Sabcs 2014 Allabstracts
Sabcs 2014 Allabstracts
Sabcs 2014 Allabstracts
P-Value
Hazard Ratio
PTEN
0.002
2.11
PIK3CA
0.007
0.69
DDR1
0.008
2.41
ARID1B
0.01
0.57
ERBB3
0.02
0.36
MAP3K1
0.01
0.5
TP53
0.04
1.48
GATA3
0.04
0.44
JAK2
0.05
2.1
Arm A (N=489)
Variable
Arm C (N=456)
HR (95% CI)
p-value
HR (95% CI)
p-value
Lymph node(-)
1.00 (ref)
<0.0001
1.00 (ref)
0.013
1-3+
4-9
10+
Nodal status
HR status
negative
1.00 (ref)
positive
0.0198
1.00 (ref)
0.42
S-TILs status
<60%
1.00 (ref)
60%
Tumor grade
0.007
1.00 (ref)
1.08 (0.42, 2.79)
0.87
Grade 1 or 2
1.00 (ref)
0.50
1.00 (ref)
0.51
Grade 3
Tumor size
0.12
0.92
Age
0.13
1 (0.98, 1.03)
0.87
Conclusions: In exploratory analyses from this subset HER2+ population from N9831, S-TILs were associated with RFS in
patients treated with chemotherapy alone, and were not shown to be associated with RFS in patients treated with chemotherapy
plus trastuzumab.
Gene %
Gene %
Gene %
CDH1 62.9
MAP3K1 7
USP9X 4.4
EPHB6 3.8
PIK3CA 44.9
BRCA2 6.6
ATR 4.2
MED12L 3.6
MLL3 15.8
ERBB2 6.4
COL22A1 4.2
PTEN 3.6
TBX3 13.2
ARID1B 5.6
MED13 4.2
ERBB3 3.2
FOXA1 9.6
ATM 4.8
NOTCH1 4.2
ROS1 3.2
MLL2 9.4
MLL 4.6
AKT1 4.2
BPTF 3.2
ARID1A 8.4
MYO5B 4.4
MYO3A 4
BRCA1 3.2
GATA3 8
EP300 4.4
NF1 4
IRS2 3.2
TP53 7.4
RUNX1 4.4
EP400 3.8
NOTCH4 3
25.3-46.1) p=0.75 & 36 pts non-basal subtypes, ORR=16.7% with C (3/18, 95%CI 3.6-41.4) & 73.7% with D (13/18, 95%CI
46.5-90.3) p<0.01. Interaction p=0.01.
Discussion
While TNT gives no evidence to support superior activity of C compared to D in unselected TNBC pts results for gBRCA+ pts
support their greater sensitivity to C than D. Neither a dichotomised MYRIAD HRD score nor 2 basal-like classifiers selected
sensitivity to C. The sensitivity to D of a non-basal-like subgroup by Prosigna PAM50 warrants independent investigation.
Endpoint
AC (%)
FEC (%)
HR
CI
p-value
DFS
83.0
82.8
1.04
0.85,1.26
0.70
OS
91.2
92.0
0.94
0.71,1.24
0.65
RFI
90.0
90.4
0.97
0.76,1.26
0.84
DRFI
92.3
93.0
0.89
0.66,1.20
0.43
Conclusions
Six cycles of the FEC-100 regimen did not result in any efficacy advantage over 4 cycles of standard AC in node-negative breast
cancer pts. As anticipated, the FEC-100 regimen resulted in greater toxicity.
Support
NCI: U10-CA-12027, -37377, -69974, -69651, -44066-26; Pharmacia & Upjohn Co.
No chemo
(n=949)
Prior Chemo
(n=1084)
Primary Analysis
(n=2033)
46
40
43
Node positive
9%
57%
35%
Tumor > 2 cm
14%
47%
32%
Grade 1
41%
14%
27%
Grade 3
7%
35%
22%
70/47/13/10
229/213/172/96
299/260/185/106
Events @ 67 months
DFS/BCFI/distant/deaths
After a median follow-up of 67 months, 5-yr DFS was 86.6% in the T+OFS group and 84.7% in the tamoxifen group (HR=0.83;
95%CI, 0.86-1.04; P=0.10). Multivariable allowance for prognostic factors suggested a greater treatment effect (HR 0.78; 95% CI,
0.62-0.98). Overall survival is not mature with 5-yr OS of 96.7% for T+OFS and 95.1% for tamoxifen (HR 0.74; 95%CI, 0.51-1.09;
P=0.13). In the no-chemotherapy cohort, one-third of events were not breast-cancer related and BCFI was > 95% with tamoxifen
alone. The cohort who remained premenopausal after chemotherapy had a 4.5% absolute improvement in 5-yr BCFI with T+OFS
vs T (table). In the chemotherapy cohort, 5-yr overall survival was 94.5% for T+OFS and 90.9% for T (HR=0.64; 95% CI,
0.42-0.96).
Prior
Chemo
Cohort
T
116/542
78.0%
T+OFS
97/542
E+OFS
80/544
82.5%
85.7%
5-yr DRFI
T
90/542
83.6%
T+OFS
82/542
84.8%
E+OFS
67/544
87.8%
Non-adherence with OFS reached 22% at 4 years. Targeted > grade 3 toxicities were reported for 31.3% of T+OFS group and
23.7% of tamoxifen group. Menopausal symptoms, depression, musculoskeletal complaints, hypertension and diabetes were
more frequent with T+OFS. Osteoporosis was reported in 5.8% for T+OFS and 3.5% for T.
Conclusions: Adding ovarian suppression to tamoxifen did not show significant benefit in the overall population in SOFT after 67
months median follow-up. However, for the cohort of women who received chemotherapy and who remained premenopausal, the
addition of ovarian suppression improved breast cancer outcomes, and further improvement was seen with the use of
exemestane plus ovarian suppression.
Year 2
Year 3
Year 4
Screening Mammograms
30670
32050
32230
27937
2706
3351
4128
3331
151
180
178
53
Cancers
11
11
13
11
PPV
7.1
6.1
8.1
17.2
4.0
3.2
3.2
3.3
% Eligible Screened
22.1
26.1
32.0
28.3
CONCLUSIONS: Based on the data collected from these sites, screening breast ultrasound in women with dense breast
parenchyma detects mammographically occult malignancy. Over the 4 years studied, the PPV improved from 7% to 17.2%
indicating that the selection of lesions biopsied was more accurate with fewer false positives. The rate of detection in the first year
was 4.0/1000 and then remained stable at 3.2/1000 in the three subsequent years. Of concern, the number of eligible women
who elect to have the additional test remains low at about 30% which is due to several factors including education and cost.
BCS Alone, CM
DCIS Score Risk
Group
Low (<39)
12.7% (9.5%,16.9%)
N=355
9.7% (6.8%,13.8%)
N=298
Int (39-54)
33.0% (23.6%,44.8%)
N=95
27.1% (17.7%,40.2%)
N=72
13.6% (8.6%,21.2%)
N=155
High (>55)
20.6% (10.3%,38.7%)
N=37
27.8% (20.0%,37.8%)
N=121
27.0% (18.2%,38.9%)
N=87
20.5% (15.1%,27.5%)
N=202
33.3% (21.9%,48.5%)
N=49
<0.001
<0.001
<0.001
<0.001
ER-positive
8.7% (2/23)
33.2% (13/39)
ER-negative
20% (2/10)
8.7% (2/23)
Overall pCR
12.2% (4/33)
24.2% (15/62)
Conclusions: Treatment with L+T (with endocrine therapy in ER+ tumors) for 24 weeks leads to doubling of the pCR rate in
women with HER2+ breast cancer without using cytotoxic chemotherapy. This approach is effective and well tolerated and
warrants study as part of a de-escalation strategy that may spare some patients the cost and toxicity of chemotherapy. Tissue
obtained on this trial will provide a valuable resource to validate correlative findings from our prior studies and discover new
biomarkers to help guide proper patient selection for treatment.
surrogates: 8.7 (7.8-9.7) for Luminal A; 8.3 (6.9-9.4) for Luminal B/HER-2neg; 9.3 years (5.9-21.1) for Luminal B/HER-2+. Similar
results were seen for RFS. ER/PR/AR using histoscores will be presented. Conclusions: a) 56% pts had T1 tumors at Dx but only
4% had BCS; b) ER was highly + in >90% but adjuvant ET given in only 77% pts; c) Male BC is usually ER+, PR+ & AR+ and of
Luminal A-like subtype (5% HER2pos & 1% TNBC); d) Significant improvement in OS over time; e) ER and PR (Allred) are
prognostic (high expression/better prognosis), less for AR, not for Ki67 nor IHC surrogates; f) In-depth characterization of
samples is ongoing. Funding: BCRF, EBCC Council, Pink Ribbon NL, BRO.
TOTAL (N=1781)
MOBC (N=110)
MEBC (N=1671)
P-VALUES
p<0.001
94
91
Scattered fibroglandular
587
38
25
23
562
40
Heterogeneously dense
715
47
67
61
648
46
Extremely dense
135
14
13
121
74
16
15
24
59
15
Mild
225
49
30
48
195
49
Moderate
116
25
13
21
103
26
Marked
47
10
42
11
p=0.29
FIBROGLANDULAR TISSUE
Predominantly fatty
91
21
12
84
23
Scattered fibroglandular
160
37
24
41
136
37
Heterogeneously dense
121
28
20
34
101
27
p=0.28
Extremely dense
57
13
14
49
13
Pathologic Node
Positive
All Cases (n=763) 234 (31%)
Sensitivity (95%
CI)
52.6 (46-59.1)
Specificity (95%
NPV (95% CI) PPV (95% CI)
CI)
Accuracy (95%
CI)
77.9 (74.1-81.3)
78.8 (75-82.1)
51.3
(44.8-57.7)
70.1 (66.7-73.3)
194 (32%)
51.0 (43.8-58.2)
80.0 (75.8-83.7)
77.8
(73.4-81.5)
54.4
(46.9-61.7)
70.8 (67-74.3)
HER2+ (n=69)
20 (29%)
65.0 (40.9-83.7)
71.4 (56.5-83)
83.3 (68-92.5)
48.1
(29.2-67.6)
69.6 (57.2-79.8)
58.8 (33.5-80.6)
75.0 (59.4-86.3)
82.5
(66.6-92.1)
p value
0.43
0.33
0.56
0.73
0.98
0.74
58 (22%)
32.8 (21.4-46.5)
79.6 (73.2-84.8)
80.4
(74.1-85.5)
31.7
(20.6-45.1)
69.1 (63.0-74.6)
2 (n=351)
124 (35%)
55.6 (46.5-64.5)
79.7 (73.8-84.6)
76.7
(70.7-81.8)
60.0
(50.4-68.9)
71.2 (66.1-75.9)
3 (n=148)
51 (34%)
66.7 (52.0-79.0)
70.1 (59.8-78.8)
80.0
(69.6-87.6)
54.0
(41.0-66.4)
68.9 (60.7-76.1)
p value
0.001
0.0003
0.11
0.72
0.02
0.93
Diagnostic performance did not vary significantly based on patient body mass index (BMI) or approximated biologic subtype, but
specificity was better for patients >age 50 (p=0.007) and sensitivity and PPV both were worse for grade 1 vs grade 2/3 tumors.
Node positivity rate at operation was 48% (51/106) for patients with a solitary abnormal node on MRI and 54% (72/134) when >1
suspicious node was seen (p=0.39). However, multiple vs solitary MRI suspicious nodes correlated with 3 positive nodes at
operation (40 [30%] vs 19 [18%], p=0.03) and pN2/pN3 disease (29 [22%] vs 12 [11%], p=0.03).
Conclusion: Axillary lymph node findings on MRI for breast cancer predict nodal status and disease volume in invasive breast
cancer patients. Tumor biologic subtype did not affect performance characteristics of preoperative MRI, but tumor grade did
influence the sensitivity and PPV of MRI. When MRI is performed in the evaluation of newly diagnosed breast cancer, axillary
findings inform pathologic nodal stage at operation regardless of tumor subtype.
|BPE|
BPE%
Conclusions
Increased BPE quantified from DCE-MRI are predictive of breast cancer risk, independent of measures of breast density and
FGT. BPE estimated from three time-point SUB sequences has a similar predictive effect of breast cancer risk, while an early
sequence (e.g., SUB 1) appears to have a larger magnitude of effect than that of a delayed sequence (e.g., SUB 3).
Clinical Relevance
Quantified BPE in breast DCE-MRI has potential for use as a biomarker of breast cancer risk and may be included to improve
breast cancer risk prediction. A single post-contrast sequence (i.e., SUB 1) may be adequate for use to estimate breast cancer
risk using breast MRI in the context of breast cancer screening for high-risk women.
Low-risk: N=47
N=5
Low-risk (2) N= 42
Intermediate-risk: N=144
N=38
High-risk: N=87
Total: N=278
Total: N= 43
Total N= 278
Table 1: The group was divided in three groups: a low- (T2N0), intermediate- (T0-2N1 and T3N0) and a high-risk group
(T0-3N2-3, T3N1 and T4). The table shows the upstaging of breast cancer patients from low- and intermediate- to the high-risk
group, and thus, after PET/CT, requiring radiotherapy.
: The patients not upstaged by PETCT showed no difference in PFS between the highrisk, intermediaterisk and lowrisk
groups (Logrank p=0.18). Due to the migration of the 43 patients from the low and intermediate group to the highrisk group,
based on PETCT findings, the PFS differed significantly between the riskgroups (Logrank p=0.04). No difference in
locoregional recurrence was seen between the lowrisk and the highrisk group (P=0.18).
Conclusion:
After upstaging with PETCT, into the highrisk group requiring radiotherapy, a significant difference is seen between the three
riskgroups. PETCT restaging may more adequately predict progression free survival. The detection occult lymphatic metastasis
with PETCT leads to upstaging in clinically unsuspected patients with primary breast cancer, enabling adequate radiotherapy
treatment.
n(%)
Peak intensity
Mean SD
p
0.50
53.8 13.5
< 50 y
30 (55.6)
65.5 25.4
50 y
24 (44.4)
61.1 21.5
1 or 2
19 (35.2)
65.8 28.0
35 (64.8)
62.4 21.2
< 30
24 (44.4)
53.8 22.4
30
30 (55.6)
71.4 21.9
Negative
13 (31.7)
58.2 20.9
Positive
41 (68.3)
80.5 24.6
Negative
47 (87.0)
62.3 24.0
Positive
7 (13.0)
71.9 20.5
NG
0.61
Ki67
0.006
ER
0.002
HER2
0.32
ER: estrogen receptor, HER2: human epidermal growth factor receptor 2, NG: nuclear grade.
Also, Thirty-eight, 7, and 9 patients had luminal, HER-2-positive and triple-negative tumors, respectively. The PI values for these
tumors were 56.8 20.9, 71.9 20.5, and 85.7 23.2, respectively. And, the PI value was significantly greater in the
triple-negative, than in luminal tumors (p = 0.001).
Table2. Relationship between signal intensity as a perfusion parameter and tumor subtypes
Tumor subtype
Subtype
Peak intensity
Mean SD
p*
Luminal
38
56.8 20.9
HER-2-positive
71.9 20.5
0.09
Triple-negative
85.7 23.2
0.001
Furthermore, PI significantly correlated with the Ki-67 value (Spearman r = 0.54, P = 0.00002).
Conclusions
These findings indicated that PI has excellent predictive value for grade malignancy in breast cancer and might help to determine
appropriate therapeutic strategies.
Key points
Contrast-enhanced ultrasonography (CEUS) enables the real-time evaluation of detailed hemodynamics in breast cancer.
Peak intensity (PI) was significantly associated with Estrogen Receptors and Ki-67 assessed by immunohistochemistry.
PI significantly correlated with the Ki-67 value, indicating that PI reflects the grade of proliferative activity in tumors.
Analyses of contrast-effect intensity will be applied to evaluate grades of malignancy and determine treatment strategies.
Number Average Size Sentinel Node Family History Prior Self Cancer Prior Screening Usg
1.2 cm
1.7 cm
1 micro-met
1.4 cm
3 macromet
0.7 cm
1.5 cm
n/a
1.2 cm
1.3 cm
DCIS Grade 2
1.3 cm
DCIS Grade 3
1.9 cm
LCIS
n/a
ADH /Pappiloma
0.5 cm
Tubular
0.4 cm
Mucinous
8.0 cm
Discussion: In our first four years of tracking patients with dense breasts, the addition of bilateral breast ultrasound diagnosed a
variety of cancers and high risk lesions. These were not visible on screening mammography and were not palpable. There were 4
with node positive disease diagnosed in high nuclear grade tumors. Most patients had no additional risk factors other than dense
breasts. In the third and fourth year, cancers were found in patients who had a prior screening usg. Their mammograms remained
negative. The cancers were small and node negative.
Conclusion: The successful treatment of breast cancer is based on several variables including type of cancer, grade, size at
diagnosis, and stage at diagnosis. We do not know at what point these lesions diagnosed with screening breast usg would have
been clinically evident, either on mammography or physical examination. One might consider that the treatment outcome would
be altered if the lesions were larger and with more positive lymph nodes.
CBE
Mammogram
Ultrasound
85
59
76
90
79
78
71
79
82
67
74
84
CONCLUSIONS
A summary of clinical studies conducted on symptomatic patients to date suggest that PI is (a) sensitive in the detection of breast
masses and b) identifies breast cancer with similar sensitivity and specificity compared with other standard imaging modalities
while being cost effective. PI may fill a gap in developing countries where little breast screening infrastructure exists. Ongoing
clinical research with PI is warranted to evaluate its potential in the early diagnosis and screening for breast cancer in the U.S.
HR (95% CI)
P value
Presentation
1.64 (0.43-6.22)
0.47
Palpable
yes vs. no
1.54 (0.36-6.69)
0.58
Lesion size
1.23 (0.34-4.40)
0.76
Mass formation
yes vs. no
1.28 (0.36-4.44)
0.7
BI RADS category
5 vs. 3, 4
2.92 (0.80-10.7)
0.11
Biopsy device
2.16 (0..64-7.30)
0.21
Nuclear grade
0.29
SUVmax
11.7 (3.70-37.0)
< 0.001
BI RADS, Breast Imaging Reporting and Data System; DCIS, ductal carcinoma in situ; IBC, invasive breast cancer; SUVmax,
maximum standardized uptake value
Conclusion: SUVmax on FDG-PET/CT is useful for predicting the underestimation of invasive breast cancer in cases of DCIS at
needle biopsy.
4-Month
Endoxifen
P-val vs.
EM/EM
Median (SD)
P-val vs.
PM/PM
P-val vs.
EM/EM
Median (SD)
P-val vs.
PM/PM
EM/EM
NA
p=0.0001
8.23 (5.09)
NA
p=0.007
EM/IM
56 8.02 (4.75)
p=0.09
p=0.002
13.11 (9.38)
p<0.0001
p<0.0001
EM/PM
74 5.72 (4.45)
p=0.0001
p=0.02
8.91 (5.28)
p=0.42
p=0.003
IM/IM
17 4.29 (4.10)
p=0.001
p=0.26
6.52 (5.53)
p=0.27
p=0.24
IM/PM
32 3.90 (3.17)
p<0.0001
p=0.48
5.82 (3.47)
p=0.0009
p=0.77
PM/PM
13 3.33 (2.89)
p=0.0001
NA
6.08 (2.57)
p=0.007
NA
Total
Age 51 (with
positive FH)
Number of patients
250
74
74
75
27
14
(5.6%)
1 (1.4%)
8 (10.8%)
4 (5.3%)
1 (3.7%)
BRCA 1
BRCA 2
All 7 BRCA1 mutation carriers had a positive family history, were between 32 and 48 years of age, and had Grade 3 IDC with
negative ER, PR, HER2 receptors (TNBC). Six BRCA2 mutation carriers had IDC with positive hormone receptors (HR) and 2
had HER2-positive disease. We found 31 VUS. One VUS (BRCA2) was seen in two sisters with breast cancer. One VUS
(BRCA2) was seen in 4 patients and another in 2 patients, while 2 VUS (BRCA1) mutations were seen in 2 sets of 2 patients. The
significance of these VUS cannot be ascertained at this time. Haplotype analysis is ongoing.
Conclusions:
This is the first large study of ethnic Lebanese Arab women with breast cancer. The prevalence of BRCA deleterious mutations in
women with breast cancer who are considered high risk of carrying a BRCA mutation is 5.6% in our total cohort, while in patients
40 with positive FH it is 10.6%. Those numbers are lower than expected from US and European populations. Tumor grade and
pathology characteristics in this patient population correlated with that previously documented for BRCA1 (TNBC) and BRCA2
(positive HR) associated breast cancers. Our data supports use of young age together with positive FH should be used to select
patients for counseling and BRCA testing in Lebanon and Arab countries with resource-sensitive guidelines. Several VUS were
found in patients and sisters with breast cancer. The finding that 94.4% of high risk patients had no deleterious BRCA mutations
suggests the need to look for alternate gene mutations and other factors that may contribute to the development of breast cancer
in these high risk patients. Conclusions regarding haplotypes and diversity will be reported at the meeting.
Breast Biopsy
Recurrence Concordant
Recurrence Discordant
ER Positive
98/127
82/95
13/95 (14%)
ER Negative
29/127
24/27
5/27 (19%)
Total ER Discordance
18/122 (15%)
HER2 Positive
19/119
11/14
3/14 (22%)
HER2 Negative
100/119
63/69
6/69 (9%)
9/83 (11%)
Conclusion: Tumor discordance of the original cancer biopsy and recurrence is not uncommon. Our pilot study demonstrated that
ER and HER2 discordance occurred in 15% and 11% of cases, respectively. Though our pilot study was limited by small sample
size, we found that IHC testing of the surgical breast and lymph node specimen may provide additional clinical information and
affect management. Of the two cases that had a positive lymph node available, one was HER2 positive and concordant with the
recurrence. Of the seven breast specimens tested, one was ER positive and concordant with the recurrence. Had IHC testing
been performed at that time of surgery, adjuvant treatment management would have been altered. Further testing of our IHC
discordant recurrence patient population will be pursued to investigate the potential benefits of surgical breast and lymph node
IHC testing.
Luminal A
Luminal B
HER2 Enriched
TNBC
Normal Weight
8 (26.67%)
15 (50%)
5 (16.67%)
2 (6.67%)
Overweight
9 (20.93%)
17 (39.53%)
2 (4.65%)
15 (34.88%)
Obese
11 (15.71%)
37 (52.86%)
3 (4.29%)
19 (27.14%)
Conclusion: Our study suggests that obesity may provide a microenvironment that support and accelerate tumor growth,
particularly in luminal breast cancer subtypes. Furthermore, the poor outcome seen in obese patients may also be in part due to
more aggressive tumor subtype.
Hormone Therapy
Baseline (n=23) 6 months (n=12) Baseline (n=14) 6 months (n=11) Baseline (n=8) 6 months (n=2)
BMI (kg/m2)
27.8
26.3
28.7
29.3
32.0
28.2
39.5
36.3
42.0
41.1
43.0
36.1
43.9
47.3
46.5
50.6
49.4
45.6
HOMA-IR*
1.75
1.77
1.21
1.81
2.10
1.18
68.2
44.1
70
53
97
88.5
1.7
1,2
1.7
1.3
2.3
2.4
12.3
6.39
9.5
6.9
11.2
10.6
*HOMA-IR was not assessed in patients known to have diabetes. ^Time to zero power quantifies the time over which patients can
sustain generating motive power as a measure of endurance.
Week long bi-axial pedometry studies at baseline and six months revealed a decrease in pt activity between 17 and 23 percent,
confirming the decrease in motive power seen in the power envelope and body composition tests. Enrollment continues to a
planned total of 75 pts. Additional follow-up with 12 month assessment, as well as QOL and serum data will be presented.
Conclusions: Pts in this study were more deconditioned than expected based on population normative data. Systemic therapy
had a profound impact on body composition with a substantial decrease in lean muscle mass with a corresponding increase in %
body fat and decrease in the ability to generate and sustain motive power. Our data suggest that most patients would not be able
to participate in or comply with commonly proposed exercise interventions and physical activity recommendations. Future
interventions need to be restructured, following the model of individualized rehabilitation with careful assessment of the pts
baseline status to improve the health of breast cancer survivors.
BC patients
controls
waist circumference
(cm)
85 12
81 10
.01
Glucose (mmol/L)
5.5 1.0
5.2 0.9
.01
Insulin (pmol/L)
60.9 50.5
47.1 28.3
.06
HOMA
2.25 2.24
1.64 1.24
.03
Triglycerides (mmol/L)
1.28 0.64
1.10 0.57
.01
BMI (kg/m2)
26.3 4.9
25.4 4.5
.26
Despite exclusion of BC patients with diabetes at study entry, 24.9% of BC survivors self reported diabetes or pre-diabetes
(1.99%/year) versus 12.6% in controls (OR 2.3, p= .0017).
Conclusion: The metabolic status of long-term BC survivors deteriorated over time and age-adjusted at LTFU were worse with
respect to a number of factors compared to the control group.
Circumference
3SD SOAC*
2SD SOAC*
Commonly-used thresholds examined
200 ml interlimb difference
10% difference
* Dylke et al, 2012; Sum of arm circumferences (SOAC); Circumference measure (Circ)
Results: For those with widespread dermal backflow, any clinical diagnostic criteria could differentiate between those with and
without lymphedema. In contrast, for those with mild to moderate dermal backflow, only the normatively-determined threshold, set
at 2 standard deviations above the norm, for arm circumference and full arm bioimpedance (Cornish et al 2001) had adequate
sensitivity and specificity. Both of these thresholds had clinically relevant positive (23 and 10 respectively) and negative (0.2 and
0.3) likelihood ratios.
Conclusion: Evidence-based diagnostic thresholds have been established for the diagnosis of secondary upper limb
lymphedema. In determining if lymphoedema is present in those with mild lymphedema, normatively-determined circumference
and bioimpedance thresholds that account for limb dominance should be used. Adoption of these evidence-based criteria will
allow, for the first time, comparison between studies, clarifying the incidence and risk factors for lymphedema, allowing the field to
make meaningful progress forward in determining who is at-risk for lymphedema and how to prevent it from developing.
Acknowledgements: Cancer Australia and National Breast Cancer Foundation.
Table 1
African American (AA) N=96
p-value
58.8 (11.6)
62.9 (13.2)
0.066
51 (53.1%)
29 (67.4%)
0.114
22 (23.2%)
18 (41.9%)
0.025
1 (4.6%)
1 (5.6%)
1.000
7 (7.6%)
3 (7.1%)
4 (57.1%)
3 (100%)
27 (28.7%)
20 (47.6%)
2 (2.2%)
1 (2.4%)
1.000
0.475
0.032
1.000
*standard deviation
No. of
Stage
patients
% having at least
one imaging test
(total)
14,113
83% (11,713)
40,464 (3.5)
55% (6,420)
11,202 (28%)
13,123
94% (12,330)
53,729 (4.4)
65% (7,983)
16,292 (30%)
The most common initial (i.e. not confirmatory) imaging modalities of the skeleton, thorax and abdomen, for both groups were,
bone scan (19%; 12,799/66,699), chest x-ray (28%; 18,789/66,699) and abdominal ultrasound (20%; 13,387/66,699) respectively.
Use of advanced imaging (isotope bone scans, CT, MRI, PET) to look at potential sites of metastasis represented 38%
(11,063/29,262) and 46% (17,180/37,437) of initial imaging tests in stage 1 and 2 BC patients respectively.
Conclusion: Imaging for distant metastatic disease in patients with stage 1 and 2 BC is commonly performed in Ontario despite
multiple guidelines recommending against this. Advanced imaging (isotope bone scans, CT, MRI, PET) is commonly used as the
initial imaging modality of choice. Of particular concern is the fact that 60% (14,403/24,043) of BC patients with early stage
disease who were imaged required some form of confirmatory imaging. These results show the importance of the recent ASCO
"Top-5" recommendation against staging imaging in such patients. The reasons for this disconnect between evidence and
practice are not fully understood, but knowledge translation strategies beyond publishing guidelines or recommendations are
required if we are to elicit a meaningful and sustained change in physician practice.
BL Mean (sd)
Post-V1 Mean(sd)
Knowledge (6-28)
20.9(2.8)
22.0 (3.0)
20.8 (3.3)
21.8(3.2)
7.4(4.1)
6.6(4.1)
6.6 ((4.0)
5.7 (3.8)
3.9 (3.9)
3.5 (3.4)
3.6 (3.7)
3.4(3.5)
36.0(15.2)
35.7(13.7)
34.6(15.0)
34.5(13.1)
p
0.007
21.5(3.1)
NSS
0,02
5.7 (3.5)
0.06
0,05
2.9 (3.5)
0.07
NSS
32.1(12.5)
NSS
42.2(3.6)
0.002
53.0(5.3)
0.008
39.5(3.(
39.8(4.0)
Post-V2 Mean(sd)
51.3(5.6)
51.5(5.7)
Patients reported several advantages to telegenetics (e.g. decreased travel burden) and few disadvantages (e.g. audio
challenges and technical glitches).Conclusions: Telemedicine delivery of cancer genetic services is feasible, identifies genetic
mutation carriers, increases knowledge, decreases anxiety and depression and is associated with high satisfaction, suggesting an
innovative model for delivery of genetic services for patients and community practices without access to local genetic providers.
