Stephen Feig, Lawrence Bassett, Jessica Leung, Haydee Ojeda-Fournier, Allis<strong>on</strong> Kurian, Elyse<br />

Love, D<strong>on</strong>na Waldenbach, Bruce L. Daniel, Edward A. Sickles<br />

Objective: To describe the impact of California <strong>Breast</strong> Density Notificati<strong>on</strong> SB 1538 legislati<strong>on</strong> <strong>on</strong><br />

California medical facilities, with emphasis <strong>on</strong> development <strong>and</strong> implementati<strong>on</strong> of policy<br />

ABSTRACTS<br />

Background: As of April 1, 2013, California State Senate Bill 1538 (SB 1538) (1) requires healthcare<br />

providers to send notificati<strong>on</strong> to all women identified with heterogeneously dense or extremely dense<br />

breast tissue <strong>on</strong> mammograms c<strong>on</strong>taining the following language: “Your mammogram shows that your<br />

breast tissue is dense. Dense tissue is comm<strong>on</strong> <strong>and</strong> is not abnormal. However, dense tissue can make it<br />

harder to evaluate the results of your mammogram <strong>and</strong> may also be associated with an increased risk of<br />

breast cancer. This informati<strong>on</strong> about the results of your mammogram is given to you to raise your<br />

awareness <strong>and</strong> to inform your c<strong>on</strong>versati<strong>on</strong>s with your doctor. Together, you can decide which screening<br />

opti<strong>on</strong>s are right for you. A report of your results was sent to your physician.” Women with n<strong>on</strong>-dense<br />

tissue <strong>on</strong> mammograms receive no notificati<strong>on</strong>. While SB 1538 does provide limited informati<strong>on</strong> to<br />

patients, the bill provides no funding for supplemental screening, <strong>and</strong> no clear guidance <strong>on</strong> the<br />

implicati<strong>on</strong>s of density for breast cancer risk or recommendati<strong>on</strong>s of when <strong>and</strong> what types of screening<br />

modalities should be recommended to patients. Figure 1 compares the screening decisi<strong>on</strong> tree in the pre-<br />

SB 1538 era (<strong>on</strong> the left) to the increasingly complex decisi<strong>on</strong> tree in the post-SB 1538 era (<strong>on</strong> the<br />

right).<br />

C<strong>on</strong>necticut legislati<strong>on</strong> (Public Act 09–41) also m<strong>and</strong>ated notificati<strong>on</strong>, but included funding for<br />

screening breast ultrasound (US). Following implementati<strong>on</strong> of the bill, screening US uptake rose in 16%<br />

of eligible women (2), but was accompanied by high false positive (FP) imaging <strong>and</strong> biopsy rates with<br />

low positive predictive values (PPV) ranging from 5.6% to 6.7% for the 3-4 cancers/1000 women<br />

detected by US (2,3,4). US was shown in other studies to have a similarly low PPV of 8.8% (5). As<br />

indicated by the required text, the letter implies that having dense breast tissue: (a) increases breast cancer<br />

risk; (b) potentially “masks” cancers; <strong>and</strong> (c) may lead to doctors’ recommending supplemental screening<br />

tests. However, the informati<strong>on</strong> necessary to interpret these implicati<strong>on</strong>s is missing, leaving patients <strong>and</strong><br />

providers to ask: How much does breast cancer risk increase Who should undergo more screening How<br />

should breast cancer risk be calculated for an individual patient Prior to implementati<strong>on</strong> of SB 1538,<br />

there was no instituti<strong>on</strong>-neutral, scientifically based website to guide women <strong>and</strong> healthcare providers<br />

regarding dense breast tissue notificati<strong>on</strong> legislati<strong>on</strong>. The California <strong>Breast</strong> Density Informati<strong>on</strong> Group<br />

(CBDIG) formed to resp<strong>on</strong>d to California SB 1538 legislati<strong>on</strong>, to review existing scientific evidence <strong>and</strong><br />

identify gaps in informati<strong>on</strong> to provide scientifically-based recommendati<strong>on</strong>s for guiding health care<br />

providers <strong>and</strong> women regarding supplemental screening after mammography that identifies<br />

59


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

heterogeneously dense <strong>and</strong> dense breast tissue, <strong>and</strong> to formulate recommendati<strong>on</strong>s when no science was<br />

available to guide policy.<br />

Materials <strong>and</strong> Methods: The CBDIG task was to review existing scientific literature to provide<br />

evidence-based recommendati<strong>on</strong>s for guiding health care providers <strong>and</strong> women regarding supplemental<br />

screening after notificati<strong>on</strong>, <strong>and</strong> to formulate recommendati<strong>on</strong>s when no science was available to guide<br />

policy. Each facility then developed their own policy to resp<strong>on</strong>d to SB 1538, which was implemented in<br />

California <strong>on</strong> April 1, 2013.<br />

Results: After breast density notificati<strong>on</strong>, Figure 2: NCCN, ACS <strong>and</strong> ACR Guidelines CBDIG<br />

recommended women <strong>and</strong> health care for <strong>Breast</strong> Cancer Screening after Normal<br />

providers to c<strong>on</strong>sider supplemental breast Screening Mammography<br />

cancer<br />

screening based <strong>on</strong> breast cancer risk<br />

assessment (6) using family history models (7,8)<br />

based <strong>on</strong> recommendati<strong>on</strong>s from nati<strong>on</strong>ally<br />

High risk (> 20% lifetime) – MRI*<br />

Intermediate risk (15-20% lifetime ) – possible MRI**<br />

recognized guidelines (7,8,9) (Figure 2). Average Risk (


P26<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

CHALLENGES IN DE-IDENTIFYING FILM-SCREEN MAMMOGRAMS PRIOR TO BREAST<br />

DENSITY ASSESSMENT FOR THE NOVICE PRACTITIONER<br />

J Jobling, C D’Este, JF Forbes<br />

School of Medicine <strong>and</strong> Public Health, University of Newcastle, NSW Australia<br />

Introducti<strong>on</strong>: Measurement of breast density <strong>on</strong> mammograms presents many challenges. Film-screen<br />

mammograms require c<strong>on</strong>versi<strong>on</strong> to digital format prior to assessment with computer software, but<br />

c<strong>on</strong>tain burned-in areas of identifying informati<strong>on</strong> that should be removed to protect participant privacy.<br />

We are undertaking a retrospective linkage study in at least 4,000 women between the lower Hunter New<br />

Engl<strong>and</strong> (NSW Australia) <strong>Breast</strong>Screen <strong>and</strong> the NSW 45 <strong>and</strong> Up Study to examine l<strong>on</strong>gitudinal changes<br />

in breast density <strong>and</strong> breast cancer risk. The BIRADS density categories are not routinely employed in<br />

Australia hence density will be assessed via the Cumulus software <strong>and</strong>/or visual methods. A simple,<br />

efficient <strong>and</strong> inexpensive method to de-identify thous<strong>and</strong>s of film-screen mammograms is needed.<br />

Coverage of identifying areas <strong>on</strong> film with st<strong>and</strong>ard sticky labels during digitisati<strong>on</strong> <strong>on</strong> an Array 2905HD<br />

laser scanner is not effective due to the strength of the laser. Our aim was to evaluate a range of software<br />

programs available for this purpose.<br />

ABSTRACTS<br />

Methods: Software capable of quickly <strong>and</strong> easily batch de-identifying digital images in DICOM format<br />

was sought via general internet searches using the terms ‘DICOM an<strong>on</strong>ymizer burned in’; searches in<br />

PubMed using similar terms were also undertaken. Software was evaluated by: 1) its capacity to easily<br />

batch de-identify DICOM images with burned-in identifying areas (i.e. digitised film-screen<br />

mammograms), 2) the ability to easily remove DICOM header informati<strong>on</strong> from digital mammograms,<br />

<strong>and</strong> 3) retenti<strong>on</strong> of detail <strong>and</strong> compatibility with the Cumulus software post-processing.<br />

Results: More than 60 free DICOM viewers <strong>and</strong> 20 de-identifiers/an<strong>on</strong>ymisers were identified<br />

(dclunie.com). Searches in PubMed found two useful, recent (


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P27 ASSOCIATION BETWEEN AUTOMATED, VOLUMETRIC BREAST DENSITY<br />

MEASURES AND BREAST CANCER IN A LARGE SCREENING POPULATION<br />

M.G.J. Kallenberg 1,2 , K. Holl<strong>and</strong> 1 , J.O.P. W<strong>and</strong>ers 3 , G.H. van Gils 3 , <strong>and</strong> N. Karssemeijer 1<br />

1 Radboud University Nijmegen Medical Centre, Nijmegen, the Neterl<strong>and</strong>s; 2 Matakina Technology,<br />

Wellingt<strong>on</strong>, New Zeal<strong>and</strong>, michiel.kallenberg@matakina.com; 3 Julius Center for Health Sciences <strong>and</strong><br />

Primary Care, University Medical Centre Utrecht, the Netherl<strong>and</strong>s<br />

PURPOSE: The associati<strong>on</strong> between breast density <strong>and</strong> breast cancer risk has been established with<br />

semi-automated, area based measurement of breast density. In this work we investigate the performance<br />

of a fully automated, volumetric method to predict breast cancer in a large screening populati<strong>on</strong>.<br />

MATERIALS AND METHOD: The data that was used in this work comprised 107,488 digital<br />

screening exams, performed in 2003-2012, from 53,975 women in the Dutch <strong>Breast</strong> Cancer Screening<br />

Program, with a mean age of 59.2 years. The set c<strong>on</strong>tained 730 biopsy proven malignant breast cancer<br />

cases that were diagnosed through the screening program (screen-detected). Interval tumors (diagnosed in<br />

between screening rounds) have not yet been identified for this screening period. For 363 cases <strong>on</strong>ly<br />

mammograms in which cancers were detected were available, while for 367 cases we also had prior<br />

screening mammograms. For the latter cases we selected the oldest mammographic examinati<strong>on</strong><br />

available. The average time between the oldest mammographic examinati<strong>on</strong> <strong>and</strong> diagnosis was 3.5 years.<br />

All images were recorded <strong>on</strong> a Hologic Selenia FFDM system, using st<strong>and</strong>ard clinical settings.<br />

For each mammographic image we assessed volumetric breast density with commercially available<br />

software (Volpara, Matakina Technology, Wellingt<strong>on</strong>, New Zeal<strong>and</strong>, versi<strong>on</strong> 1.4.0). To obtain <strong>on</strong>e<br />

density estimate per exam we averaged over the MLO <strong>and</strong> CC view <strong>and</strong> over the left <strong>and</strong> right breast.<br />

When a case c<strong>on</strong>tained pathology we averaged <strong>on</strong>ly over the n<strong>on</strong> affected side. We examined the<br />

relati<strong>on</strong>ship between volume of fibrogl<strong>and</strong>ular tissue <strong>and</strong> the risk of breast cancer by calculating odds<br />

ratios <strong>and</strong> their 95% c<strong>on</strong>fidence intervals. Exams were categorized into four groups, in such a way that<br />

the group sizes equal the group sizes <strong>on</strong>e would obtain with Volpara density grade (VDG) scoring, which<br />

is a four point scale analog to BI-RADS density scores. We used logistic regressi<strong>on</strong> to adjust for age at<br />

mammography <strong>and</strong> breast volume, which served as a surrogate marker for BMI.<br />

RESULTS: After adjustment for age <strong>and</strong> breast volume, dense volume was significantly higher in cases<br />

than c<strong>on</strong>trols (p


P28<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

LONGITUDINAL TRACKING WITH TIME OF MAMMOGRAPHIC DENSITY<br />

MEASURES THAT PREDICT BREAST CANCER<br />

Kavitha Krishnan 1,2 , Laura Baglietto 1,2 , Jennifer St<strong>on</strong>e 2 , Carmel Apicella 2 , Dallas R English 1,2 , Gianluca<br />

Severi 1,2 , Graham G Giles 1,2,3 , John Hopper 2<br />

1 Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, AUSTRALIA, 2 Centre for Molecular,<br />

Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytical epidemiology, University of Melbourne, AUSTRALIA,<br />

3 Department of Epidemiology <strong>and</strong> Preventive Medicine, M<strong>on</strong>ash University, Melbourne, AUSTRALIA<br />

Background: Mammographic density adjusted for age <strong>and</strong> body mass index (BMI) is an established risk<br />

factor for breast cancer. If density for age <strong>and</strong> BMI tracks str<strong>on</strong>gly with time from a young age then it<br />

could be possible to identify young women at high risk of breast cancer <strong>and</strong> try to offer them more<br />

effective screening regimes. This might increase early detecti<strong>on</strong> of breast cancer <strong>and</strong> hence reduce<br />

mortality due to breast cancer.<br />

ABSTRACTS<br />

Methods: Combining both the Melbourne Collaborative Cohort Study (MCCS) <strong>and</strong> the Australian <strong>Breast</strong><br />

Cancer Family Study (ABCFS), we identified 1,064 women who had an average of 4 mammograms each<br />

between the ages of 24-83 years (N=4,723 mammograms; range 1-14 mammograms per woman).<br />

Mammographic density was measured with the software Cumulus <strong>and</strong> transformed to approximate<br />

normality using the Box-Cox power formula. BMI at each mammogram was assigned using measures<br />

available at study entry <strong>and</strong> at two follow-ups for the MCCS <strong>and</strong> <strong>on</strong>e follow-up for the ABCFS based <strong>on</strong><br />

the proximity of the date at which the mammogram was taken <strong>and</strong> the attendance dates of the studies.<br />

Transformed dense area adjusted for age <strong>and</strong> BMI was fitted using mixed models with a r<strong>and</strong>om intercept,<br />

overall <strong>and</strong> separately for mammograms in the premenopausal age group 24-49 <strong>and</strong> postmenopausal age<br />

group 50-83, with no correlati<strong>on</strong> between measurements, exp<strong>on</strong>ential correlati<strong>on</strong> between measurements<br />

<strong>and</strong> r<strong>and</strong>om slope. Estimates of the correlati<strong>on</strong>s between dense areas between different ages were<br />

estimated from the best fit model identified using the log restricted-likelihood.<br />

Results: Dense area was transformed to the power of 0.2. The overall best fit model was mean dense area<br />

decreasing quadratically with age <strong>and</strong> linearly with BMI with an exp<strong>on</strong>ential correlati<strong>on</strong> structure. The<br />

correlati<strong>on</strong>s in adjusted dense area measures 2, 4, 6, 8 <strong>and</strong> 10 years apart were 0.93, 0.90, 0.89, 0.88 <strong>and</strong><br />

0.88, respectively. For mammograms in the premenopausal age group 24-49, the mean dense area<br />

decreased quadratically with age but there was no significant associati<strong>on</strong> with BMI. The correlati<strong>on</strong>s in<br />

adjusted dense area measures 2, 4, 6, 8 <strong>and</strong> 10 years apart were slightly lower at 0.85, 0.80, 0.79, 0.79 <strong>and</strong><br />

0.78, respectively. The model for the postmenopausal age group was similar to the overall model <strong>and</strong> the<br />

correlati<strong>on</strong>s were also slightly higher than the overall model, being 0.94, 0.91, 0.90, 0.89 <strong>and</strong> 0.89 at 2, 4,<br />

6, 8 <strong>and</strong> 10 years apart, respectively.<br />

C<strong>on</strong>clusi<strong>on</strong>: Mammographic dense area measurements that predict breast cancer risk are highly<br />

correlated over 10 years regardless of menopausal status.<br />

63


P29<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

MULTI-VENDOR VALIDATION OF A FULLY-AUTOMATED BREAST DENSITY<br />

ESTIMATION METHOD APPLICABLE TO BOTH RAW AND PROCESSED DIGITAL<br />

MAMMOGRAMS<br />

Brad M. Keller*, Ph.D., Diane L. Nathan, M.D., Lee D. McDaniel, M.S., Nigel Bristol, A.S., Yan Wang,<br />

Ph.D., Yuanjie Zheng, Ph.D., James C. Gee, Ph.D., Emily F. C<strong>on</strong>ant, M.D., Despina K<strong>on</strong>tos, Ph.D.<br />

Department of Radiology, Perelman School of Medicine, University of Pennsylvania<br />

<strong>Breast</strong> density is becoming increasingly recognized as an important biomarker of a woman’s breast tissue<br />

compositi<strong>on</strong> as it is known to affect both her individual risk for developing cancer as well as the<br />

sensitivity <strong>and</strong> specificity of screening mammography. In the United States, this has led to an increasing<br />

amount of legislati<strong>on</strong> m<strong>and</strong>ating that breast density be measured as part of routine clinical practice,<br />

communicated to the patient <strong>and</strong> potentially used to guide tailored screening <strong>and</strong> surveillance strategies.<br />

However, inter-reader variability in the measurement of breast density, particularly when using qualitative<br />

or semi-automated assessment strategies, is a known issue as variati<strong>on</strong>s in the interpretati<strong>on</strong> of density<br />

could lead to potential differences in risk evaluati<strong>on</strong> <strong>and</strong> any subsequent screening recommendati<strong>on</strong>s.<br />

