You are on page 1of 20

REVIEWS AND COMMENTARY • REVIEW

Novel Approaches to Screening for Breast Cancer


Ritse M. Mann, MD, PhD • Regina Hooley, MD • Richard G. Barr, MD, PhD • Linda Moy, MD
From the Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Geert Grooteplein 10, PO Box 9101, 6500 HB, Nijmegen, the
Netherlands (R.M.M.); Department of Radiology, the Netherlands Cancer Institute, Amsterdam, the Netherlands (R.M.M.); Department of Radiology and Biomedical
Imaging, Yale School of Medicine, New Haven, Conn (R.H.); Department of Radiology, Northeastern Ohio Medical University, Rootstown, Ohio (R.G.B.); Southwoods
Imaging, Youngstown, Ohio (R.G.B.); Department of Radiology, New York University Langone School of Medicine, New York, NY (L.M.); and Department of Radiol-
ogy, New York University Grossman School of Medicine, Center for Advanced Imaging Innovation and Research, Laura and Isaac Perlmutter Cancer Center, New York,
NY (L.M.). Received January 24, 2020; revision requested March 16; revision received June 24; accepted June 30. Address correspondence to R.M.M. (e-mail: Ritse.
mann@radboudumc.nl).

Conflicts of interest are listed at the end of this article.

Radiology 2020; 00:1–20 • https://doi.org/10.1148/radiol.2020200172 • Content code:

Screening for breast cancer reduces breast cancer–related mortality and earlier detection facilitates less aggressive treatment.
Unfortunately, current screening modalities are imperfect, suffering from limited sensitivity and high false-positive rates. Novel
techniques in the field of breast imaging may soon play a role in breast cancer screening: digital breast tomosynthesis, contrast
material–enhanced spectral mammography, US (automated three-dimensional breast US, transmission tomography, elastography,
optoacoustic imaging), MRI (abbreviated and ultrafast, diffusion-weighted imaging), and molecular breast imaging. Artificial intel-
ligence and radiomics have the potential to further improve screening strategies. Furthermore, nonimaging-based screening tests
such as liquid biopsy and breathing tests may transform the screening landscape.
© RSNA, 2020

Online supplemental material is available for this article.

Mammography
B reast cancer is the most common cancer in women
worldwide, with approximately 2 million women diag-
nosed each year (1). It is the second leading cause of can- Digital Breast Tomosynthesis
cer-related mortality in the United States (2). The rate is Since gaining U.S. Food and Drug Administration ap-
rising at 0.4% per year, with 1 million more cases estimat- proval in 2011, digital breast tomosynthesis (DBT),
ed for diagnosis worldwide by 2040 (1). Many randomized typically combined with two-dimensional (2D) full-
controlled trials and observational studies have shown that field digital mammography (DM) is now widely used
regular screening mammography can reduce breast cancer in the screening and diagnostic mammography setting.
mortality substantially (3,4). The image stack, comprised of thin slices of breast
Beyond traditional screen-film mammography, breast tissue, decreases tissue overlap, producing a “quasi
cancer screening and diagnosis has evolved during three-dimensional” mammogram. This allows for both
the past 20 years. Full-field digital mammography re- improved sensitivity and specificity. Multiple stud-
placed analog film mammography in the early 2000s ies show that screening DBT plus DM increases the
(5), and digital breast tomosynthesis is emerging as incremental cancer detection rate, reduces the recall
the new standard of care in screening mammography rate, and can improve cancer detection rates (Table 3)
(6). Breast US also progressed beyond its initial role in (9–14).
the differentiation of cysts versus solid masses detected Although DBT may increase the cancer detection
at mammography or physical examination (7). Breast rate, some studies show most DBT-detected cancers
MRI, first thought to be useful as a problem-solving tend to be less aggressive (15,16), raising concerns re-
tool, is now the standard supplemental screening tool garding overdiagnosis (17). Currently available data do
available for both high- and intermediate-risk women not show a significant reduction in interval cancers in
(8). women screened with DBT compared with DM (18).
With ongoing, rapid advances in technology ex- However, a recent large single-institution study demon-
pected in the next decade, it is likely that breast can- strated that more invasive cancers with poor prognoses
cer screening will be very different from our current were detected with DBT at both the first and subse-
standards. The new technology introduced into clini- quent screening rounds compared with conventional
cal practice is often first evaluated in the diagnostic DM (19). Despite the abundance of published clinical
setting for ease of measuring initial cancer specificity data and widespread clinical use, it is thus still uncer-
and sensitivity. In this review, we explore state-of-the- tain whether DBT also improves short- and long-term
art, emerging techniques for breast cancer screening outcomes for patients with breast cancer compared with
in mammography, US, MRI, and nuclear medicine DM. The multicenter randomized controlled Tomo-
(Tables 1, 2); summarize the automated analysis of ra- synthesis Mammography Imaging Screening Trial and
diologic images; and introduce modern non-imaging several similar trials are ongoing and designed to answer
screening techniques. these questions (20).
This copy is for personal use only. To order printed copies, contact reprints@rsna.org
Novel Approaches to Screening for Breast Cancer

the conventional mammogram (23–25). Postprocessing the


Abbreviations low- and high-energy image removes background signal, and the
AI = artificial intelligence, BI-RADS = Breast Imaging Reporting and recombined iodine-only image helps identify enhancing lesions.
Data System, DBT = digital breast tomosynthesis, DM = digital mam-
mography, MBI = molecular breast imaging, 2D = two-dimensional Contrast-enhanced spectral mammography is safe, and serious
adverse contrast material reactions are rare (26). The radiation
Summary dose for contrast-enhanced spectral mammography is higher
This review explores state-of-the-art techniques in mammography, than that for conventional mammography (27,28) but below
US, MRI, and automation for breast cancer screening. the U.S. Food and Drug Administration mammography safety
Essentials limits (29). The lack of a contrast-enhanced spectral mammog-
n Many potential alternative screening modalities for breast cancer raphy biopsy device for lesions seen only at contrast-enhanced
are in various states of clinical development, either as stand-alone spectral mammography is a disadvantage, and prototypes are
techniques or supplemental to mammography, each with potential under evaluation.
advantages and disadvantages.
Studies show that contrast-enhanced spectral mammogra-
n Machine learning algorithms will soon play a major role in breast
cancer screening, both in improving the quality of the screening
phy outperforms combined mammography and US for lesion
programs and assisting with increasing workload due to expanding detection (Table E1 [online]) as well as for assessment of size
screening indications. and disease extent (30–33). In comparison with MRI, most
n Adopting new screening tools requires careful investigation to contrast-enhanced spectral mammography studies show only a
determine true examination-specific benefits and improvements in small reduction of sensitivity while providing a higher specificity
breast cancer morbidity and mortality.
(34,35).
n In the next decade, breast cancer screening will move beyond tra- Although contrast-enhanced spectral mammography com-
ditional tools such as mammography, US, and MRI.
bines the disadvantages of mammography (x-ray dose and com-
pression) and breast MRI (the need for intravenous contrast
The synthetic 2D mammogram, derived from the DBT data material), studies report its potential as a screening tool. Sorin
set, permits a standard DBT screening examination to have a et al (36) reported an additional cancer detection rate of 13.1
radiation dose comparable to that with DM with similar diag- per 1000 screens over conventional mammography in a series
nostic performance (21,22). The synthetic view has a different of 611 women with intermediate breast cancer risk and dense
“look” compared with DM as the algorithms enhance bright breasts. More recently, Sung et al (37) reported an additional
spots such as calcifications and linear structures (22). six cancers detected in 904 patients at contrast-enhanced spectral
Ongoing technologic advances in DBT algorithms, such as mammography compared with low-energy images, for a supple-
deep learning–based reconstruction, will provide high-spatial- mental detection rate of 6.6 per 1000. The lower additional detec-
resolution (synthetic) images with less image noise and fewer tion rate is likely explained by 75% of patients having previously
artifacts. These advances, combined with new postprocess- undergone contrast-enhanced breast MRI (37). In a previously
ing enhancements such as the generation of thick slices and/ reported subset of this cohort (n = 307), contrast-enhanced spec-
or slabs, should allow the radiologist to manipulate the im- tral mammography compared with MRI for screening showed
ages at the clinical workstation and improve DBT accuracy two of three MRI-detected cancers (in three women) at compa-
(Fig 1). Thicker slices with improved resolution could also rable specificity (94.7% vs 94.1%) and positive predictive value
potentially decrease the interpretation time of 2D plus DBT for biopsy (15% vs 14%) (38).
mammography. Contrast-enhanced spectral mammography may provide
an advantage over breast MRI—particularly for women at
Contrast Material–enhanced Spectral Mammography moderately increased risk—due to its potential easier clinical
Contrast-enhanced spectral mammography, or contrast mam- implementation and greater accessibility for screening large pop-
mography, adds functional information to the anatomic and ulations. A multicenter trial supported by the American College
morphologic information provided with conventional (stan- of Radiology, the Contrast-enhanced Mammography Imaging
dard) mammography and DBT. Like breast MRI, contrast- Screening Trial, will compare the performance of contrast-
enhanced spectral mammography depicts (neo)vasculature enhanced spectral mammography with that of DBT by screen-
associated with breast lesions and is largely independent of ing U.S. women with average-to-intermediate risk for breast
breast density. cancer (39).
Contrast-enhanced spectral mammography requires a dual-
energy mammography system (added to several current mam- US Examination
mography units) and intravenous injection of iodinated contrast US is commonly regarded as supplemental screening technique,
material (dose, 1.5 mL/kg), followed by standard craniocaudal particularly for women with dense breasts in whom mammogra-
and mediolateral mammographic views. Each image consists of phy and DBT have a lower sensitivity. A major advantage of US
a “low-energy” acquisition, resembling a normal mammogram, is that it does not use ionizing radiation. Supplemental cancer
and a “high-energy” image, obtained by using a kiloelectron detection rates of around four per 1000 screens have consistently
volt above the k-edge of iodine to enhance the contrast material been reported, and it was shown that US may decrease the in-
signal. The low-energy image is equivalent to a standard mam- terval cancer rate (40). However, US is criticized for its relatively
mogram for the detection of masses and calcifications, replacing low specificity, leading to many recalls and biopsies for benign

2 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Table 1: Novel Imaging-based Screening Techniques in Mammography and US

Technique Current Status Role in Diagnostics Role in Screening Advantages Disadvantages