Multivariable
OR (95% CI)
pvalue
Reference
OR (95% CI)
Race
<.001
<.001
Black
White
<.001
0.880 (0.829-0.934)
<.001
0.897 (0.843-0.955)
Hispanic
<.001
0.643 (0.555-0.745)
<.001
0.701 (0.604-0.813)
Insured
<.001
Private
<.001
0.834 (0.770-0.904)
<.001
0.782 (0.700-0.874)
Medicaid
0.003
0.804 (0.698-0.926)
<.001
0.688 (0.587-0.806)
Uninsured
0.006
0.828 (0.828-0.968)
<.001
0.799 (0.718-0.890)
Location
<.001
Corporate
<.001
0.856 (0.805-0.911)
0.004
0.891 (0.822-0.964)
PCo
<.001
0.714 (0.666-0.766)
<.001
0.732 (0.680-0.787)
Age
0.002
25-39
0.150
0.849 (0.679-1.061)
0.150
0.848 (0.678-1.061)
40-49
<.001
0.895 (0.844-0.949)
<.001
0.901 (0.849-0.956)
65-89
0.083
0.928 (0.654-1.010)
<.001
0.809 (0.724-0.905)
<.001
Medicare
<.001
P CL
<.001
50-64
Conclusions: To our knowledge this is the reported largest database of MMU. Race, insurance status and location remained
independent predictors of repeat utilization. Interestingly an insured subset (MCare) was more likely to repeat utilize the van
regardless of race and location. This may reflect the average age of screening mammography however a better understanding of
the uninsured subset may be important given the intended outreach of the MMU.
FG1
FG2
FG3
Timely Diagnosis
Delay in Diagnosis
Poorly Communicated
Undergoing reconstruction
INT1
INT2
INT3
INT4
Initial Diagnosis
Treatment
Communication
Well informed about Rx plan
Not well-informed about Rx plan
Supports Utilized
Social Worker
Community Services
None
Exe
Ana (EUR)
Ana (NA)
F250
Combined
N = 168
N = 144
N = 162
N = 209
N=683
N = 88
N = 71
N = 82
N = 135
N=376
CB (%)
66 (75)
42 (59)
47 (59)
87 (64)
242 (64)
OR (%)
41 (47)
26 (37)
15 (19)
45 (33)
127 (34)
287
254
305
234
277
343
360
445
451
420
N = 80
N = 73
N = 80
N = 74
N=307
CB (%)
64 (80)
38 (52)
27 (33)
46 (62)
175 (57)
OR (%)
36 (45)
21 (29)
13 (16)
21 (28)
91 (30)
340
230
152
281
238
376
505
411
339
418
Without VM (55%)
With VM (45%)
VM
Two lines
Without VM
VM
Without VM
Three lines
VM
Without VM
3 (1-8)
6 (2-16)
11 (6-23)
15 (8-27)
23 (14-35)
26 (16-41)
First line
3 (1-8)
6 (2-16)
6 (3-15)
7 (3-16)
8 (3-17)
9 (3-17)
Second line
3 (1-6)
4 (2-10)
6 (3-12)
5 (3-11)
Third line
3 (1-5)
4 (2-9)
N (%)
Line 2
Median
N (%)
Line 3
Median
N (%)
Median
Anastrozole
339 (41.3)
135 (16.4)
113 (13.8)
Letrozole
243 (29.6)
10
165 (20.1)
109 (13.3)
Tamoxifen
114 (13.9)
140 (17.1)
159 (19.4)
Exemestane
42 (5.1)
186 (22.7)
161 (19.6)
Fulvestrant
37 (4.5)
98 (11.9)
158 (19.2)
Megestrol
23 (2.8)
80 (9.7)
104 (12.7)
*Treatment duration of other ETs (i.e., toremefene, raloxifene and combination therapy) with less than 2% of users are not
reported.
Total (N=22, %)
Clinical
palpation
US
CR
1 (4.5)
0 (0.0) 0 (0.0)
1 (4.5) 1 (4.5)
0 (0.0)
1 (4.5)
PR
17 (77.3)
12
(54.5)
9 (40.9)
9
11 (50.0)
(40.9)
15 (68.2
15 (68.2)
No change
3 (13.6)
9
(40.9)
13 (59.1)
5
9 (40.9)
(22.7)
7 (31.8)
4 (18.2)
PD
1(4.5)
1 (4.5) 0 (0.0)
1 (4.5) 1 (4.5)
0 (0.0)
2 (9.1)
Not
available/evaluable
0 (0.0)
0 (0.0) 0 (0.0)
6
0 (0.0)
(27.3)
0 (0.0)
0 (0.0)
Mammography MR
Overall
response
CR, complete response; PR, partial response; PD, progressive disease; US, ultrasonography
In analyses of biomarkers thus far, 81.8% of patients showed stationary expression of HER-2, 54.5% of patients showed
decrease in Ki-67 expression, and 27.3% of patients showed increase in ER expression from baseline to surgery by
immunothistochemistry (IHC) staining. Decreased expression of ER after surgery by IHC staining was significantly correlated with
poor clinical response (p=0.004). However, no significant differences in baseline SUVmax in FDG-PET were found between
responders and non-responders (8.8 VS 10.7, p=0.53).
Conclusion This chemo-free combination neoadjuvant therapy was feasible, with comparable efficacy outcomes and
manageable toxicities profiles. Updated data on 18F-FES PET-CT and biomarkers will be provided.
Hazard Ratio
P-value
RT to the IMNs
0.88
0.42-1.83
0.731
2D vs. CT planning
1.14
0.68-1.91
0.613
1.21
0.79-1.85
0.372
Anthracycline use
0.75
0.44-1.26
0.270
HTN
5.61
2.06-15.30
0.001
HLD
1.90
1.25-2.90
0.003
DM
3.11
1.99-4.86
<0.001
7.14 (6.85-7.52)
6.45 (6.06-6.90)
4.00 (3.69-4.33)
1.82 (1.68-1.97)
Grade 1
1.40 (1.33-1.47)
Race: White
1.38 (1.25-1.53)
1.30 (1.21-1.40)
1.15 (1.02-1.31)
Invasive Cancer
1.09 (1.04-1.14)
1.05 (1.02-1.09)
Conclusions: This comprehensive patterns of care study of HF-WBI in women 50 years old with early stage breast cancer from a
national database identified a substantial increase in the utilization of this technique with time. Advancing patient age and
treatment at academic facility were also strong predictors of delivery of HF-WBI. Disparities in utilization of HF-WBI and overall
employment of this technique, even in later years, remain suboptimal. Strategies to identify and break barriers to the adoption of
HF-WBI should be explored.
OR (95% CI)
p-value
Community
0.38 (0.35-0.42)
<0.001
Comprehensive Community
0.51 (0.48-0.53)
<0.001
Other
0.64 (0.52-0.78)
<0.001
Northeast
0.93 (0.88-0.99)
0.02
Midwest
1.04 (1.00-1.11)
0.07
West
1.69 (1.59-1.78)
<0.001
60-69
1.22 (1.16-1.29)
<0.001
70-79
1.77 (1.66-1.89)
<0.001
>=80
3.12 (2.88-3.38)
<0.001
Black
0.96 (0.89-1.05)
0.41
White Hispanic
1.23 (1.10-1.36)
<0.001
Race (White)
Other
1.23 (1.13-1.34)
<0.001
No insurance
1.16 (0.96-1.41)
0.11
Medicaid
0.97 (0.86-1.10)
0.71
Medicare
1.05 (1.00-1.11)
0.05
Other government
0.81 (0.63-1.05)
0.12
Unknown
1.22 (1.03-1.44)
0.02
30 000 - 35 000
0.95 (0.87-1.04)
0.30
35 000 - 45 999
0.97 (0.90-1.06)
0.54
>= 46 000
1.25 (1.16-1.35)
<0.001
Unknown
1.04 (0.86-1.25)
0.70
Urban
0.88 (0.83-0.95)
0.001
Rural
1.10 (0.99-1.22)
0.07
>=50 miles
1.57 (1.44-1.72)
<0.001
Unknown
1.13 (0.91-1.40)
0.27
2005
1.03 (0.92-1.14)
0.65
2006
1.11 (1.00-1.24)
0.05
2007
1.31 (1.18-1.45)
<0.001
2008
1.99 (1.80-2.19)
<0.001
2009
2,88 (2.62-3.15)
<0.001
2010
4.10 (3.62-4.64)
<0.001
2011
5.48 (4.83-6.21)
<0.001
Intermediate
0.94 (0.89-0.97)
<0.01
Poor
0.86 (0.80-0.91)
<0.001
Undifferentiated
1.03 (0.5-1.63)
0.89
Unknown
0.90 (0.82-0.99)
0.04
10-29 mm
0.90 (0.87-0.94)
<0.001
30-49 mm
0.71 (0.62-0.80)
<0.001
>=50 mm
0.94 (0.67-1.32)
0.76
Unknown
0.63 (0.42-0.95)
0.03
Urbanization (Metro)
Grade (Well-differentiated)
Conclusions
The use of hypofractionation is rising and is associated with increased travel distance and treatment at an academic center.
Further adoption of hypofractionation may be tempered by both clinical and non-clinical concerns.
Academic Center
2004
0.9%
1.7%
3.8%
2005
1.6%
2.1%
3.8%
2006
1.2%
2.0%
4.6%
2007
1.5%
2.8%
7.1%
2008
2.9%
5.3%
10.8%
2009
3.9%
8.5%
14.7%
2010
5.8%
10.7%
16.4%
2011
9.6%
13.9%
20.5%
95% CI
3.06
2.32
4.02
1.78
1.53
2.08
2.37
1.86
3.01
T2 vs. T1
0.54
0.46
0.63
0.75
0.59
0.97
0.66
0.52
0.84
2.68
1.69
4.24
N (%)
Infection
42 (11)
3 (3.3)
Seroma
40 (10.2)
Wound dehiscence
33 (8.5)
2 (0.5)
Bleeding
11 (2.8)
3 (0.8)
Skin necrosis
2 (0.5)
2 (0.5)
3.88 (3.45-4)
Heart V5
Mean Heart
Dose
Proton Plan
2.7%
(0-12%)
Conventional
Plan
34%
(7-60%)
5.9Gy (2-9.1Gy)
p-value
0.004
0.004
Mean Ventricle
Dose
Ventricle V5
31.4%
(8.7-79.8%)
Maximum LAD
dose
0.1Gy (0-4Gy)
30.5 (13.4-42.6Gy)
1.7Gy
(0.5-28.5Gy)
5.4Gy (2.4-11Gy)
44.6Gy
(35.1-54.7Gy)
27Gy
(16.8-41.9Gy)
0.004
0.004
0.04
0.02
Table 2. Cardiac dose in right-sided patients (comparison of medians of the cohort; range in parentheses)
Heart V5
Ventricle V5
Proton Plan
0.3% (0-2.9%)
0.5Gy (0-0.8Gy)
3.3% (0-1%)
0.2Gy (0-0.5Gy)
Conventional Plan
13.2% (0.4-24.7%)
2.9Gy (1-5.1Gy)
3.3% (0-7.1%)
1.2Gy (0.8-1.7Gy)
p-value
0.004
0.004
0.008
0.004
Grade 3 dermatitis developed in 4 patients (22%), which was the only grade 3 toxicity. All patients developed grade 2 dermatitis;
other acute grade 2 toxicities included fatigue (n= 6) and esophagitis (n=5). No grade 4+ toxicities developed. Dermatitis resolved
by 1 month after PT for all but 1 patient who developed cellulitis (grade 2).
Conclusion: PT for RNI after mastectomy or BCT significantly improves cardiac dose especially for left-sided patients and lung
V5 and V20 in all patients without excessive acute toxicity. PT simultaneously improves target coverage for the IMN and level II
axilla, which may positively impact long-term survival in breast cancer patients.
no ENI (n=119)
ENI (n=159)
P-value
Median
48
48
0.50
Range
27-80
27-71
MRM
90 (75.6)
2 (1.3)
BCS
29 (24.4)
157 (98.7)
IDC
116 (97.5)
153 (96.2)
Other
3 (2.5)
6 (3.8)
T1
56 (47.1)
81 (50.9)
T2
63 (53.9)
78 (49.1)
1-2
84 (70.6)
102 (64.2)
35 (29.4)
57 (35.8)
1-2
79 (66.4)
90 (56.6)
40 (33.6)
69 (43.4)
Median
19
17
Range
2-61
5-48
Positive
92 (77.3)
124 (78.0)
Negative
27 (22.7)
35 (22.0)
Positive
87 (73.1)
123 (77.4)
Negative
32 (26.9)
36 (22.6)
Positive
33 (27.7)
24 (15.1)
Negative
86 (72.3)
135 (84.9)
No
29 (24.4)
31 (21.9)
Yes
90 (75.6)
128 (78.1)
No
13 (10.9)
5 (3.1)
Yes
106 (89.1)
154 (96.9)
No
5 (15.2)
1 (4.2)
Yes
28 (84.8)
23 (95.8)
<0.001
0.74
0.55
0.30
0.11
0.17
1.00
0.48
0.01
0.38
0.01
0.19
During follow-up, 21 patients (6 in ENI and 15 in no ENI) had recurrence, and loco-regional recurrence developed in 8 patients, 6
of whom had not received ENI.
Table 2. Patterns of recurrence according to ENI and molecular subtype
No ENI
ENI
Site
Recurrence (%)
Recurrence (%)
P-value
Loco-regional-n
119
6 (5.0)
159
2 (1.3)
0.08
Luminal A
36
0 (0.0)
41
0 (0.0)
Luminal B
57
4 (7.0)
86
0 (0.0)
HER-2 enriched
12
0 (0.0)
1 (11.1)
Triple negative
14
2 (14.3)
23
1 (4.3)
119
10 (6.5)
159
5 (3.1)
Luminal A
36
2 (5.6)
41
0 (0.0)
Luminal B
57
6 (10.5)
86
2 (2.3)
HER-2 enriched
12
0 (0.0)
0 (0.0)
Triple negative
14
2 (14.3)
23
3 (13.0)
119
15 (12.6)
159
6 (3.8)
Luminal A
36
2 (5.6)
41
0 (0.0)
Luminal B
57
9 (15.8)
86
2 (2.3)
HER-2 enriched
12
0 (0.0)
1 (11.1)
Triple negative
14
4 (28.6)
23
3 (13.0)
Distnat-n
Total-n
0.06
0.01
In both univariate and multivariate analysis, ENI, adjuvant chemotherapy and endocrine therapy were the significant prognostic
factors in recurrence-free survival (RFS). ENI showed higher RFS than no ENI when they were analyzed according to molecular
subtypes, except HER-2 enriched. RFS was 96.9% at 3-years in luminal A patients with no ENI.
Conclusions: The application of ENI may improve RFS in patients with high-risk pN1 breast cancer, so ENI should be
considered in those patients. Close observation without ENI might be an option in patients with luminal A who had received
optimal systemic managements.
Negative Pathology
Positive TOUCH
66 ( 30.8%)
50 (23.4%))
Negative TOUCH
5 ( 2.3%)
93 ( 43.5%)
% (95% CI)
% (90% CI)
Sensitivity
Specificity
The training study in the UK on 18 BCS specimen results showed 50% True Positive, 39% True Negative, 11% False Positive,
6% False Negative rates. There were 5 cases (28%) of positive margins < 1 mm and the device images identified 4 with 1 case
(6%) being missed.
Results from the ongoing study will be presented.
CONCLUSIONS:
The TOUCH Imaging device is safe and efficacious.
It is easy for surgeons to use in the operating room.
This device has the potential to reduce BCS re-operation rate.
40 50 years
50 70 years
70 years
47%
60%
69%
48%
21%
14%
6%
4%
38%
29%
16%
1%
62%
68%
73%
48%
* Proportion of patients with primary breast conserving surgery, breast conserving surgery after neoadjuvant treatment and
immediate reconstruction combined with mastectomy.
Conclusions
Patient age affected both the BCS with or without neoadjuvant therapy and the percentage of immediate reconstructions after
mastectomy. Overall, more similar percentages in preservation of the breast contour are achieved for various age groups
compared to BCS alone. Therefore, combining different treatment modalities into breast contour preserving surgery provides a
more complete overview of maintaining the breast contour related to various age groups than BCS alone.
45
Educational Materials
21,079
Volunteers*
2,025
Women Interviewed
5,441
Eligible Women
643
Appointments Scheduled
581
Screening Days
15
409
"Black"
"Hispanic"
"Other"
Benefited
70%
85%
88%
54%
57%
65%
Discussion. WBHI reached the goal of increasing screening rates in this underserved population, thus showing that this type of
intervention can be effective. Educational packages presented in a face-to-face format within a door-to-door context were found
useful by most women. Our findings highlight disparities beyond a womans access to breast healthcare and education by
showing that neighborhood culture impacts response to outreach intervention and breast healthcare. This generates the
hypothesis that given equal access to care, disparities in using screening mammography will remain. Educational methods further
tailored to racial and ethnic characteristics may play a significant role in closing disparity gaps.
FNR (%)
95% CI
470
301 (64.0%)
11.3
8.0-15.4
470
311 (66.2%)
8.7
5.6-11.8
Clip in SLN
114
62 (54.4%)
6.4
1.8-15.5
Clip in ALND
29
18 (62.1%)
22.2
6.4-47.6
29
20 (69.0%)
14.3
3.0-36.3
354
207 (58.5%)
13.5
9.1-18.9
Clip location
Conclusion: Placement of a clip at initial diagnosis of node positive disease with identification of the clip during the SLN surgery
reduces the FNR. Use of IHC with inclusion of metastases <0.2mm in the definition of residual nodal disease in women with node
positive breast cancer after chemotherapy also improves the accuracy of SLN surgery. Use of one or both of these methods
should be considered when performing SLN in this setting.
Pre-Z0011
Post-Z0011
p value
Female (%)
656 (99.2)
968 (99.4)
0.86
Race (%)
0.49
White
448 (67.8)
642 (65.9)
Black
205 (31.0)
314 (32.2)
19.9
22.3
Histology (%)
0.06
0.07
434 (65.7)
663 (68.1)
30 (4.5)
62 (6.4)
ER Positive (%)
484 (73.3)
722 (74.2)
0.92
PR Positive (%)
406 (61.5)
622 (63.9)
0.61
304 (78.4)
897 (83.5)
0.01
83 (23.9)
168 (18.9)
0.07
175 (27.5)
926 (26.7)
0.71
A total of 142 patients met Z0011 criteria, with T1-2 disease, underwent partial mastectomy and had a positive SNB. There were
a similar number of patients who met Z00111 criteria in the pre- (n=63) and post-Z0011 (n=79) periods, p>0.05. In the post-Z0011
period, patients were more likely to undergo SNB alone (56 vs. 19%, p<0.001). Post-Z0011 the decrease in ALND was in those
who had immediate ALND (34% vs. 71%), with no substantive change in the delayed group (10.1 vs. 9.5%), p<0.001. Patient who
underwent SNB alone had smaller tumors (22 vs. 32mm, p<0.001), but similar histologic subtypes (p=0.60) and receptor status
(p0.15) as compared to patients who underwent SNB with ALND. Over the entire study period patients in an academic practice
were less likely to undergo SNB alone (31 vs. 43%). In the post-Z0011 period, patients cared for in an academic practice had an
increase in SNB alone from year 1 (2012) to year 2 (2013) (40% to 57%), while the non-academic practices had a decrease in
SNB alone from year 1 to year 2, (63% to 56%).
Conclusion
In this mixed practice setting there was an overall decrease in ALND in T1/T2 patients with low metastatic axillary burden
following publication of Z0011. Immediately post-Z0011 the non-academic practices more rapidly adopted SNB alone, however
these differences were no longer apparent two years following Z0011. Continued longitudinal studies will further assess the
variability in the incorporation of Z0011 among various surgical practices.
n patients
% patients
n patients
% patients
32
28
47
47
13
11
24
24
12
10
11
11
11
10
39
34
14
14
Totals
115
100
Analysis of the data revealed that tumour size was a significant predictor of low volume axillary disease (i.e. <3 nodes positive)
[p=0.02, Mann-Whitney test].
Conclusion
In this study, 46% of patients undergoing ALNC after positive staging had low volume disease (2 or less positive nodes). In the
context of recent evidence if these patients had undergone a SLNB following the positive pre-operative staging they may have
avoided a completion ALNC.
This study demonstrates tumour size to be a significant predictor of low volume axillary disease and thus may be an important
factor to consider alongside pre-operative staging when selecting patients who may be better managed by a SLNB rather than
proceeding directly to ALNC.
Age (median)
64
48
Histological Grade
ER status positive
200 (76%)
96 (65%)
49 (19%)
44 (30%)
20
17
103 (39%)
72 (49%)
Number of patients
Radiotherapy to the
axilla:163
Age (median)
64
50
59
Histological Grade
ER status positive
144 (88%)
57 (85%)
90 (96%)
10 (6%)
16 (24%)
14 (15%)
16
17
50 (75%)
72 (77%)
40 (60%)
58 (62%)
Metastasis(-)
Whole
59
Partial
10
26
Not identified
Conclusion: This new navigation method of CTLG and fluorescence imaging revealed more easy and effective to detect SLN
intraoperatively than fluorescence imaging alone. In addition, the information from CTLG may be helpful for the prediction of SLN
metastasis.
Analysis
VAB
Core SCNB
63
64
Accurate diagnosis
54 (86%)
54 (84%)
1.0
9 (56%)
17 (71%)
0.50
7 (44%)
7 (29%)
0.50
12.5%
8.7%
0.86
Both groups had significant Quality of Life (QOL) falls in Global Health (p<0.001) and Social Functioning over time (p<0.04) but
no overall difference between groups was seen. At 12 months, body image scores fell and fatigue increased in patients
undergoing surgery.
Conclusion
SCNB has equal accuracy as VAB in the firstline diagnosis of malignant microcalcification. Devicor provided the equipment to Dr
S.B. and Prof. N.B., to do this study.
Outcome
25th Percentile
Median
75th Percentile
Current Smokers
Overall Survival
0.73
0.80
0.86
Current Smokers
Recurrence-free Survival
0.67
0.73
0.78
Never Smoked
Overall Survival
0.53
0.59
0.66
Never Smoked
Recurrence-free Survival
0.51
0.59
0.66
Analysis of interaction between smoking status and gene expression values using the combined samples revealed significant
interactions between smoking status and CYP1A1, LECT2, CETN1. Molecular signatures consisting of 7-8 genes were highly
predictive for breast cancer recurrence and overall survival among smokers, with median C-index values of 0.8 and 0.73 for
overall survival and recurrence, respectively. In contrast, the median C-index values for non-smokers was only 0.59. Hence,
significant interactions between expression of crucial genes and cigarette smoking status appear to play a key role in predicting
clinical outcomes of breast carcinoma patients. Supported in part by a grant from the Phi Beta Psi Charity Trust (TSK & JLW) and
a Research of Women (ROW) grant to JLW from the EVP for Research and Innovation, University of Louisville.
SNP rsID
SNP Location
Gene
Component
rs6504950
chr17:53,056,571 STXBP4
In gene: intron 1
0.59
1.5E-23
2.6E-19
3.1E-07
-0.72
2.5E-23
2.6E-19
2.3E-10
Transcript uc010dcc
-0.42
1.5E-11
2.6E-08
-0.63
5.7E-13
1.4E-09
0.26
8.0E-10
1.2E-06
-0.48
1.3E-07
1.3E-04
4.7E-03
0.24
1.2E-06
9.1E-04
1.0E-04
Transcript uc002nfv
0.47
6.1E-08
6.5E-05
5.3E-05
Transcript uc002nfu
-0.27
5.3E-06
3.3E-03
6.3E-03
-0.45
1.4E-07
7.3E-06
1.5E-02
0.50
1.6E-07
1.5E-04
Exon 2
0.34
3.5E-06
2.3E-03
Exon 7
-0.39
1.1E-06
8.8E-04
Transcript
uc001arm
-0.39
1.1E-06
8.1E-04
In gene: exon 1
Transcript uc001eei
Transcript
uc001eeg
Exon 1:3 junction
Exon 2
rs8170
chr19:17,389,704 BABAM1
In gene: exon 8
151 kB
rs616488
146 kB
chr1:10,566,215 PEX14
In gene: intron 2
2.9E-02
rs999737
chr14:69,034,682 RAD51L1
In gene: intron 13
Exon 15
-0.33
6.2E-05
3.4E-02
Location is for hg19 build. is for effect of the breast cancer risk allele. FDR is when all exon, junction, and transcript tests are
considered together. P-values for ER- shown when < 0.05. * SNP is associated with transcript expression of two different genes.
Pattern of association with transcript does not correspond to pattern of association with breast cancer.
Conclusion: Regulation of differential transcript expression appears to be an important functional mechanism of breast cancer risk
SNPs. These associations allow us to identify likely candidate causal genes, and also specific exons and transcripts that may
mediate breast cancer risk, opening a new window into the link between risk variants, gene function, and outcome.
Luminal A
Luminal B
Her-2+
Triple Negative
P value
(n=233)
(n=79)
(n=59)
(n=86)
Age (median,IQR)
61 (54,70)
58 (50,67)
54 (46,70)
59 (48,67)
0.006
BMI (median,IQR)
27.1 (22.9,30.5)
25.7 (23.0,30.0)
26.0 (22.7,32.6)
28.1 (24.3,32.6)
0.173
Volpara density
(median,IQR)
7.18 (4.74,11.25)
8.68 (5.68,14.34)
10.25 (5.96,16.51)
7.00 (4.97,11.89)
0.002
0.183
Fatty
42 (18.0%)
15 (19.0%)
5 (8.5%)
16 (18.6%)
Scattered
103 (44.2%)
27 (34.2%)
20 (33.9%)
36 (41.9%)
Heterogeneous
74 (31.8%)
30 (38.0%)
25 (42.4%)
26 *30.2%)
Extreme
14 (6.0%)
6 (7.6%)
9 (15.2%)
7 (8.1%)
Unspecified
1 (1.2%)
1 (1.2%)
Race (n,%)
0.002
Caucasian
202 (86.7%)
65 (82.3%)
51 (86.4%)
61 (70.9%)
African American
23 (9.9%)
13 (16.5%)
6 (10.2%)
23 (26.7%)
Other
2 (0.9%)
2 (3.4%)
Unspecified
LCIS (n,%)
6 (2.6%)
1 (1.2%)
2 (2.3%)
48 (20.6%)
8 (10.1%)
8 (13.6%)
5 (5.8%)
0.004
34 (43.0%)
21 (35.6%)
39 (45.35%)
0.625
C1 (n=24)
C2 (n=48)
C3 (n=35)
P-value
64.6
56.8
51.9
<0.05
SBR
Low
High
<0.05
NPI
Low
High
<0.05
Clinicopathologic characteristics
Gene-expression signatures
PAM50
Basal-like (98%)
Basal-like (91%)
Proliferation score
Low
High ++
High +
TNBCtype
Basal-like 1, mesenchymal
Immune response
Immune response
Immune response
Immune response
Claudin-low
0%
Higher frequency
(26%)
<0.05
Biological processes
Immune response
Immunohistochemistry
CK5 and/or HER1
positive
50%
91%
<0.05
AR positive
73%
5%
<0.05
Ki-67 positive
29%
87%
<0.05
FOXA1 positive
83%
9.6%
<0.05
CONCLUSION
Heterogeneity within basal-like and IHC-TNBC is still controversial and require further research to understand the complexity of
the disease, and to identify molecular drivers that can be therapeutically targeted. Annotation discordances between IHC and
GES clearly indicate that a robust molecular subtyping method must be found. Correct molecular assignment would permit to
orientate 20 to 30% of IHC-TNBC patients towards targeted therapy (hormonotherapy or anti-HER2).