Development of an automated system to measure breast density accurately <strong>and</strong> reproducibly can alleviate<br />

such issues, resulting in objective <strong>and</strong> reproducible quantitative measures. In order to be generalizable for<br />

use in both prospective clinical practice <strong>and</strong> retrospective research settings, such a system would ideally<br />

need to be able to estimate breast density from both raw (“For Processing”) <strong>and</strong> vendor post-processed<br />

(i.e., “For Presentati<strong>on</strong>”) digital mammography images, as well as be validated <strong>on</strong> mammographic images<br />

derived from machines across multiple vendors. In this study, we dem<strong>on</strong>strate the multi-format, multivendor<br />

generalizability of a fully-automated breast percent density (PD%) estimati<strong>on</strong> algorithm recently<br />

developed by our group.<br />

In this IRB-approved, HIPAA-compliant study, we used bilateral MLO full-field digital screening<br />

mammograms in both raw <strong>and</strong> post-processed format from a total 246 women to train <strong>and</strong> validate our<br />

previously developed adaptive fuzzy c-means density estimati<strong>on</strong> computer algorithm. Data were available<br />

from two vendors: 81 studies were acquired using the Senographe Essential system (GE Healthcare); <strong>and</strong><br />

165 studies were acquired using the Selenia Dimensi<strong>on</strong>s system (Hologic, Inc.). Leave-<strong>on</strong>e-out cross<br />

validati<strong>on</strong> was used to determine algorithm agreement with a radiologist’s manual estimati<strong>on</strong> of breast<br />

PD% using the widely validated semi-automated Cumulus software <strong>on</strong> a per-woman basis.<br />

When training our density estimati<strong>on</strong> software with GE Essential digital mammograms, the algorithm was<br />

able to achieve str<strong>on</strong>g correlati<strong>on</strong> between the automated density estimates <strong>and</strong> the radiologist’s manual<br />

percent density estimates for both raw (r=0.85, p


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P30 GENETIC DETERMINANTS OF FIBROGLANDULAR BREAST TISSUE:<br />

IMPLICATIONS ON USING MAMMOGRAPHIC DENSITY MEASURES IN BREAST<br />

CANCER RISK ASSESSMENT<br />

D. K<strong>on</strong>tos 1* , B. Keller 1 , S. M. Domchek 2 , J. Chen 3 , E. H<strong>and</strong>orf 2 , M. J<strong>on</strong>es 2 , L. Boghossian 2 , K.<br />

Armstr<strong>on</strong>g 2 , E. C<strong>on</strong>ant 1<br />

University of Pennsylvania, Departments of Radiology 1 , Medicine 2 , <strong>and</strong> Epidemiology 3<br />

<strong>Breast</strong> density, typically measured as the percent area of mammographically dense tissue, is a str<strong>on</strong>g risk<br />

factor for breast cancer. Currently, the biological determinants of the associati<strong>on</strong> between breast density<br />

<strong>and</strong> the risk of breast cancer are not yet fully understood. We investigate potential genetic determinants of<br />

breast density, by examining the associati<strong>on</strong> between new measures of fibrogl<strong>and</strong>ular tissue volume,<br />

versus the st<strong>and</strong>ard percent area density measures, <strong>and</strong> a series of validated single nucleotide<br />

polymorphisms (SNPs) associated with established breast cancer susceptibility loci. Bilateral two-view<br />

full-field digital mammography (FFDM) images were retrospectively analyzed under HIPAA <strong>and</strong> IRBapproval<br />

from 433 women (59.6% Caucasian, 40.4% African American, Age 53±7yrs, Gail Risk Lifetime<br />

Risk 10.4±4.4) with a negative screening exam who had results available from a validated genetic assay<br />

for breast cancer risk assessment (deCODE <strong>Breast</strong>Cancer, deCODE genetics, Inc.).<br />

ABSTRACTS<br />

This genetic assay measures the allelic variati<strong>on</strong> for 12 validated SNPs previously associated with breast<br />

cancer risk. Fibrogl<strong>and</strong>ular breast tissue volume, both in terms of relative (%) <strong>and</strong> absolute amount (cm 3 ),<br />

was measured <strong>on</strong> a per-breast basis using validated FDA-cleared automated software (Quantra TM ,<br />

Hologic, Inc). Mammographic area percent density (PD%) was estimated with our previously validated<br />

computer algorithm based <strong>on</strong> automated intensity clustering <strong>and</strong> thresholding. The fibrogl<strong>and</strong>ular tissue<br />

measures were averaged between the left <strong>and</strong> right breasts for each woman to create corresp<strong>on</strong>ding<br />

measures <strong>on</strong> a per-woman basis. Associati<strong>on</strong>s between each SNP <strong>and</strong> the fibrogl<strong>and</strong>ular tissue measures<br />

were assessed with univariate linear regressi<strong>on</strong>. Model adjustments were c<strong>on</strong>sidered for age, ethnicity,<br />

<strong>and</strong> Gail lifetime risk. B<strong>on</strong>ferr<strong>on</strong>i correcti<strong>on</strong> was also applied to adjust for multiple comparis<strong>on</strong>s across<br />

the different models.<br />

Statistically significant associati<strong>on</strong>s (p


P31<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

FINE-MAPPING OF THREE MAMMOGRAPHIC DENSITY LOCI USING NEXT-<br />

GENERATION SEQUENCING<br />

Sara Lindström 1,2 , Brad Chapman 3 , C<strong>on</strong>stance Chen 1,2 , Rory Kirchner 3 , Stacey Gabriel 4 , Oliver<br />

Hofmann 2 , Rulla Tamimi 2,5 , Peter Kraft 1,2,6<br />

1<br />

Program in Molecular <strong>and</strong> Genetic Epidemiology, Harvard School of Public Health, Bost<strong>on</strong>, MA; 2<br />

Department of Epidemiology, Harvard School of Public Health, Bost<strong>on</strong>, MA; 3 HSPH Bioinformatics<br />

core, Harvard School of Public Health, Bost<strong>on</strong>, MA; 4 Genetic Analysis Platform <strong>and</strong> Program in Medical<br />

<strong>and</strong> Populati<strong>on</strong> Genetics, The Broad Institute of MIT <strong>and</strong> Harvard, Cambridge, MA; 5 Channing Divisi<strong>on</strong><br />

of Network Medicine, Harvard Medical School, Bost<strong>on</strong>, MA; 6 Department of Biostatistics, Harvard<br />

School of Public Health, Bost<strong>on</strong>, MA<br />

Introducti<strong>on</strong>: Recent studies have shown that single nucleotide polymorphisms (SNPs) in the breast<br />

cancer susceptibility regi<strong>on</strong>s 6q25.1 (ESR1), 10q22 (ZNF365) <strong>and</strong> 14q24 (RAD51L1) are also associated<br />

with percent mammographic density, a str<strong>on</strong>g risk factor for breast cancer. We used next-generati<strong>on</strong><br />

sequencing to characterize these regi<strong>on</strong>s <strong>and</strong> localize c<strong>and</strong>idate causal variants.<br />

Methods: As a part of a large initiative to sequence known breast cancer susceptibility regi<strong>on</strong>s, we<br />

obtained high-coverage sequence data for 598 breast cancer cases <strong>and</strong> 560 c<strong>on</strong>trols from the Nurse’s<br />

Health Study for whom we also had data <strong>on</strong> percent mammographic density. Across all three regi<strong>on</strong>s, we<br />

sequenced a total of 1.93Mb <strong>and</strong> captured 80% of the targeted regi<strong>on</strong> at a sequencing depth >20x. For<br />

each regi<strong>on</strong>, we c<strong>on</strong>ducted c<strong>on</strong>diti<strong>on</strong>al analysis as well as burden tests of rare variants to detect<br />

independent signals. We also used an approximate Bayesian approach to calculate the smallest number of<br />

SNPs needed to obtain a set (PPS) such that there is 90% probability that the causal SNP is included.<br />

Results: We identified a total of 10,274 single nucleotide variants across all three regi<strong>on</strong>s <strong>and</strong> of those,<br />

2,571 had a minor allele frequency (MAF) > 0.005. C<strong>on</strong>diti<strong>on</strong>al analysis <strong>on</strong> the original index SNPs<br />

revealed a suggestive independent signal in the ZNF365 regi<strong>on</strong> (P=0.0004). However, we were not able to<br />

narrow down the number of potentially causal SNPs. Burden tests did not reveal any excess of rare<br />

variants associated with mammographic density (P ESR1 =0.14, P ZNF365 =0.08, P RAD51L1 =0.66).<br />

C<strong>on</strong>clusi<strong>on</strong>s: We did not find any unambiguous evidence for potential causal variants in these three<br />

genetic regi<strong>on</strong>s associated with mammographic density. This study illustrates the challenges facing finemapping<br />

studies in the post-GWAS era.<br />

ABSTRACTS<br />

66


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P32 APPLICATION OF SHAPE AND APPEARANCE MODEL APPROACH TO<br />

MAMMOGRAMS<br />

Serghei Malkov 1 , Karla Kerlikowske 2 John A. Shepherd 1<br />

1 Department of Radiology <strong>and</strong> Biomedical Imaging, University of California, San Francisco, 1 Irving<br />

Street, Suite AC108, San Francisco, CA 94143-0628 ; 2 Departments of Medicine <strong>and</strong><br />

Epidemiology/Biostatistics, University of California, San Francisco, 1635 Divisadero Street, Suite 600,<br />

San Francisco, CA 94115<br />

Abstract: The extracti<strong>on</strong> of new informati<strong>on</strong> from clinical breast mammograms could help to improve<br />

breast cancer risk assessment techniques. The image shape <strong>and</strong> texture estimati<strong>on</strong> (appearance) designed<br />

for face recogniti<strong>on</strong> seems to be promising approach for applicati<strong>on</strong> in breast imaging.<br />

Purpose: The purpose of this study is to apply a shape <strong>and</strong> appearance model approach to digital<br />

mammograms. Potentially, the breast shape <strong>and</strong> appearance model parameters could give new informati<strong>on</strong><br />

valuable for breast density estimati<strong>on</strong>, breast cancer risk assessment <strong>and</strong> diagnostics.<br />

ABSTRACTS<br />

Materials <strong>and</strong> Methods: We built a shape <strong>and</strong> appearance model using 200 full field digital<br />

mammograms with calculated percent fibrogl<strong>and</strong>ular volumes (%FGV) from San Francisco<br />

Mammography Registry. The model approach was implemented according to the method of Cootes et al<br />

[1]. To build the shape model, we used the edge <strong>and</strong> grid point coordinates inside of the breast area. A<br />

Procrustes transformati<strong>on</strong> was used to remove rotati<strong>on</strong>, translati<strong>on</strong> <strong>and</strong> scale. A Principal Comp<strong>on</strong>ent<br />

Analysis (PCA) was applied to extract significant <strong>and</strong> uncorrelated comp<strong>on</strong>ents. In order to build an<br />

appearance model, first, we transformed the breast images into the mean texture image by piecewise<br />

linear image transformati<strong>on</strong>. That step aligned all texture informati<strong>on</strong> inside the reference mean image.<br />

Then, the image pixels grey-scale values are c<strong>on</strong>verted into a set of significant principal comp<strong>on</strong>ent<br />

vectors. Finally, the shape <strong>and</strong> texture feature sets of each image are created using principal comp<strong>on</strong>ent<br />

vectors. In additi<strong>on</strong>, to remove shape <strong>and</strong> texture correlati<strong>on</strong>s another PCA transform was applied to get a<br />

combined shape-appearance model. The stepwise regressi<strong>on</strong> with forward selecti<strong>on</strong> <strong>and</strong> backward<br />

eliminati<strong>on</strong> was used to estimate the outcome %FGV with shape <strong>and</strong> appearance features <strong>and</strong> other<br />

parameters including age, bmi, thickness, mAs <strong>and</strong> force.<br />

Results: We obtained the maps <strong>and</strong> pixel distributi<strong>on</strong>s of different shape <strong>and</strong> appearance PCA<br />

comp<strong>on</strong>ents. The shape <strong>and</strong> appearance scores are found to correlate moderately to breast %FGV, dense<br />

tissue <strong>and</strong> actual volumes, bmi <strong>and</strong> age. The highest Pears<strong>on</strong> correlati<strong>on</strong> coefficient is equal 0.76 for the<br />

first shape PCA comp<strong>on</strong>ent <strong>and</strong> actual breast volume. The stepwise regressi<strong>on</strong> of the outcome %FGV<br />

from shape <strong>and</strong> appearance variables, thickness, age, mAs <strong>and</strong> force, met the 0.01 significance level for<br />

entry into the model during forward selecti<strong>on</strong>, dem<strong>on</strong>strated R-square around 0.8. Eleven significant<br />

variables were selected by regressi<strong>on</strong> am<strong>on</strong>g 185 parameters.<br />

C<strong>on</strong>clusi<strong>on</strong>: We applied the shape <strong>and</strong> appearance model approach to mammograms <strong>and</strong> dem<strong>on</strong>strated its<br />

feasibility to extract a new set of variables. Further exploring <strong>and</strong> testing of this approach is necessary.<br />

References:<br />

T.F. Cootes, G.J Edwards, <strong>and</strong> C,J. Taylor "Active Appearance Models", IEEE Transacti<strong>on</strong>s <strong>on</strong> Pattern<br />

Analysis <strong>and</strong> Machine Intelligence 2001<br />

67


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P33<br />

METHODS TO ASSESS AND REPRESENT MAMMOGRAPHIC DENSITY: AN<br />

ANALYSIS OF FOUR CASE-CONTROL STUDIES<br />

Maskarinec G 1 , Woolcott CG 2 , C<strong>on</strong>roy SM 3 , Nagata C 4 , Ursin G 5 , Vach<strong>on</strong> CM 6 , Yaffe MJ 7 , Byrne C 8<br />

1 University of Hawaii Cancer Center, H<strong>on</strong>olulu, HI ; 2 Dalhousie University, Halifax, Nova Scotia, Canada ;<br />

3 Alberta Health Services-Cancer Care, Calgary, Canada ; 4 Gifu University Graduate School of Medicine,<br />

Gifu, Japan ; 5 University of Oslo, Oslo, Norway ; 6 Mayo Clinic, Rochester, Minnesota ; 7 Sunnybrook<br />

Research Institute, Tor<strong>on</strong>to, Ontario, Canada ; 8 Uniformed Services University, Bethesda, MD<br />

To achieve greater statistical power in studies of mammographic densities <strong>and</strong> breast cancer risk, it is<br />

often desirable to combine data from multiple studies. This is not straightforward due to variati<strong>on</strong> in<br />

mammographic density assessment arising from different techniques <strong>and</strong> observers. Using data from a<br />

pooled data set of 4 case-c<strong>on</strong>trol studies, we compared the measures of mammographic densities made by<br />

the original observers <strong>and</strong> readings by a single observer who re-assessed all images. In additi<strong>on</strong>, we<br />

identified predictors of differences in density readings <strong>and</strong> evaluated the strength of the associati<strong>on</strong><br />

between mammographic density <strong>and</strong> breast cancer risk using the original percent density values, the reassessed<br />

readings, <strong>and</strong> original values calibrated based <strong>on</strong> their relati<strong>on</strong>s with re-assessed measurements<br />

<strong>on</strong> a subset of mammograms.<br />

The pooled data set included 1699 cases <strong>and</strong> 2422 c<strong>on</strong>trols from California, Hawaii, Minnesota, <strong>and</strong><br />

Japan. One reader re-assessed all mammographic images for density using Cumulus software. To calibrate<br />

the original readings using reassessed values, we performed linear regressi<strong>on</strong> <strong>on</strong> 100 r<strong>and</strong>omly selected<br />

mammograms for each locati<strong>on</strong> <strong>and</strong> applied the regressi<strong>on</strong> equati<strong>on</strong> to all of the original density<br />

measurements from that locati<strong>on</strong>. The associati<strong>on</strong> between mammographic density <strong>and</strong> breast cancer risk<br />

was represented by odds ratios (OR) estimated from unc<strong>on</strong>diti<strong>on</strong>al logistic regressi<strong>on</strong> models <strong>and</strong><br />

adjusted for relevant covariates. Different representati<strong>on</strong>s of density values were used in these analyses:<br />

the approximate tertiles in the combined c<strong>on</strong>trol group (35%); 5 predefined categories<br />

(


P34<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

INTERNATIONAL POOLING PROJECT OF MAMMOGRAPHIC DENSITY<br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Anya Burt<strong>on</strong> 1 , Joachim Schuz 1 , Isabelle Romieu 1 , Norman Boyd 3 , Isabel dos Santos Silva 1, 2 , Valerie<br />

McCormack 1<br />

1 <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer, Ly<strong>on</strong>, France; 2 L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical<br />