Mammography
DBT Can be clinically Reduces masking Improves sensitiv- Widely available; further Small increase in radiation
used of fibroglandular ity by 40%, increases in resolution and dose; no evidence on re-
tissue improves specific- viewing enhancements duction in interval cancers
ity in screen­ing may improve specificity
programs with
high recall rates
Contrast- Can be clinically Shows enhancing Currently mainly Strong improvement of Need for intravenous canula
   enhanced spectral used lesions on experimental, sensitivity compared with and intravenous contrast
mammography recombined published series mammography (100%– material administration,
images are small; avail- 200%), only slightly below small increase in radiation
able for DBT sensitivity of breast MRI; dose
can be added to existing
mammography units
US
Microvascular Can be clinically Improves the May reduce the Available on most modern Cannot be used as a stand-
   imaging and/or used specificity of biopsy rate for US units alone technique (only
elastography B-mode US by benign lesions concurrently with B-mode
showing the US); learning curve and
stiffness of relatively high interreader
lesions variability
Automated Can be clinically Is an alternative to Detects cancer in Allows standardized imaging Artifacts may make image
  breast US used handheld whole 2–7 per 1000 of the whole breast; can be interpretation difficult, es-
breast US supplemental performed by technician pecially behind the nipple;
screens after and read in batches; theo- relatively long reading
negative mam- retically may be combined times; high false-positive
mography with US contrast agents rate
US transmission Experimental May be used for None; clinical data Potentially allows whole Requires imaging of the
  tomography only density estima- are too limited breast 3D imaging breast in a water bath for
tion and risk pre- without ionizing radia- coupling
diction; potential tion, contrast material, or
for noninvasive compression
lesion character-
ization
Targeted OA Can be clinically Improves the May reduce the Noninvasive assessment of Cannot be used as a stand-
  imaging used specificity of biopsy rate for vascularity and oxygen- alone technique (only
B-mode US by benign lesions ation of breast lesions concurrently with B-mode
showing the US), and SNR is limited;
microvasculature uses laser, for which pre-
of breast lesions, cautions are mandatory
including (goggles, signaling); gener-
oxygenation ates a lot of heat, cooling
status of the imaging room is
essential
Whole-breast Experimental Potential for None; clinical data Potentially allows whole Low SNR and long acquisi-
  OA imaging only noninvasive are too limited breast 3D imaging with- tion times
assessment out ionizing radiation or
of lesions in the contrast material; may be
breast combined with US (reflec-
tion and/or transmission)
Fusion imaging Experimental Potentially allows None; clinical data US scan and mammogram Lower resolution US image
only for spatially are too limited obtained in same compres- quality; US may miss
matched sion, and patient examina- posterior lesions
mammography tion time is decreased;
and US accurate lesion coregistra-
acquisitions tion
Note.—DBT = digital breast tomosynthesis, OA = optoacoustic, SNR = signal-to-noise ratio, 3D = three-dimensional.

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 3


Novel Approaches to Screening for Breast Cancer

Table 2: Novel Imaging-based Screening Techniques in MRI and Nuclear Medicine

Technique Current Status Role in Diagnostics Role in Screening Advantages Disadvantages


MRI
Abbreviated Can be clinically Shortens the duration May reduce the costs Retains high sensitivity of Intravenous contrast
  breast MRI used of breast MRI of breast MRI and breast MRI material administra-
acquisition and increase availability tion; may slightly
decreases reading reduce the specific-
time ity of breast MRI
Ultrafast Can be clinically Provides dynamic May enable preserva- Provides dynamic parameters Requires very short
  breast MRI used information on breast tion of sensitivity and with high discriminative acquisition times,
lesions without specificity of abbrevi- power between benign and which reduces the
increasing acquisition ated MRI malignant findings spatial resolution
time and may introduce
artifacts
DWI Can be clinically Shows hindered Currently as supple- Does not require intravenous Has lower sensitiv-
used diffusion of water mental technique to contrast material; improves ity than contrast-
molecules in tissue, improve specificity; as specificity of contrast- enhanced MRI;
which is indicative of stand-alone screening enhanced MRI; may have has limited value in
malignancy modality experimental higher sensitivity than small and diffuse
only mammography and/or lesions
DBT
Nuclear medicine
MBI Can be clinically Shows lesions with Improves the sensitivity Rapid interpretation and Long acquisition
used uptake of a radio- of mammography by detection of subcentimeter times; radiotracer
active technetium 100%–200% breast cancers tracer intravenous
tracer contrast material;
radiation dose 2–5
times higher than
that of mammog-
raphy
PET and/or Can be clinically Shows lesions with None; investigational High sensitivity, potential to Long acquisition
  PEM used uptake of fluorine only, limited clinical integrate screening with times; radiotracer
18 fluorodeoxyglucose data targeted drug therapy (ther- intravenous contrast
anostics) material; radia-
tion dose 10 times
higher than that of
mammography
Note.—DBT = digital breast tomosynthesis, DWI = diffusion-weighted imaging, MBI = molecular breast imaging, PEM = positron emis-
sion mammography.

lesions. Although studies on US have shown decreasing recall the Netherlands], AngioPLUS [SuperSonic Imaging, Aix-en-
and increasing positive predictive values of US screening (41), Provence, France], and microvascular flow imaging [MV-Flow;
many developments are still primarily aimed at increasing the Samsung, Seoul, Korea]), it is now possible to better document
specificity of US to improve its value as a screening modality. the microvascular distribution of vessels within lesions. Early
investigations show that this leads to better depiction of pen-
Microvascular Imaging etrating vessels and a clearer view of the internal microvascular
The use of color Doppler is one of the oldest techniques to in- distribution (42,43). This, in turn, can be used to improve le-
crease the specificity of US. Doppler color codes motion of re- sion classification and thus improve the specificity of US (44–
flectors by analyzing the frequency shift of the returning ultra- 46). For example, Zhu et al (46) reported an increase in area
sound signal. Consequently, Doppler US may be used to show under the curve from 0.70 to 0.85 when superb microvascular
flow in lesions. However, the spatial resolution and flow sensi- flow imaging was added to gray-scale US in the classification
tivity of standard Doppler US only allows for the visualization of Breast Imaging Reporting and Data System (BI-RADS) cat-
of large vessels, which are not typically present in breast lesions. egory 4 breast lesions.
With use of very low flow velocity scales and vendor-specific
algorithms to separate flow signals from artifacts caused by mo- Elastography
tion (eg, superb microvascular imaging [SMI, Canon Medical Elastography measures the stiffness of lesions and has been
Systems, Ota, Japan], microflow imaging [Philips, Eindhoven, clinically available for more than 15 years. Stiffness of malig-

4 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Table 3: Recent Large-Scale Tomosynthesis Screening Studies

Recall Rate CDR per PPV1 (%): Sensitivity Specificity


Study and No. of (%): DM vs 1000: DM vs DM vs 2D (%): DM vs (%): DM vs
Year Type of Study Women 2D + DBT 2D + DBT + DBT 2D + DBT 2D + DBT Other Findings
Marinovich Meta-analysis of 1 009 790 3.5–11.3 vs 4.5–6.4 vs NA NA NA Results varied by screen-
et al, 2018 17 studies and 4.1–8.0 5.7–8.8 ing setting; greater
(9) 413 citations improvement in DBT
(United States and CDR seen in European
Europe), includes studies (biennial screen-
paired and un- ing); greater reduction
paired studies* of absolute DBT recall
rates seen in U.S. stud-
ies (high baseline recall)
Pattacini Prospective unpaired 19 560 3.5 vs 3.5 4.5 vs 8.6 13 vs 24† NA NA DBT increased CDR of
et al, 2018 randomized trial DCIS and grade 1–2
(10) (Italy) invasive cancers smaller
than 20 mm
Zackrisson Prospective paired 14 848 2.5 vs 3.6† 6.5 vs 8.7† 25.9 vs 24.1 60.4 vs 81.1 98.1 vs 97.2 One-view DBT with re-
et al, 2018 study (Sweden); duced compression may
(11) only one-view reduce radiation dose
DBT (MLO) and screening burden;
additional cancers de-
tected with DBT only
were predominantly
invasive, but otherwise
similar to mammogra-
phy-detected cancers
Skaane Prospective paired 24 301 NA 6.3 vs 8.1 NA 54.1 vs 70.5† 94.2 vs 95.0† Additional analysis of
et al, 2019 study (Norway) synthetic 2D mam-
(12) mography plus DBT
showed no substantial
difference in sensitivity
or specificity compared
with DM plus DBT;
cancers seen only with
DBT were mostly
small, low grade, and
node negative
Hofvind Prospective unpaired 28 749 4.0 vs 3.1† 6.1 vs 6.6 15.2 vs NA NA Distribution of tumor
et al, 2019 randomized trial 21.4† characteristics did not
(13) (Norway), DBT differ significantly be-
performed only tween DM and 2D syn-
with 2D synthetic thetic mammography
mammography plus DBT for invasive
cancer or DCIS
Conant Retrospective analysis 96 269 11.2 vs 8.7† 4.4 vs 5.8† 3.9 vs 6.3† 91.5 vs 90.6 88.9 vs 91.3† Study includes age and
et al, 2019 of prospective density subgroup
(14) cohort data; multi- analysis; DBT helped
institutional study detect more smaller
(United States) node-negative invasive
cancer, especially
among women aged
40–49 years
Note.—CDR = cancer detection rate, DBT = digital breast tomosynthesis, DCIS = ductal carcinoma in situ, DM = digital mammography,
MLO = mediolateral oblique, PPV1 = positive predictive value of recall, 2D = two-dimensional.
* Unpaired study indicates that patients underwent either DBT plus DM or DM only. Paired studies indicate that patients underwent only
DBT plus DM (DM only results recorded separately).

P  .05.