ALDH1 expression in
cytoplasm
N
ALL CASES
Positive (%)
Negative (%) P
56 (35)
104 (65)
64(40)
Age (years)
96 (60)
0.867
0.022
50
94
32 (34.0)
62 (66.0)
30 (31.9)
64 (68.1)
>50
66
24 (36.4)
42 (63.6)
34 (51.5)
32 (48.5)
Tumor size
0.430
0.188
54
17 (31.5)
37 (68.5)
18 (33.3)
36 (66.7)
2-5
100
38 (38.0)
62 (62.0)
46 (46.0)
54 (54.0)
>5
1 (16.7)
5 (83.3)
1 (16.7)
5 (83.3)
0.452
0.364
80
27 (33.8)
53 (66.2)
34 (42.5)
46 (57.5)
4-9
57
18 (31.6)
39 (68.4)
19 (33.3)
38 (66.7)
>9
23
11 (47.8)
12 (52.2)
11 (47.8)
12 (52.2)
0.156
0.025
Positive
109
34 (31.2)
75(68.8)
37 (33.9)
72 (66.1)
Negative
51
22 (43.1)
29 (56.9)
27 (52.9)
24 (47.1)
0.591
0.598
Positive
112
41 (36.6)
71 (63.4)
43 (38.4)
69 (61.6)
Negative
48
15 (31.3)
33 (68.7)
21 (43.8)
27 (56.3)
0.460
0.585
Positive
43
17 (39.5)
26 (60.5)
19 (44.2)
24 (55.8)
Negative
117
39 (33.3)
78(66.7)
45 (38.5)
72 (61.5)
Multivariate Analysis
Characters
OR
95% CI
OR
95% CI
Age 50/>50
0.812
0.436-1.515
0.513
2.473
1.144-5.343
0.021
2.035
0.938-4.417
0.072
6.346
2.815-14.306
<0.001
5.655
2.477-12.910
<0.001
0.660
0.354-1.231
0.191
0.533
0.288-0.988
0.046
0.835
0.358-1.948
0.676
0.969
0.487-1.928
0.929
2.578
1.403-4.737
0.002
2.774
1.502-5.124
0.001
0.712
0.356-1.473
0.378
Doctors
Nursing Staff
Number of participants
631
120
96
92.7% (495/534)
100% (119/119)
91.7% (88/96)
PC
77.7% (490/631)
99.2% (119/120)
94.8% (91/96)
Smartphone
25.5% (161/631)
70.8% (85/120)
45.8% (44/96)
Property of media:
53.8% (276/513)
66.4% (79/119)
56.8% (54/95)
via Smartphone
24.2% (119/492
51.3% (60/117)
31.1% (28/90)
41.1% (204/497)
71.2% (84/118)
56.1% (51/91)
Statement
67
68
60
0.001<
18
20
0.01
63
66
52
0.001<
0.001<
54
58
43
0.001<
24
13
0.001<
22
25
12
0.001<
27
30
18
0.001<
Conclusion: This population of MBC patients reports a range of experiences and often conflicting beliefs about CCTs. While this
survey is limited to self-report data, there is persistent suspicion among these women about participating in CCTs. Although these
women report that they understand what a CCT is, they fear receiving a placebo and being randomized to different treatment
arms without at least the standard of care. These misconceptions persist even among people who have participated in a CCT.
These findings highlight the need for treatment decision counseling that educates patients about all their treatment options
including CCTs and for training of health care providers to better inform patients about CCTs.
Please tell us how much of a burden the following additional expenses caused you. (0=not at all to 4=very much)
3 (%)
492 46.1
492 40.9
492 39.3
492 39.7
491 37.9
489 20.6
Medical supplies
489 21.3
492 19.6
Counseling or therapy
488 19.9
Because of these expenses, registrants have: foregone vacations, celebrations, and social events (53%); sold property (12%);
refinanced their house (13%); filed for bankruptcy (5%); downsized their living accommodations (16%); liquidated their assets
(19%); depleted their savings (40%); borrowed against or used money from a retirement plan (27%); cut their grocery expenses
(42%); applied for or used public assistance (14%); chosen a treatment that is not as effective but costs less (9%); asked their
doctor if there was a less expensive treatment (19%); tried to negotiate payments with credit companies (24%); negotiated with
service providers to reduce costs (22%); used pharmaceutical assistance programs (26%); accepted money from friends or family
(39%); and cashed in a life insurance policy early (8%), among others. In order to reduce the cost of treating MBC, registrants
often or always postpone seeking psychological counseling or support (20%) and delay follow up on recommendations for
complementary treatment such as physical or occupational therapy and nutrition counseling (13%).
Conclusion: MBC places a significant financial burden on patients, which can result in patients taking measures that can
significantly impact their quality of life. Future implications for research include the development and evaluation of interventions
designed to enhance doctor-patient communication and support (e.g., financial counseling) to ensure that the financial cost of
MBC does not negatively impact the patients quality of life, course of cancer care, and health outcomes.
BEV (N=94)
BEVCAP (N=91)
N=51
N=59
Improved
15 (29.4)
17 (28.8)
Stable
26 (51.0)
34 (57.6)
N=29
N=57
Improved
11 (37.9)
12 (21.1)
Stable
12 (41.4)
30 (52.6)
N=23
N=43
7 (30.4)
16 (37.2)
Week
Week
Week
9a
18a
27a
Improved
Stable
12 (52.2)
20 (46.5)
N=15
N=35
Improved
4 (26.7)
14 (40.0)
Stable
9 (60.0)
17 (48.6)
Week
36a
aNo.
of patients with completed questionnaires at both randomization and the respective week. Only weeks with 10 pts in both
arms shown.
CONCLUSIONS The IMELDA sample size was smaller than planned but protocol adherence with PRO completion was relatively
high. Prespecified change from randomization and exploratory post hoc MMRM analyses of PROs suggest that the clinically
meaningful PFS and OS benefit from adding CAP to BEV is achieved while maintaining QoL, with no difference between BEV
and BEVCAP treatments. Responder analyses over time showed improved or stable global health status/QoL scores in the
majority of pts at each timepoint in both treatment arms.
Skin desquamation
68 (38.4)
40 (44.9)
18 (10.2)
13 (14.6)
Infection
7 (4.0)
7 (7.9)
Hematoma
3 (1.69)
3 (3.4)
6 (3.4)
6 (6.7)
Conclusion: NSM is an acceptable choice for patients with BRCA gene mutations undergoing therapeutic or prophylactic
mastectomy with no evidence of compromise to oncological safety. This report shows an acceptable complication rate, and
patients rarely required subsequent excision of the nipple-areolar complex.
95%CI
Age
<0.01
<50
50-59
0.7
0.6 - 0.8
60-69
0.7
0.6 - 0.8
70+
0.6
0.5 - 0.7
BMI (kg/m2)
<0.01
<18.5
1.8
18.5-<25
25-<30
0.8
0.7 - 0.9
30
0.7
0.6 - 0.8
Unknown
0.8
0.6 - 1.0
1.3 - 2.6
Comorbidity
0.05
1+
1.1
Race
1.0 - 1.3
0.8
0.8-1.2
Hispanic
0.8-1.3
Other
1.1
0.9-1.4
Insurance
0.16
Medicare
0.9-1.2
Self-pay/Medicaid
1.3
1-1.6
Other
0.6-1.6
0.02
1 (low)
0.9
0.8-1.1
0.9
0.8-1.1
0.9
0.8-1.0
5 (high)
0.8
0.6-0.9
1-1
Year of diagnosis
Center
0.35
<0.01
0.9
0.7-1.1
0.7
0.6-0.9
1.4
1.1-1.7
0.4
0.3-0.6
0.8
0.6-1.1
1.3
1-1.6
0.6
0.4-0.8
Clinical Stage
<0.01
T1a
T1b
0.8
0.6-0.9
T1c
0.8-1.2
T1NOS
1.1
0.9-1.4
Tumor Subtype
<0.01
HER2+HR+
HER2+HR-
1.5
1.1-1.9
HER2-HR+
0.9
0.7-1.0
HER-HR-
0.7
0.6-0.9
Grade
<0.01
Low Intermediate
High
1.2
1.1-1.4
Histology
Ductal
<0.01
Lobular
1.4
1.2-1.7
Mixed
1.3
1.1-1.5
Other
0.7
0.5-0.8
Preoperative MRI
No
Yes
1.8
<0.01
1.6-2.1
Conclusions: Among a cohort of pts with small node negative BC, 23% elected to have MAST with significant variation
associated with choice of treatment, while some of this variation is likely appropriate and clinically indicated, further studies to
assess pt understanding of the tradeoffs between BCS and MAST is warranted. These findings need to be considered in light of
the increasing number of pts who are choosing MAST/bilateral MAST.
Study
N excised
N upgraded to cancer
% upgraded
Lim, 2006
62.5
20
Kunju 2007
14
85.7
12
25
Martel 2007
63
38.1
24
38
Piubello, 2009
33
60.6
20
Chivukula, 2009
39
89.7
35
14
Senneta,2009
41
87.8
36
Hayes, 2009
100
13
Davashian, 2009
12
100
12
17
Tamasino, 2009
54
44.4
24
13
Noske, 2010
43
69.8
30
Lee, 2010
15
46.7
14
Ingegnoli, 2010
18
83.3
15
20
Noel, 2010
62
32.3
20
Flegg, 2010
NA
NA
40
Sohn, 2011
36
66.7
24
Lavoue, 2011
60
100
60
13
Rakha, 2011
24
100
24
21
Soloranzo,2011
33
84.8
28
14
Verschuur-Maes, 2011
69
34.8
24
38
Peres, 2012
128
79.7
102
10
10
Uzoaru, 2012
145
65.6
95
Bianchi, 2012
190
100
190
18
Biggar, 2012
51
100
51
Yamaguchi, 2012
17
47.1
13
Polom, 2012
20
100
20
10
Khoumais, 2013
104
90.4
94
10
11
Villa, 2013
142
85.2
121
Becker, 2013
303
78.9
239
10
Ceugnart, 2013
63
82.5
52
Uzan, 2013
35
100
35
Across 1420 patients with pure FEA in 30 studies, observed proportions with upgrade to cancer varied from 0-40% with a pooled
estimate of 11% (95% CI: 8-15%) using a random effects model. Test for heterogeneity was statistically significant (p < 0.0001, I2
statistic=58%). After excluding 7 studies with <50% (or unreported %) of all FEA cases excised, heterogeneity decreased
(p=0.07, I2=32%) and the pooled estimate of cancer upgrade was 9% (95% CI: 7-12%). For upgrade to ADH, 693 subjects with
pure FEA were analyzed from 21 studies. The percent upgraded to ADH ranged from 0-60% with significant heterogeneity (p <
0.0001, I2 = 64%). Excluding 4 studies with <50% of FEA cases excised did not improve heterogeneity (p < 0.0001 and I2 =
66%). The random effects model pooled estimate of upgrade to ADH was 16% (95% CI: 11-23%) for the 21 studies and 17%
(95% CI: 12-25%) for the subset of 17 studies.
CONCLUSION: The pooled upgrade rate of FEA to cancer was 9-11% and upgrade to ADH was 16-17%. For patients with FEA
on core needle biopsy, surgical excision should be strongly considered.
26
Pain control
17
Nausea
Urinary Retention
Cardiac Issues
Other (respiratory issue, poor mobility, social admission, fall on ward, pyrexia)
Total
62
Conclusion: Unilateral and bilateral mastectomy, with or without implant based reconstruction, is safely performed in the setting of
an AXR program. Only a small minority of patients will subsequently require hospital admission, most commonly for management
of postoperative hematoma or inadequate pain control.
All (n = 129)
p 1 vs 3; p 2 vs 3
LOS (days)
2.3 (0.84)
2.1 (0.71)
1.61 (0.54)
<0.0001; <0.0005
NU (mg)
73 (42.9)
52 (32.3)
39 (25.3)
<0.001; 0.04
LOS (days)
2.2 (0.50)
2.3 (0.63)
1.61 (0.44)
<0.001;<0.001
NU (mg)
86 (32.6)
55 (34.4)
43 (36.3)
<0.001; 0.10
LOS (days)
2.4 (0.7)
2.2 (0.72)
1.65 (0.52)
<0.009;<0.001
NU (mg)
84 (37.4)
58 (32.4)
38 (24.5)
<0.001;0.0066
TE reconstruction (n=86)
Bilateral (n=84)
19
8.9
39
19
1.6
23
3.1
11.5
3.8
0
Numb of studies
Average rate
17.23
1.79
Clinical presentation included: Palpable mass, nipple discharge, itching, mammographic mass/asymmetry or calcifications. This
study found 69.3% of papillary lesions presented as mammographic findings including nodules/asymmetry (28%), calcifications
(24%), masses (16.7%). The remaining 29.3% of the papillary lesions presented as palpable masses(14.7%), discharge (4%),
itching (2.67%), and pain/tenderness(1.3%).
Conclusion: IDPs present as a radiologic finding in 2/3 of the cases and a clinical complaint in the remaining patients. A review
of the upgrade rates in the literature shows a range of 0-39%. Although larger cores from vacuum assisted biopsies may lower
the upgrade rates, a good number of studies show rates that do not justify observation only. As concerning is the association of
papillary lesions with atypia and malignancy in 32.3% of our patients indicating a possible causal relation. Our data and review of
the literature indicate that short of a prospective randomized trial to settle this question, IDPs should be routinely excised.
Surgery
No surgery
Female (%)
16,328 (98.7)
24,750 (99.1)
13,198 (79.7)
19,328 (77.4)
T1 stage (%)
3,067 (18.5)
2,096 (8.4)
N1 stage (%)
4,173 (25.2)
5,834 (23.4)
ER Positive (%)
9,642 (58.3)
12,300 (49.3)
PR Positive (%)
7,532 (45.5)
9,346 (37.4)
414 (4.5)
745 (3.0)
Grade II (%)
4,844 (29.3)
5,400 (21.6)
11,539 (69.7)
12,461 (50.0)
6,338 (38.3)
6,937 (27.8)
Flap fixation
Mastectomy
8/14 (57%)
1/6 (17%)
0.16
Mastectomy and SN
26/42 (62%)
13/52 (25%)
<0.001
18/32 (56%)
19/34 (56%)
0.98
In contrast, flap fixation was associated with fewer seroma aspirations in all types of surgery (P=0.80 for interaction).
Effects of flap fixation on seroma aspiration stratified by operation type
Historical Control
Flap fixation
Mastectomy
6/14 (43%)
1/6 (17%)
0.35
Mastectomy and SN
19/42 (45%)
7/52 (14%)
0.001
13/32 (41%)
6/34 (18%)
0.04
Conclusion
Patients undergoing mastectomy flap fixation displayed a significant reduction in seroma formation and fewer patients were
subjected to seroma aspiration. Patients undergoing flap fixation required fewer seroma aspirations. Flap fixation is an effective
surgical technique in reducing dead space and therefore seroma formation in patients undergoing mastectomy for IBC.
Results
Relative deviation
SD
In vivo/external
13.3%
2.2%
Shielding/ no shielding
-60.2%
42.6%
Tangential axe
36.7%
24.8%
Perpendicular axe
50.6%
16.8%
Conclusions
Results about the dose as a function of depth show clearly that we cannot consider breast as equivalent to water at this energy
and may taking in account breast density .It seems to be important to spread the skin correctly from the incision, to recover
carefully the applicator and use the shielding in order to avoid secondary effects as skin necrosis and ribs failures.
Our X-ray source model allows to have realistic dose distribution wich hepls in better surgical comprehension of IORT particularly
in the set up of the applicator.
SWC
TG/ND
Patients
35
21
Mean Age
61.5
62.7
Mean BMI
25.7
25.7
Mean total drained + aspirated fluid per ME was 78 ml in the TG/No Drain group vs. 512 ml in the SWC group, an 85% reduction
(p=0.00). Four MEs required aspiration in the test group (15%) vs. 10 (25%) in the SWC group (ns). There were 3 revisions in the
SWC group and none in the test group (ns).
Outcomes
Outcomes
SWC
TG/ND
Number (mastectomies)
40
26
6.4
-100%
463
-100%
512
78
10
25% 4
15% 0.539
33% 7
27% 0.625
8% -
0% 0.273
p value
0.000
-85% Mann-Whitney
Discussion/Conclusions: Our experience suggests a clear advantage in the TG/no-drain approach in mastectomy. In addition to
the improved patient comfort and earlier discharge with TG, patients with drains had significantly higher total fluid collection and
still required more aspirations on average than the test group. Non-inferiority for minor complications was not only achieved, but
there seems to be a trend toward reduced complications in the no drain cohort. A prospective, randomized study is planned to
verify our initial impressions.
Sometimes
Often
Always
Delayed
50
51
19
Immediate implant
58
41
Immediate ADM+Implant
45
49
14
28
55
33
Immediate Autologous
29
59
22
Conclusions: Surgeons employ a variety of approaches to reconstruction in the face of PMRT, the most common approach
being delayed reconstruction. Decision-making is based upon individual surgeons perception of risks including likely delay to
adjuvant therapy and effect of PMRT on the reconstruction. Drivers appeared to be more surgeon-centred rather than
patient-based. There is awareness of a lack of evidence to support decision-making and the need for high quality studies.
Randomised clinical trials are needed to provide an evidence base for outcomes.
Subgroup
BEVCAP
(0.26-0.69)a
BEVCAP
72
90
All
53/94 (56)
33/91 (36)
0.43
<65 y
46/81 (57)
27/77 (35)
0.51 (0.32-0.82)
72
93
65 y
7/13 (54)
6/14 (43)
0.50 (0.16-1.60)
68
79
Triple negative
16/21 (76)
10/25 (40)
0.44 (0.19-0.99)
62
90
37/73 (51)
23/66 (35)
0.53 (0.31-0.89)
75
91
ER
positiveb
36/69 (52)
23/64 (364)
0.53 (0.32-0.90)
75
90
ER
negativeb
17/25 (68)
10/27 (37)
0.44 (0.20-0.99)
64
91
17/40 (43)
17/48 (35)
0.75 (0.38-1.49)
81
93
36/54 (67)
16/43 (37)
0.39 (0.22-0.71)
65
88
Visceral metastasesb
38/65 (58)
23/62 (37)
0.43 (0.26-0.73)
70
92
No visceral metastasesb
Complete or partial
Stable
responseb
diseaseb
15/29 (52)
10/29 (34)
0.76 (0.34-1.70)
76
88
36/68 (53)
24/68 (35)
0.61 (0.37-1.03)
73
89
14/22 (64)
6/20 (30)
0.22 (0.08-0.63)
68
100
Non-measurableb
3/4 (75)
3/3 (100)
0.30 (0.03-2.98)
67
67
LDH 1.5ULNb
50/89 (56)
30/85 (35)
0.49 (0.31-0.76)
72
94
3/5 (60)
3/6 (50)
1.01 (0.20-5.00)
60
44
LDH >1.5ULNb
aStratified
analysis.
bStratification
factor.
CONCLUSIONS. Combining maintenance BEV with CAP until PD after initial BEVDOC for mBC provides a statistically
significant and clinically meaningful improvement in OS (secondary endpoint), seen consistently irrespective of baseline
characteristics.
Li et al
Toxicity
Anemia
0 (0)
3 (6.3)
Neutropenia
14 (22.6)
36 (75.0)
Thrombocytopenia
9 (14.5)
10 (20.8)
Nausea/vomiting
1 (1.6)
0 (0)
Diarrhea
1 (1.6)
0 (0)
Neuropathy
3 (4.8)
0 (0)
<0.05
Chemotherapy as the third line or beyond treatment for metastatic breast cancer
Number of
Patients
ORR
(%)
median PFS
(months)
median OS
(months)
Bevacizumab+mFOLFOX6 4, 3-7
48
48.8
6.0
10.2
Aogi et al
Eribulin
21
14.3
1.9
7.3
Cortes et al
Eribulin
5, 3-6
269
14.1
2.6
10.4
Rha et al
Gemcitabine
12
17
Smorenburg et
al
Gemcitabine
23
7.8
Udom et al
Vinorelbine
3, 3-5
20
35
Rivera et al
Paclitaxel
35
20
NA
NA
Rivera et al
Paclitaxel
33
18
NA
NA
Abrams et al
Paclitaxel
172
23
NA
NA
Study
Regimens
Li et al
Median Line,
range
M2
M3
M4
M5
M1
610
M2
1096
10
21
M3
34
M4
21
M5
47
Table 1
Conclusion: In a symptomatic and follow up clinic, our study suggests that much radiologists time and x-ray exposure to the
patient could be saved by using synthetic 2D mammograms derived from the DBT data rather than using separate 2D studies.
Only if the C-view is reported M3, 4 or 5 is it necessary to review the DBT but for all these patients the DBT is already available
without further x-ray exposure or recall.
Screening Strategies
Frequency
Modeled participation
40-49 years
50-69 years
70-85 years
61-71%
74-75%
72-74%
Current practice
Variable
Annual strategy
Every year
85%
85%
85%
Biennial strategy
Every 2 years
0%
85%
0%
USPSTF
Every 2 years
85%
25-37.2%
40-49 years
50-69 years
70-85 years
21,994,479
36,820,954
12,610,766
Annual
9.2
8.6
9.6
Biennial
10.4
9.7
10.8
Annual
0.85
1.04
1.5
Biennial
1.01
1.24
1.8
Model Inputs
Women at risk
False-positive recall
rate (%)
False-positive biopsy
rate (%)
Formulas
Number of screening
mammograms
Number of
false-positive recalls
Number of
Number of screening mammograms
false-positive biopsies
x false-positive biopsy rate
Outcomes
Current practice
Annual strategy
Biennial strategy
USPSTF
6.1x107
1.6x107
(5.6x107-6.5x107)
(1.4x107-1.7x107)
2.1x107
(1.9x107-2.2x107)
Number of screening
mammograms
4.6x107 (4.3x107-5.0x107)
Number of
false-positive recalls
4.2x106 (3.4x106-5.3x106)
5.4x106
(4.1x106-6.9x106)
1.5x106
(1.1x106-1.9x106)
2.1x106
(1.5x106-2.6x106)
Number of
false-positive biopsies
5.3x105 (4.1x105-6.4x105)
6.5x105
(4.8x105-8.2x105)
2.0x105
(1.5x105-2.5x105)
2.7x105
(2.1x105-3.5x105)
Conclusion
Compared to annual screening, we estimate that following the USPSTF guidelines would result in 62% fewer false-positive recalls
and 58% fewer false-positive biopsies. The absolute magnitude is in the range of 374,000 biopsies annually. Given that these
screening strategies have been projected to be of equal benefit, more widespread adoption of the USPSTF guidelines could
decrease the patient risk and anxiety associated with recall and biopsy without impacting the benefits. Developing tools for
personalized risk assessment would facilitate adoption of the USPSTF guidelines. These tools can be tested by comparing the
personalized and annual screening strategies prospectively and can inform screening policies.
Overscreening*
Underscreening*
Breast imaging less often
than annually if age 30
Carriers
True non-carriers
Age <40: any screening MMG; age 40: MMG more often than every 2 years OR Age 50-70: MMG less
any MRI or ultrasound screening
often than every 2 years
*Definitions derived from Cancer Australia Guidelines which do not recommend for or against CBE in any group and differ from
US guidelines.
Results: Of 372 eligible participants, there were 92 mutation carriers (42 BRCA1, 50 BRCA2) and 280 true non-carriers. 1% of
carriers were overscreening, 86% were screening annually while 13% were underscreening. MRI, which is funded for mutation
carriers aged 30-50 years, was used by only 52% (13/27) of carriers aged <50 years and by 7 carriers aged >50. Underscreening
carriers were more likely to be single (OR=2.78, 95%CI=1.28-5.88, P=0.009) while those with a first degree cancer affected
relative were less likely to underscreen (OR= 0.40, 0.19-0.85, P=0.017). CBE was undertaken by 79% of carriers at least yearly;
15% had no or irregular CBE.
115/280 (41%) non-carriers were overscreening, 55% were screening appropriately and 5% were underscreening.
Underscreening non-carriers were more likely to be single (OR 3.85, 95%CI=1.00-14.9, P=0.05). Predictive of overscreening in
non-carriers was having a cancer affected first degree relative (OR=2.92, 1.55-5.51, P=0.001). Non-carriers were less likely to
utilise CBE (54%).
Conclusion: Most mutation carriers in kConFab are having regular screening MMG, but MRI is underutilised even when funded.
The reasons for low usage of MRI requires further research and is concerning given its increased sensitivity over MMG.
Overscreening is common in true non-carriers of BRCA mutations. Family cancer history and marital status may predict
inappropriate screening behaviour.
Coverage
Minimum
Maximum
North Region
12.1
0.2
27.2
Northeast Region
23.9
10.3
37.2
Southeast Region
28.5
15.5
32.5
South Region
34.6
30.3
41.0
Midwest Region
14.8
10.3
19.7
p*
0.001
* Chi-square test
Units of the Federation an estimated coverage ranged from 0.2% to 41%, the lowest in the state of Amapa and the largest in
Santa Catarina. 48% of states were in the range of 10.1% to 20% coverage. CONCLUSION: The results imply that the
contribution of SUS to mammography screening in Brazil is higher in both macro-regions and states with higher income and
better organised health system.
PgR-
>50 yrs
PgR+
50 yrs
PgR-
PgR+
PgR-
>50 yrs
PgR+
PgR-
Grade 1-2
45/607 (7.4%)
28/607 (4.6%)
Grade 3
50/263 (19.0%) 4/26 (15.4%) 69/529 (13.0%) 35/113 (31.0%) 31/263 (11.8%) 2/26 (7.7%) 44/529 (8.3%)
26/113 (23.0%)
Table 2. Results of MV model including significant main effects and interaction effects between tumor characteristics, age and
PgR status on DMFI and BCSS.
95% Confidence interval
Interactions
Hazard ratio
Lower limit
Upper limit
P-value
A. BMFI
PgR Neg. vs Pos. |-50yrs
0.784
0.395
1.556
0.4869
1.937
1.386
2.709
0.0001
B. BCSS
PgR Neg. vs Pos. |-50yrs
0.575
0.208
1.588
0.2854
2.439
1.620
3.672
<.0001
Conclusions
A negative PgR is only prognostic for DMFI and BCSS in women aged >50 at diagnosis. There was no difference of PgR for
these endpoints in patients 50 yrs at diagnosis but PR negativity is rare in this age group.
Accuracy
Sensitivity
Specificity
PPV
NPV
Microarray (Training)
89
0.96
0.89
0.98
0.98
0.96
Microarray (Validation)
44
0.91
0.80
0.97
0.92
0.90
PCR
26
0.96
0.95
1.00
1.00
0.86
IHC
28
0.89
0.86
1.00
1.00
0.89
Sensitivity and specificity of model in training (microarray, PCR, IHC) and validation datasets (microarray). PPV/NPV =
postivie/negative predictive value.
Conclusion:
A 4 gene model has been developed and validated to predict response to neoadjuvant aromatase inhibitors.
This model has been shown to work with a high degree of accuracy using both PCR and IHC technologies. Further independent
validation is currently underway.
This new test has the potential to predict accurately the benefit of endocrine therapy and has huge potential clinical value.
V/C (n=71)
Control (n=44)
TN (n=59)
5 / 13
17 / 25
1/7
4 / 14
HR+HER2- (n=56)
2 / 25
3/8
4 / 17
0/6
When the PARPi-7 High patients are added to the graduating TN subset, the OR associated with V/C is 5.12, which is
comparable to that of the TN signature (OR: 4.29), while increasing the prevalence of biomarker-positive patients by 12%.
Evaluation of PARPi-7 in the context of the graduating signature under the I-SPY 2 Bayesian model is pending.
Conclusion: Our sample size is small. Our pre-specified analysis suggests the PARPi-7 signature shows promise for predicting
response to veliparib/carboplatin combination therapy relative to control. If verified in a larger trial, this cell-line derived signature
may contribute to the selection criteria of PARP inhibitor trials in the future.
PFS p-value
OS HR (95%CI)
OS p-value
ANG2
2.3 (1.0-4.9)
.04
3.1 (1.4-7.0)
.007
IL6
1.6 (1.1-2.3)
.02
1.9 (1.3-2.8)
.001
IL8
1.9 (1.3-2.8)
.001
2.8 (1.8-4.3)
<.001
CA9
1.8 (1.2-2.7)
.006
1.9 (1.3-2.9)
.002
On C2D1, levels of all proteins, except for IL6, had significantly changed. Whereas VEGF-A, ANG2, sTIE2 and IL8 decreased
significantly, sVEGFR2 and CA9 showed a significant increase. The median relative change of both IL8 and sVEGFR2 was
significantly different between patients having CR/PR vs. SD/PD (for IL8: -19.4% vs. 22.3%, p=.001 and for sVEGFR2: 6.5 vs.
2.1%, p=.01). A large relative increase in CA9 level was associated with better PFS (HR = 0.36, 95% CI, 0.19 0.68, p=.002) and
OS (HR = 0.50, 95% CI, 0.27 0.94, p=.03). All patients had very low levels of free VEGF-A on C2D1 (median 8 pg/ml).
Conclusions
In patients with HER2-negative LR/MBC receiving first-line bevacizumab-containing chemotherapy, high baseline plasma levels
of ANG2, IL6, IL8 and CA9 indicate a high risk for poor PFS and OS. These proteins might be useful for stratification according to
prognosis. Moreover, relative decrease in IL8 and increase in sVEGFR2 on C2D1 are associated with response, whereas a large
relative increase in CA9 is associated with better PFS and OS. Changes in these proteins after one cycle might be early
indicators of efficacy of bevacizumab combined with chemotherapy.
Financial support from Roche Netherlands.