Medicine, L<strong>on</strong>d<strong>on</strong>, Engl<strong>and</strong>; 3 Ontario Cancer Institute / Princess Margaret Hospital, Tor<strong>on</strong>to, Canada<br />

Background: The <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Pooling Project of Mammographic Density is a worldwide collaborative<br />

project which aims to:<br />

(i) pool <strong>and</strong> obtain st<strong>and</strong>ardised comparable data <strong>on</strong> mammographic density (MD) from countries<br />

spanning the full breast cancer (BC) incidence range<br />

(ii) describe internati<strong>on</strong>al variati<strong>on</strong>s in overall <strong>and</strong> age-specific distributi<strong>on</strong>s of MD<br />

(iii) assess whether variati<strong>on</strong>s in MD are explained by individual-level risk factors for this marker<br />

(iv) quantify the extent to which internati<strong>on</strong>al variati<strong>on</strong>s in MD correlate with corresp<strong>on</strong>ding BC<br />

incidence rates <strong>and</strong> Pike’s model of breast tissue ageing.<br />

ABSTRACTS<br />

Methods: The project will pool original data from previously c<strong>on</strong>ducted studies worldwide. Eligible<br />

studies are those based <strong>on</strong> general populati<strong>on</strong> asymptomatic women with original analogue (ideally<br />

digitized) or digital films <strong>and</strong> breast cancer risk factor informati<strong>on</strong> for each woman. To increase betweenpopulati<strong>on</strong><br />

heterogeneity, studies spanning low to high BC risk populati<strong>on</strong>s (e.g. multi-ethnic studies)<br />

will be especially informative. From each populati<strong>on</strong>, data <strong>and</strong> an<strong>on</strong>ymized films from 200 premenopausal<br />

(ages 35-49 years) <strong>and</strong> 200 post-menopausal women (50-70 years) will be requested <strong>and</strong> sent<br />

to the study coordinators at the <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer (IARC). All films will be<br />

re-read using Cumulus or other validated methods, so that st<strong>and</strong>ardised comparable MD is obtained<br />

across studies. A validati<strong>on</strong> study of MD readings from digitised analogue, raw digital <strong>and</strong> processed<br />

digital images is being c<strong>on</strong>ducted to inform whether informati<strong>on</strong> from these images can be combined.<br />

Progress to date: Currently, 20 studies from 6 c<strong>on</strong>tinents have agreed to participate in the project,<br />

including North America, Northern <strong>and</strong> Southern Europe, South Africa, Turkey, India, Saudi Arabia,<br />

Egypt, Singapore, Brazil <strong>and</strong> Mexico. Further low risk populati<strong>on</strong>s would enhance the scientific value of<br />

the project. IARC is liaising with participating studies to put agreements in place, an<strong>on</strong>ymize images <strong>and</strong><br />

data prior to their transfers.<br />

What we hope to achieve: This internati<strong>on</strong>al perspective will investigate to what extent MD is an<br />

intermediate marker of BC risk at the populati<strong>on</strong>-level. It will examine factors that account for betweenpopulati<strong>on</strong><br />

differences in MD, i.e. target factors that could be modified to reduce MD <strong>and</strong> BC risk.<br />

Greater exposure variability will provide greater statistical power to clarify their influence <strong>on</strong> MD. We<br />

will assess age-specific variati<strong>on</strong>s in MD at ages 35-70 years, identifying critical exposure windows. The<br />

debate over which MD measure (absolute/relative, recent/cumulative) is the best indicator of BC risk will<br />

be addressed, an important issue for pers<strong>on</strong>alised risk assessment incorporating MD.<br />

Funding: Office of Research <strong>on</strong> Women’s Health (ORWH), Nati<strong>on</strong>al Cancer Institute 1R03CA167771-01.<br />

69


P35<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

AN APPROACH TO THE STANDARDIZATION OF PERCENT MAMMOGRAPHIC<br />

DENSITY TO ENABLE JOINT ANALYSIS OF DATA ACROSS MULTIPLE CENTERS<br />

V. Shane Pankratz 1 , Christopher G. Scott 1 , Matthew R. Jensen 1 , Kimberly A. Bertr<strong>and</strong> 2,3 , Rulla M.<br />

Tamimi 2,3 , Karla Kerlikowske 4 , Celine M. Vach<strong>on</strong> 5<br />

1 Department of Health Sciences Research, Divisi<strong>on</strong> of Biomedical Statistics <strong>and</strong> Informatics, Mayo Clinic<br />

College of Medicine, Rochester, MN; 2 Channing Divisi<strong>on</strong> of Network Medicine, Department of Medicine,<br />

Brigham <strong>and</strong> Women’s Hospital <strong>and</strong> Harvard Medical School, Bost<strong>on</strong>, MA; 3 Department of<br />

Epidemiology, Harvard School of Public Health, Bost<strong>on</strong>, MA; 4 Departments of Epidemiology <strong>and</strong><br />

Biostatistics <strong>and</strong> General Internal Medicine Secti<strong>on</strong>, Department of Veterans Affairs, University of<br />

California, San Francisco, CA; 5 Department of Health Sciences Research, Divisi<strong>on</strong> of Epidemiology,<br />

Mayo Clinic College of Medicine, Rochester, MN<br />

Background: Percent mammographic density (MD) is <strong>on</strong>e of the str<strong>on</strong>gest risk factors for breast cancer<br />

(BC). Because MD is often measured by a computer-assisted approach which requires human operators to<br />

set thresholds, MD measures may differ when they are assessed by different readers, or even within a<br />

single reader when measurements for a cohort are separated by l<strong>on</strong>g periods of time. This r<strong>and</strong>om<br />

variability in MD measurements across readers <strong>and</strong> “batches” introduces measurement error in<br />

epidemiologic analyses <strong>and</strong> may bias associati<strong>on</strong>s of interest. In order to integrate MD data obtained from<br />

multiple readers across several reading epochs into a single pooled analysis, we developed an approach<br />

that st<strong>and</strong>ardizes measures of MD across multiple readers <strong>and</strong> batches.<br />

Methods: Data were obtained from six cohort or case-c<strong>on</strong>trol studies, which were comprised of 3414<br />

women with breast cancer <strong>and</strong> 7199 without. All underwent screening mammography <strong>and</strong> MD was<br />

assessed from digitized film-screen mammograms using a computer-assisted threshold technique by three<br />

different readers <strong>and</strong> across varying time frames. We developed a st<strong>and</strong>ardizati<strong>on</strong> approach that relies <strong>on</strong><br />

the age-dependent trends in MD. In this approach, we regress a transformed versi<strong>on</strong> of MD <strong>on</strong> age at<br />

mammogram within each discrete set of MD reads, <strong>and</strong> extract the residuals from these regressi<strong>on</strong><br />

models. We re-center <strong>and</strong> re-scale these sets of residuals separately, such that their medians <strong>and</strong><br />

interquartile ranges were all equal to values estimated from a large set of MD reads obtained within <strong>on</strong>e<br />

reading epoch within a single cohort of individuals. We then add these st<strong>and</strong>ardized residuals to an age<br />

trend that we estimated from all currently available data. We applied this st<strong>and</strong>ardizati<strong>on</strong> approach to the<br />

data from these six studies <strong>and</strong> evaluated its effect <strong>on</strong> estimates of heterogeneity am<strong>on</strong>g the studies.<br />

Results: Prior to st<strong>and</strong>ardizati<strong>on</strong>, differences in the MD reads am<strong>on</strong>g the six studies explained 13.6% of<br />

the variability in percent MD, <strong>and</strong> we c<strong>on</strong>sistently observed evidence of significant heterogeneity am<strong>on</strong>g<br />

studies when analyzing associati<strong>on</strong>s between MD <strong>and</strong> various outcomes of interest, including breast<br />

cancer. We applied our st<strong>and</strong>ardizati<strong>on</strong> approach to the data from these studies to re-center <strong>and</strong> re-scale<br />

their percent MD measurements. The effort resulted in distributi<strong>on</strong>s of percent MD that were more similar<br />

across studies, while the rank ordering of within-study measurements was preserved. After<br />

st<strong>and</strong>ardizati<strong>on</strong>, differences am<strong>on</strong>g studies explained 1.4% of the variability in MD, <strong>and</strong> there was no<br />

l<strong>on</strong>ger evidence of significant heterogeneity am<strong>on</strong>g study sets in combined analyses of associati<strong>on</strong>s<br />

between percent MD <strong>and</strong> breast cancer status.<br />

ABSTRACTS<br />

C<strong>on</strong>clusi<strong>on</strong>: We implemented a st<strong>and</strong>ardizati<strong>on</strong> approach for MD that makes it possible to appropriately<br />

combine MD measurements obtained across multiple readers. This approach may also be used to align<br />

MD measurements obtained from studies other than those explicitly included in these analyses.<br />

70


P36<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BREAST DEVELOPMENT AT DIFFERENT BREAST TANNER STAGES: EVIDENCE<br />

FROM THE GROWTH OBESITY CHILEAN COHORT STUDY<br />

Ana Pereira 1 , John Shepherd 2 , María Luisa Garmendia 1 , Ricardo Uauy 1 , Camila Corvalan 1<br />

1 Institute of Nutriti<strong>on</strong> <strong>and</strong> Food Technology, University of Chile; 2 Department of Radiology <strong>and</strong><br />

Biomedical Imaging, University of California at San Francisco<br />

Background: Underst<strong>and</strong>ing breast development <strong>and</strong> compositi<strong>on</strong> at young ages is crucial to further<br />

investigate early determinants of breast cancer risk. A previous study carried out using Dual X-Ray<br />

Absorptiometry(DXA) in adolescents Hawaiian girls showed that absolute fibrogl<strong>and</strong>ular volume<br />

(AFGV) increased mostly until Tanner B4, while between B4 <strong>and</strong> B5 the most important increase was in<br />

breast volume (BV) rather than AFGV, thus % fibrogl<strong>and</strong>ular volume (%FGV) decreased.<br />

Objective: To describe breast compositi<strong>on</strong> (%FGV, AFGV, BV) at different Tanner stages in Chilean<br />

Girls <strong>and</strong> as a by-product to evaluate whether B4-B5 breast compositi<strong>on</strong> changes vary by menarche or<br />

adiposity.<br />

ABSTRACTS<br />

Methods: This cross-secti<strong>on</strong>al study is part of <strong>on</strong>going birth cohort (2002-2003) of 1196 Chilean<br />

children (50% female). Biannual anthropometric measurements <strong>and</strong> breast Tanner assessment are being<br />

carried out by trained pers<strong>on</strong>nel. <strong>Breast</strong> compositi<strong>on</strong> was assessed using DXA in a r<strong>and</strong>omly selected subsample<br />

of girls in Tanner B1 (n=6), B2 (n=7) <strong>and</strong> B3 (N=6, <strong>and</strong> in all girls that reached B4 (n=55) <strong>and</strong> B5<br />

(n=13) during 2012. <strong>Breast</strong> compositi<strong>on</strong> data was calculated based <strong>on</strong> a two-compartment model of<br />

adipose <strong>and</strong> fibro-gl<strong>and</strong>ular tissue using a software developed by University of California at San<br />

Francisco; test-retest precisi<strong>on</strong> was 3.5%. DXA scans of each breast were analyzed separately (although<br />

correlati<strong>on</strong> was taking into account) according to their tanner stage. We used descriptive statistics to<br />

describe progressi<strong>on</strong> of breast compositi<strong>on</strong> across all breast tanner stages, <strong>and</strong> lineal multivariate analysis<br />

to assess if changes between B4 <strong>and</strong> B5 vary by menarche <strong>and</strong> body mass index<br />

(BMI=weight(kg)/height(mt) 2 ).<br />

Results preliminary: Descriptive results are shown in Table 1. In summary, we observed that mean age,<br />

%FGV, AFGV, <strong>and</strong> BV increased across breast tanner stages. BV, AFGV <strong>and</strong> %FGV increased by 47%,<br />

68% <strong>and</strong> 11% from B4 to B5 respectively. In our linear regressi<strong>on</strong> models, we observed that changes<br />

between B4 <strong>and</strong> B5 disappeared or diminished after adjusting by menarche status; there were not<br />

statistical differences in %FGV <strong>and</strong> BV <strong>and</strong> the regressi<strong>on</strong> coefficient of AFGV diminished by half, but it<br />

remained significantly (β=28.3, 95%CI:4.7; 51.9). Further, adjusting by BMI did not modify the<br />

regressi<strong>on</strong> coefficient between B4 <strong>and</strong> B5; however BMI was inversely associated with %FGV <strong>and</strong><br />

positively with BV.<br />

Discussi<strong>on</strong>: We observed lower levels of AFGV <strong>and</strong> %FGV (all Tanner) <strong>and</strong> BV (B4 <strong>and</strong> B5) compared<br />

to previous studies. AFGV, % FGV, BV increased across all breast Tanner stages including B5; however<br />

our results suggest that measurements at time of menarche will be more critical to assess breast<br />

development. Follow-up of the remaining subjects in the cohort will help to establish the c<strong>on</strong>sistency of<br />

these findings.<br />

Table 1: Descriptive analysis of age <strong>and</strong> breast compositi<strong>on</strong> data stratified by breast Tanner stage<br />

<strong>Breast</strong> Tanner Stage N Age (mean, (sd)) %FGV (%) AFGV (cm3) BV (cm3)<br />

B1 15 121 (3.0) 30.1 22.5 77.6<br />

B2 12 118 (3.6) 22.5 26.5 125.4<br />

B3 12 124 (5.0) 36.5 80.1 251.8<br />

B4 111 124 (4.0) 39.9 89.2 237.9<br />

B5 26 127 (5.0) 44.4 150.3 348.6<br />

Funding Sources: F<strong>on</strong>decyt 3130532 , F<strong>on</strong>decyt 1020326 <strong>and</strong> WCRF 2010/245<br />

71


P37<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

DUAL X-RAY ABSORPTIOMETRY AN ALTERNATIVE METHOD TO MEASURE<br />

VOLUMETRIC BREAST DENSITY<br />

Pereira A 1 , Shepherd J 2 , Garmendia ML 1 , Galleguillos B 1 , Ralph Highnam 3, Uauy R 1 , Corvalán C 1 .<br />

1 Institute of Nutriti<strong>on</strong> <strong>and</strong> Food Technology, University of Chile; 2 Department of Radiology <strong>and</strong><br />

Biomedical Imaging, University of California at San Francisco; 3 Mātakina Technology Limited,<br />

Wellingt<strong>on</strong>, New Zeal<strong>and</strong><br />

Background: Mammographic breast density (BD) is a str<strong>on</strong>g risk factor for breast cancer; however its<br />

use in adolescent populati<strong>on</strong>s is limited due to radiati<strong>on</strong> exposure. Recently, applicati<strong>on</strong>s to measure BD<br />

have been developed for Dual X-Ray Absorptiometry (DXA) systems as a low-dose alternative method. A<br />

previous validati<strong>on</strong> study carried out in ~100 adult women showed a high correlati<strong>on</strong> (r=0.70) between<br />

volumetric breast density (%FGV) measured by DXA <strong>and</strong> area based mammographic BD. Since 2010, we<br />

have followed the mothers of 550 girls participating in the Growth <strong>and</strong> Obesity Chilean Cohort Study to<br />

assess nutriti<strong>on</strong>al determinants of increased BD. In these women, both mammograms <strong>and</strong> DXA scans of<br />

the breasts were obtained. Our aims were i) to determine the precisi<strong>on</strong> of %FGV measured by DXA, <strong>and</strong><br />

ii) to assess the equivalency of %FGV, absolute fibrogl<strong>and</strong>ular volume (AFGV), <strong>and</strong> breast volume (BV)<br />

measures made using DXA <strong>and</strong> mammograms.<br />

Methods: In this cross-secti<strong>on</strong>al study, 126 premenopausal women received both screening digital<br />

mammograms (Hologic Selenia, Bedford, MA, USA) <strong>and</strong> breast DXA scans (GE iDXA, GE healthcare,<br />

Madis<strong>on</strong>, WI). Mammographic “raw” images were used to measure volumetric BD using a fullyautomated<br />

commercial method (Volpara®, Matakina Technology, Wellingt<strong>on</strong>, New Zeal<strong>and</strong>). For the<br />

DXA scans, a left-right-left breast scanning protocol was used. The DXA analysis software was<br />

developed by University of California at San Francisco. The end result was measures of %FGV, AFGV,<br />

<strong>and</strong> BV by both mammography (Mx) <strong>and</strong> DXA. In additi<strong>on</strong>, all women underwent anthropometric<br />

measures (body mass index (BMI)=weight(kg)/height(mt) 2 ) <strong>and</strong> answered a sociodemographic<br />

questi<strong>on</strong>naire. We estimated intraclass correlati<strong>on</strong> coefficient (ICC) <strong>and</strong> the coefficient of variati<strong>on</strong><br />