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 5


Novel Approaches to Screening for Breast Cancer

Automated Breast US
In automated breast US, a wide transducer scans
the entire breast, which is imaged in two to five
overlapping views (anteroposterior, lateral, and
medial). Continuous scanning allows reconstruc-
tion of acquisition views into coronal and sagittal
planes displayed alongside the original acquisi-
tion planes (Fig 2) (57). The technique captures
images of the entire breast, eliminating the need
for technologists to assess specific findings in real
time, enabling remote (batch) reading by radiolo-
gists, and facilitating comparison of current and
previously obtained images, which is important
Figure 1: Mediolateral oblique digital breast tomosynthesis (DBT) images from a screening for screening.
examination in a 53-year-old woman. (a) A spiculated mass is seen in the right posterior breast The diagnostic quality of automated breast US
(arrow) on a routine 1-mm-thick DBT slice. (b) A high-resolution 6-mm-thick slice shows better is comparable to that of handheld US. Several
detail and sharpness of the mass (arrow) and surrounding fibroglandular tissue. Biopsy showed a studies show supplemental cancer detection rates
grade 1 infiltrating ductal carcinoma. The 6-mm-thick slice is enhanced by using machine learning
algorithms targeting calcifications, spiculations, and masses. Thicker slices also allow fewer images
after digital mammography ranging from 1.9 to
to review and may reduce radiologist reading time. (Images courtesy of Washington Radiology 7.7 per 1000 screens (58,59). However, reported
Associates, Fairfax, Va.) high false-positive rates (up to 13%) and long
reading times have tempered the enthusiasm for
automated breast US as a supplemental screening
nant breast lesions in vitro is greater than that in benign le- modality (60). Nonetheless, automated breast US is a viable
sions, with little overlap (47). Elastography supplements rou- alternative for screening handheld US in women with dense
tine US screening in women with dense breasts to characterize breasts at mammography or for high-risk women who cannot
lesions as benign or malignant, improving specificity (Fig 2). tolerate MRI. Future developments, including incorporation
It also improves sensitivity by depicting isoechoic lesions (48). of whole-breast Doppler US or elastography, may reduce the
Strain elastography compares the relative stiffness of a lesion false-positive rate and improve the value of the technique for
with other tissues in the field of view (48), so a specific numeric screening (61).
value of lesion stiffness is not measured. The technique for opti-
mal images varies by vendor (48). Two-dimensional shear-wave US Transmission Tomography
elastography provides a quantitative estimate of the lesion stiff- US transmission tomography is a currently experimental,
ness based on the speed of shear waves generated by applying prone scanning technique that uses water as the coupling
an acoustic radiation force impulse push pulse (49). A unique agent combined with low-frequency ultrasound waves (1–7
feature of breast elastography for both strain elastography and MHz) emitted by a ring-like transducer surrounding the
shear-wave elastography is that malignant breast lesions appear breast. Ultrasound waves travel through water at a speed of
larger on elastographic images than on B-mode US images, approximately 1.5 km/sec. The time required to traverse the
whereas benign breast lesions appear smaller (48). breast is dependent on the tissue type encountered by ultra-
Numerous studies have shown that both strain elastogra- sound waves, so the generated speed of sound map provides
phy and 2D shear-wave elastography can help evaluate breast classifying information of underlying tissue (62,63). The
lesions and improve lesion characterization to various degrees speed of sound is somewhat lower in fibroglandular tissue,
(49–51). A meta-analysis of strain elastography reported whereas it is higher in solid masses. Speed of sound is also
that use of the elastography-to–B-mode ratio has a negative higher in malignant compared with benign lesions and may
predictive value of 0.03, downgrading a lesion with pretest help differentiate lesions (63,64). Ultrasound waves also lose
probability of 50% malignancy to 2% (52). The addition power while traversing the breast, so attenuation can help
of elastography and Doppler to B-mode US improved the classify breast parenchymal patterns.
positive predictive value of screening US while reducing the Although in early development, US transmission tomogra-
number of false-positive findings without missing cancers (53). phy holds promise for efficacy in screening. Studies on lesion
Recent studies suggest that the combination of strain elastogra- detection and classification are currently limited. An early study
phy and shear-wave elastography can further increase sensitiv- of 71 BI-RADS category 4 lesions showed that a composite score
ity and specificity (54,55). based on speed of sound and attenuation could help differenti-
Despite being available, elastography has not been widely ate benign from malignant lesions (Fig 3) (65). US transmission
implemented due to a high learning curve for performing and tomography may also use standard reflection images showing
interpreting examinations. The recent improvements in both underlying anatomy. This combination yields a technique that
strain and shear-wave technology with the addition of quality provides a three-dimensional breast image resembling a breast
maps are aimed at overcoming these problems and enabling the MRI scan (Fig 4) (64). Several studies demonstrate the possibil-
incorporation of elastography in “best-practice” algorithms (56). ity of estimating breast density with these acquisitions (66–68),

6 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Figure 2: Images in a 61-year-old woman with a density C breast. (a) Screening mammogram shows a Breast Imaging Reporting and Data
System category 2 lesion. Patient has a history of previous remote benign biopsy of the left breast with clip placement. (b, c) Axial (b) and coronal
(c) supplemental screening US scans obtained with an automated breast scanner show a 2-cm hypoechoic mass in the left breast at 4 o’clock. (d)
Image obtained with hand-held US shows an irregular hypoechoic mass. (e) Image obtained at strain elastography shows that the mass has an
elastography–B-mode distance ratio of 1.5, which is suggestive of a malignant lesion. (f) Image obtained with two-dimensional shear-wave elastog-
raphy shows that the lesion has a maximum stiffness of more than 9.0 m/sec, which is also consistent with a malignant lesion. At biopsy, the lesion was
an invasive moderately differentiated ductal carcinoma that was estrogen receptor positive, progesterone receptor negative, and human epidermal
growth factor receptor 2 negative.

which may assist in breast cancer risk estimation. Larger studies transmission tomography with other nonionizing techniques,
are mandatory, with a particular focus on investigating specific- such as optoacoustic imaging or microwave imaging, to further
ity. Future developments include a possible combination of US increase performance.

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 7


Novel Approaches to Screening for Breast Cancer

Figure 3: (a–c) US transmission images in a


69-year-old woman with an 8-mm invasive breast
cancer at 11 o’clock in the left breast (arrow).
Speed of sound image (a) and attenuation image
(b) are combined in a composite image (c), where
values higher than 1 are indicative of malignancy
(red). (d) Three-dimensional rendering of the entire
breast shows all red voxels against a background
of voxels with a very low composite index. (Im-
ages courtesy of Vasilis Marmarelis, PhD, University
of Southern California, Dr Allen and Charlotte
Ginsburg Institute for Biomedical Therapeutics, Los
Angeles, Calif, and Transonic Imaging, Los Angeles,
Calif.)

Optoacoustic US Recent multi-institutional studies showed that optoacoustic


Optoacoustic imaging has advanced over the past 2 decades and US improves specificity for BI-RADS category 3, 4, and 5
is now poised for clinical use. Optoacoustic US, also known as masses compared with gray-scale US alone, whereas the sensi-
optoacoustic tomography and photoacoustic imaging, can help tivity of optoacoustic US was noninferior to that of gray-scale
visualize blood vessels and detect tumor-driven angiogenesis by US (71,74). Future technical advances coupled with clinical
using laser light pulses, while monitoring returning acoustic trials are required to determine clinical implementation and
stress waves, to generate the photoacoustic signal (69). Cancers the role of optoacoustic US in the screening setting.
are active compared with benign lesions, extracting more oxy-
gen and resulting in more deoxygenated hemoglobin. The use Fusion Imaging
of different wavelengths for the laser pulses allows optoacoustic Breast imaging often involves cross-correlation between
US to enable the differentiation of deoxygenated hemoglobin multiple modalities. Fusion imaging facilitates this by ac-
from oxygenated hemoglobin, which may then be color-coded. quiring multimodality information with a single device. In
The angiogenesis visible on optoacoustic US images can be cor- the screening setting, the cross-correlation between screen-
related with pathologic and molecular subtypes (70). ing US and mammographic findings may be challenging.
State-of-the-art optoacoustic US systems combine tradi- There is, consequently, ongoing interest in the fusion of
tional gray-scale US with functional optoacoustic US data mammography and US imaging. Early prototypes demon-
during real-time display (Fig 5) (71). Current clinical opto- strate the possibility of a dual-modality system that com-
acoustic US systems use a handheld probe similar to that used bines full-field DM or DBT with automated breast US
with conventional US units. The main goal of optoacoustic (75,76). With this technology, the US probe is built into
US systems is to increase the specificity of US screening. Prone the compression paddle or located on the detector side
systems in early development can image the entire breast and (75,76). The probe scans the compressed breast, acquiring
have promise as a stand-alone screening tool (Fig 6) (72,73). US images simultaneously with mammography, simplifying

8 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Figure 4: US transmission and reflection images in a 67-year-old woman who presented with a 2.1-cm grade 3 estrogen receptor–negative, progester-
one receptor–negative, and human epidermal growth factor receptor 2–positive cancer in the lower medial quadrant of the left breast (arrows). The speed
of sound image (top right) and the reflection image (bottom left) were directly obtained from the scan. These are combined to produce a so-called wafer
image (top left), or waveform-enhanced reflection. The speed of sound image is combined with the attenuation map (not shown) to create a relative tissue
stiffness map that is projected on top of the reflection image in the stiffness fusion image (bottom right). (Images courtesy of Corinne Duluk, Delphinus Medical
Technologies, Novi, Mich.)

co-registration. Although some units fail to adequately im- precontrast, early postcontrast, and delayed postcontrast T1-
age the posterior breast, early small studies show potential weighted images (81). In contradistinction, the abbreviated
in this developing technology (75,77). protocol includes a precontrast examination and one postcon-
trast T1-weighted examination, along with subtraction images
MRI Examination and maximum intensity projection images (Fig 7). In a seminal
Breast MRI offers the highest sensitivity for the detection of study, Kuhl et al (82) found equivalent diagnostic accuracy for
occult cancer, regardless of breast density (8). A review on the abbreviated and full protocols among 443 women and 606
state-of-art breast MRI was recently published in Radiology MRI studies. Both protocols enabled identification of all 11
(8). Breast MRI offers superior tissue contrast due to uptake breast cancers. In addition, the authors found similar diagnostic
of gadolinium secondary to neoangiogenesis and increased accuracies: Specificity was 94.5% for the abbreviated protocol
permeability of malignancies (78). Studies show MRI is bet- and 93.9% for the full diagnostic protocol, and positive predic-
ter than mammography and US in the detection of high- tive value was 24.4% for the abbreviated protocol and 23.4% for
grade invasive cancers (79). A recent multicenter randomized the full diagnostic protocol. Furthermore, there was a vast reduc-
clinical trial from the Netherlands, where screening MRI was tion in time for image acquisition (17 minutes vs 3 minutes) and
performed in women with extremely dense breasts, found sig- radiologist interpretation (28 seconds for the first postcontrast
nificantly fewer interval cancers in women who underwent images and 2.8 seconds for the maximum intensity projection
supplemental MRI compared with mammography alone (0.8 image alone) (82).
per 1000 screenings vs 5.1 per 1000 screenings, respectively; The basic abbreviated protocol described earlier has many
P , .001) (80). But high costs as well as the limited toler- variations. A recent review of 21 studies on various abbreviated
ability and availability of MRI scanners preclude population- breast MRI protocols performed in eight different countries
wide screening with breast MRI. and in more than 4500 women confirmed that the diagnostic
accuracy was similar to that of the full breast MRI protocol
Abbreviated Breast MRI (83). These protocols exploit the high sensitivity of MRI while
Abbreviated MRI has shorter image acquisition and interpreta- reducing acquisition and interpretation times (Table E2 [on-
tion times, which may increase the availability of breast MRI line]). Recently, a multicenter randomized trial that compared
and reduce the costs. the screening performance of abbreviated breast MRI and
The American College of Radiology accreditation require- DBT in women with dense breasts found the invasive cancer
ments for breast MRI include a T2-weighted sequence and detection rate was 11.8 per 1000 women for abbreviated breast