Multivariate*
Endpoint
p-value
p-value
PFS
HFS(Yes vs No)
0.577(0.431,0.772)
0.0002
0.558(0.409,0.760)
0.0002
HFS Grade 1
0.639(0.440,0.927)
0.0184
0.536(0.350,0.821)
0.0042
HFS Grade 2
0.481(0.321,0.720)
0.0004
0.487(0.320,0.741)
0.0008
HFS Grade 3
0.641(0.411,0.999)
0.0495
0.740(0.466,1.178)
0.2043
HFS(Yes vs No)
<.0001
0.0001
HFS Grade 1
0.0375
0.0270
HFS Grade 2
0.0048
0.0098
HFS Grade 3
0.0006
0.0015
OS
* Significant baseline factors in multivariate models: ECOG (0 vs 1-2) in all models, ER/PR status (Any positive vs Other) in both
OS models.
Conclusion:
HFS is a strong predictor for prolonged PFS and OS in mBC patients receiving BEV + CAP first-line treatment. Early occurrence
of HFS and HFS severity might be used for treatment modifications and patient motivation. Detection of biomarkers for HFS could
strengthen this approach.
Response
HR Deficient
Number (% response)
HR Non-deficient
Number (% response)
Odds Ratio
(95% CI)
PR = no (RCBII,III)
14
19
Reference: Non-deficient
PR = yes (RCB0,I)
15 (52%)
2 (9.5%)
21
21
Reference: Non-deficient
8 (28%)
0 (0%)
P-value
0.0011
0.0066
Conclusions This study demonstrates that the HRD score can be used as a tool to identify patients with breast tumors with
underlying HR deficiency, including in BRCA1/2 non-mutated tumors, that will benefit from platinum therapy.
Patients
HR
OS
p
HR
Total Population
555
1.69
<0.0001
2.43
<0.0001
Letrozole Arm
274
1.58
0.001
2.24
<0.0001
Tamoxifen Arm
261
1.92
<0.0001
2.6
<0.0001
395
1.55
<0.0001
2.45
<0.0001
In the total population with available serum (n=555), multivariate analysis for TTP revealed that high serum activin A was a
significant adverse prognostic factor (HR=1.46, p<0.001). Multivariate analysis for OS also revealed that high serum activin A was
an independent adverse prognostic factor (HR 1.78, p<0.0001).
Conclusions: Patients with high pretreatment serum activin A levels had a significantly reduced ORR, CBR, TTP and OS
compared to patients with low serum activin A. The results were similar within the letrozole or tamoxifen treatment arms, and
within the serum HER2 not-elevated subgroups. High pretreatment serum activin A level is associated with relative resistance to
hormone therapy in first-line metastatic breast cancer.
Neratinib (n=106)
HR-/HER2+ (n=28)
Control (n=62)
0/4
12 / 18
1/1
1/5
24 / 57
5 / 28
5 / 28
OR between EGFR Y1173 groups in the N relative to control arm is 10.1. When EGFR Y1173 High patients are added to the
graduating HR-/HER2+ subset, the OR associated with treatment is 3.2 and is comparable to that in the HR-/HER2+ signature
(OR: 2.1), while increasing the prevalence of biomarker-positive patients by 50%. Evaluation of EGFR Y1173 under the I-SPY 2
Bayesian model is pending.
Conclusion: Our sample size is too small to draw definitive conclusions. Our exploratory analysis reveals that HER family
phosphoproteins associate with response to N, but only phosphorylated EGFR Y1173 appears to add value to the graduating
signature. Given that this biomarker would expand the patient population that may benefit, it merits evaluation in other ongoing
trials of neratinib.
V/C (n=71)
Control (n=44)
MP1 (n=32)
MP2 (n=39)
MP1 (n=34)
MP2 (n=10)
TN (n=59)
3/8
19 / 30
3 / 13
2/8
HR+HER2- (n=56)
1 / 24
4/9
4 / 21
0/2
The OR between MP1/MP2 risk groups for predicting pCR is 9.71 in the V/C arm (p=6.63E-05), in comparison to an OR of 0.97 in
the control arm (p=1). There is a significant biomarker x treatment interaction (p=0.023), which remains upon adjusting for HR
status (p= 0.028). Based on the I-SPY 2 Bayesian model, a Phase III trial with 300 MP2 patients has a 95% predictive probability
of success. When the MP2 patients are added to the graduating TN subset, the OR associated with V/C is 4.36, which is
comparable to that of the TN signature (OR: 4.29), while increasing the prevalence of biomarker-positive patients by 10%.
Conclusion: In our exploratory analysis, MP2 suggests higher sensitivity to V/C combination therapy relative to controls. This
observation has prompted an investigation into the biological mechanisms distinguishing the MP1/MP2 subtype that may account
for this specificity.
Neratinib (n=115)
Control (n=76)
MP1 (n=74)
MP2 (n=41)
MP1 (n=59)
MP2 (n=17)
HER2- (n=105)
0 / 17
15 / 33
7 / 39
5 / 16
HER2+ (n=86)
22 / 57
4/8
5 / 20
0/1
MP2, one of the 10 eligible signatures, did not meet the graduation threshold; and MP1/MP2 did not show a significant biomarker
x treatment interaction (OR in neratinib relative to control arm = 1.25). The MP1/MP2 x treatment interaction remains
non-significant after adjustment for HR and HER2 status (p=0.54). In HER2- patients receiving neratinib, 45% (15/33) of MP2
patients achieved a pCR, compared to 0% (0/17) of MP1 patients. In the HER2- controls, there is a 31% pCR rate in MP2 (5/16)
vs. 18% in MP1 (7/39) patients (OR=2.14). This difference in performance between treatment arms appears significant (p=0.041).
90% of HER2+ patients are MP1, thus MP1/MP2 status x treatment interaction within the HER2+ subtype cannot be evaluated.
Conclusion: Within the I-SPY 2 population as a whole, MP1/MP2 stratification does not appear to be a specific biomarker of
response to neratinib relative to the control arm. The number of HER2- patients is small and precludes any definitive conclusion,
but these data motivate further investigation of the biological mechanisms distinguishing MP1 from MP2 to better understand
chemotherapy and/or neratanib responsiveness.
HR+/HER2-
HR+/HER2+
HR-/HER2+
TN
0.310 (0.159-0.607)
P value
<0.001
.039
2.454 (0.917-6.564)
P value
.074
.085
2.587 (1.015-6.595)
P value
.047
.022
2.062 (0.918-4.632)
P value
.080
.019
0.399 (0.177-0.896)
P value
.026
.119
.018
.003
<0.001
.001
<0.001
<0.001
<0.001
<0.001
Overall survival
Miller Payne grade
RCB class
RDBN level
<0.001
P value
.127
.005
.163
<0.001
HR (95% CI) 11.369 (2.618-49.375) 3.042 (0.801-11.550) 1.970 (0.805-4.821) 3.262 (2.145-4.961)
P value
ypTNM Stage
.006
.001
.102
.138
2.623 (0.851-8.080)
P value
.093
.228
.006
<0.001
<0.001
RCB, residual cancer burden; RDBN; residual disease breast and nodes.
Especially in TN, Kaplan-Meier survival curves for disease-free survival were clearly separated by all assessment methods.
However, in HR+/HER2+ and HR-/HER2+ tumors, the association of patients survival with response assessment results was
Pre-Tx
AS
ER-PCR Status
(Pre-Tx)
Negative
Positive
Total
AS 2-3
17
(94%)
1
(6%)
18
AS 4-5
9
(56%)
7
(44%)
AS 6-7
0
(0%)
21
(100%)
21
Total
26
29
55
Conclusions
Results confirm earlier reports showing substantial disagreement in ER measured by IHC vs RT-PCR in patients with lower
ER-expressing tumors
The clinical implications are that a substantial number of patients with low ER by IHC may have little to no benefit from HT
The 21-gene assay may be useful in selecting patients likely to benefit from HT
Further studies in larger cohorts are required to confirm these findings.
pathological response
CR
nouptake
11
uptake
20
Sensitivity-92%, Specificity-87%, Positive predictive value-79%, Negative predictive value-95%, LR+7, LR-0.09, LR---73.3,
accuracy-89% p value <0.000
shows T/B ratios When the T/B ratio was >2.55, the tumor can be considered to be chemo sensitive to Doxorubicin based
NACT.On the contrary, when the T/B ratio was <2.55, it is unlikely that the tumor regresses completely following Doxorubicin
based NACT. Ability of Scintimammography to assess response to NACT was done comparing post-NACT scintimammograms
with the final histopathology
T/B ratio & path response
Scintimammography
CR (tumor no)
PR(Tumor yes)
p value
T/B Ratio2.55>
18
12
<0.003
30
<0.003
EXE (nM)
17-dihydroEXE (nM)
24 hours (n=32)
EXE (nM)
17-dihydroEXE (nM)
Ki67 change
BMI
-9 (-16, -5)
28 (24, 32)
26 (23, 30)
anti-proliferative effect of EXE on BC tissue. Larger studies should determine the clinical implications of this gene on EXE
efficacy.
CC
100
100
100
CT
100
115
130
TT
100
120
140
Results:
To date 32 patients are evaluable for pharmacogenetic guided epirubicin dosing (8 CC genotypes, 14 CT genotypes and 10 TT
genotypes). All 32 patients received epirubicin100 mg/m2 in cycle one and a single patient in each of the CC and CT genotypes
experienced grade 3 febrile neutropenia and were not dose escalated. All other patients with CT and TT genotypes were dose
escalated in cycle 2 and all but two patients in the CT and TT genotypes were dose escalated in cycle 3.
The incidence of febrile neutropenia was not dose dependent as all three genotypes had similar incidence in each cycle whereas
leucopenia was genotype and dose dependent. The incidence of leukopenia increased in patients with CT and TT genotypes as
their dose was increased and cycle 3 leukopenia rates were similar to patients with the CC genotype receiving standard dose
epirubicin.
Conclusions:
Pharmacogenetic guided epirubicin dosing is well tolerated. This study is ongoing and updated data will be presented.
Characteristic
Univariate analysis
Multivariate analysis
OR
95%CI
P value
OR
95%CI
P value
N stage: N0 vs. N+
0.33
0.03-3.64
0.368
0.33
0.03-3.63
0.333
2.56
0.20-33.1
0.473
fT4
0.78
0.43-1.42
0.417
0.66
0.33-1.29
0.581
TSH
3.24
1.09-9.70
0.035
17.3
0.76-391
0.074
Table 1. Univariate and multivariate logistic regression models of baseline characteristics and TSH and fT4 predictive of pCR.
Conclusion: TSH levels at baseline were higher in breast cancer patients with pCR. Chemotherapy blunts thyroid function, and a
large decline of fT4 was associated with less side effects. These data suggest that thyroid hormones may interact with
chemotherapy to modulate treatment (side-) effects in patients with breast cancer.
Statin use
P value
No
Yes
N = 37,765
N = 3,337
Age (years)
55.0
54.2
63.8
< 0.001
BMI (kg/m2)
25.4
25.2
27.1
< 0.001
< 0.001
Premenopausal
40.2 (16,506)
43.1 (16,272)
7.0 (234)
Perimenopausal/unknown
3.3 (1,349)
3.5 (1,304)
1.4 (45)
Postmenopausal
56.6 (23,247)
53.5 (20,189)
91.6 (3,058)
Model 2
Model 3
Model 4
8.01
7.93
7.93
7.94
Statin use
Volumetric percent density (%)
No
Yes
6.89
7.73
7.74
7.66
P value
< 0.001
0.001
0.001
< 0.001
No
56.8
57.1
57.1
57.0
Yes
60.7
57.6
57.5
58.0
P value
< 0.001
0.31
0.32
0.06
None
8.01
7.93
7.93
7.94
Lipophilic
6.92
7.73
7.77
7.69
Hydrophilic
6.46
7.32
7.34
7.26
P value
< 0.001
< 0.001
< 0.001
< 0.001
None
56.8
57.1
57.1
57.0
Lipophilic
60.8
57.7
57.7
58.1
Hydrophilic
58.2
55.0
54.8
55.3
P value
< 0.001
0.20
0.66
0.21
Statin type
Volumetric percent density (%)
Model 1: adjusted for age Model 2: Model 1 + BMI Model 3: Model 2 + menopausal status, HRT use, parity, age at menarche,
education level, smoking, alcohol consumption and benign breast disease Model 4: Model 3 + low-dose aspirin and metformin
use
Conclusions:
Statin use was associated with a lower mammographic percent density, although no evidence was found for an effect of statins
on absolute dense volume. The observed interaction between statin and HRT use requires further investigation.
1983-1992
1993-2002
2003-2011
p value
0.1
0.1
0.2
0.2
<0.001
<50
0.0
0.0
0.1
0.05
<0.001
50+
0.2
0.4
0.6
0.7
<0.001
Hodgkins
0.0
0.002
0.003
0.002
<0.001
NHL-B-cell
0.04
0.1
0.2
0.2
<0.001
DLBCL
0.03
0.1
0.1
0.1
0.001
Follicular
0.005
0.02
0.03
0.04
0.062
Marginal Zone
0.0
0.0
0.03
0.1
<0.001
0.0
0.001
0.004
0.01
<0.001
All
Age
Lymphoma
NHL-T-cell
By age, the greatest incidence increase was for women 50 and older (0.02/100,000 in 1973-82 to 0.68/100,000 in 2003-11,
p<0.001). Both B-cell marginal zone lymphoma and T-cell lymphoma have had steep increases in incidence from 1983-1992 to
2003-2011 (0/100,000 to 0.06/100,000, p<0.001 and 0.001/100,000 to 0.008/100,000, p<0.001, respectively). The incidence of
DLBC and follicular lymphomas arising in the breast rose from 1973-1992 and subsequently stabilized. Age-adjusted, all-cause
mortality has dropped significantly since 1992, as has cause-specific mortality (Table 2).
Table 2: Primary Breast Lymphoma-Mortality and Initial Local Therapy
Age-adjusted, 5-year Mortality (% deceased)
1973-1982
1983-1992
1993-2002
2003-2011
p value
All-cause
51%
54%
33%
22%
<0.001
Cause-specific
43%
43%
25%
15%
<0.001
Both
26%
26%
23%
12%
0.274
Radiation only
8%
11%
18%
31%
<0.001
Surgery only
59%
47%
34%
16%
<0.001
None
4%
14%
21%
40%
<0.001
For the entire cohort, no local therapy and radiation therapy, as initial local treatment, has increased over time. Surgery alone has
decreased substantially.
Conclusions: Primary breast lymphoma remains a rare diagnosis; however the incidence has risen markedly over the last four
decades, with B-cell marginal zone and T-cell lymphomas increasing significantly in the most recent time period. Further research
is required to better understand the factors contributing to these trends. Survival gains over time suggest either that lymphoma
treatments have improved or that the excess cases are treatment responsive or non-survival-threatening.
Prior to 2004
After 2004
p-value
60.1
60.3
<0.001
20.1
20.2
<0.001
1.3
1.2
<0.001
36.8
34.6
<0.001
41.1
42.2
<0.001
41.7
53.5
<0.001
Conclusions: These data demonstrate that while early detection may be increasing the number of low grade node-negative
tumors in older patients, our use of systemic therapy has become more liberal. Increasing use of hormonal therapy may be due to
more aggressive guidelines and a lower threshold for ER-positivity; increasing use of chemotherapy may be the result of more
efficacious drugs with lower toxicity being available to treat limited disease. Nonetheless, implications in terms of cost vs. benefit
of our more aggressive approach may warrant further evaluation.
Characteristic
Overall (n=306)
De novo (n=142)
Recurrent (n=164)
57 (27-86)
54 (27-83)
59 (30-86)
White
233 (76)
105 (74)
128 (78)
Black
54 (18)
28 (20)
26 (16)
Other
8 (3)
3 (2)
5 (3)
Not available
11 (4)
6 (4)
5 (3)
Not Hispanic
257 (84)
120 (85)
137 (84)
Hispanic
31 (10)
17 (12)
14 (9)
Not available
18 (6)
5 (4)
13 (8)
Pre-menopausal, n (%)
61 (20)
42 (30)
19 (12)
209 (68)
90 (63)
119 (73)
Race, n (%)
Ethnicity, n(%)
ER and PR negative
92 (30)
50 (35)
42 (26)
Unknown
5 (2)
2 (1)
3 (2)
2.0 (1-7)
2.0 (1-6)
2.0 (1-7)
Bone
141 (46)
76 (54)
65 (40)
Liver
120 (39)
63 (44)
57 (35)
Nodes
89 (29)
49 (35)
40 (24)
Lung
87 (28)
38 (27)
49 (30)
Chest wall
43 (14)
12 (9)
31 (19)
CNS
30 (10)
9 (6)
21 (13)
Conclusions
The proportion of patients with de novo MBC appears to have increased over time to 46% in SystHERs, compared with 33% in
RegistHER, a registry study that enrolled 1023 patients with HER2-positive MBC from 20032006 (Yardley et al, 2014), and
predated the broad use of HER2-targeted therapy in the (neo)adjuvant setting. The DFI for patients with recurrent HER2-positive
MBC also appears to be longer (median DFI 43.6 months in SystHERs vs. 32.6 months in registHER). We hypothesize that the
proportion of patients with de novo MBC within the metastatic population is higher due to the use of advanced screening
techniques which allow better detection of metastases, and due to a reduction in recurrences related to the availability of
HER2-targeted adjuvant therapy. Changes in the population characteristics of patients with HER2-positive MBC may impact
treatment strategies and have trial design implications. Updated data from approximately 500 patients are expected by the time of
presentation.
South
Incidence
6.1 (0.42)
16.57 (0.53)
10.5 (6-15.2)
<0.05
Mortality
7.8 (0.46)
9.56 (0.43)
2 (1-3.2)
<0.05
Center
Incidence
12.9 (0.27)
26.9 (0.33)
7.6 (5.7-9.6)
<0.05
Mortality
14 (0.29)
14.4 (0.25)
0.3 (0-0.6)
<0.05
North
Incidence
24.2 (0.64)
26.8 (0.54)
1 (-1.6-3.7)
NS
Mortality
17.6 (0.57)
18.8 (0.48)
0.7 (0-1.3)
<0.05
Conclusions: BC trends in Mexico show a continuous increase in incidence and mortality from 2001 to 2011, which could reflect
population growth, ageing, lifestyle modifications and changes in access to diagnosis and treatment. These changes could be an
expression of epidemiological transition in developing countries such as Mexico. A significant growth in both incidence and
mortality was found in both the Center and the South. In the North, incidence rates remained unchanged while mortality rates had
a significant rise, which was comparable to that of the other regions. One possible unexplored explanation for this observation
could be the recent wave of drug-related violence and high criminality rates in the north of the country, which may perhaps cause
underreporting of cases and disrupt availability of medical attention and access to healthcare in an otherwise developed and
wealthy region.
2013
Preoperative MDT
81%
96%
Postoperative MDT
90%
98%
80%
85%
6.1%
5.0%
25%
20%
10%
12%
15%
19%
39%
44%
MDT= Multi-disciplinary team meeting; PST= Primary Systemic Treatment; DCIS= Ductal Carcinoma In Situ
Conclusions
The continuous cycle of registration and providing feedback by clinical auditing provides a powerful tool for quality monitoring and
improving breast cancer care. Improvements of monodisciplinairy surgical and pathological aspects of care have been reached in
a relatively short time period. However, for more complicated multidisciplinary issues like the use of primary systemic treatment
and immediate reconstruction, detailed analyses of the variation between hospitals is needed to further improve these aspects of
breast cancer care.
Univariate Analysis
Perceived High Risk-%
meeting guidelines
Fruit/Veg Intake (>=5 svg
daily)
Multivariate Analysis
p-value OR (95% CI) p-value
4.42%
0.5679
1.14
(0.78-1.66)
0.5110
95.13%
0.3093
0.84
(0.61-1.17)
0.3076
27.74%
0.1465
1.02
(0.87-1.21)
0.7921
4.90%
de novo MBC
HR+/HER2-
38118
95.1 (94.8-95.3)
1351
52.5 (49.8-55.1)
38.7 (35.9-41.3)
HR+/HER2+
6920
94.6 (94.1-95.2)
450
55.1 (50.4-59.6)
45.3 (36.5-50.5)
Unclassified
10813
91.2 (90.6-91.7)
894
26.0 (23.1-28.9)
11.9 (9.7-13.8)
HR-/HER2+
3589
88.4 (87.3-89.4)
333
37.5 (32.3-42.7)
23.1 (19.2-27.4)
TN
7083
84.3 (83.4-85.1)
357
17.7 (13.9-21.9)
12.7 (11.2-14.1)
HER2-neg*
45550
93.3 (93.1-93.6)
1745
44.8 (42.4-47.1)
30.9 (28.6-33.6)
HER2-pos*
10635
92.5 (92.0-93.0)
797
47.6 (44.0-51.0)
33.6 (30.7-37.1)
* Pts with available HER2 data, but missing HR data were included
Total
(N=657)
HR+/HER2(N=365)
HR-/HER2(N=118)
HR+/HER2+
(N=111)
HR-/HER2+
(N=63)
n(%)
n(%)
n(%)
n(%)
n(%)
265(40)
145(40)
37(31)
50(45)
33(52)
- Bone
283(43)
167(46)
49(42)
46(41)
21(33)
- Brain*
105(16)
45(12)
32(27)
17(15)
11(17)
- Liver*
171(26)
76(21)
40(34)
35(32)
20(32)
- Lung*
127(19)
64(18)
36(31)
11(10)
16(25)
494(75)
277(76)
97(82)
76(68)
44(70)
614(93)
349(96)
98(83)
109(98)
58(92)
- CT*
422(69)
204(58)
92(94)
78(72)
48(83)
- BIO*
124(20)
1(<1)
4(4)
68(62)
51(88)
- HT*
210(34)
169(48)
9(9)
31(28)
1(2)
293(48)
159(46)
45(46)
71(65)
18(31)
26(4)
10(3)
8(8)
5(5)
3(5)
-- Discontinuation/gap*
174(28)
105(30)
36(37)
15(14)
18(31)
-- Censored*
121(20)
75(21)
9(9)
18(17)
19(33)
*p<0.05. Not all groups are mutually exclusive. Percentages may not total 100% due to rounding or overlap.
CONCLUSION: In this study of commercially insured mBC women, initial therapy choices were generally consistent with NCCN
guidelines for three of the four subtypes, while the largest subtype of HR+/HER2- women had a higher than expected utilization of
CT. Future analyses of study data will investigate appropriateness of these initial treatments and their impact on clinical and
economic outcomes.
MBC
HER2+
HER2-
HER2+
HER2-
No
5898 (33.4%)
52467 (61.0%)
317 (22.6%)
1519 (43.7%)
Yes
11463 (64.8%)
32403 (37.7%)
1063 (75.9%)
1894 (54.5%)
322 (1.8%)
1128 (1.3%)
20 (1.4%)
60 (1.7%)
Any chemotherapy
Unknown
3546 (76.9%)
24794 (89.8%)
Yes
953 (20.7%)
2381 (8.6%)
Unknown
110 (2.4%)
429 (1.6%)
1943 (42.2%)
19296 (69.9%)
Yes
2556 (55.5%)
7879 (28.5%)
110 (2.4%)
429 (1.6%)
Unknown
* not applicable
1975-84
2 vs 1 [+1]**
4 vs 4 [0]
8 vs 5 [+3]
13 vs 3 [+10]
2000-09
50 vs 44 [+6]
52 vs 46 [+6]
52 vs 41 [+11]
50 vs 33 [+17]
1975-84
42 vs 37 [+5]
80 vs 66 [+14]
2000-09
1975-84
7 vs 9 [-2]
13 vs 14 [-1]
28 vs 29 [-1]
44 vs 46 [-2]
2000-09
51 vs 54 [-3]
65 vs 68 [-3]
72 vs 89 [-17]
90 vs 110 [-20]
IN-SITU
INVASIVE
MORTALITY
Regimen Type
Total, n
No chemo
14024
3.3 (3.6)
29.3
Reference
A-based
7465
4.8 (3.0)
27.7
0.96 (0.88-1.04)
CMF
4389
6.0 (3.9)
29.1
1.04 (0.95-1.14)
T-based
1030
2.8 (1.8)
39.4
0.91 (0.74-1.12)
Other
1140
5.1 (4.0)
31.2
1.04 (0.89-1.22)
Gene
ER-C vs ER+C
ER-T vs ER+T
ER-C vs RM
ER+C vs RM
DHRS2
2.4 (0.034)*
0.16 (0.042)*
6.0 (0.014)*
1.3
HMGCS2
2.9 (0.050)*
0.15 (0.0095)*
6.5 (0.006)*
2.2
UGT2B7
2.2 (0.004)*
0.77
1.8
1.0
UGT2B11
2.4 (0.019)*
0.11 (0.0068)*
9.6 (0.013)*
2.0
UGT2B28
2.3 (0.009)*
0.15 (0.018)*
2.8 (0.029)*
1.5
GLYATL1
2.9 (0.010)*
0.37
4.3 (0.026)*
1.4
GSTT2
1.1
0.37 (0.040)*
1.5
1.3
ALOX15B
1.6
0.56
2.5 (0.016)*
1.5
SERHL
1.8
0.29
4.9 (0.0047)*
1.4
ER-C: ER- CUB; ER+C: ER+ CUB; ER-T: ER- tumor; ER+T: ER+ tumor; RM: reduction mammoplasty
High vs
Average
High vs
control
Average
vs control
Age(yr)
meanSD 41.129.86
51.037.98
50.529.09
0.001
<0.001
0.141
PD (%)
meanSD 52.9012.93
45.5216.04
41.5215.80
<0.001
(<0.001*)
<0.001
(<0.001*)
<0.001
(<0.001*)
PD_median
(%)
<50
442 (41.5%)
567 (59.4%)
1414 (69.7%)
<0.001
<0.001
<0.001
50
624 (58.5%)
387 (40.6%)
615 (30.3%)
<25
26 (2.4%)
106 (11.1%)
329 (16.2%)
<0.001
<0.001
<0.001
25 <50
416 (39.0%)
461 (48.3%)
1085 (53.5%)
50 <75
564 (52.9%)
360 (37.7%)
570 (28.1%)
75
60 (5.6%)
27 (2.8%)
45 (2.2%)
PD_quartile
(%)
25%PD<50%
(<25%(ref))
TNBC (control(ref))
Non-TNBC
Luminal
Non-Luminal
HR 1.91, 95%CI
1.50-2.43
HR 1.77, 95%CI
1.38-2.26
HR 3.04, 95%CI
2.00-4.64
50%PD<75%
HR 8.85, 95%CI
4.45-17.61
HR 3.81, 95%CI
2.98-4.89
HR 3.89, 95%C
I3.03-5.00
HR 5.08, 95%CI
3.32-7.80
75%PD
HR 16.25, 95%CI
6.97-37.87
HR 4.09, 95%CI
2.61-6.41
HR 4.09, 95%CI
2.61-6.43
HR 8.16, 95%CI
4.41-15.07
Among the 1066 high risk group, 81.5% (869) undergone BRCA testing and 70(6.6%) had BRCA1, 78(7.3%) had BRCA2
mutations without significant difference in density. Similar strong magnitude association of mammographic density was observed
in both BRCA mutated/non-mutated tumors and among subtypes. The ongoing GWAS and whole exome analysis of this
population-subset will give insight into the tumor etiology and how density could stratify breast cancer risk for personalized
screening especially in high risk population.
No. of Participants
ER+
Her2+
BRCA1/2 +
BMI
Cayman Islands
66
51.4
29/44
3/43
1/27
30.3
Barbados
89
46.6
52/73
9/73
4/87
27.5
Dominica
60
52.2
9/13
2/7
0/46
28.7
Jamaica
137
48.6
69/105
28/105
28.7
45.5
4/6
1/5
1/5
27.4
Haiti
34
48.5
NA
NA
NA
24.9
TAH-BSO
Parity
Mean Age
p-value
Performed
71
53
0.005
Not Performed
304
48
Nulliparous
60
42
0.0001
Pregnancy
Multiparous
319
50
None
48
43
More than 1
331
50
0.0001
Conclusions: This population-based study provides an insight into pattern of risk factors both genetic and environmental of
breast cancer incidence and subtype across the Caribbean. In conclusion 1) genetic causes of breast cancer appear rare outside
of the Bahamas, 2) fertility factors appear important in the development of breast cancer, 3) TAH-BSO is common as both a form
of contraception and because of the high incidence of fibroids in the Caribbean and it may be protective, 4) BMI may impact on
breast cancer development and 5) screening mammography is rare and the vast majority of mammography performed is
diagnostic in nature.
Number of Pts
10 yr OS(%)
p-value
10 yr DFS(%)
p-value
<4 weeks
774
69.6
ref
60.9
ref
4-8 weeks
2263
71.5
0.1351
64.7
0.1043
>8-12 weeks
742
77.3
0.0002
72.6
0.0001
>12 weeks
35
77.0
0.4531
73.9
0.2124
However, in multivariate analysis there was no significant association between any sTTC time periods and either outcome (see
table below). Covariates which did show a significant association are listed in the table.