(precisi<strong>on</strong>) of DXA after repositi<strong>on</strong>ing, c<strong>on</strong>sidering obesity (BMI>30), age (over 36 years) <strong>and</strong> BV (less<br />

than 654 cm 3 ). Using linear regressi<strong>on</strong> models we estimated the correlati<strong>on</strong> coefficient <strong>and</strong> R 2 between<br />

volumetric BD measured with DXA (left image) <strong>and</strong> with Mx (MLO left image).<br />

Results: The mean age was 37.1 years (sd=6.5) <strong>and</strong> 31.6% were obese. Mean time between<br />

mammographic <strong>and</strong> DXA measurements was 9 m<strong>on</strong>ths (sd=5.6). The ICCs for %FGV, AFGV <strong>and</strong> BV<br />

after repositi<strong>on</strong> the left breast were over 0.9 <strong>and</strong> the precisi<strong>on</strong> for %FGV, 3.7%; however the latter<br />

declined to 5% in obese women <strong>and</strong> women with breast volumes less than 654 cm 3 . Mean values for<br />

DXA <strong>and</strong> Mx are shown in Table 1. The correlati<strong>on</strong> coefficients for %FGV, AFGV <strong>and</strong> BV between DXA<br />

<strong>and</strong> Mx were all over 0.8 <strong>and</strong> the R 2 were 0.6, 0.7 <strong>and</strong> 0.8, respectively. The R 2 of %FGV increased to 0.8<br />

if the time between both exams was less than six m<strong>on</strong>ths.<br />

ABSTRACTS<br />

Discussi<strong>on</strong>: DXA is a precise method to measure volumetric BD <strong>and</strong> it correlates better to volumetric<br />

mammography measurements compared to area based models. These results suggest that DXA is a<br />

reliable method to measure volumetric BD that can be used to c<strong>on</strong>tinuously m<strong>on</strong>itor BD in adult women<br />

in follow-up studies<br />

Table 1: Mean <strong>and</strong> st<strong>and</strong>ard deviati<strong>on</strong>(sd) of breast compositi<strong>on</strong> using DXA <strong>and</strong> Mx<br />

<strong>Breast</strong> Compositi<strong>on</strong> Data DXA (mean(sd)) Mx (mean(sd))<br />

%FGV(%) 30.9(10.5) 10.2(5.2)<br />

AFGV(cm 3 ) 214.4(98.8) 67.6(34.7)<br />

BV(cm 3 ) 732.8(315.8 767.9(415.6)<br />

Funding sources: F<strong>on</strong>decyt 3130532 <strong>and</strong> 11100238, WCRF 2010/245 , Ellis<strong>on</strong> Medical Foundati<strong>on</strong> Grant to Ana Pereira<br />

72


P38<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

AUTOMATED DENSITY RISK SCORING WITH LEARNED HIERARCHICAL<br />

FEATURES<br />

K Petersen 1 , M Lillholm 2 , P Diao 2 , J Marques 2 , N Karssemeijer 3 , M Nielsen 1,2<br />

1 University of Copenhagen, Dept. of Comp. Science; 2 Biomediq A/S; 3 Radboud University, Nijmegen<br />

Medical Centre<br />

Purpose: Radiodense fibrogl<strong>and</strong>ular tissue is an established risk factor for breast cancer. Density<br />

estimati<strong>on</strong> is comm<strong>on</strong>ly performed using the interactive threshold software Cumulus, which measures the<br />

percental area of dense breast tissue in a mammogram. However, for large studies, this method is too<br />

laborious <strong>and</strong> expensive. We propose a novel machine learning based method called multiscale<br />

c<strong>on</strong>voluti<strong>on</strong>al neural network (MSCNN) for scoring dense breast tissue.<br />

ABSTRACTS<br />

Material <strong>and</strong> Methods: The density scores were evaluated <strong>on</strong> digitized film-based mammograms from<br />

the Dutch screening program (age 58 ± 5.7 years), c<strong>on</strong>sisting of the left <strong>and</strong> right mediolateral oblique<br />

views (LMLO <strong>and</strong> RMLO) of 495 women (123 interval cancers, 122 screen-detected cancers, <strong>and</strong> 250<br />

c<strong>on</strong>trols). The mammograms were taken 2-4 years prior to diagnosis of interval cancers <strong>and</strong> 4 years prior<br />

to screen-detected cancer. The MSCNN was trained <strong>on</strong> labeled RMLO views, for which density labels<br />

from expert Cumulus scoring were available. Testing was performed <strong>on</strong> both LMLO <strong>and</strong> RMLO views,<br />

i.e., 990 mammograms in total. To score all views, two-fold cross-validati<strong>on</strong> was employed. MSCNNs are<br />

multiscale multi-layer networks for learning a deep hierarchy of discriminative features. These features<br />

are learned directly from raw intensity values of the mammograms, <strong>and</strong> used for discriminating dense<br />

from n<strong>on</strong>-dense tissues using a multinomial logistic regressi<strong>on</strong>. MSCNNs allow to efficiently learn<br />

features in a data-driven fashi<strong>on</strong> by sharing parameters <strong>and</strong> modeling the topology of images. They<br />

circumvent the modeling <strong>and</strong> selecti<strong>on</strong> of h<strong>and</strong>crafted density features.<br />

Results: MSCNN scores for RMLO (R1) <strong>and</strong> LMLO (L1) were compared to the expert's Cumulus scores<br />

<strong>on</strong> RMLO (R2) by evaluating the area under the ROC curve (AUC), the p-value of a ranksum test, <strong>and</strong><br />

Pears<strong>on</strong>'s R to R2. R1 performed very similar to R2 in separating interval cancers from c<strong>on</strong>trols (R1:<br />

AUC=0.60, p=5.3e-3 / R2: AUC=0.60, p=2.6e-3) <strong>and</strong> in discriminating all cancers from c<strong>on</strong>trols (R1:<br />

AUC=0.56, p=4.1e-2 / R2: AUC=0.56, p=2.7e-2). The separati<strong>on</strong> of screen cancers was n<strong>on</strong>-significant<br />

for R1 <strong>and</strong> R2. Even L1 (which was trained <strong>on</strong> RMLO <strong>on</strong>ly) performed comparable for interval cancers<br />

(L1: AUC=0.58, p=1.9e-2). For screen cancers <strong>and</strong> all cancers the discriminati<strong>on</strong> based <strong>on</strong> L1 was n<strong>on</strong>significant.<br />

The correlati<strong>on</strong> between all cancers <strong>and</strong> c<strong>on</strong>trol scores was R=0.79 for R1 vs. R2 <strong>and</strong> R=0.75<br />

for L1 vs. R2.<br />

C<strong>on</strong>clusi<strong>on</strong>: Automated MSCNN density scores based <strong>on</strong> RMLO views (R1) performed very similar to<br />

expert's scores (R2) in predicting breast cancer <strong>and</strong> showed a str<strong>on</strong>g correlati<strong>on</strong> to R2. MSCNN scores for<br />

LMLO views were well correlated with R2, <strong>and</strong> were predictive for interval cancers.<br />

73


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P39 ASSURE - ADAPTING BREAST CANCER SCREENING STRATEGY USING<br />

PERSONALISED RISK ESTIMATION<br />

Bram Platel, Ritse Mann, Nico Karssemeijer<br />

Radboud University Nijmegen Medical Centre, The Netherl<strong>and</strong>s<br />

ASSURE c<strong>on</strong>sists of 10 project partners from 7 countries with leading expertise in the field of breast<br />

imaging, with the Radboud University Nijmegen Medical Centre as the coordinating partner. The project<br />

started in December 2012 <strong>and</strong> is supported by the European Commissi<strong>on</strong> under the 7th Framework<br />

Programme.<br />

Approximately 1 in 8 women develop breast cancer during their lifetime. Screening programs have been<br />

introduced, decreasing the mortality rate <strong>and</strong> allowing for less radical treatment opti<strong>on</strong>s for early detected<br />

cancers. Unfortunately not all cancers are detected in screening. Approximately 30% of breast cancers are<br />

detected between screening rounds. This c<strong>on</strong>stitutes a need for improved cancer screening.<br />

The ASSURE project promotes going from a <strong>on</strong>e-size-fits-all approach to a pers<strong>on</strong>alised screening<br />

protocol. Nowadays all women undergo the same screening protocol, independent of the density of their<br />

breast tissue <strong>and</strong> the risk to develop breast cancer. Almost all women make use of the same diagnostic<br />

modality, X-ray mammography.<br />

Unfortunately the sensitivity of mammographic screening is seriously impaired in women with dense<br />

breasts. Fibrogl<strong>and</strong>ular <strong>and</strong> stromal tissue look equally bright as tumours <strong>on</strong> mammography images, this<br />

causes tumours to remain masked for radiologists <strong>and</strong> thus breast cancer to remain undetected.<br />

One task of the project is to estimate pers<strong>on</strong>al risk depending <strong>on</strong> breast density, age, gene mutati<strong>on</strong>s,<br />

family <strong>and</strong>/or pers<strong>on</strong>al history, etc., <strong>and</strong> based <strong>on</strong> this risk, propose an optimal, cost effective,<br />

pers<strong>on</strong>alized screening strategy.<br />

Another task is to improve the use of MRI <strong>and</strong> Automatic <strong>Breast</strong> UltraSound (ABUS) as additi<strong>on</strong>al breast<br />

cancer screening modalities, by optimizing protocols, enhance reading workflow, <strong>and</strong> by offering better<br />

diagnostic performance through software soluti<strong>on</strong>s.<br />

Expected Results & Impact<br />

Effective breast cancer screening substantially reduces mortality <strong>and</strong> maintains the quality of life of<br />

women. Additi<strong>on</strong>ally, less disfiguring treatments are required. A new stratificati<strong>on</strong> protocol <strong>and</strong> new<br />

screening tools will result from this project. The screening process will be optimised in resp<strong>on</strong>se to the<br />

increased awareness regarding the low sensitivity of current breast cancer screening in women with dense<br />

breasts. Our estimati<strong>on</strong>s show that a reducti<strong>on</strong> of interval cancers of at least 30% is achievable, assuming<br />

the efforts in this project lead to a sensitivity for dense breast comparable to that of n<strong>on</strong>-dense breasts.<br />

Furthermore, the risk stratificati<strong>on</strong> enabled by this project will benefit a very low risk group (10-15% of<br />

women) as their screening interval can be further optimized (<strong>and</strong> increased) to limit the adverse effects<br />

due to the radiati<strong>on</strong> inherent with X-ray mammography screening.<br />

ABSTRACTS<br />

The ASSURE project will be an excellent opportunity for the participating SMEs (small <strong>and</strong> mediumsized<br />

enterprises) to join forces with leading research instituti<strong>on</strong>s <strong>and</strong> nati<strong>on</strong>al screening experts.<br />

Exploitable results from this project are expected to include (1) new <strong>and</strong> validated measures for breast<br />

density, (2) a quality assurance system for breast cancer screening with ABUS <strong>and</strong> MRI, (3) new<br />

sequences for breast cancer screening with MRI <strong>and</strong> (4) dedicated screening software for high-throughput<br />

ABUS <strong>and</strong> MRI reading.<br />

74


P40<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

BREAST DENSITY INFORM: NEW YORK STATE AND FEDERAL INITIATIVES<br />

J o A n n P u s h k i n; CoFounder, D.E.N.S.E.; Founder, D.E.N.S.E. NY; (Density Educati<strong>on</strong> Nati<strong>on</strong>al<br />

Survivors' Effort)<br />

Introducti<strong>on</strong>: <strong>Breast</strong> density compromises the effectiveness of a mammogram <strong>and</strong> is recognized as an<br />

independent risk factor for breast cancer ii . These facts are especially alarming as 40% iii of women of<br />

mammography age have dense breasts <strong>and</strong> 95% iv of women have no knowledge of their own breast<br />

density. However, as recently as four years ago breast density disclosure was not required through state<br />

or federal law. Based <strong>on</strong> the ultrasound “find rate” in C<strong>on</strong>necticut, the first state to pass a density inform<br />

law in 2009, it is estimated that each year at least 2,500 New York State women with dense breasts<br />

receive “normal/negative” mammogram results - but actually have invasive breast cancer that would be<br />

found by ultrasound screening.<br />

Purpose: To provide NYS women informati<strong>on</strong> about their own breast density, I sought the introducti<strong>on</strong> of<br />

the NYS <strong>Breast</strong> Density Inform bill <strong>and</strong> cofounded D.E.N.S.E. (TM) (Density Educati<strong>on</strong> Nati<strong>on</strong>al<br />

Survivors’ Effort) to support legislati<strong>on</strong>.<br />

ABSTRACTS<br />

Method: Advocacy included: initiating bill introducti<strong>on</strong>, providing input <strong>on</strong> bill language, resp<strong>on</strong>ding to<br />

oppositi<strong>on</strong> c<strong>on</strong>cerns, building a coaliti<strong>on</strong> of radiological experts, advocates <strong>and</strong> supportive organizati<strong>on</strong>s,<br />

meeting with Governor Cuomo’s health counsel, organizing a capital Lobby Day <strong>and</strong> reaching out to NYS<br />

legislature.<br />

Result: On July 23, 2012, New York’s <strong>Breast</strong> Density Inform Bill was signed into law by Governor<br />

Cuomo having received unanimous bi-partisan support. NY became the first state to unambiguously tell<br />

women if they have dense breasts. NYS law requires a woman with dense tissue (>50% density) be<br />

informed:<br />

1. Her mammogram shows that her breast tissue is dense<br />

2. Dense tissue can compromise a mammogram<br />

3. Dense tissue may be associated with an increased risk of breast cancer<br />

4. To discuss the value of additi<strong>on</strong>al screening with her doctor<br />

As a result, New York women are now aware about their breast density <strong>and</strong> its associated risks.<br />

Multi-state Status: Six states v have m<strong>and</strong>atory <strong>Breast</strong> Density Inform Laws, each with a different density<br />

notificati<strong>on</strong>. Further almost half the country has density legislati<strong>on</strong> pending. As a result, women living in<br />

neighboring states may get very different density notificati<strong>on</strong>s.<br />

Federal Soluti<strong>on</strong>: A nati<strong>on</strong>al st<strong>and</strong>ardized <strong>Breast</strong> Density Inform notificati<strong>on</strong> is necessary to rectify this<br />

growing inequity. For this reas<strong>on</strong> an amendment to the Federal Mammography Quality St<strong>and</strong>ards Act to<br />

require the inclusi<strong>on</strong> of breast density in the letter women receive after their mammogram, <strong>and</strong> a Federal<br />

<strong>Breast</strong> Density Inform Bill have been initiated, the latter of which has 45 co-sp<strong>on</strong>sors <strong>and</strong> is scheduled for<br />

a 2013 re-introducti<strong>on</strong> to the House.<br />

Through intensive advocacy work <strong>and</strong> with bi-partisan support it is possible to enact breast density inform<br />

legislati<strong>on</strong> <strong>and</strong> make breast density awareness a nati<strong>on</strong>al priority.<br />

75


P41<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

CIRCULATING INSULIN-LIKE GROWTH FACTOR-1, INSULIN-LIKE GROWTH<br />

FACTOR BINDING PROTEIN-3, GENETIC POLYMORPHISMS, AND<br />

MAMMOGRAPHIC DENSITY IN PREMENOPAUSAL MEXICAN WOMEN: RESULTS<br />

FROM THE ESMAESTRAS COHORT<br />

Sabina Rinaldi 1 , Carine Biessy 1 , M Hern<strong>and</strong>ez 1 , F Lesueur 1,3 , Isabel dos Santos Silva 4 , Megan S Rice 5,6 ,<br />

Martin Lajous 2 , RuyLopez-Ridaura 2 , Gabriela Torres-Mejia 2 , Isabelle Romieu 1<br />

1 <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer (IARC), Ly<strong>on</strong>, France; 2 Instituto Naci<strong>on</strong>al de Salud<br />

Publica, Cuernavaca, Mexico; 3 INSERM, U900, Institut Curie, Paris, France; 4 L<strong>on</strong>d<strong>on</strong> School of<br />

Hygiene & Tropical Medicine, L<strong>on</strong>d<strong>on</strong>, United Kingdom; 5 Department of Epidemiology, Harvard School<br />

of Public Health, Bost<strong>on</strong>, MA, USA; 6 Channing Divisi<strong>on</strong> of Network Medicine, Brigham & Women’s<br />

Hospital, Bost<strong>on</strong>, MA, USA<br />

Background: The insulin-like growth factor (IGF) axis plays an essential role in the development of the<br />

mammary gl<strong>and</strong>. Higher circulating levels of IGF-I have been associated with an increased risk of breast<br />

cancer. However, results from studies of IGF-1 <strong>and</strong> mammographic density have been inc<strong>on</strong>sistent.<br />

Methods: We c<strong>on</strong>ducted a cross-secti<strong>on</strong>al analysis am<strong>on</strong>g 593 premenopausal Mexican women in the<br />

ESMaestras cohort study to examine the associati<strong>on</strong>s between serum IGF-I, IGFBP-3, <strong>and</strong> the IGF-<br />