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 9


Novel Approaches to Screening for Breast Cancer

Figure 5: Optoacoustic images in two patients. (a) Conventional B-mode image in a 44-year-old woman shows an oval mass with indistinct margins
and parallel orientation (arrow) consistent with a Breast Imaging Reporting and Data System (BI-RADS) category 4a lesion. This mass could be misinterpreted
as a BI-RADS category 3 lesion. (b) Simultaneously obtained optoacoustic map demonstrates absence of internal vascularity or deoxygenation. Vessels are
parallel to capsule, not perpendicular, and are gently curved and gradually tapered. These findings are consistent with benign findings. Core needle biopsy
showed a fibroadenoma. (Images courtesy of Tchaiko Parris, MD, RadNet, Long Beach, Calif.) (c) Conventional B-mode US scan in a 56-year-old woman
shows an oval mass with partially circumscribed margins and a parallel orientation and mixed posterior acoustic enhancement (arrow). (d) Optoacoustic
map in same patient shows internal vascularity with mostly deoxygenated signal (red), consistent with a hypervascular BI-RADS category 4 mass. Core
needle biopsy showed a human epidermal growth factor receptor 2–enriched infiltrating ductal carcinoma. (Images courtesy of Minh Nguyen, MD, Solis
Women's Health, Plano, Tex.)

issues must be addressed supporting implementa-


tion of this examination into clinical workflow. In
the United States, there is no Current Procedural
Terminology code for an abbreviated MRI exami-
nation. In addition, Borthakur et al (85) analyzed
the activity times from 70 abbreviated breast MRI
studies and 736 full MRI screening studies, con-
cluding that the realized gains in patient flow rate
were lower than expected for an abbreviated MRI
examination. These results are not entirely unex-
pected because of workflow considerations includ-
ing setup, patient positioning, and room turnover.
Figure 6: Images in a 44-year-old woman who presented with a 19-mm invasive ductal car-
cinoma grade 3 in the right breast (arrow). (a) Maximum intensity projection image of the contrast Ultrafast Breast MRI
material–enhanced MRI scan with a color overlay of average enhancement intensity. (b) Large field- An ultrafast sequence can help document fast ar-
of-view (approximately 35 cm2) optoacoustic image obtained with a photoacoustic mammoscope by terial enhancement and fast venous drainage of
using a 1064-nm yttrium-aluminum-garnet laser of the area within the dashed box in a. Tumor is well
visible and of similar size and shape as in the MRI scan. (Images courtesy of Srirang Manohar, PhD,
breast cancers. The temporal resolution of an ul-
University of Twente, Enschede, the Netherlands; adapted with permission from reference 73.) trafast protocol is typically less than 10 seconds
per frame and may be incorporated into abbrevi-
ated or full breast MRI protocols. These sequences
MRI versus 4.8 per 1000 women for DBT, a difference of seven often use various k-space view sharing and compressed sensing
per 1000 women (P = .002) (84). Abbreviated breast MRI will techniques where the central region of k-space is sampled con-
likely be increasingly used as a screening tool, but operational tinuously. However, the outer region is only partially sampled,

10 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

enhancement outperformed the BI-RADS wash-out evaluation


(area under the curve, 0.829 vs 0.692). Abe et al (88) found the
initial enhancement rate and signal enhancement ratio af-
ter aortic enhancement was higher for malignant than benign
lesions. Additional studies using various ultrafast techniques
showed high reproducibility of early wash-in temporal kinetic
parameters. Hence, ultrafast breast MRI allows for increasing
the specificity of (abbreviated) breast MRI, without increasing
the scanning time. The most modern sequences even obtain a
diagnostic spatial resolution with the potential as a stand-alone
screening technique. van Zelst et al (89) showed similar sensi-
tivity and increased specificity by reading only ultrafast images
compared with a full diagnostic MRI protocol.

Diffusion-weighted Imaging
Diffusion-weighted imaging can help visualize and quantify
random movement of water molecules in tissue, influenced by
tissue microstructure and cell density. It does not require the
use of an intravenous contrast agent. Breast cancers show de-
creased water diffusion due to increased cell density, leading to
higher signal intensity on diffusion-weighted images. Multiple
studies show that diffusion-weighted imaging can improve the
differentiation between benign and malignant lesions (90,91).
Given the concerns of gadolinium deposition in the brain and
the general burden of the need for intravenous access (92),
there is growing interest to develop a screening noncontrast
MRI examination. The use of high b values with background
suppression seems to be a viable screening technique (93).
Preliminary studies suggest unenhanced MRI with diffusion-
weighted MRI may provide higher sensitivity than screening
mammography for the detection of breast malignancies (94).
However, current diffusion-weighted imaging techniques are
not sensitive enough to replace contrast-enhanced breast MRI
(95,96), particularly because the sensitivity for subcentimeter
and nonmass lesions is limited.

Nuclear Medicine Techniques


Advances in nuclear medicine, using dedicated gamma or PET
breast scanners, provide functional breast imaging techniques
that, in turn, may allow the use of specific cellular information
to assist in the diagnosis, staging, and treatment of breast can-
cer. Although current American College of Radiology practice
guidelines do not recommend use of molecular breast imaging
(MBI) or PET for routine screening (97), further technologic
advances may enhance these techniques to allow a role in breast
Figure 7: Images in a 44-year-old woman with BRCA1 gene mutation cancer screening.
and history of left breast cancer. The patient had undergone lumpectomy
and radiation therapy in 2008 and presented for high-risk surveillance.
(a) Precontrast axial MRI scan demonstrates no abnormalities. (b) First
MBI Examination
contrast-enhanced axial MRI scan and (c) first subtraction postcontrast MBI, which includes gamma-specific breast imaging, uses dedi-
MRI scan demonstrate an irregular heterogeneously enhancing mass (ar- cated breast-specific high-spatial-resolution gamma camera sys-
row) in right breast. Biopsy yielded a moderately differentiated estrogen tems combined with intravenous technetium 99m (99mTc) sesta-
receptor–positive, progesterone receptor–positive, human epidermal mibi to image subcentimeter cancers in projections comparable
growth factor receptor 2–negative invasive carcinoma that is well seen
with an abbreviated breast MRI protocol.
to mammography (98,99). Dual-panel MBI systems that use
cadmium zinc telluride detectors demonstrate improved sensi-
allowing for simultaneous high temporal resolution with a tivity, energy resolution, spatial resolution, and lesion detection
minimal decrease in spatial resolution (Fig 8) (86–88). Mann compared with the single-panel breast-specific gamma imaging
et al (87) found that the maximum slope of malignancy in early systems with sodium or cesium iodide crystals (99,100).

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 11


Novel Approaches to Screening for Breast Cancer

reactions (metallic taste, rash, flushing), making it a good sup-


plemental screening examination for women with elevated risk
and those with dense breasts. The incremental cancer detection
rate across multiple studies in asymptomatic women is 8–16 per
1000 women screened, with most cancers being invasive and tu-
mor size ranging from 2 mm to 5 cm (99–101). The high cancer
detection rate of MBI is in range of those of other vascular imag-
ing techniques (Table 4).
Despite being available for at least 15 years, MBI has not
gained widespread adoption primarily due to concerns regard-
ing whole-body radiation dose. With technical improvements,
radiation dose has decreased over time. The effective dose of 8
mCi of 99MTc sestambi is approximately 2.4 mSv, compared
with 0.5–1.2 mSv for mammography and DBT (100). Al-
though MBI dose is thus still two- to fivefold that with mam-
mography, it has a much higher cancer yield (99,100). Current
dosage for breast-specific gamma imaging studies are higher
than MBI requirements (103). Novel image processing tech-
niques may allow further decrease in both radiation dose and
scanning time (104). MBI-directed biopsy is also available for
lesions occult with other modalities. Beyond the high supple-
mental cancer detection rate, MBI also has the advantage of
being cost-effective and well-tolerated by patients, as it requires
minimal breast compression.

Breast PET Imaging


Breast PET employs detectors positioned close to the breast
and measures fluorine 18 (18F) fluorodeoxyglucose uptake.
18
F fluorodeoxyglucose is a positron-emitting glucose analog
that accumulates in metabolically active cells, which is usually
the case for cancer cells. Patients must fast 4–6 hours as blood
glucose should be within normal limits before 18F fluorode-
oxyglucose injection. Patients must also wait 60–90 minutes
after the injection to allow for optimal tracer uptake before
imaging. Breast PET systems use detectors so that patients may
be imaged in the upright position with minimal compression
(similar to mammography and often referred to as positron
emission mammography or PEM) or the prone position with
no compression (100).
Breast PET systems are primarily used for the evaluation of
Figure 8: Images in a 41-year-old woman who is a carrier for the patients with newly diagnosed breast cancer and for treatment
ataxia-telangiesctasia mutated gene, ATM, and who presented for high- planning. However, an early study published by Yamamoto
risk screening MRI. (a) Ultrafast time-resolved angiography with stochastic
trajectories, or TWIST, axial MRI scan with 4-second temporal resolution
et al (105) in 2016 showed that positron emission mammog-
after contrast material injection demonstrates early wash-in of contrast ma- raphy may have a potential role in breast cancer screening. In
terial in a mass in the right breast (thin arrow) at first time frame (4 seconds that study, which comprised 265 asymptomatic and symptom-
after aortic enhancement). Note the contrast material in the thoracic aorta atic patients, the overall recall rate, positive predictive value, and
(thick arrow). (b) Second frame (8 seconds after aortic enhancement) cancer detection rate were 8.3%, 27.3%, and 2.3%, respectively.
shows marked enhancement of the mass (arrow). (c) Routine postcontrast
axial MRI scan shows an irregular heterogeneous mass (arrow) within
Although the spatial resolution was only 2.4 mm, the smallest
marked background parenchymal enhancement. cancer reported in that study measured 7 mm.
Despite techniques used to lower 18F fluorodeoxyglucose
A complete MBI study consists of standard craniocaudal and dose, the radiation exposure, costs, and patient time to prepare
mediolateral oblique views of the breast, with acquisition times for the examination currently limit the utility of breast PET im-
of 7–10 minutes for each projection (Fig 9) (99,101). 99mTc aging for widespread screening. Ongoing investigation of novel
sestamibi is sequestered by mitochondria (increased in cancer PET tracers with improved dosimetry and pharmacologic safety
cells) and influenced by blood flow and angiogenesis (102). Pa- plus development of new imaging techniques may allow this
tients are imaged approximately 5–10 minutes after injection. technology to be more widely used in clinical practice for diag-
MBI has a long patient safety history and only rare mild adverse nosis as well as treatment selection (106).

12 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Figure 9: Images in a 45-year-old woman with bilateral saline implants who presented for breast screening. Molecular breast imaging (MBI) was recommended
for supplemental screening on the basis of breast density and family history of breast cancer. (a, b) Left mediolateral oblique image (a) with implant displacement
view (b) from screening tomosynthesis examination were interpreted as negative (the biopsy clip is from a previously biopsied fibroadenoma). (c, d) Left mediolateral
oblique (c) and craniocaudal (d) views from screening MBI performed the same day as mammography demonstrate focal uptake in the upper posterior left breast (ar-
row). Pathologic examination confirmed invasive ductal carcinoma, grade 2, with an extensive 19-mm ductal carcinoma in situ component. (Images courtesy of Carrie
Hruska, PhD, May Clinic, Rochester, Minn.)