OS p-value
DFS p-value
<4 weeks
ref
ref
4-8 weeks
0.7762
0.6764
>8-12 weeks
0.4251
0.1396
>12 weeks
0.6861
0.9312
Age
<0.0001
<0.0001
BMI
0.0238
0.0298
Menopause Status
0.0126
0.1226
Performance Status 2
0.0256
0.1665
Surgery Type
<0.0001
<0.0001
Pathological Stage 3
0.0037
0.0229
<0.0001
<0.0001
ER Negative
<0.0001
<0.0001
Time to Adjuvant
Chemotherapy Regimen
<0.0001
<0.0001
The fact that the statistical significance of sTTC in univariate analysis was lost when covariates related to DFS and OS were
accounted for in a multivariate analysis suggested there might be a relationship between sTTC and the risk of adverse outcomes.
A disease risk score analysis was therefore carried out, but there was no indication of an advantage to an earlier sTTC within
disease risk categories.
Conclusions: Within the context of chemotherapy given within 12 weeks, we were unable to demonstrate an effect on survival
based on time to adjuvant chemotherapy in our multivariable analysis. Those treated later did not do significantly worse than
those treated earlier. Significant covariates which effected survival were consistent with predictors of poor prognosis (younger
age, poorer performance status, increased disease burden and receptor negativity). There is a potential relation of patient risk to
time to treatment which requires further study.
TP(n=61)
TE(n=60)
P(2 test)
Median age,years(range)
53.5(35-68)
51.8(32-69)
0.614
61(100%)
60(100%)
27(44.3%)
25(41.7%)
0.773
ER/PR-positive
42(68.9%)
43(71.7%)
0.735
HER2-positive
18(29.5%)
15(25.0%)
0.578
33(58.1%)
35(55.2%)
0.639
Visceral
40(65.6%)
42(70.0%)
0.602
Lymph
25(41.0%)
28 (46.7%)
0.529
Lobular
12(19.7%)
11(18.3%)
0.851
Ductal
44(72.1%)
42(70.0%)
0.796
Other
5(8.2)
7(11.7%)
0.523
Yes
28(45.9%)
25(41.7%)
0.639
No
33(54.1%)
35(58.3%)
Yes
13(21.3%)
11 (18.3%)
No
48(78.7%)
49(81.7%)
Metastatic sites
Histology
Adjuvant radiotherapy
0.681
Results: After a median follow-up of 21.3 months, there was no significant difference between the two treatment arms in objective
response rate(ORR) (68.5 % vs. 73.3%, respectively;). And both the progression-free survival (P=0.704)and overall survival
(P=0.403)were very similar between the two arms. Both regimens were well tolerated. The main toxicities were
myelosuppression, gastrointestinal reactions and alopecia. There was more grades 34 alopecia and more nausea with
TE(p<0.05). For the QLQ-C30 questionnaire statistically significant changes after treatment were fatigue(0.031) in TP,
nausea/vomiting(p=0.041) and lose of appetite(p=0.048) in TE.
Table 2 Summary of adverse events per patient; p-values are given overall
TP (n=61)
TE (n=60)
P value
Adverse events
Grades 1-4
Grades 3-4
Grades 1-4
Grades 3-4
Neutropenia
46
13
38
0.149
Leukopenis
50
11
40
0.054
Anemia
0.981
Thrombocytopenia
0.714
Lymphocytopenia
0.391
Vomiting
15
20
0.289
Nausea
27
41
0.010
Diarrhea
0.714
Alopecia
44
15
60
31
0.000
Cardiotoxicity
0.523
Hypersensitivity reaction
ALT increase
0.523
Peripheral neurotoxicity
20
19
0.895
Trial
Median OS, mo
260 q3w
229
15.0
second line
260 q3w
131
13.0
300 q3w
76
27.7
100 qw 3/4
76
22.2
150 qw 3/4
74
33.8
Median OS, mo
Subgroups With Aggressive Disease Features
VDM
SDFI
CA0123
(first line)
260 q3w
74
15.1
42
14.6
CA0243
(first line)
300 q3w
61
27.7
20
16.6
100 qw 3/4
60
19.6
21
19.1
150 qw 3/4
qw 3/4, weekly for the first 3 of 4 weeks.
1
59
32.1
14
18.6
Grade 3/4
Leukopenia
3 (25%)
Neutropenia
10 (83%)
Febrile Neutropenia
3 (25%)
Hypokalemia
1 (8%)
0 (0%)
PR
5 (41%)
SD24 weeks
2 (17%)
SD<24 weeks
3 (25%)
PD
2 (17%)
Clinical Benefit
7 (58%)
10 (83%)
OR
95% CI
p value
> 50
8500
Reference
< 50
7186
1.27
Ductal
12,926
Reference
Lobular
837
0.65
0.52 - 0.80
<0.001
Mixed ductal/lobular
577
0.71
0.57 - 0.89
0.003
Other
1346
0.82
0.72 - 0.94
0.003
872
Reference
5222
1.09
0.90 - 1.33
0.363
9592
1.76
1.45 - 2.13
<0.001
ER/PR+, Her2-
6738
Reference
ER/PR+, Her2+
2662
2.62
2.37 - 2.90
<0.001
ER/PR-, Her2+
1757
4.03
3.58 - 4.53
<0.001
Age
<0.001
1.19 - 1.37
<0.001
Histology
<0.001
Grade
<0.001
Molecular type
<0.001
ER/PR-, Her2-
4529
2.32
2.12 - 2.55
<0.001
T1
2147
Reference
T2
7167
0.71
0.64 - 0.79
<0.001
T3
3513
0.48
0.43 - 0.54
<0.001
T4
2859
0.50
0.44 - 0.57
<0.001
N0
5713
Reference
N1
7307
0.77
0.72 - 0.84
<0.001
N2
1646
0.68
0.60 - 0.78
<0.001
N3
1020
0.70
0.60 - 0.82
<0.001
Clinical T stage
<0.001
Clinical N stage
<0.001
CT + ZA
Odd's Ratio
95% C.I.
pCRb/total
Percentage
pCRb/total
Percentage
Total population
48/373
12.9
58/363
16.0
1.34
0.86-2.08
Postmenopausal patients
11/130
8.5
24/141
17.0
2.72
1.15-6.42*
Pre/perimenopausal patients
34/232
14.7
33/214
15.4
1.03
0.59-1.79
13/70
18.6
21/67
31.3
1.99
0.85-4.66
pCR/total
Percentage
pCR/total
Percentage
Total population
28/279
10.0
33/274
12.0
1.31
0.73-2.34
Postmenopausal patients
7/90
7.8
14/103
13.6
2.48
0.80-7.69
Pre/perimenopausal patients
19/178
10.7
18/163
11.0
1.00
0.49-2.08
9/52
17.3
15/51
29.4
1.80
0.63-5.21
pCR in breast
Overview of pCRb and pCR in both treatment arms in the total population and predefined subgroups
Conclusion:
This meta-analysis shows that no overall benefit from the addition of ZA to neoadjuvant CT. However, as has been seen in the
adjuvant setting, the addition of ZA to CT in postmenopausal women with early breast cancer appears to specifically increase the
effects of CT with increases in pCRb and pCR. Further translational research is warranted to explore the mechanism of these
observations and the potential treatment interaction with menopause.
Febrile neutropenia
No. of trials
RR (95%CI)
No. of trials
RR (95%CI)
Neoadjuvant/Adjuvant setting
1.21 (1.06-1.38)
0.004
1.27 (1.06-1.52)
0.008
Metastatic setting
1.21 (0.82-1.79)
0.34
1.38 (0.79-2.42)
0.26
Combination therapy
1.18 (1.04-1.35)
0.01
1.27 (1.06-1.52)
0.008
1.47 (1.02-2.12)
0.04
1.38 (0.79-2.42)
0.26
Treatment setting
(-)Arthralgia N(%)
(+)Arthralgia N(%)
p-value
Type of AI Used
Anastrozole
204
112(55.9)
92(45.1)
Letrozole
131
83(63.4)
48(36.6)
Exemestane
36
23(64.9)
13(36.1)
151
79(52.3)
72(47.7)
40
23(57.5)
17(42.5)
155
102(65.8)
53(34.2)
Adjuvant
348
200(57.5)
148(42.5)
Metastatic
22
17(77.3)
5(22.7)
112
55(49.1)
57(50.9)
171
96(56.1)
75(43.9)
0.19
Menopause timing
0.055
Type of therapy
0.067
40
29(72.5)
11(27.5)
(-) Therapy
218
118(54.1)
100(45.9)
(+) Therapy
127
81(63.8)
41(32.3)
4 (1-20)
3 (1-14)
(n= 17 evaluable)
(n= 34 evaluable)
CR
1 (6%)
PR
6 (35%)
3 (9%)
SD
6 (35%)
16 (47%)
PD
4 (24%)
15 (44%)
ORR (95%CI)
41% (18-67%)
9% (1.9-23.7%)
5.0 (0.1-12.8)
3.3 (1.4-5.1)
In an exploratory analysis, ORR in cohort A was higher in PARPi nave pts: 64% (7/11 pts).
Grade (G) 3-4 related adverse events occurred in >5% pts were myelosuppression (neutropenia 69%, febrile neutropenia 7%,
thrombocytopenia 14%); G3 fatigue 7%, transient transaminase increase 19% (G4: 2%), nausea 6% and dyspnea 6% (3 pts, 2 of
them due to pneumonitis). One patient with massive liver involvement and impaired function died on C1D4 due to multiorgan
failure possibly related to the study drug.
Conclusions: PM01183 has promising activity in pretreated MBC pts with BRCA mutation. Safety profile appears to be mostly
predictable and with non-cumulative toxicity. Treatment-related neutropenia was manageable with G-CSF and/or dose reduction.
After futility analysis, targeted activity has been met in cohort A (BRCA+), and recruitment will continue up to 53 evaluable
BRCA+ pts.
1 (2.4)
17 (40.5)
16 (38.1)
3 (7.1)
Not evaluable
5 (11.9)
18 (42.9)
95% CI
27.7, 59.0
24 (57.1)
95% CI
41.0, 72.3
Conclusions: This phase II study suggests that eribulin in combination with capecitabine is efficacious in patients with MBC, with
a safety and tolerability profile consistent with previous data. Phase III studies of this combination in patients with MBC are
warranted.
Stage IV
variables
N (%)
N (%)
p value
chemotherapy = no
14 (2%)
27 (25%)
<.001
radiation = no
59 (10%)
59 (55%)
<.001
surgery = no
1 (.2%)
52 (49%)
<.001
taxanes = no
270 (46%)
50 (63%)
.005
non-anthracycline regimen
108 (18%)
27 (34%)
.001
397 (66%)
70 (65%)
.933
Response, %
0.4
15.5
14.4
53.6
52.7
26.4
28.9
Not evaluable
4.5
3.6
Conclusions: In this exploratory, pooled analysis, magnitude of benefit from single-agent eribulin did not differ between the ILC
and IDC cohorts. While there was a limited numbers of pts with ILC, response rates, PFS, and OS were similar for the two pt
groups. The results with eribulin for advanced ILC contrast with data for other agents in early-stage settings, where ILC is
generally less responsive to chemotherapy than IDC. These findings may, however, underline changes in the disease biology
after exposure to previous therapies or changes inherent to disease progression.
Target Therapy
Objective Response
Mutations
Everolimus
NO
ERBB2, PIK3CA
Everolimus
Yes
Everolimus
Yes
PI3KCA, FGFR
Everolimus
Yes
AKT1, PI3KCA
Everolimus
Yes
AKT1
Everolimus
Yes
AKT1, FGFR
Everolimus
Yes
AKT1. FGFR
Everolimus
No
ERBB2, PI3K
Table1
Mutations Prevalence
Mutation
Prevalence
TP53
83,30%
PIK3CA
60,60%
FGFR
33%
CCND1
25%
PTEN
25%
BRCA2
16%
AKT
50%
Table2
CONCLUSION
. NGS was able to find genomic alterations in the majority of breast cancer tissues;
. Most patients did not have "actionable" mutations for which a specific therapy could be offered;
. We are just at the dawn of exploring this strategy and NGS can effectively contribute for increasing the understanding of the
disease.
REFERENCES
1. Jeffrey. Ross, Massimo Cristofanilli et al, JClin Oncol 3, 2013 (suppl; abster 1009).
Total
Luminal
Luminal/HER2
HER2
Triple negative
Luminal
18 (66.7)
2 (8.3)
1(4.2)
3(12.5)
6/24 (25.0)
Luminal/HER2
1 (14.3)
6 (85.7)
1/7 (14.3)
HER2
1 (100.0)
1/1 (100.0)
Triple negative
1 (16.7)
1 (16.7)
4(66.7)
2/6 (33.3)
20
10
10/38 (26.3)
Changes in management included the addition of trastuzumab in patients with gain of HER2 (n=3), the use of chemotherapy in
those with loss of HR (n=6) and provision of endocrine therapy for those with gaining HR (n=3). Response rate of discordant
group and concordant group were 10.0% and 21.4%, respectively (p=1.000). Clinical benefit rate of discordant group and
concordant group were 40.0% and 67.9%, respectively (p=0.150). There is no significant difference of time to progression (TTP)
between the discordant and concordant groups (169.5days vs.319.5days, p= 0.081).
CONCLUSION
Patients with receptor discordance tended towards worse response rate and shorter TTP, leading to the poor prognosis of the
recurrent breast cancer patients with receptor discordance.
CK+CD45- CTCs
pCR (n=6)
RCB-I (n=2)
RCB-II (n=5)
RCB-III (n=4)
P-value
Negative (n=9)
1(11)
2(22)
3(33)
3(33)
0.146 0.057*
Positive (n=8)
5(62)
0(0)
2(25)
1(13)
All data are no. of patients (%) Two did not have an RCB status available, and 1 did not have a T0 sample. * Comparing pCR
status (pCR vs no-pCR) between CTCs negative and positive.
CTC at D1C2
CEC Baseline
CEC at D1C2
37 (21%)
>20
>20
65 (39%)
<5
45 (26%)
>20
20
25 (15%)
<5
1 (<1%)
20
>20
27 (16%)
<5
<5
90 (52%)
20
20
49 (30%)
Total
173
166
CTC number at baseline line and CTC D1C2 were correlated (p<0.01) (Spearman test). There was no correlation between CEC
at baseline or at D1C2 with CTC or CTC changes. Final analysis will be completed when 206 couples for both CTC and CEC at
baseline and D1C2 will be available. Accrual is still ongoing.
Conclusion: this 22% rate of failure to reduce CTC < 5 after one cycle of first line CT in a homogeneously bevacizumab-treated
cohort of MBC patients did not differ from previous series. As second lines of chemotherapy do not improve the poor prognosis of
this group of patients according to the SWOG 500 study results (Smerage et al, JCO 2014), trials of novel therapeutic agents
Patient
Group/N
Tumor
status
HT
Status
CTC
ER+/million
(meanSD)
ER+ HST
Metastatic Sensitive 130379260109
sensitive/4
CTC
CTC ER+GREB-1
ER-/million(mean Acetylation+(mean
SD)
SD)
PREDICTED
ESTROGEN
RESPONSE
336594
0.95*0.08
0.015*0.02
Sensitive
ER+>ER-
ER+ HST
Metastatic Resistant
resistant/2
82429.7
73346421
0.87*0.13
1*
Resistant
ER+<ER-
ERMetastatic Resistant
metastatic/1
18758
21498
1*
ND
Resistant
ER+<ER-
34000000
89500000000126600000000
Sensitive
ER+>ERactive
GREB-1
0*
0*
ERprimary/2
Healthy
controls/2
None
None
0.50.7071
Her 2
n (%)
OR ( 95% CI)
0.562
0.038
No
19
7 (36.8)
ref
Yes
1.75 [0, 4]
12
9 (75.0)
n (%)
OR ( 95% CI)
CellSearch
Her 2
p-value
p-value
0.704
p-value
0.056
No
19
6 (41.2)
ref
Yes
12
8 (34.7)
Conclusions: The enumeration of CTCs showed strong prognostic significance in MBC raising interest in a more accurate
molecular characterization to achieve the possibility for a dynamic molecular monitoring. Introducing novel methodologies should
demonstrate comparable detection rate for epithelial cells. This new microfluidic system showed accuracy and flexibility to move
to the next phase of molecular testing with the intent of assessing the value as liquid biopsy.
No CLS-B, N=48
CLS-B +, N=52
Glucose, mg/dL
73.2 (8.0)
84.3 (37.6)
0.04
Insulin, mU/L
4.3 (2.1)
5.6 (2.9)
0.01
105.9 (31.0)
119.4 (32.4)
0.04
71.9 (15.8)
62.1 (16.1)
0.003
Triglycerides, mg/dL
69.2 (26.3)
104.9 (50.6)
<0.001
Leptin, pg/mL
12.0 (10.1)
22.6 (19.7)
<0.001
Adiponectin, ng/mL
13.7 (5.2)
10.4 (5.4)
0.002
hsCRP, ng/mL
1.0 (1.4)
2.3 (2.7)
0.003
Conclusions: Breast WAT inflammation, which we have previously linked to increased aromatase activity, is associated with
biochemical changes that occur in the metabolic syndrome, a risk factor for BC. Statin use is more common in patients with
breast WAT inflammation and metabolic syndrome. Clinically, statin use may be a surrogate identifier of a population that is at
increased baseline risk of BC. These findings may account for the variability in results of prior studies examining statin use and
breast cancer risk due to elevated risk in users compared to non-users.
Grade
Description
no TSM
scanty TSM
II
III
IV
Score (No(%))
0
II
p-value
III
IV
Histological subtype
0.0883
IDC
148
3 (2%)
64 (43%)
58 (39%)
20 (14%)
3 (2%)
ILC
31
0 (0%)
21 (68%)
7 (23%)
3 (10%)
0 (0%)
Mixed
0 (0%)
0 (0%)
7 (88%
1 (13%)
0 (0%)
Mitosis
0-10
0.0005
143
2 (1%)
76 (53%)
50 (35%)
15 (10%)
0 (0%)
10-20
19
1 (5%)
9 (47%)
7 (37%)
1 (5%)
1 (5%)
>20
25
0 (0%)
0 (0%)
15 (60%)
8 (32%)
2 (8%)
Grade
0.0226
45
0 (0%)
22 (49%)
19 (42%)
4 (9%)
0 (0%)
II
98
2 (2%)
53 (54%)
31 (32%)
12 (12%)
0 (0%)
III
44
1 (2%)
10 (23%)
22 (50%)
8 (18%)
3 (7%)
pT
0.2312
93
1 (1%)
49 (53%)
32 (34%)
10 (11%)
1 (1%)
71
1 (1%)
27 (38%)
30 (42%)
11 (15%)
2 (3%)
21
1 (5%)
9 (43%)
10 (48%)
1 (5%)
0 (0%)
0 (0%)
0 (0%)
0 (0%)
2 (100%)
0 (0%)
pN
0.1376
92
2 (2%)
44 (48%)
39 (42%)
6 (7%)
1 (1%)
95
1 (1%)
41 (43%)
33 (35%)
18 (19%)
2 (2%)
LVI
0.8007
No
162
3 (2%)
73 (45%)
64 (40%)
19 (12%)
3 (2%)
Yes
25
0 (0%)
12 (48%)
8 (32%)
5 (20%)
0 (0%)
ER
Negative
0 (0%)
0 (0%)
1 (25%)
3 (75%)
0 (0%)
Positive
179
3 (2%)
83 (46%)
70 (39%)
20 (11%)
3 (2%)
PR
0.4849
Negative
25
0 (0%)
10 (40%)
10 (40%)
5 (20%)
0 (0%)
Positive
156
3 (2%)
73 (47%)
60 (38%)
17 (11%)
3 (2%)
Her-2
0.0903
Negative
166
3 (2%)
79 (47%)
62 (37%)
20 (12%)
3 (2%)
Positive
0 (0%)
0 (0%)
6 (75%)
2 (25%)
0 (0%)
Focality
0.8170
Unifocal
1141
1 (1%)
66 (47%)
55 (39%)
16 (11%)
3 (2%)
Multifocal
46
2 (4%)
19 (41%)
17 (37%)
8 (17%)
0 (0%)
Age at diagnosis
0.8936
<55
60
1 (2%)
18 (31%)
35 (57%)
6 (10%)
0 (0%)
55-65
56
2 (4%)
32 (57%)
17 (30%)
4 (7%)
1 (2%)
>65
77
0 (0%)
34 (44%)
29 (38%)
14 (18%)
0 (0%)
For both CD 68 and CD163, the tumor introduces important clustering or correlation, whereas the host does not introduce any
additionale correlation. TAM recruitment is therefore primarily determined by tumor-related characteristics.
Department of Health.
DCIS (n=348)
Age at entry
59 (55, 63)
60 (56, 65)
BMI (kg/m2)
Adiponectin (ng/mL)
Leptin (pg/mL)
L/A ratio
Glucose (mg/dL)
89 (81, 97)
93 (86, 103)
Insulin (U/mL)
HOMA index
Future investigations in both trials will assess whether adiponectin is also associated with BC events.
Acknowledgments: J.Forbes and T.Howell, Co-Chairmen,IBIS-II Steering Committee.
BCI+TS
% Patients
10 year DR
% Patients
10 year DR
Low Risk
53.7%
5.3%
54.7%
5.2%
Intermediate Risk
29.3%
15.5%
29.2%
16.5%
High Risk
17.0%
28.0%
16.2%
27.3%
In cross stratification analysis between BCI and BCI+TS, no significant re-classification was observed (Table 2), however 14.5%
of BCI and 9.7% of BCI+TS intermediate risk patients were re-stratified as low risk.
Re-classification of BCI and BCI+TS for all patients
BCI
BCI+TS
Low
Intermediate
High
Total
Low
358
30
388 (54.7%)
Intermediate
20
163
23
206 (29.1%)
High
14
98
115 (16.2%)
Total
381 (53.7%)
207 (29.2%)
121 (17.1%)
709
Discussion: Integration of tumor size, but not grade, statistically enhanced the prognostic ability of BCI to predict 10 year DR risk
in patients with ER+, LN- early stage breast cancer. However, there was limited clinical impact on risk stratification, indicating that
the prognostic information provided by these clinicopathological factors is effectively captured by BCI alone.
Variables
n (%)
2cm
51 (31)
> 2cm
112 (67)
Negative
79 (47)
Positive
72 (43)
1,2
128 (77)
27 (16)
Low
129 (77)
High
38 (22)
Univariate
Multivariate
P value
P value
HR (95% CI)
Tumor size
1 (reference)
0.264
0.228
2.75 (0.57-23.5)
Node status
1 (reference)
0.003
0.002
5.25 (1.35-34.7)
Grade
1 (reference)
0.485
0.547
1.57 (0.32-6.04)
0.012
4.94 (1.44-19.4)
No recurrence(%)
Recurrence(%)
Univariate
HR
Mitosis
Multivariate
p
HR
0.025
p
0.007
14/10HPF
170(96.0)
7(4.0)
59/10HPF
33(84.6)
6(15.4)
4.259
0.009
4.297
0.014
10/10HPF
15(88.2)
2(11.8)
3.461
0.122
12.211
0.007
Size
0.035
0.029
2cm
75(98.7)
1(1.3)
136(89.5)
16(10.5)
8.599
0.037
5.630
0.010
>5cm
51(98.1)
1(1.9)
1.564
0.752
0.360
0.519
Operation
0.957
0.922
WLE
229(93.1)
17(6.9)
VABS
23(92.0)
2(8.0)
1.247
0.769
1.396
0.814
Mastectomy
13(92.9)
1(7.1)
1.055
0.959
1.472
0.744
Margin
0.846
Clear
200(93.5)
14(6.5)
Close/Involvement
39(92.9)
3(7.1)
1.132
0.773
1.253
In the risk stratification for LR, PTBs sized 2 - 5cm with 10 mitoses / 10 HPF had the highest LR rate (60%) compared with all
other groups (p < 0.001).
Conclusions Only PTB with 2 - 5 cm and frequent mitoses, is it recommended to follow to ascertain a wide excision and clear
margin of 1 cm, it necessary by means of a 2nd surgery could be considered to avoid the risk of LR in this distinct group.
58
59
207 (68.3)
39 (83)
PM
96 (31.7)
8 (17)
301
47
Mean SD
17.3 7.38
24.7 8.53
IDC
248 (82.4)
44 (93.6)
ILC
50 (16.6)
3 (6.4)
Age
Mean
M status
OncotypeDx score
Histology
Stage
T1a
2 (0.7)
2 (4.2)
T1b
49 (16.2)
9 (19.1)
T1c
174 (57.5)
25 (53.3)
T2
74 (24.5)
11 (23.4)
T3
2 (0.7)
Lumpectomy
198 (65.6)
30 (63.8)
Mastectomy
102 (33.8)
17 (36.1)
Yes
93 (30.4)
32 (68.1)
No
210 (69.6)
15 (31.9)
AI
167 (55.4)
30 (65.2)
Tam
89 (29.2)
9 (19.5)
36 (10.9)
4 (8.8)
Surgery
Chemotherapy
Endocrine tx
N= number, SD= standard deviation, M= menopausal, PM= premenopausal, IDC= invasive ductal carcinoma, ILC= invasive
lobular carcinoma, AI= aromatase inhibitor, Tam= tamoxifen, = statistically significant
ER+/PR+ N(%)
ER+/PR- N(%)
P value
ER+/PR+
226 (91.9)
12 (31.6)
< 0.001
ER+/PR-
20 (8.1)
24 (63.2)
ER-/PR-
2 (5.3)
ODX 25
ODX RS>25
Total
ERS < 21
120
124
ERS(21-30)
11
19
ERS(> 30)
Total
132
17
149
We also presented the comparison of ERS and observed RS based on original risk categories.
Table2 (Original risk categories )
Toranomon cohort
ODX RS<18
ODX RS 18-30
ODX RS>30
Total
ERS(<18)
66
33
99
ERS(18-30)
17
25
44
ERS(>30)
Total
83
59
149
Conclusions: We developed an ERS model to find those patients most likely to benefit from ODX RS results in clinical practice
setting. Although further external and prospective validation is mandatory, preliminary result supports the usefulness of this ERS
model.
Stage
-0.378
<0.0001
217
HbA1C
0.004
0.9731
69
The negative correlation means that higher levels of serum albumin are associated with lower stages. Serum albumin at
diagnosis had a significant effect on the probability of survival (p=0.0005). For every unit increase in serum albumin, the odds of
survival increase (OR=4.938, 95% CI: 2.023, 12.050). Alternatively, for each unit decrease the odds of dying increase by about
five fold. Hypoalbuminemia has a significant effect on survival (p=0.0020). When serum albumin is >3.5, the odds of death
decrease (OR=0.176 95% CI: 0.058, 0.528). Neither HbA1c nor age had a significant effect on survival (P=0.7635, 0.0858
respectively).
Odds Ratios for probability of survival
95% CI
OR
Lower Limit
Upper Limit
p-value
Serum Albumin
4.938
2.023
12.050
0.0005
HbA1C
1.092
0.614
1.942
0.7635
Age
0.974
0.946
1.004
0.0858
Conclusion: In the present study, a low serum albumin level at the time of diagnosis, either as a continuous or categoric variable,
was significantly associated with poorer survival in patients with breast cancer. Furthermore, a lower serum albumin is associated
with a higher stage of breast cancer. In contrast, glycated hemoglobin (HbA1C) at the time of diagnosis did not have a significant
effect on survival.
Intrinsic
Subtype
DFS (% at 5yr)(pCR
pCR(Tumour)(%) pCR(Node)(%) tumor vs pPR
tumor)(pvalue)
DFS (% at 5yr)(pCR
node vs pPR
node)(pvalue)
OS (% at
OS (% at 5yr)(pCR
5yr)(pCR Node vs
Tumour vs pPR
pPR node)(p
tumor)(p value)
value)
Luminal A 26
26
100 vs 67 (0.08)
88 vs 70 (0.49)
100 vs 72 (0.1)
85 vs 75 (0.7)
Luminal B 23
38
68 vs 56 (0.9)
87 vs 32 (0.11)
80 vs 57 (0.9)
85 vs 35 (0.35)
Her-2
Type
39
41.6
90 vs 44 (0.003)
83 vs 45 (0.009)
87 vs 59 (0.02)
90 vs 66 (0.03)
Basal
31.5
59
85 vs 50 (0.004)
46 vs 50 (0.26)
83 vs 50 (0.02)
975 vs 60 (0.19)
Multivariable*
HR
P-value
95% CI for HR
HR
P-value
95% CI for HR
TM
- (ref.)
- (ref.)
- (ref.)
BCS
0.443
0.0318
0.211-0.932
No
- (ref.)
- (ref.)
- (ref.)
Yes
4.783
<0.0001
2.163-10.578
Positive
2.726
0.3269
0.367-20.244
Negative
- (ref.)
- (ref.)
- (ref.)
1&2
- (ref.)
- (ref.)
- (ref.)
3.793
0.0004
1.806-7.969
- (ref.)
- (ref.)
- (ref.)
- (ref.)
- (ref.)
- (ref.)
2.283
0.1697
0.703-7.416
1.784
0.3380
0.546-5.833
12.967
<0.0001
4.303-39.083
9.912
<0.0001
3.265-30.096
Positive
4.294
0.0032
1.631-11.303
2.587
0.0603
0.960-6.971
Negative
- (ref.)
- (ref.)
- (ref.)