1/IGFBP-3 ratio <strong>and</strong> mammographic density. Our initial models adjusted for age at mammogram, IGF-<br />

1/IGFBP-3 batch, state (Jalisco/Veracruz), family history of breast cancer (no, yes), history of benign<br />

breast disease (no, yes), age at menarche (


P42<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

ADJUSTING FOR INTER-OBSERVER VARIATION IN VISUAL ASSESSMENT OF<br />

PERCENTAGE BREAST DENSITY ON A CONTINUOUS SCALE AND DETERMINING<br />

WOMEN AT HIGH RISK OF DEVELOPING BREAST CANCER RISK<br />

Jamie C. Sergeant 1 , Matthew Sperrin 2 , Lawrence Bardwell 3 , Iain Buchan 2 , D. Gareth Evans 4 , Anth<strong>on</strong>y<br />

Howell 4 , Susan M. Astley 1<br />

1 Centre for Imaging Sciences, Institute for Populati<strong>on</strong> Health, University of Manchester, Manchester, UK;<br />

2 Centre for Health Informatics, Institute for Populati<strong>on</strong> Health, University of Manchester, Manchester,<br />

UK;<br />

3 Department of Mathematics <strong>and</strong> Statistics, University of Lancaster, Lancaster, UK<br />

4 Nightingale Centre <strong>and</strong> Genesis Preventi<strong>on</strong> Centre, University Hospital of South Manchester,<br />

Manchester, UK<br />

ABSTRACTS<br />

Aim: Assessment of percentage breast density <strong>on</strong> a c<strong>on</strong>tinuous visual analogue scale (VAS) is a method<br />

str<strong>on</strong>gly associated with risk of developing breast cancer <strong>and</strong> readily applicable in breast screening<br />

programms due to its simplicity. However, it is subject to inter-observer variati<strong>on</strong> which may affect<br />

patient outcomes if density is used in risk stratificati<strong>on</strong>. We present a novel method of adjusting<br />

observers’ disparate VAS breast density estimates to make them directly comparable <strong>and</strong> we examine the<br />

effect of applying our method in a clinical c<strong>on</strong>text where women may be classified as being at high risk of<br />

developing breast cancer.<br />

Method: Our approach to adjusting observers’ individual estimates of breast density c<strong>on</strong>sists of two<br />

stages. Firstly, all observers are transformed <strong>on</strong>to a comm<strong>on</strong> distributi<strong>on</strong>. Sec<strong>on</strong>dly, we correct for<br />

different observers having seen different case mixes. We apply our method to a subset of data from the<br />

Predicting Risk Of Cancer At Screening (PROCAS) study in which women are classified as being at high<br />

risk of developing breast cancer if their 10-year risk, computed using the Tyrer-Cuzick risk model, is at<br />

least 8% (following UK Nati<strong>on</strong>al Institute for Health <strong>and</strong> Care Excellence (NICE) guidelines), or if their<br />

Tyrer-Cuzick 10 year risk is at least 5% <strong>and</strong> their VAS breast density places them in the top decile of<br />

densities. C<strong>on</strong>senting high risk women are informed of their status <strong>and</strong> offered a c<strong>on</strong>sultati<strong>on</strong> with a<br />

clinician to discuss their risk. In the current substudy a total of 13,694 screening cases with Full-Field<br />

Digital Mammography (FFDM) images acquired <strong>on</strong> GE Senographe Essential systems were assessed by<br />

13 experienced mammographic readers. The number of women classified as high risk before <strong>and</strong> after the<br />

applicati<strong>on</strong> of our method of adjustment was determined.<br />

Results: Of the 13,694 women, 165 (1.2%) were classified as high risk due to having a Tyrer-Cuzick 10-<br />

year breast cancer risk of at least 8%. In additi<strong>on</strong>, 1,125 (8.2%) of the women had a Tyrer-Cuzick risk of<br />

at least 5% but less than 8%, making them potentially high risk, depending <strong>on</strong> their VAS density. Before<br />

adjustment of densities, 126 (11.2%) of the 1,125 women were classified as high risk, which increased to<br />

147 (13.1%) after the densities had been adjusted. After adjustment, 35 women had been reclassified from<br />

n<strong>on</strong>-high risk to high risk (3.5% of those initially classified as n<strong>on</strong>-high risk) <strong>and</strong> 14 women had been<br />

reclassified from high risk to n<strong>on</strong>-high risk (11.1% of those initially classified as high risk).<br />

C<strong>on</strong>clusi<strong>on</strong>: We have presented a novel adjustment of VAS breast density estimates to account for interobserver<br />

variati<strong>on</strong>. Applying our method <strong>on</strong> data from the PROCAS study had a substantial impact <strong>on</strong><br />

identifying high risk women based <strong>on</strong> their Tyrer-Cuzick 10-year risk estimate <strong>and</strong> breast density. The<br />

reclassificati<strong>on</strong> of women as high or n<strong>on</strong>-high risk after adjustment highlights the need to c<strong>on</strong>sider<br />

correcting for inter-observer variati<strong>on</strong> if VAS breast density assessment is used in assessing breast cancer<br />

risk for the purposes of stratificati<strong>on</strong> <strong>and</strong> pers<strong>on</strong>alized screening.<br />

77


P43<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

VISUAL ANALOGUE SCALE ASSESSMENT OF PERCENTAGE BREAST DENSITY: A<br />

STUDY OF AGREEMENT BETWEEN OBSERVERS AND WITH INTERACTIVE<br />

THRESHOLDING<br />

Jamie C. Sergeant 1 , Mary Wils<strong>on</strong> 2 , Nicky Barr 2 , Ursula Beetles 2 , Caroline Boggis 2 , Sara Bundred 2 , Megan<br />

Bydder 2 , Soujanya Gadde 2 , Emma Hurley 2 , Anil Jain 2 , Yit Lim 2 , Liz Lord 2 , Valerie Reece 2 , D. Gareth<br />

Evans 2 , Anth<strong>on</strong>y Howell 2 , Susan M. Astley 1<br />

1 Centre for Imaging Sciences, Institute for Populati<strong>on</strong> Health, University of Manchester, Manchester, UK:<br />

2 Nightingale Centre <strong>and</strong> Genesis Preventi<strong>on</strong> Centre, University Hospital of South Manchester,<br />

Manchester, UK<br />

Aim: The measurement of percentage breast density <strong>on</strong> a visual analogue scale (VAS) has been shown to<br />

have an associati<strong>on</strong> with breast cancer risk comparable to that of interactive thresholding software such as<br />

Cumulus (Sunnybrook Health Sciences Centre, Tor<strong>on</strong>to, Canada). While interactive thresholding is too<br />

labour intensive to implement in a breast screening programme for the purposes of prospective risk<br />

estimati<strong>on</strong>, the quick <strong>and</strong> simple VAS method is more well-suited to this applicati<strong>on</strong>. However, for VAS<br />

density measurement to be satisfactorily used for this purpose, there must be comparability between<br />

estimates produced by different observers. We examine the agreement between a group of observers<br />

assessing VAS breast density <strong>and</strong> their agreement with Cumulus.<br />

Method: 120 screening cases were sampled from the Predicting Risk Of Cancer At Screening<br />

(PROCAS) study, a large clinical study based in Manchester, UK, which aims to predict the risk of breast<br />

cancer when women attend routine breast screening. The cases all had Full-Field Digital Mammography<br />

(FFDM) images acquired <strong>on</strong> GE Senographe Essential equipment. The breast density of the cases was<br />

independently assessed by 12 experienced mammographic readers using a separate VAS for each<br />

radiographic projecti<strong>on</strong> (craniocaudal <strong>and</strong> mediolateral oblique) of both breasts. In additi<strong>on</strong>, a trained <strong>and</strong><br />

validated Cumulus user also estimated the density for each image. Assessment of inter-observer<br />

agreement <strong>and</strong> agreement with Cumulus was performed using the Bl<strong>and</strong>-Altman limits of agreement<br />

approach <strong>and</strong> the c<strong>on</strong>cordance correlati<strong>on</strong> coefficient (CCC). All density estimates were also discretised<br />

to the four BI-RADS breast compositi<strong>on</strong> categories <strong>and</strong> agreement <strong>on</strong> this ordinal scale measured with<br />

Cohen’s weighted kappa.<br />

Results: The maximum observed difference between estimates by two readers assessing the same case<br />

was 67.75 percentage points, while the mean difference between pairs of readers ranged from 0.76 to<br />

28.58 percentage points <strong>and</strong> the mean difference from Cumulus ranged from 0.38 to 22.98 percentage<br />

points. 95% limits of agreement between reader pairs, between which 95% of differences are expected to<br />

lie, were (-6.96, 18.62) at their narrowest <strong>and</strong> (-59.13, 1.97) at their widest, while between readers <strong>and</strong><br />

Cumulus they ranged from (-32.55, -3.29) to (-49.71, 3.94). Pairwise CCC values ranged from 0.44 to<br />

0.92, while the overall CCC across the 12 readers was 0.70. C<strong>on</strong>cordance with Cumulus ranged from 0.48<br />

to 0.87. Pairwise kappa values for the BI-RADS classificati<strong>on</strong> ranged from 0.37 to 0.84 for the 12 readers,<br />

with a mean of 0.66. Kappa values between the readers <strong>and</strong> Cumulus ranged from 0.40 to 0.80.<br />

ABSTRACTS<br />

C<strong>on</strong>clusi<strong>on</strong>: A lack of agreement was found between the readers visually assessing breast density <strong>and</strong> a<br />

comparable lack of agreement was also found between the readers <strong>and</strong> Cumulus. This group of readers<br />

had varying levels of experience of VAS breast density assessment, but were all routinely performing<br />

estimati<strong>on</strong> in the PROCAS study. The current study suggests that for VAS density to be used for risk<br />

stratificati<strong>on</strong> <strong>and</strong> pers<strong>on</strong>alised screening, inter-observer variati<strong>on</strong> must be accounted for, either through<br />

st<strong>and</strong>ardised training or by applying a correcti<strong>on</strong> to results after reading has been performed.<br />

78


P44<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

LARGE INTERNATIONAL STUDY REVEALS NOVEL ASSOCIATIONS BETWEEN<br />

COMMON BREAST CANCER SUSCEPTIBILITY VARIANTS AND RISK-<br />

PREDICTING MAMMOGRAPHIC DENSITY MEASURES<br />

Jennifer St<strong>on</strong>e 1 , Deborah Thomps<strong>on</strong> 2 , Isabel dos Santos Silva 3 , Christopher Scott 4 , Melissa Southey 5 ,<br />

Laura Baglietto 6 , Fergus Couch 4 , Sara Lindström 7,8 , Jingmei Li 9 , Gretchen Gierach 10 , Julie A Douglas 11 ,<br />

Javier Benitez 12 , Peter Fasching 13 , Graham Giles 6 , Marina Pollan 12 , J<strong>on</strong>ine Figueroa 10 , Kristen Stevens 4 ,<br />

Norman F. Boyd 14 , Per Hall 9 , Douglas F. East<strong>on</strong> 2 , Rulla M. Tamimi 7,8,15 , John L. Hopper 1 <strong>and</strong> Celine<br />

Vach<strong>on</strong> 4 for the DENSNP <strong>and</strong> Markers of Density (MODE) c<strong>on</strong>sortia<br />

ABSTRACTS<br />

1 Centre for Molecular, Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytic Epidemiology, School of Populati<strong>on</strong> Health,<br />

The University of Melbourne, Melbourne, Australia; 2 Centre for Genetic Epidemiology, Department of<br />

Public Health <strong>and</strong> Primary Care, University of Cambridge, Cambridge, UK; 3 Faculty of Epidemiology<br />

<strong>and</strong> Populati<strong>on</strong> Health, L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical Medicine, L<strong>on</strong>d<strong>on</strong>, UK; 4 Department of<br />

Health Sciences Research, Mayo Clinic, Rochester, MN, USA; 5 Genetic Epidemiology Laboratory<br />

(GEL), Department of Pathology, The University of Melbourne, Melbourne, Australia; 6 Cancer<br />

Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia; 7 Program in Molecular <strong>and</strong><br />

Genetic Epidemiology, 8 Department of Epidemiology , Harvard School Of Public Health, Bost<strong>on</strong>, MA,<br />

USA; 9 Department of Medical Epidemiology <strong>and</strong> Biostatistics, Karolinska Institutet, Stockholm, Sweden;<br />

10<br />

Horm<strong>on</strong>al <strong>and</strong> Reproductive Epidemiology Branch, Nati<strong>on</strong>al Cancer Institute, Rockville, MD ; 11<br />

Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI; 12 Centro<br />

Naci<strong>on</strong>al de Investigaci<strong>on</strong>es Oncologicas, Madrid, Spain; 13 Erlangen University Perinatal Center,<br />

Department of Gynaecology <strong>and</strong> Obstetrics, Erlangen University Hospital, Friedrich Alex<strong>and</strong>er<br />

University of Erlangen-Nuremberg, Erlangen, Germany; 14 Campbell Family Institute for <strong>Breast</strong> Cancer<br />

Research, Ontario Cancer Institute, Tor<strong>on</strong>to, Ontario, Canada; 15 Channing Divisi<strong>on</strong> of Network<br />

Medicine, Brigham <strong>and</strong> Women’s Hospital, Bost<strong>on</strong>, MA<br />

Background: Mammographic density adjusted for age <strong>and</strong> body mass index (BMI) is a heritable<br />

predictor of breast cancer risk but the underlying biological processes are unclear. To date, 77 comm<strong>on</strong><br />

variants have been found to be associated with breast cancer susceptibility.<br />

Methods: We examined the associati<strong>on</strong>s between these variants <strong>and</strong> mammographic density measures<br />

(absolute dense area, percent dense area <strong>and</strong> absolute n<strong>on</strong>-dense area) adjusted for study, age <strong>and</strong> BMI<br />

using mixed linear models using 10,727 women from the internati<strong>on</strong>al DENSNP <strong>and</strong> Markers Of DEnsity<br />

(MODE) c<strong>on</strong>sortia.<br />

Results: We found str<strong>on</strong>g support for previously reported associati<strong>on</strong>s of rs10995190 (in the regi<strong>on</strong> of<br />

ZNF365), rs2046210 (ESR1) <strong>and</strong> rs3817198 (LSP1) with adjusted absolute <strong>and</strong> percent dense areas (all p<br />


P45<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

META-ANALYSIS OF TWELVE GENOME-WIDE ASSOCIATION STUDIES (GWAS)<br />

IDENTIFIES NOVEL GENETIC LOCI ASSOCIATED WITH MAMMOGRAPHIC<br />

DENSITY PHENOTYPES<br />

Sara Lindström 1,2 , Deborah Thomps<strong>on</strong> 3 , Andrew D. Paters<strong>on</strong> 4 , Jingmei Li 5 , Gretchen L. Gierach 6 , Jennifer<br />

St<strong>on</strong>e 7 , Julie A. Douglas 8 , Isabel dos Santos Silva 9 , Javier Benitez 10 , Christopher Scott 11 , Peter A. Fasching 12 ,<br />

Laura Baglietto 13 , Melissa Southey 7 , Graham Giles 13 , Marina Pollan 10 , J<strong>on</strong>ine Figueroa 6 , Fergus Couch 11 , John<br />

L. Hopper 7 , Per Hall 5 , Douglas F. East<strong>on</strong> 3 , Norman F. Boyd 14 , Celine M. Vach<strong>on</strong> 11† , <strong>and</strong> Rulla M. Tamimi 1,2,15†<br />

†<br />

for the Markers of Density (MODE) c<strong>on</strong>sortium Co-Last Authors<br />

1 Program in Molecular <strong>and</strong> Genetic Epidemiology; 2 Department of Epidemiology , Harvard School Of Public<br />

Health, Bost<strong>on</strong>, MA, USA; 3 Centre for Genetic Epidemiology, Department of Public Health <strong>and</strong> Primary Care,<br />

University of Cambridge, Cambridge, UK; 4 Program in Genetics <strong>and</strong> Genome Biology, The Hospital for Sick<br />

Children, Tor<strong>on</strong>to, Ontario, Canada; 5 Department of Medical Epidemiology <strong>and</strong> Biostatistics, Karolinska<br />

Institutet, Stockholm, Sweden; 6 Horm<strong>on</strong>al <strong>and</strong> Reproductive Epidemiology Branch, Nati<strong>on</strong>al Cancer Institute,<br />

Rockville, MD ; 7 Centre for Molecular, Envir<strong>on</strong>mental, Genetic <strong>and</strong> Analytic Epidemiology, School of<br />

Populati<strong>on</strong> Health, The University of Melbourne, Melbourne, Australia; 8 Department of Human Genetics,<br />

University of Michigan Medical School, Ann Arbor, MI; 9 Faculty of Epidemiology <strong>and</strong> Populati<strong>on</strong> Health,<br />