Table 4: Comparison of Vascular-based Screening Techniques

Parameter Abbreviated MRI Contrast-enhanced SM MBI PET and/or PEM Optoacoustic US


Indication Elevated risk, dense Elevated risk, dense Elevated risk, dense Potential for screening Potential to enhance
Breasts breasts breasts handheld or
automated US
Intravenous contrast Gadolinium-based Standard iodinated Technetium 99 Fluorine 18 None
material agent contrast material sestamibi fluorodeoxyglucose
FDA approved Yes Yes Yes Yes No
Clinical use Yes Yes, but investigational Yes, but investigational Yes, but investigational Investigational, not
for screening for screening for screening FDA approved
Supplemental CDR 19 per 1000 13 per 1000 screened 8–16 per 1000 23 per 1000 Unknown
screened, range: screened (single study)
9–36 prevalence
screening
Other considerations No CPT code (full Potential to be easily Dedicated equipment Dedicated equipment, Laser light
protocol only) accessible and radiation dose radiation dose, and technology
potential use of other
breast-specific tracers
Note.—CDR = cancer detection rate, CPT = current procedural terminology, FDA = U.S. Food and Drug Administration, MBI = molecu-
lar breast imaging, PEM = positron emission mammography, SM = spectral mammography.

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 13


Novel Approaches to Screening for Breast Cancer

Table 5: Status of Current AI Algorithms

AI Use Current Performance Clinical Role Experimental Strategies


Mammography On par with human readers for In use as an aid for concurrent reading Preselection of normal mammograms;
lesion detection and of mammograms to improve human independent second reader of screening
classification performance mammograms; risk prediction for the
development of breast cancer
DBT On par with human readers for In use as an aid for concurrent reading Independent reading of DBT examina-
lesion detection and of mammograms to improve human tions; creation of “intelligent” synthetic
classification performance and decrease reading mammograms that highlight potential
time abnormalities
Handheld US On par with human readers for In use to aid in lesion classification Incorporation of multiparametric data,
lesion classification detected by human readers including Doppler and elastography;
extraction of quantitative data for lesion
classification
Automated breast Below human performance for In use to prevent overlook errors and Improvement of detection and classification
US lesion detection and decrease reading time algorithms; creation of intelligent sum-
classification mary images
MRI Below human performance for Experimental only Incorporation of multiparametric data, in-
lesion detection and cluding dynamics, T2-weighted imaging,
classification and DWI; improvement of detection and
classification algorithms; enhanced image
reconstruction; creation of intelligent
summary images
Note.—AI = artificial intelligence, DBT = digital breast tomosynthesis, DWI = diffusion-weighted imaging.

Artificial Intelligence and Machine Learning personalized when deep learning risk models include electronic
The application of machine learning to medical images has health records (117).
been performed for decades. Until recently, however, the per- Although breast US is often used as a supplemental screening
formance of these algorithms for lesion detection and classi- technique, image acquisition is not standardized compared with
fication (commonly referred to as computer-aided diagnosis) mammography and MRI. Typically, operators store static images
has been below human standards. In recent years, increased of lesions deemed relevant, and the assessment of the remaining
computer power, along with mathematical advances, has en- breast (when performed) is usually not captured. The result is
abled the use of complex multilayered (deep) neural networks, that the sensitivity of AI for US is much more operator depen-
leading to a markedly improved performance of machine in- dent than for mammography. Current algorithms mainly focus
terpretation of highly standardized imaging tasks (Table 5). It on the classification of lesions pointed out by the operator, where
is widely believed that these deep learning algorithms will soon AI achieves human-like performance (118,119), and may aid in
play a major role in screening, both to improve the quality of radiologist decision support to improve reader performance and
the screening programs and to assist with the increasing work- decrease interreader variability (Fig 12) (120). The use of such
load that originates from ever-expanding screening indications. an algorithm (S-detect; Samsung, Seoul, Korea) in real time
Research using deep learning algorithms has focused on may improve the performance of practicing breast radiologists,
screening mammography because there is a simple binary out- leading to an improvement in specificity and reduction in false-
come (cancer vs not cancer) and the large number of screening positive assessments (121).
mammograms permits leverage of big data sets as well as the Because the performance of automated breast US is stan-
potential for important clinical impact (improved mortality) dardized, it is possible to use AI not only for lesion classifi-
(107). Several studies show that diagnostic accuracies of cur- cation but also for lesion detection. One commercial applica-
rent algorithms for mammography approach that of radiologists tion developed for this purpose (QVCAD; Qview, Los Altos,
(Fig 10) (108–110). DBT studies report similar performance Calif ) generates a composite image of each automated breast
(Fig 11) (111). Furthermore, early studies suggest that when US view, with potential lesions displayed as dark areas. This
artificial intelligence (AI) models predict a very low likelihood system allows for reduced reading time while also providing
of malignancy, the algorithms can triage and interpret these potential for improved performance, particularly for less expe-
mammograms (112,113) to save time and resources. Beyond rienced readers (122,123). Underlying AI algorithms for breast
lesion detection and classification, other potential applications US require further improvements to increase their value and
for deep learning models include assessment of mammographic accuracy in screening.
breast density (114) and prediction of breast cancer risk by in- Like mammography, breast MRI is a standardized technique
corporating the normal mammographic parenchymal pattern well suited for automated analysis and often used for staging
(density, texture, etc) (115,116). Risk assessment may be further in patients with newly diagnosed breast cancer. There is active

14 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Figure 10: Screening mammograms in a 72-year-old asymptomatic woman. (a) Mediolateral view of the right breast dem-
onstrates multiple masses (black arrows), with the largest mass (white arrow) located in the superior breast. (b) The deep learning
model identified the superior mass as suspicious (color overlay in red). There was no heat map overlying the other masses (c–e).
(d, e) Patch-wise attention scores are highly concentrated on the two region-of-interest patches (0.6 and 0.38) that contain the
superior mass. The deep learning model predicted this mass was highly suspicious for carcinoma. Pathologic examination yielded
poorly differentiated invasive ductal carcinoma.

interest in MRI radiomics, using quantified characteristics of the relatively small data sets. Once much larger case sets become
gray-scale distribution in cancers and the surrounding tissue for available, AI algorithms should achieve human-like perfor-
classification and to predict eventual response to therapy as well mance for breast lesion detection and classification.
as to provide a short- and long-term prognosis (124,125). To en- For further reference, interested readers are referred to more
hance specificity of automated lesion classification, deep learn- extensive reviews on AI for breast imaging that have recently
ing algorithms can differentiate between benign and malignant been published (107,131).
breast lesions with acceptable accuracy (126,127). Although per- Most studies on AI and breast imaging are retrospective. In-
formance is still below human standards, concurrent use of such creasing the generalizability of the results requires prospective
algorithms may prevent false-positive recalls and may someday studies in different patient populations. Investigators should fol-
improve the accuracy of MRI screening. low guidelines to increase the reproducibility of their results (132).
Preliminary studies also report on deep learning–based au-
tomated detection algorithms for breast cancer at screening Non-Imaging Novel Screening Techniques
MRI (128–130). Although these systems improve upon pre- A liquid biopsy helps detect circulating tumor components in
viously reported machine learning approaches, an acceptable blood or plasma (133). Growing cancers actively or passively
sensitivity is only achieved at high false-positive rates (currently shed debris into the bloodstream, including cell-free RNA,
more than one per MRI), which reduces their use in clinical tumor DNA, and dead and living cancer cells. Although con-
practice. However, current studies are conducted by using centrations are usually low, modern amplification techniques

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 15


Novel Approaches to Screening for Breast Cancer

Figure 11: Images from digital breast tomosynthesis of the right breast. Images in top row are mediolateral oblique views, images in bottom row are craniocaudal
views. The tomosynthesis stack is summarized in synthetic mammograms in a, b, e, and f. Original tomosynthesis slices are shown in c, d, g, and h. There is a subtle irregular
mass (red circle), which was sampled for biopsy and proven to be a 13-mm invasive cancer not otherwise specified (estrogen receptor positive, progesterone receptor
positive, human epidermal growth factor receptor 2 negative) present in the upper outer quadrant. This was detected by an artificial intelligence system that marks the find-
ing on the synthetic images and on the tomosynthesis slices (b, c, f, and g). Clicking on the mark in the synthetic image will take the reader immediately to the most suspi-
cious slice in the tomosynthesis stack. (Images courtesy of Nico Karssemeijer, PhD, Radboud University Medical Center, Nijmegen, the Netherlands, Screenpoint Medical,
Nijmegen, the Netherlands.)

permit detection of RNA and DNA remnants, which may were detected in only 59% of patients (136). Another study tar-
contain carcinogenic mutations. Molecular profiling may al- geting multiple tumors showed a median sensitivity of 70% using
low assessment of tumor heterogeneity in patients with breast a combination of circulating tumor DNA and cancer-associated
cancer (134). Circulating tumor DNA can also help predict proteins. Unfortunately, only 33% of asymptomatic breast cancers
therapy response and prognosis (135). A major advantage is were found (137), requiring more sensitive tests. More frequent
that liquid biopsy is repeatable and used for the evaluation of somatic mutations occur with advancing age, and these mutations
changes over time. do not necessarily all cause cancer (138). Consequently, the risk of
Screening with liquid biopsy is more challenging. In one study false-positive results requires mitigation before the liquid biopsy
of patients with stage I and II breast cancer, somatic mutations technique has real merit for screening. Further advancements and

16 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

Figure 12: Images in a 59-year-old woman who presented with a mass detected at screening mammography. Pathologic examination
showed infiltrating ductal carcinoma, grade 2. A, Gray-scale US scan in the radial plane demonstrates an oval heterogeneous, but pre-
dominately hyperechoic, mass in the posterior right upper outer breast (arrow). B, Artificial intelligence decision support software graded this
lesion as suspicious (risk alignment: Breast Imaging Reporting and Data System [BI-RADS] category 4A–4B). C, Diagram shows results of an
independent reader evaluation. Three of 15 readers (20%) initially classified this lesion as BI-RADS category 2 or 3; all readers classified this
lesion as BI-RADS category 4 or higher after being shown the decision support (DS) result.