- (ref.)
- (ref.)
- (ref.)
Positive
6.107
<0.0001
2.905-12.837
4.494
<0.0001
2.105-9.594
Breast surgery
Presence of LVI
RM
NG
AJCC Stage
Ki-67
p53
Negative
- (ref.)
- (ref.)
- (ref.)
- (ref.)
- (ref.)
- (ref.)
BCS, breast-conserving surgery; LVI, lymphovascular invasion; NG, nuclear grade; RM, resection margin; TM, total mastectomy;
NG, LVI and total mastectomy were not significant in multivariable Cox analysis;* Cox-proportional hazard regression model.
In this study, we have shown that overexpression of p53 and Ki-67 could be used to discriminate low-risk luminal A subtype in
breast cancer. Therefore, using the combination of p53 and Ki-67 expression in discriminating low-risk luminal A breast cancer
may improve the prognostic power and provide the greatest clinical utility.
Gene
% of Total Mutations
BRCA1
21.2%
BRCA2
19.0%
CHEK2
11.3%
ATM
9.7%
PALB2
7.2%
NBN
6.6%
APC
5.6%
BARD1
3.3%
PMS2
2.6%
BRIP1
2.3%
MSH6
2.0%
TP53
1.5%
MSH2
1.3%
RAD51C
1.3%
CDH1
1.0%
RAD51D
1.0%
CDKN2A
1.0%
PTEN
1.0%
MLH1
0.5%
Biallelic MUTYH
0.3%
SMAD4
0.3%
Conclusions: Testing patients using a 25-gene panel identified 234 mutations outside of BRCA 1 and BRCA 2 (157). That is a
149% increase in mutations identified over BRCA 1 and BRCA 2 testing alone. Mutations in moderately penetrant breast cancer
genes (including CHEK2, ATM, PALB2 and NBN) comprised 34.8% of total mutations and 6.4% of mutations were in LS genes.
Additionally, panel testing identified mutations in more than one gene in 4.0% of patients, which would not have been identified by
single-syndrome testing. Panel testing provides a broader understanding of hereditary cancer in breast cancer patients both by
identifying mutations in more genes and identifying patients with mutation in more than one gene. This approach can provide
more guidance both for management of the patient and the patient s family members.
General Anxiety
General Depression
Event Anxiety
Cancer Worry
Knowledge (K) Total
K-Inheritance
K-Benefits
K-Limitations
Baseline
Post V1
Post V2
Mean (SD)
Mean (SD)
Mean (SD)
6.8 (3.9)*
6.1 (4.0)*
7.2 (3.5)*
6.1 (3.6)*
2.6 (3.0)
2.3 (2.6)
2.6 (2.9)
2.3 (2.6)
37.1 (9.6)
37.7 (9.5)
37.0 (8.9)
37.3 (8.5)
18.3 (15.7)
16.9 (14.1)
18.4 (15.4)
15.7 (14.1)
61.8 (6.1)**
63.9 (6.4)**
62.1 (6.7)**
64.1 (6.8)**
29.5 (3.2)
30.0 (3.2)
29.7 (3.5)
29.8 (3.3)
12.0 (1.4)
12.3 (1.8)
12.0 (1.4)
12.4 (1.7)
20.3 (3.2)**
21.6 (2.8)**
20.4 (3.4)**
21.9 (3.0)**
5.8 (4.2)*
2.9 (3.6)
37.3 (9.4)
6.6 (14.7)
66.3 (6.9)**
30.3 (3.7)
12.4 (1.9)
23.6 (2.6)**
Satisfaction
42.8 (3.8)
42.9 (3.6)*
Uncertainty
Perceived Utility
7.5 (4.3)
6.9 (4.6)
7.7 (4.0)
6.5 (4.5)
41.4 (2.6)*
6.7 (4.6)
37.2 (7.9)
37.7 (7.0)*
33.8 (8.6)*
*p,0.05, **p<0.001
Conclusion: With a tiered-binned counseling model, patients experience increased knowledge. Uptake of panel testing varies by
prior testing and potentially by patient affective factors. Most patients do not experience negative psychological responses,
although this may vary by test result. Declines in satisfaction and perceived utility may also vary by test result and may reflect the
current unclear utility and uncertainty of multiplex testing.
2010
p-value
0.006
0.276
Uninsured
11.8
22.8
Medicaid
59.5
Medicare
28.7
7.0
Private
15.7
18.8
Other
35.4
Education
0.031
0.463
< Grade 12
8.7
7.9
High School/GED
17.2
Some college
19.0
Associates
34.5
19.5
Bachelors
34.2
5.6
Masters/Prof/Doctorate
38.3
42.5
Region
p-value
0.077
0.122
Northeast
35.3
30.3
Midwest
17.0
6.1
South
3.2
29.1
West
5.8
10.7
Age
0.449
0.463
<55
26.2
14.0
>55
12.2
21.1
Conclusion: More people undergoing genetic testing for breast cancer risk in 2010 were concerned that this would adversely
affect their health insurance than in 2005, and there was less variation in this perception based on insurance, education and
region than in the earlier period. We conclude that, despite the passage of significant U.S. legislation including GINA and PPACA,
nearly 1 in 5 people undergoing genetic testing remained concerned about potential risk to their health insurance coverage.
Accurate and early identification of those people carrying a cancer genetic mutation is crucial to cancer prevention and treatment
efforts; our results suggest a dire need for improved education regarding federal protections for genetic testing so that any
barriers to seeking genetic counseling are eliminated.
Offered testing
Actual testing
Gr 1
72
27 (37.5%)
15 (55.5%)
5 (33.3%)
Gr 2
53
22 (41.5%)
16 (72.3%)
8 (50%)
The most common reason for genetic testing was patients with primary breast cancer age < 45 years. Patients who met criteria
but were not offered testing were those with age < 45 years and triple negative breast cancer. 5 patients in Gr 1 were seen by the
genetic counsellor post treatment and all 5 were tested.
Conclusion A greater number of women with newly diagnosed breast cancer were identified and offered genetic testing with an
onsite genetic counsellor. The precentage of individuals tested increased with an onsite counsellor.
Caucasians
African Americans
33 (21-60)
39.5 (26-74)
39.0 (38.0-40.0)
43.9 (41.3-46.5)
Black
Hispanic
Asian/Pacific Islander
American Indian
OR (95% CI)
OR (95% CI)
OR (95% CI)
OR (95% CI)
OR (95% CI)
Stage 1
1.00
1.20 (1.12,1.29)
Stage 2
1.00
Stage 3
1.00
Stage 4
1.00
Confidence intervals that include 1.00 indicate that the odds of IDC were no different than the reference category
CONCLUSION
The incidence of IDC is not the same for all race/ethnicities and depends on the stage of disease. APIs are at an increased risk
for IDC in all except for the highest stage of disease.
52 (28-77)
64% (50-82%)
105 (48%)
48 (22%)
80 (37%), 242mg/m2
217 (100%)
TRC occurred in 33 patients (15.2%). In multivariate models, lower baseline LVEF (OR 3.23 for a 5% lower LVEF, [95% CI
1.96-5.33], p<0.0001) and greater absolute decline in LVEF from T1 to T2 (OR 3.25 for each 5% drop in LVEF, [95%CI
1.83-5.79], p<0.0001] were each independent predictors of TRC. NT pro-BNP increased from T1 to T2 (median =1.31pmol/L,
p<0.0001) and then fell to baseline levels. Troponin I increased from T1 to T2 in 64.5% of patients (p<0.0001) and remained
above baseline 6 months post-A (p<0.0001). NT pro-BNP and troponin I at T1 were not predictive for TRC. There were trends for
the absolute increase in NT-pro BNP and troponin I from T1 (pre-A) to T2 (post-A) to be associated with risk of TRC (p=0.08 and
0.09) in univariate analyses. Germline SNPs were not predictive of TRC.
Conclusion: This is the largest prospective study analyzing predictors for TRC, and the first studying immune-related SNPs in this
context. Consistent with the literature, lower LVEF at baseline increases risk of TRC. Previously unreported, the absolute
decrease in LVEF after A is also predictive for TRC. The persistent elevation of troponin I 6 months after completing A
demonstrates chronic cardiac stress during therapy. Ongoing modeling analyses (to be presented at the meeting) will determine
whether longitudinal changes in serum biomarkers can predict subsequent development of TRC.
Ratio
HER2 copy#
CEP17 copy#
HER2 IHC
ER/PR
yAJCC
RCB class
Patient 1 (AC+TH)
2.2
3.9
1.8
1+
+/+
ypT0 N0(i+)
Patient 2 (TCH)
2.2
3.8
1.7
1+
+/-
ypT0 N0
Patient 3 (TCH)
2.2
3.8
1.7
2+
+/+
ypTis N0
Patient 4 (AC+TH)
2.3
3.0
1.3
+/+
ypT2 N2a
Patient 5 (TCH)
2.2
3.7
1.7
1+
-/-
ypT1c N0
Patient 6 (TCH)
2.2
3.9
1.8
2+
-/-
ypTis N0
Patient 7 (TC)
2.0
3.4
1.7
-/-
ypT0 N0
Patient 8 (AC+T)
2.2
3.6
1.7
1+
+/-
ypT1c N0
Patient 9 (TC)
2.1
3.9
1.9
1+
-/-
ypT0 N0
Patient 10 (AC)
2.5
3.4
1.4
1+
+/+
ypT1b N1a
Patient 11 (TAC)
2.4
3.6
1.5
2+
-/-
ypT3 N1mi
Patient 12 (AC)
2.0
3.3
1.6
1+
-/-
ypT1c N1a
demonstrated a good response to neoadjuvant trastuzumab-based chemotherapy and further support the recent modifications of
ASCO/CAP guidelines to determine HER2 amplification.
22/10 trial.
Participation Status
Number of Centers
Closed
17
Not Participating
28
643
No response/Unknown
21
850
Yes, participating
96
5333
Activated
67
4215
Not Activated
29
1118
Totals
148
6843
Because the original BIG 1-98 informed consent indicated life-long follow-up, only three countries required patient re-consent in
order to participate. At least one LTFU data submission has occurred for 73% of patients participating in the LTFU (May 2014).
Conclusion
Long-term follow-up for a large-scale clinical trial is feasible, but challenging. The methods used for BIG 1-98 LTFU will be
described and the status will be updated at the meeting.
Hb
H2O
-31.0%
-22.3%
-5.5%
-4.4%
-2.3%
-0.3%
P value
0.02
0.01
0.015
Conclusions: Two-week DOT change is an early predictor of response to NACT as measured by the RCB score. We found
significant associations between the RCB index with 2-week changes in HbO, Hb, and H2O. Significantly different changes in
DOT parameters were associated with the other RCB classifications. Ki-67 changes and baseline MVD were not statistically
significantly associated with DOT parameters. We are analyzing static and dynamic DOT data on the remaining pts. Additional pts
are being recruited to evaluate DOTs predictive ability by tumor subtype.
s'(/cm)
cancer
normal breast
cancer
normal breast
760nm
0.078
0.063
9.58
9.71
800nm
0.071
0.05
9.23
9.4
830nm
0.084
0.063
9.07
9.22
There was no difference in reduced scattering coefficient (s) between breast cancer tissue and contra-lateral normal
breast(760nm: cancer;9.58, normal;9.71, 800nm: cancer:9.23, normal;9.40. 830nm; cancer;9.07, normal;9.22). The tHb of breast
cancer tissue was significantly high, compared with normal breast (cancer:32.314.6, normal breast;22.08.6, p=0.001). There
was no difference in oxygen saturation (SO2) between breast cancer tissue and contra-lateral normal breast (cancer:73.24.3,
normal breast;73.65.9, p=0.31).
The tHb and SO2 between breast cancer tissue and contra-lateral normal breast
cancer
normal breast
p-value
tHb(M)
32.314.6
22.08.6
p<0.001
SO2(%)
73.24.3
73.65.9
p=0.32
Conclusion: Absorption coefficient (a) and tHb increased in breast cancer, whereas reduced scattering coefficient (s) and
oxygen saturation did not.
<14%
14%
Discordance rate %
Kappa
Sign.
<14%
56
13.1
0.723
<0.001
14%
13
30
Table 2. Discordance between whole slide and core biopsies 1-3 and 4-6 for Ki67 result
Whole slide
<14%
Core 1-3
Core 4-6
Discordance rate %
14%
<14%
33
14%
30
<14%
31
14%
30
Kappa
Sign.
20.3
0.595
<0.001
20.8
0.584
<0.001
Conclusion
Tumor heterogeneity results in substantial variation of Ki67 scores between TMA cores and whole slides that may result in
considerable differences in distinguishing luminal A from luminal B ER-positive, HER2-negative tumors. This may have far
reaching consequences for the choice for (neo)adjuvant treatment.
Mean % (SD)
Tumor ER Status
P Value
0.05
Negative
14
27.4 (13.9)
Positive
60
19.3 (13.6)
0.34
Negative
57
21.8 (14.9)
Positive
17.7 (5.7)
0.05
No
53
19.7 (13.6)
Yes
12
28.5 (14.7)
HER2 status was not known in some cases and was excluded from statistical calculations.
Conclusion: This computer assisted image analysis study confirms ER expression in NBR increases with age and menopausal
status in women with BC. We report, for the first time, a significant association between ER expression in normal breast
epithelium with ER negative and triple negative cancers in post-menopausal women. Our study suggests that ER expression in
normal epithelium may play a role in development of hormone receptor negative breast cancers.
2+ Cases (All)
57/707 (8.1%)
37/507 (7.3%)
-------Strong
-------1/8 (12.5%)
-------Moderate
-------16/136 (11.8%)
-------Weak
-------20/363 (5.5%)
Conclusion: 2013 guidelines will identify a higher % (1.1% for FISH) of eligible patients for targeted therapy. In this series, 2+ IHC
cases with reflex FISH testing also had higher Amp rates by 2013 guidelines (8.1% vs 6.9%). Evaluating these rates over time will
help assess the effect of the change in the scoring criteria. Amp rates for Eq (2+) IHC cases with 10-30% strong and moderate
membrane staining are similar, although only the former group is classified as positive (3+) following new guidelines and would
not be retested by FISH. Cases with less than 30% membrane staining may represent a biological spectrum that is not well
represented by current IHC testing guidelines, and FISH confirmation on these cases may be justified.
Type II
Cases
Errors
Errors
SPC Rate
Practice A
2,205
0.54%
Practice B
617
0.00%
Practice C
173
0.00%
Practice D
162
Practice E
142
Practice F
128
Practice G
118
Total
3,545
1
2
0.62%
1.41%
0.78%
0.00%
10
0.45%
Fig 2
Type I
Type II
Cases
Errors
Errors
SPC Rate
Lab A
1,435
0.42%
Lab B
967
0.31%
Lab C
430
Lab D
260
Lab E
182
Lab F
84
Lab G
78
Lab H
71
Lab I
20
Lab J
Lab K
0.00%
Lab L
0.00%
Lab M
0.00%
Lab N
0.00%
Total
3,545
0.00%
2
0.77%
0.00%
1
1.19%
0.00%
2.82%
0.00%
2
10
22.22%
0.45%
45
(29-88)
Apocrine
28
7.1
93
23.7
Central scar/necrotizing
34
8.7
Clear Cell
29
7.4
Infiltrating ductal ca
71
18.1
14
3.6
43
11.0
Metaplastic ca
24
6.1
Trabecular ca
33
8.4
other histologies
23
5.9
Histology
Grade
1
0.8
31
7.9
358
91.3
0.1-2 cm
196
50
> 2.0 cm
195
49.6
not stated
0.3
251
64
1-3
82
20.9
4-9
33
8.4
10+
22
5.6
not evaluated
1.0
0-15
77
19.6
15.1-30
65
16.6
>30%
247
63.3
not done
0.5
Yes
102
26.0
No
238
60.7
unknown
52
13.3
Tumor Size
Nodal status
Ki-67
Adjuvant Chemotherapy
Screening MRI
Screening
ultrasound
33% (556/1669)
Self-detected
mass
Physician physical
exam
p value
4% (33/786)
4% (5/112)
<0.001
IN SITU
% DCIS
INVASIVE
Molecular type
<0.001
85% (45/53)
85% (33/39)
61% (393/641)
75% (63/84)
ER/PR+,
Her2+
7% (63/907)
2% (1/53)
5% (2/39)
11% (67/641)
7% (6/84)
4% (2/53)
6% (40/641)
6% (5/84)
9% (5/53)
10% (4/39)
22% (141/641)
12% (10/84)
Grade
<0.001
28% (280/983)
45% (25/56)
42% (15/36)
12% (73/633)
20% (17/87)
53% (517/983)
46% (26/56)
42% (15/36)
49% (313/633)
49% (43/87)
19% (186/983)
9% (5/56)
16% (6/36)
39% (247/633)
31% (27/87)
Tumor size
<0.001
T1
78% (874/1113)
81% (53/65)
77% (31/40)
38% (284/753)
49% (52/107)
T2
18% (205/1113)
14% (9/65)
20% (8/40)
47% (354/753)
41% (44/107)
T3
3% (30/1113)
5% (3/65)
3% (1/40)
12% (89/753)
6% (6/107)
T4
1% (4/1113)
3% (26/753)
5% (5/107)
Within Stroma
Sample
CD8 density
CD4 density
PDL1 H-score
CD8 density
CD4 density
PDL1 H-score
50
63
1301
207
34
30
591
892
38
122
60
2211
1331
10
305
3833
106
34
808
131
75
10
1189
1591
110
177
546
21
585
1143
17
3918
68
121
859
161
74
4834
2570
196
For patients not achieving a pathologic complete response (pCR), the density of both the CD8 (p=.03) and CD4 (p=.05) infiltrates
in the stroma were significantly greater than in the tumor. For patients achieving a pCR, there was no significant difference in the
densities of stromal and intratumoral CD8 (p=.11) or CD4 (p=.75) infiltrates suggesting that T cell infiltration into the tumor from
the stroma is critical.
Conclusion: Multispectral imaging allows different immune cell phenotypes to be visualized and quantified simultaneously in the
same tissue section enabling further study of the relationships and distribution of these cells within the tumor and tumor
microenvironment, and their spatial distribution and proximity to the tumor cells. This technology will enable improved
understanding of the immune infiltrate in breast tumors thereby facilitating the rational design and use of immunotherapeutic
tumor size
Nodal status
Grade
ER atatus
PR status
HER2 status
rs10030044
Variables
n (%)
2cm
210 (36)
>2cm
376 (64)
Negative
369 (63)
Positive
216 (37)
1,2
426 (72)
123 (21)
Positive
440 (75)
Negative
145 (25)
Positive
377 (64)
Negative
208 (35)
Positive
55 (9)
Negative
324 (55)
TT+GT
479 (81)
GG
109 (19)
Univariate (P value)
Multivariate (P value)
<.0001
<.0001
2.268 (1.615-3.181)
1 (reference)
<.0001
<.0001
3.117 (1.984-4.954)
1 (reference)
0.0001
0.970
0.994 (0.731-1.356)
1 (reference)
<.0001
0.069
1.666 (0.962-2.893)
1 (reference)
0.0005
0.224
1.399 (0.812-2.380)
1 (reference)
<.0001
0.0008
0.420 (0.261-0.690)
1 (reference)
0.0282
0.0082
1,860 (1.181-2.853)
The SNP, ZNF423 rs8060157, was not associated with prognosis in this study.
Conclusion: We show that the genotype GG at CTSO rs10030044 is an independent factor indicating poor prognosis in
ER-positive breast cancer patients receiving adjuvant endocrine therapy.
MDA-MB-231
231DasB
p value
0.40 0.04
0.87 0.07
0.003
>10
2.1 0.1
0.0001
1.9 0.1
1.2 0.1
0.003
138.6 1.0
97.2 6.6
0.007
13.2 0.8
11.5 1.3
0.147
23.1 2.2
20.9 2.1
0.289
94.5 2.0
95.9 7.6
0.834
No significant change in sensitivity to carboplatin or to the EGFR inhibitors gefitinib and neratinib was observed. However, the
231-DasB cells demonstrated a significant increase in sensitivity to the c-Met inhibitor, CpDA, with an IC50 value of 2.1 0.1 M
compared to an IC50 greater than 10 M in the parental MDA-MB-231 cells. Treatment of MDA-MB-231 cells with dasatinib (100
nM) blocked phosphorylation of Src kinase. In contrast, dasatinib treatment (100 nM) did not decrease p-Src levels in the
231-DasB cells. p-Met levels were significantly increased in 231-DasB cells relative to MDA-MB-231. Treatment with 2 M CpdA
decreased p-Met and p-Src in both 231-DasB and MDA-MB-231 cells. Other key receptor tyrosine kinases (EGFR, HER2, HER3,
HER4, IGFIR and IR) show no significant changes in phosphorylation in 231-DasB cells compared to MDA-MB-231.
Constitutive activation of p-Src through increased c-Met signalling may be a potential mechanism of resistance and suggests that
combined treatment with dasatinib and a c-Met inhibitor may block the development of acquired resistance.
IHC4
RMH
Dako
Dako
0.92
Leica
0.89
0.95
Ventana
0.93
0.96
%DRprob
Leica
RMH
Dako
Leica
0.97
0.95
0.98
0.98
0.98
0.98
0.98
Mean
Median difference
RMH
Dako
RMH
9.9%
Dako
8.4%
1.0%
Leica
8.9%
0.9%
0.5%
Ventana
9.1%
0.6%
0.5%
Leica
0.6%
Table 2. Showing for each center, the mean %DRprob score and median value obtained when the absolute difference was
calculated between matched scores
Conclusion
The IHC4+C algorithm is tolerant of variation in staining and ER-scoring method used. Although additional comparative studies
are required to confirm them, these data support the use of IHC4+C in routine clinical practice outside its institute of origin.
Sample DCIS
ID
content
Invasive
Carcinoma
content
Difference in
Macrodissected ESR1 mRNA
[DDCT]
Difference in
Difference in PGR
HER2 mRNA
mRNA [DDCT]
[DDCT]
Difference in KI67
mRNA [DDCT]
10%
30%
>80%
0,65
0,26
0,65
0,55
70%
10%
>80%
0,43
0,35
0,38
-0,19
40%
10%
>80%
-0,44
-0,54
-0,17
0,25
30%
5%
>80%
0,46
-0,98
0,3
0,59
10%
50%
>80%
1,32
0,95
0,7
1,16
20%
10%
>80%
0,16
0,32
-0,14
0,58
25%
10%
>80%
1,69
1,32
1,26
-0,56
70%
15%
>80%
0,16
-0,42
-0,07
-0,31
40%
10%
>80%
0,46
-0,13
0,47
1,09
10
40%
30%
>80%
0,52
0,27
0,28
0,28
Conclusion
The performance of the MammaTyper diagnostic assay does not appear to be affected by fluctuations in the TCC of the original
FFPE specimens under the presence of increased amounts of DCIS. Similar findings have been previously reported in a research
setting (Kotoula, Virchows Arch 2013). Effective RNA extraction and optimal PCR output normalization provide sufficient
robustness for tolerating up to 8-fold TCC specimen changes, independent of the presence of DCIS. Our analysis suggests that
extra time spent on macro-dissection of specimens for routine RT-qPCR assays could be avoided with safety when using the
MammaTyper RT-qPCR kit.
88%
90%
38%
16%
90%
92%
6%
45%
52%
93%
77%
100%
25%
71%
In general, highly effective agents to preventing cancers (e.g., tamoxifen) also prevented normal mammary gland proliferation
after only two weeks of treatment, while inactive agents (e.g., naproxen) had minimal effects on normal gland proliferation. The
effects of the agents on established mammary cancers (in which the agents were given for 7 days to rats bearing small
MNU-induced mammary cancers) showed similar correlations. Additional biomarkers, as well as the proliferation change and
efficacy of these agents in rats fed a high-fat (Western) diet, will also be presented. In conclusion, determining the effect of a
potential chemopreventive agent on cell proliferation following short-term treatment appears to be an effective method for
predicting its efficacy in preventing mammary cancers. These data further confirm that Ki67 measurements are useful in Phase II
prevention trials as a biomarker of agent efficacy.
Q2
Q3
Q4
Pts Screened
1025
1434
5251
7344
196
290
1166
1569
51
75
444
355
15
15
17
80
10
Conclusion: The program provides a comprehensive breast cancer identification and reduction option to a large number of
women. It also provides the primary provider a service and resources regarding breast cancer risk identification and prevention.
RW
%change
Other radiologists
%change
Cumulus
%change
% Dense volume
change^
Quantra
%change
Volpara
%change
40+
40+
15
30+
30+
14
10
20+
12
13
20+
20
20
10+
35
30
10+
17
26
0+
49
81
43
0+
16
19
0-
18
0-
16
15
Number
105
105
98
98
99
_____
RW = Ruth Warren. CUMULUS was measured by JS, a trained operator of the software. * Percentage point change eg if density
changed from 40% to 30% this was regarded as a 10% change.^ Reduction in dense volume expressed as a percentage of
baseline dense volume. We used Quantra version 2.0 and Volpara version 1.4.5
Conclusions Whilst RW remained consistent with previous density estimation it was not possible to use a pool of other expert
readers to predict change. Cumulus gave similar results to RW but is difficult to use in practice. There was good agreement
between the two objective volumetric measures used (r=0.5) and since these are automated they may be suitable for clinical
practice. However their relationship to the long term breast cancer preventative effect of tamoxifen needs to be established.
Reference 1 Tamoxifen-induced reduction in mammographic density and breast cancer risk reduction: a nested case-control
study. Cuzick J, Warwick J, Pinney E, Duffy JW, Cawthorn S, Howell A, Forbes JF, Warren RM. J Natl Cancer Inst. 2011 May 4;
103 (9): 744-52.
Premalignant subgroup
Metformin arm
Placebo arm
p-value*
LCIS (n=9)
15 (5-15)
5 (4-6)
0.1
DH (n=69)
3 (1-4)
3 (1-4)
0.5
12 (8-20)
10 (7-24)
0.9
10 (7-16)
10 (6-17)
0.9
33 (25-55)
40 (32-40)
0.2
HER2+ve (n=22)
22 (11-32)
35 (30-40)
0.06
HER2-ve (n=58)
16 (10-20)
17 (8-26)
0.7
ER+ve/HER2+ve (n=15)
12 (7-18)
32 (27-42)
0.004
ER+ve/HER2-ve (n=53)
16 (10-20)
15 (8-22)
0.8
PR+ve/HER2+ve (n=12)
18 (12-18)
32 (24-44)
0.02
PR+ve/HER2-ve (n=48)
16 (10-20)
12 (8-20)
0.6
p-interaction
0.2
0.04
0.001
0.05
*Wilcoxon rank-sum test; p-interaction between treatment and DCIS grade or HER2 status from a linear regression model,
adjusted for age and BMI.
The effect of metformin on DCIS was different by HER2 status (p-interaction=.04) and, among this molecular subtype, by ER and
PR status (p-interaction=.001 and .02, respectively). In HER2+ve DCIS, metformin decreased Ki67 by 40% overall (p=.06), by
over 60% in ER+ve/HER2+ve DCIS (p=.004). and by 45% in PR+ve/HER2+ve DCIS (p=.02) There was no effect of metformin in
HER2-ve DCIS, regardless of ER or PR status. Metformin did not affect Ki67 in DH overall, but showed a trend towards a
decrease in women with abdominal adiposity (p-interaction=.05).
Conclusions: Window of opportunity pre-surgical models provide insight into a drugs preventive potential by targeting
intraepithelial proliferations adjacent to invasive cancer. The model shows a high prevalence of preinvasive lesions (field
cancerization) in apparently normal breast tissue adjacent to breast cancer. Metformin selectively decreased Ki67 in HER2+ve
DCIS, particularly in the ER+ve subgroup, in line with a selective inhibitory effect on the HER2 pathway observed both in in vitro
and in vivo preclinical models. Our results provide the background for a phase III trial of metformin in HER2+ve DCIS
(ISRCTN16493703, Supported by AIRC, LILT and Health Ministry 2009-RF-153222).
56 (range 38-81)
58 (range 38-71)
54 (range 39-81)
7 (range 1-25)
6 (range 2-25)
8 (range 1-21)
Self-detected abnormality
23 (92%)
12 (92%)
11 (92%)
Screening mammogram
1 (4%)
2 (16%)
Not known
1 (4%)
1 (8%)
17 (68%)
10 (77%)
7 (58%)
-Sub-areolar
7 (41%)
6 (40%)
1 (8%)
-UOQ/axillary tail
2 (12%)
2 (16%)
-Lower/inframmamary crease
2 (12%)
1 (10%)
1(8%)
6 (35%)
3 (30%)
3 (25%)
1 (4%
1 (8%)
4 (16%)
1 (8%)
3 (25%)
3 (12%)
2 (15%)
1 (8%)
Presentation
No of OR (95% CI) I2
P for
studies
statistics heterogeneity
(%)
Nonluminal (ER,
PR)
Cohort
0.84 (0.72
0.97)
50
0.06
Cohort, adjusted*
0.88 (0.74
1.06)
42
0.18
Casecontrol
13
0.76 (0.67
0.86)
59
0.004
Cohort
0.74 (0.62
0.88)
0.46
Cohort, adjusted*
0.81 (0.62
1.04)
Casecontrol
0.73 (0.64
0.84)
12
0.34
Triplenegative
Luminal (ER+
and/or PR+)
Cohort
0.98 (0.89
1.07)
75
<0.001
Cohort, adjusted*
1.04 (0.98
1.10)
0.75
Casecontrol
18
0.82 (0.76 69
<0.001
0.89)
, negative; +, positive; ER, estrogen receptor; PR, progesterone receptor
I2 statistics show % of total variation across studies that is due to heterogeneity rather than random variation (chance).