L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical Medicine, L<strong>on</strong>d<strong>on</strong>, UK; 10 Centro Naci<strong>on</strong>al de Investigaci<strong>on</strong>es<br />

Oncologicas, Madrid, Spain; 11 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA;<br />

12 Erlangen University Perinatal Center, Department of Gynaecology <strong>and</strong> Obstetrics, Erlangen University<br />

Hospital, Friedrich Alex<strong>and</strong>er University of Erlangen-Nuremberg, Erlangen, Germany;<br />

13 Cancer<br />

Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia; 14 Campbell Family Institute for <strong>Breast</strong><br />

Cancer Research, Ontario Cancer Institute, Tor<strong>on</strong>to, Ontario, Canada; 15 Channing Divisi<strong>on</strong> of Network<br />

Medicine, Brigham <strong>and</strong> Women’s Hospital, Bost<strong>on</strong>, MA<br />

Mammographic density is <strong>on</strong>e of the str<strong>on</strong>gest risk factors for breast cancer <strong>and</strong> has been shown to have a<br />

heritable comp<strong>on</strong>ent that remains poorly understood. In a previous meta-analysis of five genome-wide<br />

associati<strong>on</strong> study (GWAS) of mammographic density in 4,887 women within the MODE c<strong>on</strong>sortium, we<br />

identified comm<strong>on</strong> variants in the breast cancer susceptibility locus ZNF365 associated with percent<br />

mammographic density. We have now extended our meta-analysis to include seven additi<strong>on</strong>al GWAS of<br />

percent mammographic density as well as report results from the first GWAS of dense area <strong>and</strong> n<strong>on</strong>-dense<br />

area. We c<strong>on</strong>ducted a meta-analysis of 12 GWAS of percent mammographic density, dense area, <strong>and</strong> n<strong>on</strong>-dense<br />

area <strong>on</strong> a mammogram in almost 8,000 women of European descent. Ten of the studies used linear regressi<strong>on</strong><br />

treating the mammographic density phenotype as a quantitative trait adjusting for age, body mass index (BMI)<br />

<strong>and</strong> menopausal status. Two studies selected women based <strong>on</strong> the extreme categories of mammographic<br />

density (<strong>on</strong>e using percent mammographic density <strong>and</strong> the other dense area) adjusted for age, BMI <strong>and</strong><br />

menopausal status. The differences in study design (extreme sampling vs. c<strong>on</strong>tinuous trait) did not allow us to<br />

perform meta-analysis based <strong>on</strong> the estimated effect size in each study as units of density measurement were<br />

not comparable across studies. Therefore, we performed a combined test for each SNP by combining p-values<br />

<strong>and</strong> the directi<strong>on</strong> of associati<strong>on</strong> for each study, weighted by the square-root of the sample size <strong>and</strong> the study<br />

specific inflati<strong>on</strong> factor. We c<strong>on</strong>ducted replicati<strong>on</strong> of SNPs with P


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P46 NON-INVASIVE OPTICAL ASSESSMENT OF BREAST DENSITY AND<br />

IDENTIFICATION OF HIGH-RISK SUBJECTS<br />

Paola Tar<strong>on</strong>i, 1 * Giovanna Quarto, 1 Ant<strong>on</strong>io Pifferi, 1,2 Lorenzo Spinelli, 2 Aless<strong>and</strong>ro Torricelli, 1 Francesca<br />

Ieva, 3 Anna Maria Pagan<strong>on</strong>i, 3 Francesca Abbate, 4 Nicola Balestreri, 5 Sim<strong>on</strong>a Menna, 4 Enrico Cassano, 4 <strong>and</strong><br />

Rinaldo Cubeddu 1,2<br />

1 Dipartimento di Fisica, Politecnico di Milano, Piazza Le<strong>on</strong>ardo da Vinci 32, 20133 Milano, Italy; 2 Istituto di<br />

Fot<strong>on</strong>ica e Nanotecnologie, C<strong>on</strong>siglio Nazi<strong>on</strong>ale delle Ricerche, Piazza Le<strong>on</strong>ardo da Vinci 32, 20133 Milano,<br />

Italy; 3 Dipartimento di Matematica, Politecnico di Milano, Piazza Le<strong>on</strong>ardo da Vinci 32, 20133 Milano, Italy;<br />

4 European Institute of Oncology, <strong>Breast</strong> Imaging Unit, Via G. Ripam<strong>on</strong>ti, 435, 20141 Milano, Italy ;<br />

5 European Institute of Oncology, Department of Radiology, Via G. Ripam<strong>on</strong>ti, 435, 20141 Milano, Italy<br />

We propose the assessment of breast density by n<strong>on</strong>-invasive optical means using time domain diffuse optical<br />

spectroscopy.<br />

Optical techniques can provide functi<strong>on</strong>al <strong>and</strong> structural informati<strong>on</strong> <strong>on</strong> biological tissue in an absolutely n<strong>on</strong>invasive<br />

way, <strong>and</strong> they have already been successfully applied to the characterizati<strong>on</strong> of breast tissue in vivo.<br />

We have further exploited the potential of diffuse optical spectroscopy operating in the time domain to assess<br />

both tissue compositi<strong>on</strong> in terms of key c<strong>on</strong>stituents <strong>and</strong> scattering parameters that are related to the<br />

microscopic structure of tissue <strong>and</strong> specifically to breast density.<br />

ABSTRACTS<br />

Time domain multi-wavelength (635-1060 nm) optical mammography was performed <strong>on</strong> 147 subjects, <strong>and</strong><br />

average breast tissue compositi<strong>on</strong> (water, lipid, collagen, oxy- <strong>and</strong> deoxyhemoglobin) <strong>and</strong> scattering<br />

parameters (amplitude a <strong>and</strong> slope b) were estimated using the diffusi<strong>on</strong> approximati<strong>on</strong> to the radiative transfer<br />

theory to model phot<strong>on</strong> propagati<strong>on</strong> in tissue. Mammographic density was classified through BI-RADS<br />

categories.<br />

The dependence of optically derived tissue compositi<strong>on</strong> <strong>and</strong> scattering parameters <strong>on</strong> BI-RADS categories was<br />

investigated. All parameters show highly significant difference for BI-RADS category 2 vs 3, <strong>and</strong> 3 vs 4 (n<strong>on</strong>parametric<br />

Wilcox<strong>on</strong> test, p < 0.005), while for most parameters the difference between category 1 <strong>and</strong> 2 is not<br />

statistically significant. Specifically, increasing breast density corresp<strong>on</strong>ds to progressively increasing average<br />

amounts of water <strong>and</strong> collagen, while the lipid c<strong>on</strong>tent decreases gradually. An increase with BI-RADS<br />

category is also observed in both scattering amplitude a <strong>and</strong> slope b, in agreement with differences in<br />

microscopic structure expected between fatty <strong>and</strong> fibrogl<strong>and</strong>ular tissue. The blood parameters (i.e. total<br />

hemoglobin c<strong>on</strong>tent tHb <strong>and</strong> oxygenati<strong>on</strong> level SO 2 ) are less sensitive, with <strong>on</strong>ly tHb showing a slight increase<br />

with mammographic density.<br />

The linear correlati<strong>on</strong> between optically derived parameters was also investigated. In particular, lipid c<strong>on</strong>tent<br />

shows str<strong>on</strong>g negative correlati<strong>on</strong> with both water <strong>and</strong> collagen c<strong>on</strong>tent. Moreover, positive (negative)<br />

correlati<strong>on</strong> exists between scattering amplitude a <strong>and</strong> c<strong>on</strong>centrati<strong>on</strong>s of water <strong>and</strong> collagen (lipid). Such<br />

observati<strong>on</strong>s are c<strong>on</strong>sistent with the hypothesis that the major c<strong>on</strong>tributi<strong>on</strong> to scattering comes from<br />

fibrogl<strong>and</strong>ular tissue comp<strong>on</strong>ents.<br />

To develop a procedure for the identificati<strong>on</strong> of high-risk women, the mammographic density was<br />

dichotomized, comparing subjects in BI-RADS categories 1 to 3 to subjects in category 4. The p-values of the<br />

Wilcox<strong>on</strong> test showed that tHb, lipid, water, collagen, a e b are significantly different in the two populati<strong>on</strong>s<br />

c<strong>on</strong>sidered (at least p < 0.001), while SO 2 is not. The best regressi<strong>on</strong> logistic model for the risk probability was<br />

found via a stepwise variables selecti<strong>on</strong> minimizing the AIC (Akaike Informati<strong>on</strong> Criteri<strong>on</strong>) <strong>and</strong> resulted to<br />

depend <strong>on</strong> collagen c<strong>on</strong>tent <strong>and</strong> scattering parameters. The model provides a total misclassificati<strong>on</strong> error of<br />

12.3%, corresp<strong>on</strong>ding to a simple kappa of 0.84, which compares favorably with the reproducibility of BI-<br />

RADS measures am<strong>on</strong>g radiologists <strong>and</strong> even intra-radiologist.<br />

These results qualify the optical technique as a potential c<strong>and</strong>idate for n<strong>on</strong>-invasive, relatively simple, <strong>and</strong><br />

operator-independent assessment of breast-density related risk.<br />

81


P47<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

RETROSPECTIVE ANALYSIS OF BREAST DENSITY IN TRIPLE NEGATIVE BREAST<br />

CANCERS<br />

Gopal R Vijayaraghavan, Nancy Restighini, Rebecca Hultman, Jilliane Sotello <strong>and</strong> Srinivasan<br />

Vedantham.<br />

Dept of Radiology, UMass Medical School, 55 lake avenue north, Worcester, MA-01655.<br />

Introducti<strong>on</strong>: <strong>Breast</strong> density is generally c<strong>on</strong>sidered a surrogate marker of breast tissue proliferati<strong>on</strong>.<br />

Multiple studies have shown a 4-5 fold increased relative risk of breast cancer in women with dense<br />

breasts. Younger women tend to have denser breasts <strong>and</strong> triple negative tumors are more comm<strong>on</strong> in<br />

younger women. Triple negative breast tumors account for approximately 20% of all breast cancers <strong>and</strong><br />

are generally c<strong>on</strong>sidered more aggressive with a poorer prognosis. We retrospectively analyzed our data<br />

for any correlati<strong>on</strong> between breast density <strong>and</strong> triple negative tumors, their clinical stage <strong>and</strong> node<br />

positivity.<br />

Materials <strong>and</strong> Methods: We retrospectively analyzed mammograms of all triple negative tumors<br />

operated in our institute over a 3 year period from Jan 2009- Dec 2011. These mammograms were<br />

reviewed <strong>on</strong> a secur view hologic work stati<strong>on</strong> for breast density <strong>and</strong> categorized from 1- 4 based <strong>on</strong> ACR<br />

BI-RADS category. From the hospital medical records demographic data such as patient age, clinical<br />

stage of the tumor, histology <strong>and</strong> nodal status were also obtained <strong>and</strong> tabulated <strong>on</strong> an excel spread sheet.<br />

There were 147 triple negative tumors. Records of mammograms in 4 cases were not retrievable. <strong>Breast</strong><br />

density was tabulated in 143 cases <strong>and</strong> compared with a c<strong>on</strong>trol arm of 132 BI-RAD’s 4 <strong>and</strong> 5 cases from<br />

a different study. Nodal status was available in 128 cases. 12 patients had a significant family history of<br />

breast cancer <strong>and</strong> 3 patients had a positive BRCA test. Given small numbers correlati<strong>on</strong> to breast density<br />

in this cohort was not evaluated.<br />

Results: Of the 143 evaluable cases, there were 3 patients in BI-RADS breast density category 1, 59 in<br />

category 2, 73 in category 3 <strong>and</strong> 8 in category 4. The mean age of presentati<strong>on</strong> was 60 years, the median<br />

was 61 years <strong>and</strong> the range from 29-93 years. Ductal histology was noted in 110 cases, lobular in 28 cases<br />

<strong>and</strong> 9 cases had other histologies. 78 tumors were clinically Stage 1, 49 Stage 2, 8 Stage 3 <strong>and</strong> 2 Stage 4<br />

tumors were documented. 6 of the cases were documented as Tx. 36 patients had positive nodes at surgery<br />

<strong>and</strong> 92 patients with negative nodes in the 128 evaluable cases.<br />

Table 1: Correlati<strong>on</strong> (Spearman rho) between breast density, tumor stage <strong>and</strong> nodal status<br />

"TNC-BD" "Stage" "Node status"<br />

"TNC-BD" 1 0.24629* 0.14925<br />

"Stage" 0.24629* 1 0.46524*<br />

"Node status" 0.14925 0.46524* 1<br />

* p0.79, Mann-Whitney test)<br />

between women diagnosed with triple-negative cancers <strong>and</strong> the c<strong>on</strong>trol group (BI-RADS 4/5 subjects).<br />

Statistically significant <strong>and</strong> positive correlati<strong>on</strong>s were observed between BI-RADS breast density <strong>and</strong><br />

tumor stage, <strong>and</strong> between tumor stage <strong>and</strong> nodal involvement.<br />

C<strong>on</strong>clusi<strong>on</strong>: In our study triple negative breast cancers did not display any unique correlati<strong>on</strong> to breast<br />

density. The findings were no different from those described in literature for other breast cancers.<br />

82


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

P48<br />

A FULLY-AUTOMATED METHOD FOR QUANTIFYING FIBROGLANDULAR<br />

TISSUE AND BACKGROUND PARENCHYMAL ENHANCEMENT IN BREAST MRI<br />

Sh<strong>and</strong><strong>on</strong>g Wu, Susan P. Weinstein, Emily F. C<strong>on</strong>ant, Despina K<strong>on</strong>tos<br />

Department of Radiology, University of Pennsylvania, Philadelphia PA USA<br />

Purpose: The goal is to provide a methodological framework for automated quantitative analysis of the<br />

fibrogl<strong>and</strong>ular tissue (FGT) <strong>and</strong> background parenchymal enhancement (BPE) in breast magnetic<br />

res<strong>on</strong>ance imaging (MRI) for breast cancer risk assessment <strong>and</strong> risk reducti<strong>on</strong> interventi<strong>on</strong>s in high-risk<br />

women.<br />

Background: <strong>Breast</strong> MRI provides 3D scanning <strong>and</strong> therefore can eliminate the tissue superpositi<strong>on</strong><br />

problem in mammography. Recent studies suggest that FGT <strong>and</strong> BPE are associated with breast cancer<br />

risk assessment, specifically for high-risk women. However, currently FGT <strong>and</strong> BPE are mainly evaluated<br />

qualitatively by visual assessment, which suffers from subjective results <strong>and</strong> inter- <strong>and</strong> intra-reader<br />

variability.<br />

ABSTRACTS<br />

Method: Our fully automated method for quantifying FGT <strong>and</strong> BPE includes three steps (Fig. 1). First,<br />

an integrated scheme of synergistic edge extracti<strong>on</strong> has been developed to detect chest wall line so that<br />

segment the whole breast. Sec<strong>on</strong>d, the FGT is estimated based <strong>on</strong> our proposed fuzzy-c-means (FCM)-<br />

Atlas method. Third, BPE is estimated through identifying the enhancing voxels in the DCE subtracti<strong>on</strong><br />

(sub) images by measuring the relative intensity change relative to the intensity in the pre-c<strong>on</strong>trast (pre)<br />

images. We define an enhancement ratio R = Intensity_sub / Intensity_pre × 100 <strong>and</strong> the voxels that have<br />

a greater value than a predefined threshold are identified as the enhancing voxels. Based <strong>on</strong> the<br />

segmentati<strong>on</strong>, we compute four measures: absolute volume of FGT (|FGT|), percentage of |FGT| relative<br />

to the whole breast volume (FGT%), absolute volume of BPE (|BPE|), <strong>and</strong> percentage of |BPE| relative to<br />

the whole breast volume (BPE%). Our method has been evaluated using a representative dataset of 60<br />

bilateral breast MRI scans (120 breasts) through comparing to the manual segmentati<strong>on</strong> obtained from<br />

two experienced breast imaging radiologists by using our customized user-interactive software<br />

(“MRwizard V.1.0”).<br />

Results: The inter-reader Pears<strong>on</strong> correlati<strong>on</strong> coefficient (r) is 0.95 for FGT% <strong>and</strong> 0.96 for |FGT|. When<br />

compared to the average of the two readers, the proposed FGT segmentati<strong>on</strong> method achieves a<br />

correlati<strong>on</strong> of r=0.88 for FGT% <strong>and</strong> r=0.84 for |FGT|; the bilateral correlati<strong>on</strong> between left breasts <strong>and</strong><br />

right breasts is r=0.90 for FGT% <strong>and</strong> r=0.86 for |FGT|. For BPE-related measures, the inter-reader<br />

correlati<strong>on</strong> is r=0.80 for |BPE | <strong>and</strong> r=0.78 for BPE%, respectively. We parameterize the enhancement<br />

ratio threshold ranging from 0% to 200% <strong>and</strong> the corresp<strong>on</strong>ding BPE is estimated. It is found that there is<br />

a range, i.e., 30% - 80%, rather than a single value of the enhancement ratio that high correlati<strong>on</strong>s (r = 0.8<br />

to 0.9) are achieved between the automated BPE estimati<strong>on</strong> <strong>and</strong> the manual segmentati<strong>on</strong> in terms of the<br />