more clinical data are needed to provide more extensive tumor is a paid consultant for Screenpoint Medical and Transonic Imaging; institution
has grants/grants pending with Siemens Healthineers, Bayer Healthcare, Medtronic,
analysis for liquid biopsy to become a reliable and cost-effective BD, Seno Medical, Screenpoint Medical, Identification Solutions, and Elswood;
means of screening and diagnosis (139). receives payment for lectures including service on speakers bureaus from Bayer
Healthcare. Other relationships: disclosed no relevant relationships. R.H. Activities
Conclusion related to the present article: receives support for travel or speaking engagements
from Hologic. Activities not related to the present article: receives honorarium for
Breast cancer screening in the next decade will move beyond consulting from Hologic; receives honoraria and travel expenses for speaking en-
the traditional familiar tools including mammography, US, gagements from Holigic. Other relationships: disclosed no relevant relationships.
R.G.B. Activities related to the present article: disclosed no relevant relationships.
and MRI. Ideally, the variety of new imaging options, com- Activities not related to the present article: institution received equipment grants
bined with artificial intelligence and/or neural networks, will from Philips Ultrasound, Siemens Healthineers, Mindray, and Samsung Ultra-
be enhanced by the integration of new screening protocols di- sound; receives payment for lectures including service on speakers bureaus from
Philips Ultrasound, Mindray, and Siemens Healthineers; receives royalties from
rected toward more personalized and precision medicine. The Thieme Publishers; receives travel/accommodations/meeting expenses unrelated to
“one size fits all” approach of past decades may no longer be activities listed from Philips Ultrasound and Mindray. Other relationships: disclosed
relevant. Primary and supplemental screening tools will likely no relevant relationships. L.M. Activities related to the present article: disclosed no
relevant relationships. Activities not related to the present article: is paid to be on
progress beyond screening mammography plus US or MRI for advisory boards at Lunit and iCAD; has grants/grants pending from Siemens. Other
women with dense breasts and/or elevated breast cancer risk. relationships: disclosed no relevant relationships.
New screening benchmarks and development of efficient can-
cer registries are crucial to enhance our ability to track efficacy References
and performance of new screening tools and treatments in a 1. Cancer Tomorrow. International Agency for Research on Cancer. http://
gco.iarc.fr/tomorrow/graphicisotype?type=0&population=900&mode=popu
timely and relevant manner. lation&sex=2&cancer=39&age_group=value&apc_male=0&apc_female=0.
Accessed January 5, 2020.
Disclosures of Conflicts of Interest: R.M.M. Activities related to the present ar- 2. Breast Cancer Statistics. Centers for Disease Control and Prevention. https://
ticle: disclosed no relevant relationships. Activities not related to the present article: www.cdc.gov/cancer/breast/statistics/index.htm. Accessed January 6, 2020.

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 17


Novel Approaches to Screening for Breast Cancer

3. Simon S. Facts & Figures 2019: US Cancer Death Rate Has Dropped 27% 26. Zanardo M, Cozzi A, Trimboli RM, et al. Technique, protocols and adverse
in 25 Years. American Cancer Society. https://www.cancer.org/latest-news/ reactions for contrast-enhanced spectral mammography (CESM): a systematic
facts-and-figures-2019.html. Published January 8, 2019. Accessed January review. Insights Imaging 2019;10(1):76.
6, 2020. 27. Jeukens CR, Lalji UC, Meijer E, et al. Radiation exposure of contrast-enhanced
4. Tabár L, Dean PB, Chen TH, et al. The incidence of fatal breast cancer spectral mammography compared with full-field digital mammography.
measures the increased effectiveness of therapy in women participating in Invest Radiol 2014;49(10):659–665.
mammography screening. Cancer 2019;125(4):515–523. 28. James JR, Pavlicek W, Hanson JA, Boltz TF, Patel BK. Breast Radiation
5. Pisano ED, Gatsonis C, Hendrick E, et al. Diagnostic performance of digi- Dose With CESM Compared With 2D FFDM and 3D Tomosynthesis
tal versus film mammography for breast-cancer screening. N Engl J Med Mammography. AJR Am J Roentgenol 2017;208(2):362–372.
2005;353(17):1773–1783 [Published correction appears in N Engl J Med 29. Phillips J, Mihai G, Hassonjee SE, et al. Comparative Dose of Contrast-
2006;355(17):1840.]. Enhanced Spectral Mammography (CESM), Digital Mammography, and
6. Chong A, Weinstein SP, McDonald ES, Conant EF. Digital Breast Tomo- Digital Breast Tomosynthesis. AJR Am J Roentgenol 2018;211(4):839–846.
synthesis: Concepts and Clinical Practice. Radiology 2019;292(1):1–14. 30. Dromain C, Thibault F, Muller S, et al. Dual-energy contrast-enhanced digital
7. Hooley RJ, Scoutt LM, Philpotts LE. Breast ultrasonography: state of the mammography: initial clinical results. Eur Radiol 2011;21(3):565–574.
art. Radiology 2013;268(3):642–659. 31. Cheung YC, Lin YC, Wan YL, et al. Diagnostic performance of dual-energy
8. Mann RM, Cho N, Moy L. Breast MRI: State of the Art. Radiology contrast-enhanced subtracted mammography in dense breasts compared
2019;292(3):520–536. to mammography alone: interobserver blind-reading analysis. Eur Radiol
9. Marinovich ML, Hunter KE, Macaskill P, Houssami N. Breast Cancer 2014;24(10):2394–2403.
Screening Using Tomosynthesis or Mammography: A Meta-analysis of 32. Tagliafico AS, Bignotti B, Rossi F, et al. Diagnostic performance of contrast-
Cancer Detection and Recall. J Natl Cancer Inst 2018;110(9):942–949. enhanced spectral mammography: Systematic review and meta-analysis. Breast
10. Pattacini P, Nitrosi A, Giorgi Rossi P, et al. Digital Mammography versus Digital 2016;28:13–19.
Mammography Plus Tomosynthesis for Breast Cancer Screening: The Reggio 33. Patel BK, Garza SA, Eversman S, Lopez-Alvarez Y, Kosiorek H, Pockaj
Emilia Tomosynthesis Randomized Trial. Radiology 2018;288(2):375–385. BA. Assessing tumor extent on contrast-enhanced spectral mammography
11. Zackrisson S, Lång K, Rosso A, et al. One-view breast tomosynthesis versus versus full-field digital mammography and ultrasound. Clin Imaging
two-view mammography in the Malmö Breast Tomosynthesis Screening 2017;46:78–84.
Trial (MBTST): a prospective, population-based, diagnostic accuracy study 34. Lee-Felker SA, Tekchandani L, Thomas M, et al. Newly Diagnosed Breast
Lancet Oncol 2018;19(11):1493–1503 [Published correction appears in Cancer: Comparison of Contrast-enhanced Spectral Mammography and
Lancet Oncol 2019;20(1):e9.]. Breast MR Imaging in the Evaluation of Extent of Disease. Radiology
12. Skaane P, Bandos AI, Niklason LT, et al. Digital Mammography versus 2017;285(2):389–400.
Digital Mammography Plus Tomosynthesis in Breast Cancer Screening: 35. Fallenberg EM, Schmitzberger FF, Amer H, et al. Contrast-enhanced spec-
The Oslo Tomosynthesis Screening Trial. Radiology 2019;291(1):23–30. tral mammography vs. mammography and MRI - clinical performance in a
13. Hofvind S, Holen ÅS, Aase HS, et al. Two-view digital breast tomosyn- multi-reader evaluation. Eur Radiol 2017;27(7):2752–2764.
thesis versus digital mammography in a population-based breast cancer 36. Sorin V, Yagil Y, Yosepovich A, et al. Contrast-Enhanced Spectral Mam-
screening programme (To-Be): a randomised, controlled trial Lancet On- mography in Women With Intermediate Breast Cancer Risk and Dense
col 2019;20(6):795–805 [Published correction appears in Lancet Oncol Breasts. AJR Am J Roentgenol 2018;211(5):W267–W274.
2019;20(7):e346.]. 37. Sung JS, Lebron L, Keating D, et al. Performance of Dual-Energy Contrast-
14. Conant EF, Barlow WE, Herschorn SD, et al. Association of Digital Breast enhanced Digital Mammography for Screening Women at Increased Risk
Tomosynthesis vs Digital Mammography With Cancer Detection and Recall of Breast Cancer. Radiology 2019;293(1):81–88.
Rates by Age and Breast Density. JAMA Oncol 2019;5(5):635–642. 38. Jochelson MS, Pinker K, Dershaw DD, et al. Comparison of screening CEDM
15. Bahl M, Mercaldo S, Dang PA, McCarthy AM, Lowry KP, Lehman CD. and MRI for women at increased risk for breast cancer: A pilot study. Eur J
Breast Cancer Screening with Digital Breast Tomosynthesis: Are Initial Radiol 2017;97:37–43.
Benefits Sustained? Radiology 2020;295(3):529–539. 39. Contrast Enhanced Mammography Imaging Screening Trial (CMIST).
16. Kim JY, Kang HJ, Shin JK, et al. Biologic Profiles of Invasive Breast Cancers American College of Radiology. https://www.acr.org/Research/Clinical-
Detected Only With Digital Breast Tomosynthesis. AJR Am J Roentgenol Research/CMIST. Accessed January 5, 2020.
2017;209(6):1411–1418. 40. Ohuchi N, Suzuki A, Sobue T, et al. Sensitivity and specificity of mam-
17. Lång K. The Coming of Age of Breast Tomosynthesis in Screening. Radiology mography and adjunctive ultrasonography to screen for breast cancer in the
2019;291(1):31–33. Japan Strategic Anti-cancer Randomized Trial (J-START): a randomised
18. Hovda T, Holen ÅS, Lång K, et al. Interval and Consecutive Round Breast controlled trial. Lancet 2016;387(10016):341–348.
Cancer after Digital Breast Tomosynthesis and Synthetic 2D Mammogra- 41. Weigert JM. The Connecticut Experiment; The Third Installment: 4 Years
phy versus Standard 2D Digital Mammography in BreastScreen Norway. of Screening Women with Dense Breasts with Bilateral Ultrasound. Breast
Radiology 2020;294(2):256–264. J 2017;23(1):34–39.
19. Conant EF, Zuckerman SP, McDonald ES, et al. Five Consecutive Years of 42. Zhan J, Diao XH, Jin JM, Chen L, Chen Y. Superb Microvascular Imaging-A
Screening with Digital Breast Tomosynthesis: Outcomes by Screening Year new vascular detecting ultrasonographic technique for avascular breast masses:
and Round. Radiology 2020;295(2):285–293. A preliminary study. Eur J Radiol 2016;85(5):915–921.
20. TMIST / EA1151 Study: Tomosynthesis Mammographic Imaging Screening 43. Yongfeng Z, Ping Z, Wengang L, Yang S, Shuangming T. Application of
Trial. American College of Radiology. https://www.acr.org/-/media/ACR/ a Novel Microvascular Imaging Technique in Breast Lesion Evaluation.
Files/Research/TMIST-Site-Process-Summary.pdf?la=en. Accessed January Ultrasound Med Biol 2016;42(9):2097–2105.
7, 2020. 44. Jung HK, Park AY, Ko KH, Koh J. Comparison of the Diagnostic Performance
21. Svahn TM, Houssami N, Sechopoulos I, Mattsson S. Review of radiation of Power Doppler Ultrasound and a New Microvascular Doppler Ultrasound
dose estimates in digital breast tomosynthesis relative to those in two-view Technique (AngioPLUS) for Differentiating Benign and Malignant Breast
full-field digital mammography. Breast 2015;24(2):93–99. Masses. J Ultrasound Med 2018;37(11):2689–2698.
22. Bae MS, Moon WK. Is Synthetic Mammography Comparable to Digital 45. Park AY, Seo BK, Woo OH, et al. The utility of ultrasound superb microvas-
Mammography for Detection of Microcalcifications in Screening? Radiology cular imaging for evaluation of breast tumour vascularity: comparison with
2018;289(3):639–640. colour and power Doppler imaging regarding diagnostic performance. Clin
23. Fallenberg EM, Dromain C, Diekmann F, et al. Contrast-enhanced Radiol 2018;73(3):304–311.
spectral mammography: Does mammography provide additional clinical 46. Zhu YC, Zu DM, Zhang Y, et al. A comparative study on superb microvas-
benefits or can some radiation exposure be avoided? Breast Cancer Res Treat cular imaging and conventional ultrasonography in differentiating BI-RADS
2014;146(2):371–381. 4 breast lesions. Oncol Lett 2019;18(3):3202–3210.
24. Lalji UC, Jeukens CR, Houben I, et al. Evaluation of low-energy contrast- 47. Krouskop TA, Wheeler TM, Kallel F, Garra BS, Hall T. Elastic moduli
enhanced spectral mammography images by comparing them to full-field of breast and prostate tissues under compression. Ultrason Imaging
digital mammography using EUREF image quality criteria. Eur Radiol 1998;20(4):260–274.
2015;25(10):2813–2820. 48. Barr RG. Sonographic breast elastography: a primer. J Ultrasound Med
25. Francescone MA, Jochelson MS, Dershaw DD, et al. Low energy mam- 2012;31(5):773–783.
mogram obtained in contrast-enhanced digital mammography (CEDM) 49. Berg WA, Cosgrove DO, Doré CJ, et al. Shear-wave elastography improves
is comparable to routine full-field digital mammography (FFDM). Eur J the specificity of breast US: the BE1 multinational study of 939 masses.
Radiol 2014;83(8):1350–1355. Radiology 2012;262(2):435–449.