Arbitrarily,I2 percentages may be interpreted as follows: 25%, low; 50%, moderate; and 75, high heterogeneity.
*Results adjusted at least for age, body mass index, parity, and family history of breast cancer.
Conclusion:
Breastfeeding is a powerful strategy to reduce the risk of several aggressive breast cancer subtypes, with a relative risk reduction
of approximately 10% to 20%, depending on receptor status. To maximize breastfeeding use for the longterm health of mothers
and babies, it is important to remove barriers in the home, community, and workplace as well as provide targeted education and
support before and after delivery.
NEPA + DEX
Cycle 1
74.3% (N = 724)
66.6% (N = 725)
Cycle 2
68.5% (N = 485)
57.1% (N = 434)
Cycle 3
65.7% (N = 423)
52.7% (N = 348)
Cycle 4
63.6% (N = 375)
50.6% (N = 300)
Conclusions: This multiple cycle analysis indicates that NEPA, a novel, fixed-dose antiemetic combination, more effectively
demonstrates sustained control of CINV over multiple cycles than oral PALO. As females with breast cancer represent a
particularly challenging population in terms of emesis control, it is especially crucial that antiemetic recommendations are followed
to allow these patients to maintain their quality of life and continue their treatment plan over multiple cycles of chemotherapy.
NEPA offers effective guideline-recommended prophylaxis in a convenient single dose.
P-value
35.24.5
37.05.1
0.03
34.84.5
34.94.7
0.88
BMI (kg/m2)
22.83.7
23.13.9
0.56
34
48
0.02
1.91.9
0.62
<2 67
66
0.59
2-5 31
30
>5 2
31
37
0.44
1-2 40
39
1.00
3 57
59
82
77
0.27
35
33
0.78
89
87
1.00
5.02.1
6.93.6
<0.001
Histologic grade
The median follow-up after diagnosis was 4.9 years in COS and 6.2 years in the control group. In the COS group, the hazard ratio
(HR) for recurrence after IVF was 0.77 (95% CI: 0.28, 2.13) and the survival was not compromised compared with controls
(P=0.61). In the COS group, survival was not different between patients with ER-positive and ER-negative breast cancer (P=0.75)
and between patients who underwent COS before and after tumor resection (P=0.56). The survival was also not different
between patients who pursued COS before and after chemotherapy (P=0.57).
Conclusion(s): Here we presented the largest prospective data with longest follow up on the safety of ovarian stimulation in
women with breast cancer. COS with letrozole and gonadotropins for FP is unlikely to cause substantially increased recurrence
risk in breast cancer, even in patients who have not yet undergone breast surgery. Larger studies are needed to confirm the
findings from the subgroup analysis.
Support: Supported by NIH RO1 HD053112.
BPI
mean
score
Baseline
score
3.75
2.57*
2.59
2.30
2.11*
1.85*
Three
month
score
4.25
4.52
6.08*
4.68
4.13
3.87
Six month
score
4.50
4.83*
5.32*
6.37*
5.07
4.22
Nine month
4.00
score
5.10
5.11
5.13
5.75*
5.04
Twelve
month
score
5.37*
5.48
4.86
5.66
6.21*
4.25
Start LLLT
End LLLT
N improved
3.89
2.16*
66 (71%)
Mean OM score
6.60
2.78*
75 (80.6%)
5.14
1.64*
20 (90.9%)
11 (11.8%)
60 (64.5%)*
60 (64.5%)
Pain scores were available for 22 (of the 93) patients. * p < 0.0001 (t-test or chi-square, as appropriate).
Conclusion
This retrospective analysis showed that LLLT, a standard management strategy for OM in head and neck cancer, significantly
reduced the severity of chemotherapy-induced oral mucositis and relieved pain in patients with breast cancer. This is the first
study in this population. Further research, preferably high-quality randomized controlled trials, is warranted to better investigate its
usefulness in this population.
Follow-Up (n=13)
MeanSD
QoL*+
112.8817.74
103.1119.26
110.9614.40
LTEL*+
45.1114.61
14.874.78
34.5619.51
34.636.7
36.477.47
35.744.56
Muscle mass
41.33.16
38.788.51
41.482.01
Strength Index*+
2.60.83
2.120.69
2.320.58
CMS*+
45.8911.07
31.479.87
53.7612.42
LMS*+
93.0727.3
69.0024.94
114.6524.29
PC*+
32.584.96
27.083.73
32.117.10
FACT-F *+
135.9418.20
124.0024.20
138.2417.49
48.494.83
45.615.82
49.752.75
SF-36 Psychological
Dimension*+
43.008.11
37.0312.58
44.337.33
Depression*+
6.837.83
12.5510.77
5.885.19
VARIABLES
*Significant differences were found between control and intervention group. + Significant differences were maintained between
baseline and follow-up assessments.
No differences taking into account confounders were observed.Variable changes were maintained in FU participants assessed
after 6 months. Correlation between QoL and LTEL (r=0.52; p=0.008) and significant weight loss (22=6.08; p=0.048) were
observed.
Conclusion. These results suggest that a specifically designed BC exercise program increases LTEL, which correlates to a better
QoL. This may reduce psychological and physical side effects of systemic treatment in patients with early BC that have recently
finished treatments, even producing lifestyle changes in BC patients that could be long lasting.
Phrophylaxis
Prophylaxis decision relative to guidelines
>20%
250
53.8
10-20%
170
36.5
<10%
45
9.7
Primary
372
79.8
Secondary
94
20.2
Undertreated
50
10.8
Correct
289
62.1
Overtreated
126
27.1
54y (28-79)
32mo (0-131).
10 (2-67)
49%/51%
24%/55%/49%
31%/47%/21%
30%/35%/33%
Inpatient mortality was 18%. Among the 16 patients who had non-ambulatory status at admission only 2 (12%) were able to walk
after treatment. Overall survival was 13,6 months. Overall survival according to the Tokuhashi index was 1,5mo high risk, 13,6mo
intermediate risk and 23,3mo low risk.
The following prognostic factors for inferior OS were identified on univariate analysis: high risk Tokuhashi score - HR 3,7 (p
0,0001); PS4 - HR 3,2 (p 0,0004); Presence of visceral metastasis - HR 4,0 (p 0,0001); interval between symptoms and
radiotherapy >14d - HR 2,5 (p 0,01). On multivariate analysis, a high risk Tokuhashi score was statistically associated with inferior
survival.
Conclusion: Metastatic spinal cord compression is an oncological emergency associated with significant morbidity and mortality.
Life expectancy is a key factor for therapeutic planning. In the limited resource setting of the Brazilian public health system, the
validation of prognostic factors is essential to guide the clinician on referring the patient to a tertiary cancer care center or to
provide palliative care avoiding burden for debilitated patients resulting from multiple daily trips to the radiation oncology
department and painful positioning on the treatment couch .This study validated the Tokuhashi index as a useful prognostic tool in
this population.
RB
SB
SCC
1.31 (1.05, 1.63) 2.41 (1.80, 3.23) 1.85 (0.68, 5.05) 4.26 (1.38, 13.19)
P value
0.0159
< 0.0001
0.2322
0.0120
1.20 (0.97, 1.48) 2.25 (1.72, 2.95) 1.11 (0.41, 3.00) 4.74 (2.15, 10.44)
P value
0.1003
< 0.0001
0.8432
0.0001
1.35 (1.09, 1.69) 2.30 (1.70, 3.10) 2.86 (1.09, 7.47) 2.26 (0.45, 11.37)
P value
0.0070
< 0.0001
0.0326
0.3232
Includes patients with baseline BPI score 8; HR = hazard ratio; CI = confidence interval.
Conclusions: In patients with advanced breast cancer, SREs are associated with an increase in pain interference. Effective
treatments that prevent SREs may reduce the burden of pain on patients daily functioning.
Category, n (%)
Improved
Stable
Worsened
2 (7)
16 (55)
11 (38)
Physical functioning
9 (31)
13 (45)
7 (24)
Role functioning
14 (48)
10 (35)
5 (17)
Emotional functioning
9 (31)
14 (48)
6 (21)
Cognitive functioning
2 (7)
13 (45)
14 (48)
Social functioning
10 (35)
13 (45)
6 (21)
Fatigue
16 (55)
5 (17)
8 (28)
8 (28)
16 (55)
5 (17)
Pain
15 (52)
8 (28)
6 (21)
Dyspnea
9 (31)
18 (62)
2 (7)
Insomnia
12 (41)
15 (52)
2 (7)
Appetite loss
7 (24)
16 (55)
6 (21)
Constipation
5 (17)
18 (62)
6 (21)
Diarrhea
2 (7)
21 (72)
6 (21)
Median time to deterioration in global health status/QoL was 5.06 m (responders, 8.54 m; nonresponders, 3.71 m; hazard
ratio=0.60, P=0.22). In linear mixed models, responders (n=16) performed better than nonresponders (n=40) in role functioning
(P=0.011), emotional functioning (P=0.031), fatigue (P=0.007), pain (P=0.047), insomnia (P=0.018), and appetite loss (P=0.032).
Mean symptom scores were significantly correlated with corresponding adverse event rates for nausea and vomiting, dyspnea,
appetite loss, constipation, and diarrhea; Spearman rank correlation coefficients ranged from 0.31 to 0.54. For QLQ-BR23 at
cycle 6, symptom scores were mostly stable; more pts had worsening in body image and systemic therapy side effects than had
improvement and more pts had improvement in breast and arm symptoms than had worsening. Responders also had longer time
to symptom deterioration.
Conclusions: In this study of first-line eribulin treatment for HER2- MBC, a majority of pts had stable or improvement in QoL
scales. Responders to eribulin were more likely than nonresponders to have stable or improved QoL.
Category, n (%)
Symptom Scale
Improved
Stable
Worsened
GHS/Qol
13 (29.5)
23 (52.3)
8 (18.2)
Physical
10 (22.7)
26 (59.1)
8 (18.2)
Role
13 (29.5)
20 (45.5)
11 (25.0)
Emotional
17 (38.6)
18 (40.9)
9 (20.5)
Cognitive
6 (13.6)
18 (40.90
20 (45.5)
Social
12 (27.3)
18 (40.9)
14 (31.8)
Fatigue
14 (31.8)
8 (18.2)
22 (50.0)
Nausea/vomiting
9 (20.5)
24 (54.5)
11 (25.0)
Pain
21 (47.7)
14 (31.8)
9 (20.5)
Dyspnea
12 (27.3)
19 (43.2)
13 (29.5)
Conclusions: Given the improvements in pain and in arm and breast symptoms, long median time to deterioration in
functioning/symptom scales in this analysis, and the tumor response rates and safety profile in the primary analysis, combination
eribulin/TRAS may be an acceptable treatment option for locally recurrent or HER2+ MBC and merits further study in larger
clinical trials.
A (n=40)
AV (n=38)
IC (n=43)
12 (30)
13 (34)
18 (42)
11.9
12.3
18.4
57.7
37.3
52.1
CNS lesions
3 (8)
6 (14)
3 (8)
2 (5)
CNS lesions
27(68)
27 (71)
31 (72)
17 (43)
19 (50)
26 (61)
(A), 57%/24% (AV; G4 AEs were mainly neutropenias) and 14%/7% (IC) of pts; there were no treatment-related G5 events.
Conclusions: Approximately one third of pts with HER2+ MBC benefited from the assigned treatments and two thirds had CNS
lesions controlled per RECIST in each group. Objective response in CNS was infrequent (0 to 14%) with all treatments. Overall,
AEs were manageable in this heavily pretreated pt population.
Lin et al. Breast Cancer Res Treat 2012;133:10571065.
All Grades
Grade 3
DIARRHEA
45 (71.4%)
2 (3.2%)
NAUSEA
33 (52.4%)
NEUTROPENIA
29 (46.0%)
17 (27.0%)
RASH
29 (46.0%)
7 (11.1%)
FATIGUE
28 (44.4%)
STOMATITIS
19 (30.2%)
DECREASED APPETITE
15 (23.8%)
1 (1.6%)
VOMITING
15 (23.8%)
In Part 1, the MTD was not reached, the maximum administered dose (MAD) of GDC-0941 was 100 mg, a dose-limiting AE was
subclavian vein thrombosis. In Part 2, Arm A, the MTD was 250 mg, Grade 3 febrile neutropenia, bacteremia, and rash were
DLTs. In Arm B and Arm C, the MAD was 260 mg, Grade 3 rash was a DLT in both arms. No differences in the PK of either GDC
0941 or paclitaxel were observed when administered in combination compared to historical single-agent data. Best tumor
response by RECIST was as follows: Part 1, 1 (5%) complete (CR) and 4 (21%) partial responses (PR); Arm A, 1 (6%) CR and 3
(17%) PR; Arm B, 7 (47%) PR; Arm C, 1 (11%) PR.
Conclusions:
GDC-0941 was generally well-tolerated in combination with paclitaxel with or without bevacizumab or trastuzumab at dose ranges
of up to 330 mg at the "5+2" dosing schedule. Anti-tumor activity has been observed in combination with paclitaxel with and
without bevacizumab or trastuzumab. Updated biomarker data and associations with clinical outcomes will be presented.
2ND-LINE TREATMENT
A1 (n = 20)
A3 (n = 20)
B1 (n = 20)
20
17
14
Complete response
Partial response
16
12
Stable disease
Progressive disease
Not assessed
80
88
50
TUMOR RESPONSE
Evaluable pts, n
Best response, n
PROGRESSION-FREE SURVIVAL
Median, months
14.5
18.5
8.6
95% CI
6.920.6
10.421.9
3.714.4
Evaluable pts, n
16
15
Median, months
12.6
16.6
8.3
95% CI
4.320.2
8.2not estimable
DURATION OF RESPONSE
Tumor
Efficacy
4.116.8
results for B3 are not reported due to
Conclusion: Interim results from this phase 1b study of pts with HER2+ locally recurrent or MBC suggest that adding trebananib
to paclitaxel and trastuzumab or capecitabine and lapatinib is tolerable and may improve antitumor activity.
Scenario
Time to
Recurrence(REC) Met
sites
Endocrine
Rx
Another
NSAI
Fulvestrant Exemestane
(F)
(EXE)
Targeted
Strategy
CT
Other
EXE +
Everolimus
(EVE)
Single
agent(SA)/
combo
10%
28%
6%
35%
14%
7%
7%
46%
5%
28%
7%
6%
25%
35%
18%
17%
3%
5%
Figure 2: PPrefs for next Rx following adjuvant anastrozole and then letrozole (LET) at first failure.
Scenario
Time to REC after TX with LET
Met sites
S3, N=187 5 mths N-VIS
Endocrine
Rx
TS
CT
Other
EXE
EXE +
EVE
SA/Combo
42%
6%
37%
7%
9%
46%
6%
36%
5%
9%
65%
5%
21%
3%
6%
EXE
EXE +
EVE
SA/Combo
Other/CLT
N/O
3%
40%
55%
2%
Time to REC on F
S6, N= 186 6 mths N-VIS, VIS
N/O - not offered
Conclusion:
MOPs PPrefs for management of pts with ER+, HER2- mBC are dictated by time to REC, number of RECs, and
presence/absence of VIS disease. EXE and EVE has substantial traction in all scenarios studied. Since these PPref data were
acquired, OS findings from the BOLERO-2 trial of EXE alone or EXE + EVE have been presented. PPrefs using these case
scenarios will be studied at 2 additional events prior to SABCS, allowing more robust data and insights into the early impact of the
BOLERO-2 survival data on PPrefs to be available at the meeting.
Treatment
Comparison
HR(95%CI)DFS HR(95%CI)LRR
B-13
(n=1,084)
N(-) / ER(-)
MF v No Adj Rx
<0.001
B-14
(n=4,028)
N(-) / ER(+)
TAM v Placebo
<0.001
B-19
(n=1,074)
N(-) / ER(-)
CMF v MF
<0.001
B-20
(n=2,299)
N(-) / ER(+)
CMF/MF+TAM v
TAM
<0.001
TAM v Placebo
0.10
Log rank
p-value
B 27
(n=2,346)
Operable T1-3
N0-1
ACT v AC
(neoadj)
0.015
B-28
(n=3,036)
N(+)
ACP v AC
0.24
B-30
(n=5,240)
N(+) / HER2(-)
ACT v AT/TAC
0.23
B-31
(n=2,063)
N(+) / HER2(+)
0.02
N events (% events)
333 (15.2%)
134 (24.3%)
< .001
138 (6.3%)
64 (11.6%)
< .001
386 (17.6%)
148 (26.8%)
< .001
Research derived from an IRB approved protocol at Naval Medical Center Portsmouth, VA. The views expressed in this abstract
are those of the authors and do not necessarily reflect the official policy or position of the Department of the Army, Department of
the Navy, Department of Defense or the United States Government. Dr. C.G. and Dr. K.M. are members of the U.S. military. This
work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that 'Copyright protection under this title is not
available for any work of the United States Government.' Title 17 U.S.C. 101 defines a United States Government work as a work
prepared by a military service member or employee of the United States Government as part of that person's official duties.
LB (%)
LHER2 (%)
HER2+
TN (%)
pCR
12.5
42.5
27.5
17.5
48
4.5
No pCR
100
42.3
29.2
8.8
14.6
37
31.5
p Value
<0.001
=0.001
All but 19 HER2+ pts (84) received H obtaining pCR in 38% of cases regardless chemotherapy type (A-based 35% vs Not A38%)
The median follow-up was 45 months (range 1-166 ms).
The 4y-RFS and OS were better in which achieved pCR than those did no (RFS 92 vs to 74%; p=0.0014 and OS 95 vs 78%;
p=0.0074).
Median Ki67 in pretreated core biopsy was 40 compared to 27% in post-NC RT. Patients with high (>30%) post-NC Ki67 levels
showed significantly higher risk for disease relapse (4 y-RFS 60%; p=0.0019) and death (4y OS 71%; p=0.018) compared with
patients with <15% (4y-RFS 93 and OS 88%) or >15-30 Ki67 levels (4y-RFS 83 and OS 82%) .
CONCLUSIONS:
According to literature data, pts achieving pCR after NC showed better RFS and OS compared to no pCR pts. The pCR rate was
significantly higher in aggressive subtypes (HER2 and TN). In HER2 disease, pCR was achieved by using chemo + H,
irrespective of A-addition. Interestingly high pre-NC KI67 levels seem to predict the possibility obtaing pCR, while post-NC Ki67
levels seem to be of prognostic value in pts who do not receive pCR.
PR
Path Downst
No Path Downst
ANG 2
2396+/-650
3455+/-1394
0.08
3184+/-1610
2818+/-768
0.6
VEGF
110+/-125
77+/-66
0.57
94+/-63
88+/-121
0.9
VEGFR2
10570+/-225
12110+/-4394
0.35
12140+/-4321
10980+/-3196
0.55
Values of biomarkers are average in ng/ml Standard Deviation. Differences analyzed by students t-test.
Abreviations: SD= stable disease. PR= partial response. p = p value. Path. Downst.= Pathological downstaging.
Furthermore, there was a significant decrease in VEGFR2 mean levels after one month of treatment (p=0.0046): 122843449
ng/ml at baseline; 81483216 ng/ml at one month and 77323052 ng/ml at 6 months. Differences between basal and one month
determination were significant (p<0.01), but no differences were seen between 1 month and 6 months, showing a relevant early
decrease of VEGFR2 plasma levels. In contrast, levels of VEGF did not change significantly over time and had no association
with clinical outcomes.
Conclusions Baseline ANG2 levels have a promising predictive value of response in this phase I trial of neoadjuvant
combination of sunitinib plus exemestane. There is a significant early decrease in VEGFR2 with treatment with sunitinib. These
results should be validated in further studies to improve the selection of patients for antiangiogenic+hormonal therapy.
Mean
Standard Deviation
Median
Range
IL-6 (pg/ml)
4.3
5.1
3.0
0-48.0
CRP(g/ml)
5.7
7.9
2.8
0.1-48.4
D-dimer(g/ml)
0.7
0.6
0.6
0.1-3.3
Spearman Coefficient
p value
MOS vs D-dimer
-0.21
<0.01
IADL vs IL-6
-0.27
<0.01
0.16
0.04
0.19
0.02
TUG vs D-dimer
0.15
0.06
TUG vs IL-6
0.26
<0.01
TUG vs CRP
0.23
<0.01
SOX10+ (%)
FR+ (%)
ER+/PR+/HER2+ (n=13)
1 (7.7%)
0 (0.0%)
0 (0.0%)
ER+/PR+/HER2- (n=92)
6 (6.5%)
7 (7.6%)
2 (2.2%)
ER+/PR-/HER2+ (n=10)
0 (0%)
5 (50.0%)
0 (0.0%)
ER+/PR-/HER2- (n=19)
8 (42.1%)
10 (52.6%)
5 (26.3%)
HER2+ (n=30)
1 (3.3%)
6 (20.0%)
0 (0.0%)
ER-/PR-/HER2- (n=25)
10 (40.0%)
13 (52.0%)
6 (24.0%)
Conclusion:
SOX10 and FR were frequently expressed in triple negative breast cancers and in progesterone receptor negative breast
cancers. Our data suggests that there may be different mechanisms by which SOX10 and FR are implicated in aggressive
breast cancers. These findings may help achieve a better understanding of the two different pathways involving stem cells
(SOX10) and growth factors (FR), their potential prognosis and their therapeutic management in the future.
MELK GE
Survival Probability
95%CI
RFS
<7
73.3
64.7-80.1
0.0112
51.8
43.2-59.8
DMFS
OS
<7
75.3
66.8-82.0
53.5
44.8-61.5
<7
84.3
76.3-89.7
62.7
53.9-70.4
0.0081
0.0002
RU-486
CDB-2914
CDB-4124
50
0.8
0.56
0.7
250
0.72
0.63
0.68
1000
0.84
0.64
0.6
50
0.95
0.76
250
0.9
0.83
0.74
1000
0.96
0.85
0.71
Premenopausal condition
Postmenopausal condition
Premenopausal and postmenopausal hormone concentrations stimulated spheroid growth by two- fold and 1.7 fold higher,
respectively, when compared to the vehicle control (0.1% DMSO) at14 days.
In the premenopausal condition, RU-486 showed moderate inhibition of spheroid growth at all three concentrations; CDB-2914
was more efficient at inhibition than CDB-4124 at 50 nM. At 250 nM and 1000 nM, CDB-2914 and CDB-4124 showed similar
efficacy.
In the postmenopausal condition, RU-486 showed very minor inhibition on spheroid growth at all three concentrations; CDB-2914
showed significantly higher inhibition than CDB-4124 at 50 nM. At 250 nM and 1000 nM, CDB-4124 was more efficient than
CDB-2914.
Conclusions: Our results indicate that T47D spheroids will grow in postmenopausal hormone concentrations, but growth is
enhanced in premenopausal hormone conditions. RU-486 did not produce effective inhibition at postmenopausal hormone levels;
however pharmacological concentrations (50, 250 and 1000 nM) of both CDB-2914 and CDB-4124 efficiently decrease the
spheroid growth induced by both premenopausal and postmenopausal hormone levels. These data suggest that low doses of
CDB-2914 and CDB-4124 should be further investigated for ER/PR positive breast cancer prevention and therapy.
Regional recurrence
no. of events
rate (%)
no. of events
rate (%)
no. of events
rate (%)
2003 (n=8933)
185
2,40%
86
1,11%
227
3,08%
2004 (n=9048)
181
2,35%
83
1,07%
189
2,51%
2005 (n=9055)
144
1,84%
75
0,95%
190
2,49%
2006 (n=7970)
131
1,87%
50
0,70%
146
2,05%
Overall (n=35.006)
641
2,12%
294
0,96%
752
2,55%
(a)Local recurrence (ipsilateral in-breast recurrence + new primary). Rates represent Kaplan Meier estimates
The LR-rate was lower with breast conserving surgery (BCS) vs. amputation (1.8% vs. 2.5%), T1a-b vs. T1c-T2 tumors (2.0% vs.
2.5%), ER+ vs. ER- tumors (1.8% vs. 3.5%) and inversely related with age (highest in pts. <35 yrs: 2.9%). LR rate seemed
independent of HER2 status.
The 5-year RR-rate was 0.9% for N0 patients, and decreased from 1.0% to 0.7% over time. The risk of RR after amputation
decreased from 1.8% to 0.9% over time, but was higher than after BCS (1.6% vs. 0.6%). Overall, the RR-rate was highest in the
N>1 group (1.4%) and the triple negative group (2.0%).
The CBC-rate was lower for patients who received chemotherapy (CT) than for patients who did not (1.6% vs. 3.1%). The
CBC-rate only decreased over the years in the CT-group (3.7% to 2.5%).
Conclusions
Loco-regional recurrence rates have decreased substantially in recent years and have become very low. For the vast majority of
patients the risk of LR is substantially lower than the risk of CBC and the risk of RR is rarely larger than 1.0%. These low rates
might reflect improvements in systemic treatment.
BMI >30
Luminal A
1105 (76%)
820 (75%)
470 (70%)
Luminal B
355 (24%)
278 (25%)
199 (30%)
Total cohort
BMI <25
BMI >30
BMI <25
BMI >30
luminal B
BCSS: breast cancer specific survival, BMI: body mass index, CI: confidence interval, DMFI: distant metastasis free interval, HR:
hazard ratio
Conclusion
Normal weight patients have a reduced risk of developing distant metastases and of BC-related death if the tumor is PR positive
compared to PR negative BC. No difference between PR positive and PR negative cases was observed in overweight BC
patients. This BMI-dependent prognostic effect of PR was limited to luminal B BC patients.
luminal A
luminal B
Metastases
BC-related death
PR positive
110/2103 (5%)
61/2103 (3%)
PR negative
16/267 (6%)
9/267 (3%)
PR positive
120/786 (15%)
75/786 (10%)
PR negative
39/138 (28%)
27/138 (20%)
62 (33)
33 (36)
41 (22)
17 (9)
17 (18)
21 (23)
25 (27)
30 (32)
For ATX-treated pts, rates of HFS are shown in Table 1. With regard to maximum grade of HFS, there was a trend of a lower rate
of grade 23 HFS for CES 823C/G compared to C/C genotype (33% vs 64%, Fishers exact p=0.059). The median cumulative
dose of X until grade 1 HFS was 98.7 g/m2 (95%CI 82.1115). CES 823C>G was not associated with the cumulative dose of X
until grade 1 HFS or dose reduction.
Conclusions
Our results indicate that CYP2C8 416G>A and FGD4 2044-236G>A might be predictive markers for paclitaxel-induced
neurotoxicity, whereas reported associations of other SNPs with toxicity could not be confirmed.
1Hertz et al. Ann Oncol 2013, 2De Graan et al. Clin Cancer Res 2013, 3Leandro-Garcia et al. Clin Cancer Res 2012, 4Baldwin et
al. Clin Cancer Res 2012, 5Caronia et al. Clin Cancer Res 2011
Financial support from Roche Netherlands.
8 (12.1)
10 (30.3)
18 (9.0)
CHT to HT 11 (10.9)
9 (13.6)
0 (0.0)
20 (10.0)
17 (25.7)
10 (30.3)
38 (19.0)
TOTAL
11 (10.9)
Treatment decisions changed for 19.0% of all patients: 10.0% and 9.0% of patients went from CHT to HT and HT to CHT,
respectively. The percentage of patients who received chemotherapy in the low, intermediate and high risk groups was 5%, 36%
and 88%, respectively. Both the central and each local laboratory analyzed the samples using IHC. We found 60% concordance
between central IHC and Prosigna intrinsic subtypes (Kappa=0.2365). Prosigna results were consistent across labs (Kappa =
0.89).
Conclusions: The Prosigna test can be reliably performed in hospital laboratories to provide useful information beyond standard
clinical-pathological variables that oncologists can use to optimize adjuvant treatment decisions in clinical practice. Subtype
determined using IHC is not an interchangeable proxy for subtype determined by Prosigna.
*Two last authors have contributed equally to the study.