4 BPE-related measures (i.e., |BPE| <strong>and</strong> BPE% by 2 readers). Our fully automated method runs timeefficiently<br />

at ~5 minute for each 3D MR scan (56 slices), compared to ~65 minutes needed for the manual<br />

segmentati<strong>on</strong>.<br />

C<strong>on</strong>clusi<strong>on</strong>s: We have developed <strong>and</strong> validated a fully automated method for quantifying FGT <strong>and</strong> BPE<br />

in breast MRI. It has been used successfully as a quantitative imaging biomarker for assessing the<br />

resp<strong>on</strong>se of risk-reducing salpingo-oophorectomy in BRCA1/2 mutati<strong>on</strong> carriers <strong>and</strong> can also be used to<br />

support other clinical applicati<strong>on</strong>s.<br />

83


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

DCE Sub<br />

(a) (b) (c)<br />

(d) (e) (f)<br />

Figure 1. Examples illustrating the workflow of the proposed method. (a) A tomographic slice of a breast MR image<br />

with the segmented breast outlined by the red c<strong>on</strong>tour. (b) The segmnted fibrogl<strong>and</strong>ular tissue (FGT) regi<strong>on</strong>s (green)<br />

within the breast. (c) Three-dimensi<strong>on</strong>al (3D) volume of the breast (red) <strong>and</strong> the segmented FGT (green). (d) The DCE-<br />

MRI subtracti<strong>on</strong> images are used to estimate the background parenchymal ehnhancement (BPE). (e) The translated<br />

breast mask (red), the FGT (green) over the subtracti<strong>on</strong> image shown in (d) with the idenfitied BPE (purple). (f) 3D<br />

volume of the breast (red), the FGT (green), <strong>and</strong> the BPE (purple).<br />

ABSTRACTS<br />

84


2013 PARTICIPANTS<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

PARTICIPANTS<br />

First Name Last Name Affiliati<strong>on</strong> Email address<br />

Irene Acerbi UCSF irene.acerbi@ucsfmedctr.org<br />

Luana Act<strong>on</strong> Kaiser Permanente Divisi<strong>on</strong> of Research luana.act<strong>on</strong>@kp.org<br />

Carmel Apicella University of Melbourne capic@unimelb.edu.au<br />

Valentina Assi Queen Mary, University of L<strong>on</strong>d<strong>on</strong> v.assi@qmul.ac.uk<br />

Sue Astley U-Manchester sue.astley@manchester.ac.uk<br />

Alfred Au UCSF alfred.au@ucsfmedctr.org<br />

Marje Bakker<br />

Julius Center for Health Sciences <strong>and</strong><br />

Primary Care, University Medical Center<br />

Utrecht, the Netherl<strong>and</strong>s<br />

m.f.bakker-8@umcutrecht.nl<br />

Desiree Basila UCSF, SPORE Advocacy dlbasila@gmail.com<br />

Tricia Benjamin Palo Alto Medical Foundati<strong>on</strong> benjamt@pamf.org<br />

Kimberly Bertr<strong>and</strong> Harvard School of Public Health kbertran@hsph.harvard.edu<br />

Rives Bird VuCOMP, Inc. kayla.svec@vucomp.com<br />

Archie Bleyer Pediatric & Young Adult Oncology ableyer@gmail.com<br />

Norman Boyd Ontario Cancer Institute boyd@uhnres.utor<strong>on</strong>to.ca<br />

Dejana Braithwaite UCSF DBraithwaite@epi.ucsf.edu<br />

Kathy Br<strong>and</strong>t Mayo Clinic br<strong>and</strong>t.kathy@mayo.edu<br />

Adam Brentnall Queen Mary University of L<strong>on</strong>d<strong>on</strong> a.brentnall@qmul.ac.uk<br />

Allis<strong>on</strong> Bunch Sutter Health buncha@sutterhealth.org<br />

Barry Burgdorf VuCOMP, Inc. kayla.svec@vucomp.com<br />

Anya Burt<strong>on</strong><br />

<str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong><br />

Cancer<br />

burt<strong>on</strong>a@fellows.iarc.fr<br />

Sherry Butler Kaiser Permanente<br />

Celia Byrne<br />

Uniformed Services University of the<br />

Health Sciences<br />

celia.byrne@usuhs.edu<br />

Nancy Cappello Are You Dense Advocacy nancy@areyoudense.org<br />

Adrienne Castillo Kaiser Permanente-Divisi<strong>on</strong> of Research adrienne.castillo@kp.org<br />

Ariane Chan Matakina Technology e.Chan@matakina.com<br />

Biao Chen Hologic, Inc. biao.chen@hologic.com<br />

Chira Chen-Tanyolac UCSF chira.chen-tanyolac@ucsf.edu<br />

Ann Johnst<strong>on</strong> Cloud<br />

Mailman School of<br />

Columbia University<br />

Public Health,<br />

asj2117@columbia.edu<br />

Deborah Collyar UCSF & Patient Advocates In Research Deborah@tumortime.com<br />

Amy Colt<strong>on</strong> Are You Dense, Inc abcolt<strong>on</strong>@comcast.net<br />

Emily C<strong>on</strong>ant University of Pennsylvania emily.c<strong>on</strong>ant@uphs.upenn.edu<br />

Alice Couwenberg<br />

University Medical Center, Utrecht, the<br />

Netherl<strong>and</strong>s<br />

alice.couwenberg@gmail.com<br />

Margo Cusack Sutter Health cusackm@sutterhealth.org<br />

Lance Dai U-Systems Lance.Dai@ge.com<br />

Laura Damiano UCSF lauradamiano2706@gmail.com<br />

Hatef Darabi Karolinska Institutet hatef.darabi@ki.se<br />

Allen Davies U-Systems Alan.G.Davies@ge.com<br />

Shaheenah Dawood Harvard shaheenah@post.harvard.edu<br />

Judy Dean Santa Barbara Women's Imaging judy@judydeanmd.net<br />

RosaAnna DeFilippis UCSF rosa-anna.defilippis@ucsf.edu<br />

Caroline Dickens <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer dickensc@fellows.iarc.fr<br />

Isabel dos Santos Silva<br />

L<strong>on</strong>d<strong>on</strong> School of Hygiene <strong>and</strong> Tropical<br />

isabel.silva@lshtm.ac.uk<br />

Medicine<br />

Jennifer Drukteinis Moffitt Cancer Center jendruk@yahoo.com<br />

85


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

First Name Last Name Affiliati<strong>on</strong> Email address<br />

Meg Durbin Palo Alto Medical Foundati<strong>on</strong> durbinm@pamf.org<br />

Merete Ellingjord-Dale<br />

University of Oslo, Institute of Basic<br />

Medical Sciences, Dept. of Nutriti<strong>on</strong><br />

ellingjord-dale@medisin.uio.no<br />

Sarah Els<strong>on</strong> UCSF Sarah.Els<strong>on</strong>@ucsf.edu<br />

Julius Center for Health Sciences <strong>and</strong><br />

Marleen Emaus<br />

Primary Care, University Medical Center m.j.emaus@umcutrecht.nl<br />

Utrecht, the Netherl<strong>and</strong>s<br />

Natalie Engmann<br />

Mailman School of Public Health, Columbia<br />

University<br />

ne2227@columbia.edu<br />

Luis Estevez UCSF lestevez@ucsf.edu<br />

Bo Fan UCSF bo.fan@ucsf.edu<br />

Peter Fasching University Hospital Erlangen, German peter.fasching@uk-erlangen.de<br />

J<strong>on</strong>athan Fish Bay Imaging Associates shmd@gmail.com<br />

Henri Folse Archimedes, Inc. henri.folse@archimedesmodel.com<br />

June F<strong>on</strong>g Sutter Health F<strong>on</strong>gjy@sutterhealth.org<br />

Jacqueline Frost Sutter Health frostjv@sutterhealth.org<br />

Barbara Fuhrman University of Arkansas for Medical Sciences bjfuhrman@uams.edu<br />

Mary Luisa Garmendia<br />

Institute of Nutriti<strong>on</strong> <strong>and</strong> Food Technology,<br />

University of Chile<br />

mgarmend@gmail.com<br />

Berta Geller U-Verm<strong>on</strong>t berta.geller@uvm.edu<br />

Karthik Ghosh Mayo Clinic, Rochester, MN ghosh.karthik@mayo.edu<br />

Gretchen Gierach Nati<strong>on</strong>al Institute of Health gierachg@mail.nih.gov<br />

William Goods<strong>on</strong> Sutter Health whgjgg@sbcglobal.net<br />

Deborah Gord<strong>on</strong> UCSF gord<strong>on</strong>d@dahsm.ucsf.edu<br />

Rebecca Graff Harvard School of Public Health rgraff@mail.harvard.edu<br />

Lori Greene Sutter Health greenlx@sutterhealth.org<br />

Sarah Griffith Sutter Health GriffiSZ@sutterhealth.org<br />

Arun Guruajan VuCOMP, Inc. kayla.svec@vucomp.com<br />

Laurel Habel Divisi<strong>on</strong> of Research/Kaiser Permanente laurel.habel@kp.org<br />

Jennifer Harvey U-Virginia jah7w@virginia.edu<br />

Tyler Haskell Office of Supervisor S. Joseph Simitian Tyler.Haskell@bos.sccgov.org<br />

Ralph Highnam Matakina <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g><br />

Ralph.Highnam@VolparaDensity.co<br />

m<br />

John Hipwell UCL j.hipwell@ucl.ac.uk<br />

Solveig Hofvind Cancer registry of Norway Solveig.Hofvind@kreftregisteret.no<br />

John Hopper University of Melbourne j.hopper@unimelb.edu.au<br />

Carrie Hruska Mayo Clinic hruska.carrie@mayo.edu<br />

Scott Huntley Tractus sphuntley@aol.com<br />

Ella<br />

Huszti<br />

Campbell Family Institute for <strong>Breast</strong> Cancer<br />

Research, Ontario Cancer Institute<br />

ehuszti@uhnresearch.ca<br />

Debra Ikeda Stanford dikeda@stanford.edu<br />

Jessie Jacobs U-Systems jjacob@u-systems.com<br />

Abra Jeffers Stanford University abra@stanford.edu<br />

Judy Jobling University of Newcastle (Australia) judy.jobling@anzbctg.org<br />

B<strong>on</strong>nie Joe UCSF B<strong>on</strong>nie.Joe@ucsf.edu<br />

Ella J<strong>on</strong>es UCSF ella.j<strong>on</strong>es@ucsf.edu<br />

Leah Josephs Advocate maaravi.leah@gmail.com<br />

Katherine Jue Kaiser Permanente Katherine.F.Jue@kp.org<br />

Jane Kakkis Orange Coast <strong>Breast</strong> Center askdrjane123@yahoo.com<br />

Celia Kaplan UCSF celia.kaplan@ucsf.edu<br />

Leah Karliner UCSF leah.karliner@ucsf.edu<br />

Nico Karssemeijer Radboud University Nijmegen Medical n.karssemeijer@rad.umcn.nl<br />

PARTICIPANTS<br />

86


PARTICIPANTS<br />

Centre<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

First Name Last Name Affiliati<strong>on</strong> Email address<br />

Leila Kazemi UCSF leila.kazemi@ucsf.edu<br />

Patricia Keeley U-Wisc<strong>on</strong>sin pjkeely@facstaff.wisc.edu<br />

Mallika Keralapura U-Systems mkeralapura@u-systems.com<br />

Karla Kerlikowske UCSF karla.kerlikowske@ucsf.edu<br />

Philip Kivitz Stanford University Medical Center pghsinc@aol.com<br />

Julia<br />

Knight<br />

Samuel Lunefeld Research Institute of<br />

Mount Sinai Hospital<br />

knight@lunenfeld.ca<br />

Despina K<strong>on</strong>tos U-Pennsylvania Despina.K<strong>on</strong>tos@uphs.upenn.edu<br />

Kavitha Krishnan Cancer Epidemiology Centre kavitha.krishnan@cancervic.org.au<br />

Ashwini Kshirsagar Hologic akshirsagar@hologic.com<br />

Jiy<strong>on</strong> Lee NYU JIYON_L@hotmail.com<br />

Vivian Lee Advocate vlee@aquapartners.net<br />

Jessica Leung Sutter Health LeungJW@sutterhealth.org<br />

Martin Lillholm Biomediq A/S / University of Copenhagen mli@biomediq.com<br />

Sara Lindstrom Harvard University slindstr@hsph.harvard.edu<br />

Amir Mahmoudzadeh UCSF amirpasha.mahmoudzadeh@ucsf.edu<br />

Kunlanat Makboom<br />

Suphanburi College of Public Health,<br />

Thail<strong>and</strong><br />

kmakbo<strong>on</strong>@ucdavis.edu<br />

Serghei Malkov UCSF Serghei.Malkov@ucsf.edu<br />

Ori Maller UCSF Ori.Maller@ucsfmedctr.org<br />

Lisa<br />

Martin<br />

Campbell Family Institute for <strong>Breast</strong> Cancer<br />

Research<br />

lmartin@uhnres.utor<strong>on</strong>to.ca<br />

Gertraud Maskarinec U-Hawaii Cancer Center gertraud@cc.hawaii.edu<br />

Valerie McCormack <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer mccormackv@iarc.fr<br />

Valerie McGuire Stanford vmcguire@stanford.edu<br />

Diana Miglioretti UC-Davis dmiglioretti@ucdavis.edu<br />

James M<strong>on</strong>aco VuCOMP, Inc. kayla.svec@yahoo.com<br />

Elizabeth Morris Memorial-Sloan Kettering Cancer Center Morrise@mskcc.org<br />

Sibel Narin Hologic sibel.narin@hologic.com<br />

Kevin Nguyen University of Melbourne nguk@unimelb.edu.au<br />

Mads Nielsen University of Copenhagen madsn@diku.dk<br />

Lawrence Oliver Boca Rat<strong>on</strong> Regi<strong>on</strong>al Hospital kloliver@gmail.com<br />

Elissa Ozanne UCSF elissa.ozanne@ucsfmedctr.org<br />

Deng Pan UCSF deng.pan@ucsf.edu<br />

Shane Pankratz Mayo Clinic, Rochester MN pankratz.vern<strong>on</strong>@mayo.edu<br />

Bahram Parvin Lawrence Livermore Nati<strong>on</strong>al Laboratory parvin.bahram@gmail.com<br />

Ana<br />

Pereira<br />

University of Chile/Institute of Nutriti<strong>on</strong> <strong>and</strong><br />

Food Technology<br />

apereira@inta.uchile.cl<br />

Dan Perisho S<strong>on</strong>oCine, Inc. dperisho@s<strong>on</strong>ocine.com<br />

Kersten Petersen University of Copenhagen ten@biomediq.com<br />

Suzanne P<strong>on</strong>ik U-Wisc<strong>on</strong>sin suzp<strong>on</strong>ik@gmail.com<br />

Mark Powell Marin Women's Study mpowell@marincounty.org<br />

Lee Ann Prebil Marin County DHHS lprebil@cal.berkeley.edu<br />

Andrew Prokop UCSF <strong>and</strong>rew.prokop@ucsf.edu<br />

JoAnn Pushkin D.E.N.S.E., D.E.N.S.E. NY Dense-NY@opt<strong>on</strong>line.net<br />

Dave Reinhart S<strong>on</strong>oCine Dreinhart@s<strong>on</strong>ocine.com<br />

Richard Reitherman Orange Coast <strong>Breast</strong> Center reithermanbreastmri@yahoo.com<br />

Deborah Rhodes Mayo Clinic jsgould@charter.net<br />

Megan Rice Harvard/Brigham <strong>and</strong> Women's Hospital nhmsr@channing.harvard.edu<br />

Peter Richard Sutter Health PCRSF@YAHOO.COM<br />

Lisa Rivera-Serra U-Systems riveraserra@aol.com<br />

87


88<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Jimmy Roehrig Matakina jimmy.roehrig@pacbell.net<br />

First Name Last Name Affiliati<strong>on</strong> Email address<br />

Richard Rosene VuCOMP, Inc. kayla.svec@vucomp.com<br />

Eva Rubin M<strong>on</strong>tgomery Radiology Associates Evarubin@aol.com<br />

Daniel Rubin Stanford University dlrubin@stanford.edu<br />

B<strong>on</strong>nie Rush <strong>Breast</strong> Imaging Specialists brush4info@Aol.com<br />

Susan Sams<strong>on</strong> Advocate ssams<strong>on</strong>@pacbell.net<br />

Valerie Sankatsing Erasmus MC, The Netherl<strong>and</strong>s v.sankatsing@erasmusmc.nl<br />