18 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020


Mann et al

50. Barr RG, Destounis S, Lackey LB 2nd, Svensson WE, Balleyguier C, Smith 76. Larson ED, Lee WM, Roubidoux MA, et al. Preliminary Clinical Experi-
C. Evaluation of breast lesions using sonographic elasticity imaging: a mul- ence with a Combined Automated Breast Ultrasound and Digital Breast
ticenter trial. J Ultrasound Med 2012;31(2):281–287. Tomosynthesis System. Ultrasound Med Biol 2018;44(3):734–742.
51. Destounis S, Arieno A, Morgan R, et al. Clinical experience with elasticity 77. Schaefgen B, Heil J, Barr RG, et al. Initial results of the FUSION-X-US
imaging in a community-based breast center. J Ultrasound Med 2013;32(2): prototype combining 3D automated breast ultrasound and digital breast
297–302. tomosynthesis. Eur Radiol 2018;28(6):2499–2506.
52. Barr RG, De Silvestri A, Scotti V, et al. Diagnostic Performance and Accuracy 78. Taylor JS, Tofts PS, Port R, et al. MR imaging of tumor microcirculation:
of the 3 Interpreting Methods of Breast Strain Elastography: A Systematic promise for the new millennium. J Magn Reson Imaging 1999;10(6):903–907.
Review and Meta-analysis. J Ultrasound Med 2019;38(6):1397–1404. 79. Sung JS, Stamler S, Brooks J, et al. Breast Cancers Detected at Screening MR
53. Lee SH, Chung J, Choi HY, et al. Evaluation of Screening US-detected Imaging and Mammography in Patients at High Risk: Method of Detection
Breast Masses by Combined Use of Elastography and Color Doppler US Reflects Tumor Histopathologic Results. Radiology 2016;280(3):716–722.
with B-Mode US in Women with Dense Breasts: A Multicenter Prospective 80. Bakker MF, de Lange SV, Pijnappel RM, et al. Supplemental MRI
Study. Radiology 2017;285(2):660–669. Screening for Women with Extremely Dense Breast Tissue. N Engl J Med
54. Seo M, Ahn HS, Park SH, et al. Comparison and Combination of Strain 2019;381(22):2091–2102.
and Shear Wave Elastography of Breast Masses for Differentiation of Benign 81. Breast magnetic resonance imaging (MRI) accreditation program require-
and Malignant Lesions by Quantitative Assessment: Preliminary Study. J ments. American College of Radiology. https://www.acraccreditation.org/
Ultrasound Med 2018;37(1):99–109. modalities/breast-mri. Accessed January 3, 2020.
55. Han J, Li F, Peng C, et al. Reducing Unnecessary Biopsy of Breast Lesions: 82. Kuhl CK, Schrading S, Strobel K, Schild HH, Hilgers RD, Bieling HB.
Preliminary Results with Combination of Strain and Shear-Wave Elastog- Abbreviated breast magnetic resonance imaging (MRI): first postcontrast
raphy. Ultrasound Med Biol 2019;45(9):2317–2327. subtracted images and maximum-intensity projection—a novel approach to
56. Barr RG. Breast Elastography: How to Perform and Integrate Into a “Best- breast cancer screening with MRI. J Clin Oncol 2014;32(22):2304–2310.
Practice” Patient Treatment Algorithm. J Ultrasound Med 2020;39(1):7–17. 83. Leithner D, Moy L, Morris EA, Marino MA, Helbich TH, Pinker K. Ab-
57. Rella R, Belli P, Giuliani M, et al. Automated Breast Ultrasonography (ABUS) breviated MRI of the Breast: Does It Provide Value? J Magn Reson Imaging
in the Screening and Diagnostic Setting: Indications and Practical Use. Acad 2019;49(7):e85–e100.
Radiol 2018;25(11):1457–1470. 84. Comstock CE, Gatsonis C, Newstead GM, et al. Comparison of Abbrevi-
58. Giuliano V, Giuliano C. Improved breast cancer detection in asymptomatic ated Breast MRI vs Digital Breast Tomosynthesis for Breast Cancer Detec-
women using 3D-automated breast ultrasound in mammographically dense tion Among Women With Dense Breasts Undergoing Screening. JAMA
breasts. Clin Imaging 2013;37(3):480–486. 2020;323(8):746–756.
59. Brem RF, Tabár L, Duffy SW, et al. Assessing improvement in detection of 85. Borthakur A, Weinstein SP, Schnall MD, Conant EF. Comparison of Study
breast cancer with three-dimensional automated breast US in women with Activity Times for “Full” versus “Fast MRI” for Breast Cancer Screening. J
dense breast tissue: the SomoInsight Study. Radiology 2015;274(3):663–673. Am Coll Radiol 2019;16(8):1046–1051.
60. Butler RS. Invited Commentary: Handheld or Automated-Staying Focused 86. Heacock L, Gao Y, Heller SL, et al. Comparison of conventional DCE-
on the Goals of Screening US, with Response from Drs van Zelst and Mann. MRI and a novel golden-angle radial multicoil compressed sensing method
RadioGraphics 2018;38(3):683–687. for the evaluation of breast lesion conspicuity. J Magn Reson Imaging
61. Wang Y, Nasief HG, Kohn S, et al. Three-dimensional Ultrasound Elastic- 2017;45(6):1746–1752.
ity Imaging on an Automated Breast Volume Scanning System. Ultrason 87. Mann RM, Mus RD, van Zelst J, Geppert C, Karssemeijer N, Platel B. A
Imaging 2017;39(6):369–392. novel approach to contrast-enhanced breast magnetic resonance imaging
62. Khodr ZG, Sak MA, Pfeiffer RM, et al. Determinants of the reliability of for screening: high-resolution ultrafast dynamic imaging. Invest Radiol
ultrasound tomography sound speed estimates as a surrogate for volumetric 2014;49(9):579–585.
breast density. Med Phys 2015;42(10):5671–5678. 88. Abe H, Mori N, Tsuchiya K, et al. Kinetic Analysis of Benign and Malig-
63. Li C, Duric N, Littrup P, Huang L. In vivo breast sound-speed imaging with nant Breast Lesions With Ultrafast Dynamic Contrast-Enhanced MRI:
ultrasound tomography. Ultrasound Med Biol 2009;35(10):1615–1628. Comparison With Standard Kinetic Assessment. AJR Am J Roentgenol
64. Ranger B, Littrup PJ, Duric N, et al. Breast ultrasound tomography versus 2016;207(5):1159–1166.
MRI for clinical display of anatomy and tumor rendering: preliminary results. 89. van Zelst JCM, Vreemann S, Witt HJ, et al. Multireader Study on the
AJR Am J Roentgenol 2012;198(1):233–239. Diagnostic Accuracy of Ultrafast Breast Magnetic Resonance Imaging for
65. Zografos G, Liakou P, Koulocheri D, et al. Differentiation of BIRADS-4 Breast Cancer Screening. Invest Radiol 2018;53(10):579–586.
small breast lesions via Multimodal Ultrasound Tomography. Eur Radiol 90. Shi RY, Yao QY, Wu LM, Xu JR. Breast Lesions: Diagnosis Using Diffusion
2015;25(2):410–418. Weighted Imaging at 1.5T and 3.0T-Systematic Review and Meta-analysis.
66. Duric N, Boyd N, Littrup P, et al. Breast density measurements with ul- Clin Breast Cancer 2018;18(3):e305–e320.
trasound tomography: a comparison with film and digital mammography. 91. Baltzer P, Mann RM, Iima M, et al. Diffusion-weighted imaging of the
Med Phys 2013;40(1):013501. breast-a consensus and mission statement from the EUSOBI Interna-
67. Sak M, Duric N, Littrup P, et al. Using Speed of Sound Imaging to Char- tional Breast Diffusion-Weighted Imaging working group. Eur Radiol
acterize Breast Density. Ultrasound Med Biol 2017;43(1):91–103. 2020;30(3):1436–1450.
68. O'Flynn EAM, Fromageau J, Ledger AE, et al. Ultrasound Tomography 92. McDonald RJ, McDonald JS, Kallmes DF, et al. Intracranial Gado-
Evaluation of Breast Density: A Comparison With Noncontrast Magnetic linium Deposition after Contrast-enhanced MR Imaging. Radiology
Resonance Imaging. Invest Radiol 2017;52(6):343–348. 2015;275(3):772–782.
69. Manohar S, Dantuma M. Current and future trends in photoacoustic breast 93. Bickelhaupt S, Laun FB, Tesdorff J, et al. Fast and Noninvasive Character-
imaging. Photoacoustics 2019;16:100134. ization of Suspicious Lesions Detected at Breast Cancer X-Ray Screening:
70. Dogan BE, Menezes GLG, Butler RS, et al. Optoacoustic Imaging and Gray- Capability of Diffusion-weighted MR Imaging with MIPs. Radiology
Scale US Features of Breast Cancers: Correlation with Molecular Subtypes. 2016;278(3):689–697.
Radiology 2019;292(3):564–572. 94. Amornsiripanitch N, Bickelhaupt S, Shin HJ, et al. Diffusion-weighted MRI
71. Neuschler EI, Butler R, Young CA, et al. A Pivotal Study of Optoacoustic for Unenhanced Breast Cancer Screening. Radiology 2019;293(3):504–520.
Imaging to Diagnose Benign and Malignant Breast Masses: A New Evalua- 95. Baltzer PA, Benndorf M, Dietzel M, Gajda M, Camara O, Kaiser WA. Sen-
tion Tool for Radiologists. Radiology 2018;287(2):398–412. sitivity and specificity of unenhanced MR mammography (DWI combined
72. Asao Y, Hashizume Y, Suita T, et al. Photoacoustic mammography capable with T2-weighted TSE imaging, ueMRM) for the differentiation of mass
of simultaneously acquiring photoacoustic and ultrasound images. J Biomed lesions. Eur Radiol 2010;20(5):1101–1110.
Opt 2016;21(11):116009. 96. Pinker K, Moy L, Sutton EJ, et al. Diffusion-Weighted Imaging With Apparent
73. Heijblom M, Piras D, Brinkhuis M, et al. Photoacoustic image patterns of Diffusion Coefficient Mapping for Breast Cancer Detection as a Stand-Alone
breast carcinoma and comparisons with Magnetic Resonance Imaging and Parameter: Comparison With Dynamic Contrast-Enhanced and Multipara-
vascular stained histopathology. Sci Rep 2015;5(1):11778. metric Magnetic Resonance Imaging. Invest Radiol 2018;53(10):587–595.
74. Menezes GLG, Pijnappel RM, Meeuwis C, et al. Downgrading of Breast 97. Expert Panel on Breast Imaging; Mainiero MB, Moy L, et al. ACR
Masses Suspicious for Cancer by Using Optoacoustic Breast Imaging. Radiol- Appropriateness Criteria® Breast Cancer Screening. J Am Coll Radiol
ogy 2018;288(2):355–365. 2017;14(11S):S383–S390.
75. Padia K, Douglas TS, Cairncross LL, Baasch RV, Vaughan CL. Detecting Breast 98. Huppe AI, Mehta AK, Brem RF. Molecular Breast Imaging: A Comprehensive
Cancer with a Dual-Modality Device. Diagnostics (Basel) 2017;7(1):E17. Review. Semin Ultrasound CT MR 2018;39(1):60–69.