Karen Sein Sutter Health seink@sutterhealth.org<br />

John Shepherd UCSF john.shepherd@ucsf.edu<br />

Ed Sickles UCSF edward.sickles@ucsfmedctr.org<br />

Weiva Sieh Stanford wsieh@stanford.edu<br />

Mahvash Sigaroudinia UCSF mahvash.sigaroudinia@ucsf.edu<br />

Joe Simitian Santa Clara County Supervisor C<strong>and</strong>ace.Joy@bos.sccgov.org<br />

Patty Smith Imaging <strong>and</strong> EEG/ECG Services patty_smith@elcaminohospital.org<br />

Brian Sprague University of Verm<strong>on</strong>t bsprague@uvm.edu<br />

Nicole Stendell-Hollis U-Minnesota nstendel@umn.edu<br />

Jennifer St<strong>on</strong>e U-Melbourne st<strong>on</strong>ej@unimelb.edu.au<br />

Natasha Stout<br />

Harvard Medical School <strong>and</strong> Harvard<br />

Pilgrim Health Care Institute<br />

natasha_stout@hms.harvard.edu<br />

Dick Tabbutt VuCOMP, Inc. kayla.svec@vucomp.com<br />

Rulla Tamimi Harvard Medical School rulla.tamimi@channing.harvard.edu<br />

Paola Tar<strong>on</strong>i Politecnico di Milano paola.tar<strong>on</strong>i@polimi.it<br />

Mary Beth Terry<br />

Mailman School of Public Health, Columbia<br />

University<br />

mt146@columbia.edu<br />

Jeff Tice UCSF jtice@medicine.ucsf.edu<br />

Amy Trentham-Dietz U-Wisc<strong>on</strong>sin trentham@wisc.edu<br />

Melissa Troester UNC School of Public Health troester@unc.edu<br />

Celine Vach<strong>on</strong> Mayo Clinic vach<strong>on</strong>.celine@mayo.edu<br />

Carla Van Gils University Medical Center Utrecht C.vangils@umcutrecht.nl<br />

Nicolien van Ravesteyn Erasmus MC, Dept. of Public Health n.vanravesteyn@erasmusmc.nl<br />

Robert Van Uitert iCAD, Inc. rvanuitert@icadmed.com<br />

JoAnn Venticinque <strong>Breast</strong> Cancer C<strong>on</strong>necti<strong>on</strong>s joan@bcc<strong>on</strong>necti<strong>on</strong>s.org<br />

Gopal Vijayaraghavan UMass Medical School gopalvijay@gmail.com<br />

Marvella Villasenor Kaiser Permanente Divisi<strong>on</strong> of Research Marvella.A.Villasenor@kp.org<br />

Marina Vor<strong>on</strong>chikhina UCSF marina.vor<strong>on</strong>chikhina@ucsf.edu<br />

Hanneke W<strong>and</strong>ers University Medical Center Utrecht j.o.p.w<strong>and</strong>ers@umcutrecht.nl<br />

Valerie Weaver UCSF valerie.weaver@ucsfmedctr.org<br />

Lani Wegrzyn Harvard School of Public Health LRW@mail.harvard.edu<br />

Jeff Wehnes VuCOMP, Inc. kayla.svec@vucomp.com<br />

Dana Whaley Mayo Clinic whaley.dana@mayo.edu<br />

Alice Whittemore Stanford University alicesw@stanford.edu<br />

Dorota Wisner UCSF dorota.wisner@ucsf.edu<br />

Anna Wu USC annawu@usc.edu<br />

Martin Yaffe Sunnybrook Research Institute, U Tor<strong>on</strong>to martin.yaffe@sri.utor<strong>on</strong>to.ca<br />

Alex<strong>and</strong>ria Y<strong>on</strong>kers Sutter Health y<strong>on</strong>kera@sutterhealth.org<br />

Wei Zhang qview medical inc. wzhang@qviewmedical.com<br />

Jianxin Zhao UCSF jianxin.zhao@ucsf.edu<br />

Elad Ziv UCSF elad.ziv@ucsf.edu<br />

Judy Jobling University of Newcastle (Australia) judy.jobling@anzbctg.org<br />

B<strong>on</strong>nie Joe UCSF B<strong>on</strong>nie.Joe@ucsf.edu<br />

Ella J<strong>on</strong>es UCSF ella.j<strong>on</strong>es@ucsf.edu<br />

Leah Josephs Advocate maaravi.leah@gmail.com<br />

PARTICIPANTS


PARTICIPANTS<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Katherine Jue Kaiser Permanente Katherine.F.Jue@kp.org<br />

First Name Last Name Affiliati<strong>on</strong> Email address<br />

Jane Kakkis Orange Coast <strong>Breast</strong> Center askdrjane123@yahoo.com<br />

Celia Kaplan UCSF celia.kaplan@ucsf.edu<br />

Leah Karliner UCSF leah.karliner@ucsf.edu<br />

Nico<br />

Karssemeijer<br />

Radboud University Nijmegen Medical<br />

Centre<br />

n.karssemeijer@rad.umcn.nl<br />

Leila Kazemi UCSF leila.kazemi@ucsf.edu<br />

Patricia Keeley U-Wisc<strong>on</strong>sin pjkeely@facstaff.wisc.edu<br />

Mallika Keralapura U-Systems mkeralapura@u-systems.com<br />

Karla Kerlikowske UCSF karla.kerlikowske@ucsf.edu<br />

Philip Kivitz Stanford University Medical Center pghsinc@aol.com<br />

Julia<br />

Knight<br />

Samuel Lunefeld Research Institute of<br />

Mount Sinai Hospital<br />

knight@lunenfeld.ca<br />

Despina K<strong>on</strong>tos U-Pennsylvania Despina.K<strong>on</strong>tos@uphs.upenn.edu<br />

Kavitha Krishnan Cancer Epidemiology Centre kavitha.krishnan@cancervic.org.au<br />

Ashwini Kshirsagar Hologic akshirsagar@hologic.com<br />

Jiy<strong>on</strong> Lee NYU JIYON_L@hotmail.com<br />

Vivian Lee Advocate vlee@aquapartners.net<br />

Jessica Leung Sutter Health LeungJW@sutterhealth.org<br />

Martin Lillholm Biomediq A/S / University of Copenhagen mli@biomediq.com<br />

Sara Lindstrom Harvard University slindstr@hsph.harvard.edu<br />

Amir Mahmoudzadeh UCSF amirpasha.mahmoudzadeh@ucsf.edu<br />

Kunlanat Makboom<br />

Suphanburi College of Public Health,<br />

Thail<strong>and</strong><br />

kmakbo<strong>on</strong>@ucdavis.edu<br />

Serghei Malkov UCSF Serghei.Malkov@ucsf.edu<br />

Ori Maller UCSF Ori.Maller@ucsfmedctr.org<br />

Lisa<br />

Martin<br />

Campbell Family Institute for <strong>Breast</strong> Cancer<br />

Research<br />

lmartin@uhnres.utor<strong>on</strong>to.ca<br />

Gertraud Maskarinec U-Hawaii Cancer Center gertraud@cc.hawaii.edu<br />

Valerie McCormack <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> Agency for Research <strong>on</strong> Cancer mccormackv@iarc.fr<br />

Diana Miglioretti UC-Davis dmiglioretti@ucdavis.edu<br />

James M<strong>on</strong>aco VuCOMP, Inc. kayla.svec@yahoo.com<br />

Elizabeth Morris Memorial-Sloan Kettering Cancer Center Morrise@mskcc.org<br />

Sibel Narin Hologic sibel.narin@hologic.com<br />

Kevin Nguyen University of Melbourne nguk@unimelb.edu.au<br />

Mads Nielsen University of Copenhagen madsn@diku.dk<br />

Lawrence Oliver Boca Rat<strong>on</strong> Regi<strong>on</strong>al Hospital kloliver@gmail.com<br />

Deng Pan UCSF deng.pan@ucsf.edu<br />

Shane Pankratz Mayo Clinic, Rochester MN pankratz.vern<strong>on</strong>@mayo.edu<br />

Ana<br />

Pereira<br />

University of Chile/Institute of Nutriti<strong>on</strong> <strong>and</strong><br />

Food Technology<br />

apereira@inta.uchile.cl<br />

Dan Perisho S<strong>on</strong>oCine, Inc. dperisho@s<strong>on</strong>ocine.com<br />

Kersten Petersen University of Copenhagen ten@biomediq.com<br />

Suzanne P<strong>on</strong>ik U-Wisc<strong>on</strong>sin suzp<strong>on</strong>ik@gmail.com<br />

Mark Powell Marin Women's Study mpowell@marincounty.org<br />

Lee Ann Prebil Marin County DHHS lprebil@cal.berkeley.edu<br />

Andrew Prokop UCSF <strong>and</strong>rew.prokop@ucsf.edu<br />

JoAnn Pushkin D.E.N.S.E., D.E.N.S.E. NY Dense-NY@opt<strong>on</strong>line.net<br />

Dave Reinhart S<strong>on</strong>oCine Dreinhart@s<strong>on</strong>ocine.com<br />

Richard Reitherman Orange Coast <strong>Breast</strong> Center reithermanbreastmri@yahoo.com<br />

Deborah Rhodes Mayo Clinic jsgould@charter.net<br />

89


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Megan Rice Harvard/Brigham <strong>and</strong> Women's Hospital nhmsr@channing.harvard.edu<br />

First Name Last Name Affiliati<strong>on</strong> Email address<br />

Peter Richard Sutter Health PCRSF@YAHOO.COM<br />

Lisa Rivera-Serra U-Systems riveraserra@aol.com<br />

Jimmy Roehrig Matakina jimmy.roehrig@pacbell.net<br />

Richard Rosene VuCOMP, Inc. kayla.svec@vucomp.com<br />

Eva Rubin M<strong>on</strong>tgomery Radiology Associates Evarubin@aol.com<br />

Daniel Rubin Stanford University dlrubin@stanford.edu<br />

B<strong>on</strong>nie Rush <strong>Breast</strong> Imaging Specialists brush4info@Aol.com<br />

Susan Sams<strong>on</strong> Advocate ssams<strong>on</strong>@pacbell.net<br />

Valerie Sankatsing Erasmus MC, The Netherl<strong>and</strong>s v.sankatsing@erasmusmc.nl<br />

Karen Sein Sutter Health seink@sutterhealth.org<br />

John Shepherd UCSF john.shepherd@ucsf.edu<br />

Ed Sickles UCSF edward.sickles@ucsfmedctr.org<br />

Weiva Sieh Stanford wsieh@stanford.edu<br />

Mahvash Sigaroudinia UCSF mahvash.sigaroudinia@ucsf.edu<br />

Joe Simitian Santa Clara County Supervisor C<strong>and</strong>ace.Joy@bos.sccgov.org<br />

Patty Smith Imaging <strong>and</strong> EEG/ECG Services patty_smith@elcaminohospital.org<br />

Brian Sprague University of Verm<strong>on</strong>t bsprague@uvm.edu<br />

Christine Stavem<br />

Deputy Chief of Staff, Supervisor S. Joseph<br />

Simitian<br />

cstavem@yahoo.com<br />

Nicole Stendell-Hollis U-Minnesota nstendel@umn.edu<br />

Jennifer St<strong>on</strong>e U-Melbourne st<strong>on</strong>ej@unimelb.edu.au<br />

Natasha Stout<br />

Harvard Medical School <strong>and</strong> Harvard<br />

Pilgrim Health Care Institute<br />

natasha_stout@hms.harvard.edu<br />

Dick Tabbutt VuCOMP, Inc. kayla.svec@vucomp.com<br />

Rulla Tamimi Harvard Medical School rulla.tamimi@channing.harvard.edu<br />

Paola Tar<strong>on</strong>i Politecnico di Milano paola.tar<strong>on</strong>i@polimi.it<br />

Mary Beth Terry<br />

Mailman School of Public Health, Columbia<br />

University<br />

mt146@columbia.edu<br />

Jeff Tice UCSF jtice@medicine.ucsf.edu<br />

Amy Trentham-Dietz U-Wisc<strong>on</strong>sin trentham@wisc.edu<br />

Melissa Troester UNC School of Public Health troester@unc.edu<br />

Celine Vach<strong>on</strong> Mayo Clinic vach<strong>on</strong>.celine@mayo.edu<br />

Carla Van Gils University Medical Center Utrecht C.vangils@umcutrecht.nl<br />

Nicolien van Ravesteyn Erasmus MC, Dept. of Public Health n.vanravesteyn@erasmusmc.nl<br />

Robert Van Uitert iCAD, Inc. rvanuitert@icadmed.com<br />

JoAnn Venticinque <strong>Breast</strong> Cancer C<strong>on</strong>necti<strong>on</strong>s joan@bcc<strong>on</strong>necti<strong>on</strong>s.org<br />

Gopal Vijayaraghavan UMass Medical School gopalvijay@gmail.com<br />

Marvella Villasenor Kaiser Permanente Divisi<strong>on</strong> of Research Marvella.A.Villasenor@kp.org<br />

Marina Vor<strong>on</strong>chikhina UCSF marina.vor<strong>on</strong>chikhina@ucsf.edu<br />

Hanneke W<strong>and</strong>ers University Medical Center Utrecht j.o.p.w<strong>and</strong>ers@umcutrecht.nl<br />

Valerie Weaver UCSF valerie.weaver@ucsfmedctr.org<br />

Lani Wegrzyn Harvard School of Public Health LRW@mail.harvard.edu<br />

Jeff Wehnes VuCOMP, Inc. kayla.svec@vucomp.com<br />

Dana Whaley Mayo Clinic whaley.dana@mayo.edu<br />

Alice Whittemore Stanford University alicesw@stanford.edu<br />

Dorota Wisner UCSF dorota.wisner@ucsf.edu<br />

Anna Wu USC annawu@usc.edu<br />

Martin Yaffe Sunnybrook Research Institute, U Tor<strong>on</strong>to martin.yaffe@sri.utor<strong>on</strong>to.ca<br />

Alex<strong>and</strong>ria Y<strong>on</strong>kers Sutter Health y<strong>on</strong>kera@sutterhealth.org<br />

Jianxin Zhao UCSF jianxin.zhao@ucsf.edu<br />

PARTICIPANTS<br />

90


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

Elad Ziv UCSF elad.ziv@ucsf.edu<br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

NOTES<br />

91


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

NOTES<br />

92


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

NOTES<br />

93


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

NOTES<br />

94


LET US KNOW WHAT YOU THINK<br />

6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

Thank you for attending the 6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong> <strong>and</strong> <strong>Breast</strong> Cancer<br />

Risk Assessment.<br />

Your input regarding our program is critical to the success of future events. Feel free to add you<br />

c<strong>on</strong>tact informati<strong>on</strong>, or to remain an<strong>on</strong>ymous.<br />

Would you please take a few moments to answer the following:<br />

1. What was your most significant learning experience<br />

2. Which presenters would you like to hear again – were the most compelling<br />

FEEDBACK<br />

3. What specific topic(s) pertaining to breast density would you like presented that were not<br />

included at this meeting What topic(s) would you like to have more informati<strong>on</strong> presented<br />

4. What did you think of the facilities<br />

5. What feedback can you give us to help us make the next c<strong>on</strong>ference an even greater success<br />

6 Will you tell your colleague(s) of the benefit from attending the meeting<br />

7. Would charging $150 registrati<strong>on</strong> fee impact your ability or desire to attend the next <strong>Breast</strong><br />

Density <str<strong>on</strong>g>Workshop</str<strong>on</strong>g><br />

95


6 th <str<strong>on</strong>g>Internati<strong>on</strong>al</str<strong>on</strong>g> <str<strong>on</strong>g>Workshop</str<strong>on</strong>g> <strong>on</strong> <strong>Breast</strong> <strong>Densitometry</strong><br />

<strong>and</strong> <strong>Breast</strong> Cancer Risk Assessment<br />

•<br />

ii Centers for Disease C<strong>on</strong>trol <strong>and</strong> Preventi<strong>on</strong> (2013), <strong>Breast</strong> Cancer Risk Factors. Retrieved from<br />

http://www.cdc.gtors.ht<br />

iii D’Orsi, CJ,Bassett LW, Berg WA et al. <strong>Breast</strong> Imaging <strong>and</strong> Reporting <strong>and</strong> Data System, BI-RADS:<br />

Mammography, 4th ed. Rest<strong>on</strong>, VA: American College of Radiology, 2003<br />

iv Harris Interactive Nati<strong>on</strong>al Survey, 5/2010<br />

v CT, TX, VA, NY, CA <strong>and</strong> HI<br />

Affix<br />

Postage<br />

Here<br />

Alice LaRocca<br />

San Francisco Coordinating Center<br />

185 Barry Street<br />

Lobby 5, Suite 5700<br />

San Francisco, CA 94107<br />

Please tear out this form, complete reverse, fold into thirds, add postage <strong>and</strong> mail. Thank you<br />

96

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!