Radiology: Volume 00: Number 0— 2020 n radiology.rsna.org 19


Novel Approaches to Screening for Breast Cancer

99. Hruska CB. Molecular Breast Imaging for Screening in Dense Breasts: State of 120. Barinov L, Jairaj A, Becker M, et al. Impact of Data Presentation on Physician
the Art and Future Directions. AJR Am J Roentgenol 2017;208(2):275–283. Performance Utilizing Artificial Intelligence-Based Computer-Aided Diag-
100. Narayanan D, Berg WA. Dedicated Breast Gamma Camera Imaging and Breast nosis and Decision Support Systems. J Digit Imaging 2019;32(3):408–416.
PET: Current Status and Future Directions. PET Clin 2018;13(3):363–381. 121. Cho E, Kim EK, Song MK, Yoon JH. Application of Computer-Aided Di-
101. Rauch GM, Adrada BE. Comparison of Breast MR Imaging with Molecular agnosis on Breast Ultrasonography: Evaluation of Diagnostic Performances
Breast Imaging in Breast Cancer Screening, Diagnosis, Staging, and Treatment and Agreement of Radiologists According to Different Levels of Experience.
Response Evaluation. Magn Reson Imaging Clin N Am 2018;26(2):273–280. J Ultrasound Med 2018;37(1):209–216.
102. Taillefer R. Clinical applications of 99mTc-sestamibi scintimammography. 122. van Zelst JCM, Tan T, Clauser P, et al. Dedicated computer-aided detec-
Semin Nucl Med 2005;35(2):100–115. tion software for automated 3D breast ultrasound; an efficient tool for the
103. Berg WB, Leung JT, eds. Diagnostic Breast Imaging: Breast. 3rd ed. Phila- radiologist in supplemental screening of women with dense breasts. Eur
delphia, Pa: Elsevier, 2019; 190–201. Radiol 2018;28(7):2996–3006.
104. Tao AT, Hruska CB, Conners AL, et al. Dose Reduction in Molecular Breast 123. Xu X, Bao L, Tan Y, Zhu L, Kong F, Wang W. 1000-Case Reader Study of
Imaging With a New Image-Processing Algorithm. AJR Am J Roentgenol Radiologists’ Performance in Interpretation of Automated Breast Volume
2020;214(1):185–193. Scanner Images with a Computer-Aided Detection System. Ultrasound Med
105. Yamamoto Y, Tasaki Y, Kuwada Y, Ozawa Y, Inoue T. A preliminary report Biol 2018;44(8):1694–1702.
of breast cancer screening by positron emission mammography. Ann Nucl 124. Grimm LJ, Mazurowski MA. Breast Cancer Radiogenomics: Current Status
Med 2016;30(2):130–137. and Future Directions. Acad Radiol 2020;27(1):39–46.
106. Miladinova D. Molecular Imaging in Breast Cancer. Nucl Med Mol Imaging 125. Pinker K, Chin J, Melsaether AN, Morris EA, Moy L. Precision Medicine
2019;53(5):313–319. and Radiogenomics in Breast Cancer: New Approaches toward Diagnosis
107. Geras KJ, Mann RM, Moy L. Artificial Intelligence for Mammography and and Treatment. Radiology 2018;287(3):732–747.
Digital Breast Tomosynthesis: Current Concepts and Future Perspectives. 126. Truhn D, Schrading S, Haarburger C, Schneider H, Merhof D, Kuhl C.
Radiology 2019;293(2):246–259. Radiomic versus Convolutional Neural Networks Analysis for Classification
108. Rodríguez-Ruiz A, Krupinski E, Mordang JJ, et al. Detection of Breast of Contrast-enhancing Lesions at Multiparametric Breast MRI. Radiology
Cancer with Mammography: Effect of an Artificial Intelligence Support 2019;290(2):290–297.
System. Radiology 2019;290(2):305–314. 127. Dalmiş MU, Gubern-Mérida A, Vreemann S, et al. Artificial Intelligence–
109. Wu N, Phang J, Park J, et al. Deep Neural Networks Improve Radiolo- Based Classification of Breast Lesions Imaged With a Multiparametric Breast
gists’ Performance in Breast Cancer Screening. IEEE Trans Med Imaging MRI Protocol With Ultrafast DCE-MRI, T2, and DWI. Invest Radiol
2020;39(4):1184–1194. 2019;54(6):325–332.
110. McKinney SM, Sieniek M, Godbole V, et al. International evaluation of an 128. Dalmış MU, Vreemann S, Kooi T, Mann RM, Karssemeijer N, Gubern-
AI system for breast cancer screening. Nature 2020;577(7788):89–94. Mérida A. Fully automated detection of breast cancer in screening MRI
111. Conant EF, Toledano AY, Periaswamy S, et al. Improving Accuracy and using convolutional neural networks. J Med Imaging (Bellingham)
Efficiency with Concurrent Use of Artificial Intelligence for Digital Breast 2018;5(1):014502.
Tomosynthesis. Radiol Artif Intell 2019;1(4):e180096. 129. Herent P, Schmauch B, Jehanno P, et al. Detection and characteriza-
112. Rodriguez-Ruiz A, Lång K, Gubern-Merida A, et al. Can we reduce the tion of MRI breast lesions using deep learning. Diagn Interv Imaging
workload of mammographic screening by automatic identification of 2019;100(4):219–225.
normal exams with artificial intelligence? A feasibility study. Eur Radiol 130. Zhou J, Zhang Y, Chang KT, et al. Diagnosis of Benign and Malignant Breast
2019;29(9):4825–4832. Lesions on DCE-MRI by Using Radiomics and Deep Learning With Con-
113. Yala A, Schuster T, Miles R, Barzilay R, Lehman C. A Deep Learning sideration of Peritumor Tissue. J Magn Reson Imaging 2020;51(3):798–809.
Model to Triage Screening Mammograms: A Simulation Study. Radiology 131. Reig B, Heacock L, Geras KJ, Moy L. Machine learning in breast MRI. J Magn
2019;293(1):38–46. Reson Imaging 2019. 10.1002/jmri.26852. Published online July 5, 2019.
114. Lehman CD, Yala A, Schuster T, et al. Mammographic Breast Density 132. Bluemke DA, Moy L, Bredella MA, et al. Assessing Radiology Research
Assessment Using Deep Learning: Clinical Implementation. Radiology on Artificial Intelligence: A Brief Guide for Authors, Reviewers, and Read-
2019;290(1):52–58. ers—From the Radiology Editorial Board. Radiology 2020;294(3):487–489.
115. Dembrower K, Liu Y, Azizpour H, et al. Comparison of a Deep Learning 133. Alimirzaie S, Bagherzadeh M, Akbari MR. Liquid biopsy in breast cancer:
Risk Score and Standard Mammographic Density Score for Breast Cancer A comprehensive review. Clin Genet 2019;95(6):643–660.
Risk Prediction. Radiology 2020;294(2):265–272. 134. Yates LR, Gerstung M, Knappskog S, et al. Subclonal diversification
116. Yala A, Lehman C, Schuster T, Portnoi T, Barzilay R. A Deep Learning of primary breast cancer revealed by multiregion sequencing. Nat Med
Mammography-based Model for Improved Breast Cancer Risk Prediction. 2015;21(7):751–759.
Radiology 2019;292(1):60–66. 135. Lee JH, Jeong H, Choi JW, Oh HE, Kim YS. Liquid biopsy prediction of
117. Akselrod-Ballin A, Chorev M, Shoshan Y, et al. Predicting Breast Cancer axillary lymph node metastasis, cancer recurrence, and patient survival in
by Applying Deep Learning to Linked Health Records and Mammograms. breast cancer: A meta-analysis. Medicine (Baltimore) 2018;97(42):e12862.
Radiology 2019;292(2):331–342. 136. Phallen J, Sausen M, Adleff V, et al. Direct detection of early-stage cancers
118. Ciritsis A, Rossi C, Eberhard M, Marcon M, Becker AS, Boss A. Automatic using circulating tumor DNA. Sci Transl Med 2017;9(403):eaan2415.
classification of ultrasound breast lesions using a deep convolutional neural 137. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable
network mimicking human decision-making. Eur Radiol 2019;29(10):5458– cancers with a multi-analyte blood test. Science 2018;359(6378):926–930.
5468. 138. Adjiri A. DNA Mutations May Not Be the Cause of Cancer. Oncol Ther
119. Fujioka T, Kubota K, Mori M, et al. Distinction between benign and ma- 2017;5(1):85–101.
lignant breast masses at breast ultrasound using deep learning method with 139. Underwood JJ, Quadri RS, Kalva SP, et al. Liquid Biopsy for Cancer: Review
convolutional neural network. Jpn J Radiol 2019;37(6):466–472. and Implications for the Radiologist. Radiology 2020;294(1):5–17.

20 radiology.rsna.org n Radiology: Volume 00: Number 0— 2020

You might also like