RFS, unadjusted
Hazard Ratio*
P Value
0.779
0.315
0.48-1.27
1.073
0.769
0.67-1.73
0.978
0.934
0.58-1.65
0.968
0.913
0.54-1.75
0.921
0.798
0.49-1.73
1.038
0.913
0.53-2.01
Table 2
Estrogen Positive Carcinomas
TC Quartile
OS, unadjusted
Hazard Ratio*
P Value
0.824
0.581
0.42-1.64
1.224
0.535
0.65-2.21
RFS, unadjusted
1.564
0.164
0.83-2.94
0.654
0.299
0.29-1.46
0.813
0.602
0.37-1.77
0.891
0.778
0.40-1.98
RFS, unadjusted
Hazard Ratio*
P Value
1.090
0.839
0.78-2.49
0.708
0.414
0.31-1.62
0.488
0.138
0.19-1.26
3.048
0.031
1.10-8.41
1.212
0.739
0.39-3.76
0.820
0.759
0.23-2.91
TNM Stage
Number of Patients
90
113
IA
18993
90
112222
IA
2246
90
21211
IIA
5101
90
122112
IIA
4019
90
31111
IIB
682
90
131111
IIIA
82
58
2232221
IIB
1103
58
22322213
IIB
92
58
3331111
IIIA
128
58
14211111
IIIC
64
**Abbreviated table. Prognostic Factor Combination in order from left to right: T, N, grade, ER status, PR status, age, race,
histological type. * T1 = 1; T2 = 2; T3 = 3; N0 = 1; N1= 2; N2 = 3; N3 = 4; Grade 1 = 1; Grade 2 = 2; Grade 3 = 3; ER+ = 1; ER- =
2; PR+ = 1; PR- = 2; Age <= 50 = 1; Age >50 = 2;
Conclusions:
Integrating new prognostic factors into the TNM always changed the outcome. Survival rates, therefore, are relative and depend
on the selection of prognostic factors. Adding new factors selected different cohorts from the population which had a
heterogeneous population of cancer survivors. These cohorts usually had different survival rates compared with the overall
population from which they were drawn. Integrating combinations of prognostic factors revealed frequent crossover of stage
groups at 10 years, which is a violation of a staging system and could impact the interpretation of clinical trials.
Hazard ratio
p value
Overall
0.77
0.52-1.16
0.21
ER positive
0.60
0.31-1.16
0.13
ER negative
0.82
0.47-1.43
0.49
Broup B
Group C
Chemotherapy (%)
No Mammaprint
122
61 (47%)
Mammaprint
19
4 (21%)
15/3
89%
No Mammaprint
1229
507 (41%)
MammaPrint
226
75 (33%)
125/74
86%
No MammaPrint
397
209 (53%)
MammaPrint
62
26 (42%)
34/23
84%
p-value
0.019
0.016
0.117
Conclusion
The proportion of patients who receive ACT decreased when a GS was used in predefined cohorts of patients for whom
ambivalence exists regarding the use of ACT. The majority of patients for whom a GS was used did not fit these predefined
categories and in subsets an inverse relation was seen: the use of a gene signature was associated with a higher chance of
receiving ACT.
reduced survival. The thrombin pathway may provide a novel therapeutic target, particularly in ER negative, HER2 positive breast
cancer.
HR
95%CI
0.157
ypT-stage
<0.0001
ypN-stage
0.035
pCR (T0/isN0)
0.027
Lymphovascular invasion
0.040
Subtype
<0.0001
Histologic grade
0.001
0.954
1.00
0.973
1.01
0.55-1.86
0.797
1.12
0.48-2.59
Conclusion: LNR is not superior to ypN-stage in predicting clinical outcome of breast cancer after neoadjuvant chemotherapy.
N dead/total
2124/2311
Median OS, m
15
De novo MBC
N dead/total
Median OS, m
593/699
26
P-value
<.0001
821/912
24
290/364
34
<.0001
PR known
414/477
20
251/319
34
<.0001
PR unknown
407/435
28
39/45
33
0.1315
110/126
15
0.0001
TNBC
All
368/391
9
HER2+
All
935/1008
11
193/209
17
0.0061
ER neg
268/278
58/60
9.5
0.2191
ER+
667/730
15
135/149
25
0.0088
OS was longest for denovo HR+/HER2 negative (34m) and shortest for HER2+/ER negative (7m) and TN (9m) relapsers. The
difference in OS for relapsed vs denovo HER2+ disease was significant for HR+ but not HR negative cases, likely due to small
numbers in the latter subset.
Conclusion: Relapsers experience shorter OS than their denovo biomarker counterparts, possibly due to the selective pressure of
adjuvant therapy on disease biology. When restricted to pts who received systemic therapy for MBC, OS figures may be higher.
Novel therapies may decrease the total number of relapsers, and improve OS in MBC for all, but are unlikely to narrow the OS
gap between relapsed and denovo groups. Trials exploring therapies for MBC of all biomarker types should therefore stratify by
stage at initial diagnosis.
Metabolic Syndrome
p-value
Intermediate 21-gene RS
Low 21-gene RS
0.30
0.004
Yes
14(24%)
24(32%)
23(22%)
21(46%)
No
44(76%)
50(68%)
81(78%)
25(54%)
Diabetes
0.24
0.002
Yes
12(21%)
22(30%)
18(17%)
19(41%)
No
46(79%)
52(70%)
86(83%)
27(59%)
Hypertension
0.28
0.003
Yes
36(62%)
39(53%)
52(50%)
35(76%)
No
22(38%)
35(47%)
52(50%)
11(24%)
Obesity
p-value
0.27
0.004
Yes
35(60%)
38(51%)
51(49%)
34(74%)
No
23(40%)
37(49%)
54(51%)
12(26%)
coming years. It is also possible that multigene assays currently in use for prognosis and prediction may need refinement in the
presence of MS and/or its components.
58 (27-88)
Median RS (Range)
22 (18-31)
ER+ (%)
262 (99.6)
PR+ (%)
234 (89)
10 (3.8)
Grade (%)
1
84 (32.8)
136 (53.1)
36 (14.1)
Histology (%)
Ductal
187 (71.1%)
Lobular
45 (17.1%)
Mixed
31 (11.8%)
1.6 cm (0.2-6.5)
238 (90.8)
24 (9.2)
Conclusions:
With a median follow-up of 45 months, our data indicate that the realized distant recurrence rate for patients with an intermediate
range RS was less than 5%. These data do not exclude the possibility that some patients with an intermediate RS may derive a
small incremental benefit from the addition of adjuvant chemotherapy. Larger, randomized prospective trials like TAILORx should
provide additional guidance for the management of this patient population.
DFS
OS
Definition A ( 10%)
AR positive
122 (79)
AR negative
32 (21)
Definition B (IRS 3)
AR positive
97 (63)
AR negative
57 (37)
DFS and OS did not differ between both endocrine treatment arms in a Cox regression model tested for interaction between AR
expression and endocrine treatment.
Conclusion: In this pilot study patients with AR positive disease had a numerically better DFS and OS compared to AR negative
patients, but only prolongation of OS in patients with 10% AR positive tumor cells was statistically significant. AR expression did
not influence outcome between tamoxifen and anastrozole treated patients, but based on the small number of events, this results
have to be interpreted with caution. This data will be confirmed in a larger proportion of patients treated within the ABCSG-12 trial.
P Value
Age (continuous)
1.03 (1.01-1.04)
0.0004
0.99 (0.94-1.04)
0.74
0.70 (0.49-0.99)
0.042
Brain metastasis
1.09 (0.77-1.56)
0.63
0.98 (0.61-1.58)
0.94
1.66 (1.17-2.35)
0.005
Conclusions: In the treatment era of trastuzumab and lapatinib, 8% of patients within this cohort with HER2+ MBC lived more
than 10 yrs. Analysis of current standard clinical and pathologic characteristics are not predictive of survival duration. Identifying
factors associated with prolonged survival may provide insights for individualizing treatment selection.
Overall
35 to 54
55 to 74
75+
*Probabilistic sensitivity analysis using 10,000 second-order parameter samples with a 20 year time horizon
Chinese (n=234)
Caucasian (n=1,492)
P Value
Risk
low
high
low
high
low
high
Gail
137 (58.6%)
97 (41.4%)
107 (63.7%)
61 (36.3%)
884 (59.2%)
608 (40.8%)
0.509
BCSC
211 (90.2%)
23 (9.8%)
152 (90.5%)
16 (9.5%)
864 (58.0%)
626 (42.0%)
0.000
PUMC
167 (71.7%)
66 (28.3%)
108 (64.7%)
59 (35.3%)
781 (52.6%)
704 (47.4%)
0.000
TC N=26,266
TCH N=9,105
AC/AC-T N=15,522
78.1
74.5
73.6
Age 65
15.1
11.5
11.4
Cardiovascular disease
16.8
14.6
16.6
Diabetes
11.8
10.2
11.1
Liver disease
1.9
2.2
2.0
Lung disease
2.0
1.5
1.9
Renal disease
1.3
0.9
1.0
Osteoarthritis
7.0
6.1
6.0
Thyroid disorder
13.0
11.9
10.6
33.2
33.9
33.7
24.6
23.2
22.9
12.5
11.2
11.1
4+
7.8
6.1
5.9
21.9
10.4 (NA)
25.5
12.7 (NA)
26.4
11.6 (NA)
Chronic comorbidities
Group
N Events
p-value
NP
8478
42
1.00 (ref)
<0.0001
PD
4229
54
AH
703
NP
8478
232
1.00 (ref)
PD
4229
151
AH
703
74
<0.0001
Increase
Decrease
Stay nondense
n; OR (95% CI)
n; OR (95% CI)
n; OR (95% CI)
Vitamin D*
291
Vitamin D**
291
Vitamin D***
211
Vitamin D****
211
*Short-term: adjusted for age, BMI, race and menopausal status; **Short-term: additional adjustment for highest education level,
annual household income; ***Long-term: adjusted for age, BMI, race and menopausal status; ****Long-term: additional
adjustment for highest education level, age of first childbirth, time intervals from the first to the last mammogram
Discussion: Although vitamin D supplementation was not associated with short-term changes in MD, we did observe a trend
toward an association with long-term change among high-risk women. If replicated in larger studies, our study gives added
evidence that MD changes may need longer observation time.
FH+ Mean(SD)
FH-Mean(SD)
By parent/guardian report
n=437
n=431
Anxiety
46.7 (30.3)
49.1 (29.1)
Depression
51.2 (27.2)
52.8 (27.3)
Somatization
43.9 (30.7)**
50.3 (30.7)**
Internalizing
46.1 (29.6)*
50.8 (29.1)*
Daughter reported
n=228
n=222
Anxiety
40.2 (28.5)
42.4 (29.5)
Depression
32.4 (26.2)
32.4 (27.7)
Somatization
36.5 (25.5)
44.0 (27.7)
Internalizing
33.1 (25.5)
34.0 (26.7)
IBC Worry
2.0 (3.4)**
1.2 (2.4)**
ABC Worry
3.8 (5.8)**
2.0 (4.3)**
Perceived Stress
4.8 (2.8)
4.6 (2.8)
n=219
n=209
85 (39%)**
27 (13%)**
57 (26%)
54 (26%)
19 (9%)
35 (17%)
58 (26)
93 (45)
* p<0.05, ** p<0.01
Conclusions: Pre-adolescent girls from BC families have lower somatization and internalizing behaviors by parent report, but
higher self-reported BC worry. Daughter general anxiety, depression and BC worry are associated with corresponding mother
affect. Some girls from BC families are aware of their increased risk and related research suggests this may increase with age.
Understanding how PSA and BC worry changes over time and the impact on health and risk behaviors can inform interventions to
optimize responses to growing up in families at familial and genetic risk for breast cancer.
Time
IBE
Invasive IBE
IBE
Invasive IBE
At 5 years
At 7 years
At 10 years
At 12 years
Controls n=274
p-value
2 (3%)
6 (2%)
0.81
7 (10%)
25 (9%)
0.68
9 (13%)
24 (9%)
0.30
4 (1-19)
3 (1-41)
0.42
11 (1-129)
6 (1-342)
0.40
Cell Type
A
A/B
B
24 (33%)
45 (63%)
3 (4%)
99 (36%)
145 (53%)
30 (11%)
0.70
Nuclear Grade
1
14 (19%)
54 (75%)
58 (21%)
189 (69%)
0.54
2
3
4 (6%)
27 (10%)
Necrosis
6 (2%)
NA
Calcifications
27 (38%)
114 (42%)
0.57
Ductal Extension
60 (83%)
249 (91%)
0.09
ALH
12 (17%)
31 (11%)
0.19
HER2 negative
HR (95% CI)
Reference 1.0
0.53 (0.31-0.90)
Ref
1.18 (0.77-1.82)
Ref
1.64 (0.92-2.90)
Ref
0.76 (0.39-1.50)
Ref
Conclusions: In this long term follow-up DCIS cohort positive HER2 status in the primary DCIS predicted a statistically lower risk
of IBCR. This effect was seen from ten years after primary surgery and onwards. The risk of an in situ IBE was increased and the
risk of an invasive IBE was decreased if the primary DCIS was HER2 positive. All together, this challenges the role of HER2 as a
driving force of the progression from in situ to invasive cancer.
95% CI
1-year
96%
89%-100%
2-year
73%
57%-92%
3-year
49%
32%-75%
Conclusions: This retrospective analysis of clinical outcomes of Stage III TN-IBC treated with contemporary anthracycline/taxane
regimes is consistent with previously reported outcomes using historical NAC regimens. (Li, et al, the Oncologist 2012). These
dismal rates of 49% 3-year OS from diagnosis and 58% 1-yr DFS following MRM demand more active investigation into novel
targeting agents which can be combined with standard NAC specifically for the treatment of TN-IBC; a disease that has no known
therapeutic target. For this reason DFCI is actively investigating the role of inhibiting the JAK2/STAT3 pathway using ruxolitinib in
conjunction with standard weekly paclitaxel followed by AC as NAC for TN-IBC. Clinical trial information: NCT01796197.
No surgery (%)
White race
78.4
68.7
T1 stage
15.8
7.4
N1 stage
27.5
38.6
ER +
72.5
57.6
PR +
57.2
45.2
Her2 +
1.4
3.7
0.25
Grade II
34.7
26.3
76.6
56.2
Radiation therapy
37.8
32.3
Year of diagnosis
P-value
1988-1992
8.1
7.4
2008-2011
25.2
36.9
Population
Overall stable
disease
Overall
23
2 (9%)
11 (48%)
4 (17%)
48%
"Luminal"
0 (0%)
2 (50%)
1 (25%)
50%
16
2 (13%)
9 (56%)
3 (19%)
56%
HER2
positive
TNBC
0 (0%)
0 (0%)
0 (0%)
0%
Conclusions:
2B3-101 alone or with trastuzumab is safe and well tolerated and shows intra- and extracranial anti-tumor activity in heavily
pretreated BC pts. A 12-week PFS rate of 56% in HER2+ BCBM was observed, which warrants further clinical studies. An
international, multicenter, randomized, controlled phase IIb study in HER2+ BCBM is being planned.
NCT01386580, sponsored by to-BBB technologies BV.
Points
KPS
80-100
13
<70
Hazard Ratio
0.41 (0.32-0.52)
<0.001
10
0.49 (0.32-0.74)
<0.001
HER2
0.58 (0.43-0.77)
0.007
Luminal A
0.63 (0.48-0.77)
0.002
Basal
0
0.65 (0.56-0.77)
<0.001
0.51 (0.38-0.67)
<0.001
0.68 (0.53-0.88)
0.003
12
2-3
>3
Leptomeningeal Disease
No
10
Yes
Primary Controlled
Yes
No
>/=50
Both
0.75 (0.59-0.96)
0.02
0.76 (0.61-0.96)
0.02
The new GPA consists of four groups: unfavorable, intermediate 1, intermediate 2 and favorable with OS of 2.7, 9.1, 18.5 and
29.5 months respectively.
New GPA and Conventional GPA
New GPA
Group
Number of points
Number
Median OS (months)
Unfavorable
</=21
84 (19%)
2.7
Intermediate-1
22-36
153 (34%)
9.1
Intermediate-2
37-45
133 (30%)
18.5
Favorable
>45
78 (17%)
29.5
Conventional GPA
Unfavorable
0-1
55 (12%)
4.8
Intermediate-1
1.5-2
130 (29%)
9.1
Intermediate-2
2.5-3
148 (33%)
12.9
Favorable
3.5-4
116 (26%)
20.4
Conclusions:
A new GPA for BCBM is proposed.
Number of points
KPS
80-100
</= 70
>/= 2
Leptomeningeal Disease
No
Yes
Hazard Ratio
0.31 (0.21-0.48)
<.0001
0.42 (0.28-0.64)
<.0001
0.43 (0.26-0.71)
0.001
The new GPA consists of three groups: unfavorable, intermediate and favorable with OS of 2.7, 9.1 and 13.6 months respectively.
New GPA
GPA
Number of points
Number
Median OS ( months)
Favorable
36 (27%)
13.6
Intermediate
4-5
59 (45%)
9.1
Unfavorable
</= 3
36 (27%)
2.7
<0.0001
Conclusions:
A novel GPA for TBCBM is proposed.
overall
Arm A
Arm B
Arm C
ER+
ER-
n tot
pCR rate %
LP
17
59
non-LP
88
27
LP
40
non-LP
27
26
LP
50
non-LP
28
21
LP
83
non-LP
33
33
LP
33
non-LP
59
25
LP
11
73
non-LP
29
31
p
0.011
0.52
0.15
0.022
0.67
0.02
Overall, 71 of the 121 CherLOB patients had residual disease at surgery: for 54 of them, paired pre-treatment and post-treatment
TIL were available. No significant changes in ItTIL and StrTIL levels were observed before and after treatment. However, six
cases presented a LP phenotype on the residual disease; all but one of them started from a non-LP pre-treatment phenotype and
ECOG PS
Menopausal status
Tumor size, cm
Subgroup
All (N=2591)
<40 (N=484)
40<65 (N=1868)
65 (N=239)
0 (N=2388)
1 (N=192)
Post (N=1250)
Pre (N=1341)
0<2 (N=939)
OS HR (95% CI)
0.94 (0.751.18)
0.88 (0.511.54)
0.89 (0.681.17)
1.50 (0.733.05)
0.98 (0.771.25)
0.68 (0.311.49)
1.16 (0.831.62)
0.79 (0.581.08)
1.17 (0.691.98)
25 (N=1514)
0.93 (0.701.23)
5 (N=132)
0 (N=1640)
0.84 (0.451.58)
0.79 (0.561.13)
1.01 (0.651.56)
1.14 (0.751.73)
13 (N=638)
Adjuvant CT
4 (N=313)
Anth (N=947)
Anth + tax (N=1508)
Tax (N=136)
ER and PgR negative (N=2453)
ER and/or PgR low (N=138)
Surgery
0.83 (0.541.28)
1.03 (0.781.37)
0.63 (0.241.66)
0.93 (0.731.17)
1.42 (0.464.34)
0.93 (0.671.30)
0.95 (0.691.31)
Conclusions: The final OS analysis after 293 deaths showed no significant benefit from the addition of BEV to standard adjuvant
CT for early TNBC. Updated IDFS results were similar to the primary IDFS results, showing no difference between treatments.
Late-onset AEs were rare in both groups. 5-year IDFS and OS rates suggest that the prognosis for pts with TNBC is better than
previously thought.
$ pCR in all breast tumours AND absence of disease in ax LNs in all breast tumours
DFEC
Bev+DFEC
% (95%CI)
% (95%CI)
(n=66/393)
(n=87/388)
p*
44% (36-54)
52% (34-69)
7% (4-11)
6% (3-10)
(n=114/394)
(n=138/388)
56% (47-65)
73% (54-87)
19% (14-24)
* Adjusted for stratification variables. $ Primary endpoint for the ARTemis trial
Conclusions: ARTemis showed a significant improvement in both pCR and MRD rates with the addition of bev to D-FEC. ER-neg
and ER-weak pos / HER2-neg breast cancer pts appeared to benefit most from bev, whilst pCR and MRD rates in ER-strong pos
pts were lower and did not appear to benefit from bev. Our results are similar to those reported in Geparquinto and CALGB
40603.
Median
Pts with PFS events/Total
2nd-line PFS, Unstratified HR (95% CI)
No. of pts (%)
mo
Subgroup
CT
BEVCT
CT BEVCT
All
0.75 (0.61-0.93)a
56/60 (93)
2.1 4.9
0.59 (0.40-0.88)
0.84 (0.67-1.05)
<6
61/69 (88)
3.9 5.1
0.62 (0.43-0.90)
0.82 (0.65-1.03)
1.5ULN
0.77 (0.62-0.96)
>1.5ULN
36/40 (90)
36/37 (97)
2.1 5.8
0.73 (0.46-1.16)
Taxane
25/32 (78)
26/32 (81)
3.2 6.9
0.55 (0.31-0.98)
0.84 (0.68-1.06)
Vinorelbine
22/24 (92)
2.4 6.5
0.41 (0.22-0.75)
Capecitabine
0.92 (0.72-1.19)
choiceb
45/56 (80)
54/68 (79)
27/27 (100)
BEV-free interval, wk
aStratified. bStratification
Non-capecitabine
86/101 (85)
79/96 (82)
3.2 6.5
0.55 (0.40-0.75)
0.82 (0.64-1.04)
>6
65/81 (80)
0.72 (0.51-1.00)
74/98 (76)
4.4 7.6
factor.
CONCLUSIONS: PFS was statistically significantly improved with the addition of BEV to 2nd-line CT in BEV-pretreated pts,
meeting the primary objective of TANIA. This effect was seen consistently within subgroups based on stratification factors. Within
the limitations of exploratory subgroup analyses with small sample sizes, further subgroup analyses may suggest some potential
inconsistencies in treatment effect. These hypothesis-generating observations require further exploration of potential differences
in disease and pt characteristics in TANIA, which may have influenced physicians CT choice.
White
74
80
60,519/ 65,425
Black
17
10
13,611/ 8,474
Hispanic
4,219/ 3,748
None
4,792/ 1,792
Medicaid
10
8,326/ 3,911
Medicare
39
35
32,614/ 28,843
Private
40
54
32,614/ 43,916
>31%
16
11
12,609/ 8,962
23-30.9%
17
15
14,169/ 11,895
18-22%
16
15
12,991/ 12,058
12-17.9%
23
24
18,854/ 19,717
Race/ Ethnicity*
Insurance*
<12%
24
30
19,168/ 24,606
<$28,000
12
9,330/ 6,274
$28,000-32,999
13
11
10,422/ 8,718
$33,000-38,999
17
16
14,188/ 13,199
$39,000-48,999
23
23
18,585/ 18,658
>$49,000
31
37
25,269/ 30,391
64
66
51,832/ 54,019
11
8,855 /7,007
2+
3,178 /1,548
Household Income*
(per year)
Comorbidity*
* p-value < .0001 **Observed # of patients / % of all stages in category x total # of stage IV patients (Expected)
Residual
Disease Status
Cancer Burden
TNBC (AR
subtype)
TNBC (BL 1)
Bone/liver/lymph node
recurrence at 135 days
TNBC (BL 1)
TNBC (BL 2)
Luminal B
Progression during
chemotherapy
Luminal HER2 3
Conclusions: We observed substantial evolutionary changes in residual breast tumors remaining after NAC. Our findings suggest
that a comprehensive assessment of the mutational landscape that has evolved during NAC can inform drug development in high
risk breast cancer patients.
Cohort 1
50%
(90% CI 33.8-66.2%)
57.5%
(95% CI 40.9-73.0)
7.4 months
Cohort 2
22.2%
(90% CI 11.2-37.1%)
42.2%
(95% CI 27.7-57.8)
5.3 months
As we previously reported (Wagle et al, ASCO 2014), across 57 metastatic tumors, significant recurrently mutated genes were
TP53 (n=30; 53%) and PIK3CA (n=19; 33%). The frequency of mutant TP53 and PIK3CA was not significantly different from 119
primary, treatment-nave HER2+ tumors sequenced in the TCGA study (50%, p=0.8 and 27%, p=0.5, respectively). Recurrent
copy number alterations were also similar to TCGA data.
Comparing the 38 pts who received any prior T with the 19 pts who did not, there was no significant difference in the incidence of
mutant TP53 (53% vs 53%, p=1.0) and PIK3CA (37% vs 26%, p=0.6).
We identified mutations in the HER2 kinase domain in 4/38 pts who received prior T (11%), as compared to 0/19 T-nave pts.
HER2 kinase domain mutations have been identified in 2% of HER2-negative cancers but <1% of primary HER2+ cancers. 3 of
the mutations were L755S, which has been shown to be resistant to L and sensitive to irreversible HER2 inhibitors. The 4th
mutation was D742N, a novel kinase domain mutation. None of the 4 pts with HER2 kinase domain mutations had an objective
response, though 1 pt had stable disease for 29 weeks.
An analysis comparing paired archival primary tumors and baseline metastatic biopsies from 36 pts to identify genomic alterations
acquired or enriched in the metastatic tumors will be presented.
Conclusions: We present an analysis of the genomic landscape of HER2+ MBC, including comparisons between matched
primary tumors and metastatic biopsies. Somatic HER2 kinase mutations in pts with HER2+ MBC treated with prior T suggests
that these mutations may be involved in resistance to T, and may predict poor response to additional anti-HER2 therapy with
combined L and T. Novel therapeutic approaches may be required for these pts.
Gene
Mutation
Metastasis (MAF)
Plasma (MAF)
PTEN
p.Q171E
YES (27.5%)
YES (25.9%)
SMAD4
p.E394*
NO
YES (10.9%)
NO
NO
4
6
PIK3CA
p.H1047R
YES (20.5-47.7%)
YES (4.6%)
TP53
p.V274A
YES (35.6-56.1%)
YES (20.7%)
IDH2
p.R140R
YES (28.2)
YES (0.5%)
10
PIK3CA
p.H1047R
YES (19.7%)
NO
11
PIK3CA
p.E453K
YES (4.6-17.8%)
YES (2.8%)
11
PIK3CA
p.E453K
YES (13.1-23%)
YES (3.4%)
14
PIK3CA
p.H1047R
YES (24.8%)
NO
14
PIK3CA
p.H1047R
YES (0-13.8%)
YES (0.5%)
16
TP53
p.Y103*
YES (59.8-86.3%)
YES (49%)
17
NO
NO
19
NO
NO
20
PIK3CA
p.E545K
NO
YES (14.3%)
30
TP53
p.M237K
YES (27.6-50%)
YES (51.8%)
37
TP53
p.H193L
YES (61.6-82.9%)
YES (55.5%)
38
AKT1
p.E17K
YES (26-68.2%)
YES (10%)
38
TP53
p.R248W
YES (23.6-56.8%)
YES (5.9%)
39
TP53
p.R136H
YES (7.5%)
NO
NO
NO
40
MAF: Mutant allele frequency; For patients with multiple metastases samples at the same time point, the MAF range is provided.
Patients 11 & 14 had 2 timepoints with synchronous metastases/plasma samples.
0-18 months
18-30 months
30-42 months
42-54 months
>54 months
1920
1150
842
673
543
401
770
170
86
53
44
Percentage (%)
58.5
67.0
20.2
12.8
9.8
11.0
1944
1294
979
782
618
481
650
153
109
69
45
Percentage (%)
52.8
50.3
15.6
13.9
11.2
9.4
1.26 (1.11-1.43) 1.33 (1.17-1.52) 1.29 (1.01-1.65) 0.92 (0.67-1.25) 0.87 (0.59-1.29) 1.17 (0.74-1.86)
Anastrozole
Women at risk
Placebo
Women at risk
Tamoxifen Only
AI Only
No Hormones
HR (95% Cl)
HR (95% Cl)
HR (95% Cl)
HR (95% Cl)
Ischemic
1.00 (ref)
Stroke
1.00 (ref)
Other CVD
1.00 (ref)
COMPOSITE CVD
1.00 (ref)
Based on a subset of 7,982 patients who underwent breast irradiation, the risk of CVD overall was greater among women who
used AIs only and received left-sided irradiation (HR=1.21, 95% CI: 1.02-1.44).
Tamoxifen Only
AI Only
No Hormones
HR (95% Cl)
HR (95% Cl)
HR (95% Cl)
HR (95% Cl)
1.00 (ref)
1.00 (ref)
Discussion
These results indicate that variation exists in the type of CVD events that occur in breast cancer patients receiving AIs in
comparison to tamoxifen users. For example, the risk of ischemic disease or stroke was not elevated in those who used AIs only
versus TAM users. However, overall CVD events were greater in women who used AIs only (or sequentially after TAM),
especially if they received left-sided breast irradiation. While these observational study results require cautious interpretation, they
provide a basis for comparing the benefits and risks of endocrine treatments.
Blood Draw
Blood Pressure
Injection
Number of flights
None
91.2% (1796/1969)
83% (1633/1967)
97.6% (1923/1969)
72% (1461/2031)
1 or more
8.8% (173/1969)
17% (334/1967)
2.4% (46/1969)
28% (570/2031)
Conclusions:
In our patient population, non-treatment related risk factors including blood draws, blood pressures, and injections in the at-risk
arm, and flying were not significantly associated with increases in arm volume. This data can be used to help improve and refine
patient education regarding the importance of risk-reducing practices after breast cancer treatment.
PD
PR (Cycle
completed)
SD (Cycle
completed)
NE
1A DL1 3 80 mg daily
2 (confusion, ALT)
1 (22)
1 (a)
3 (ALT, rash)
2 (13,3 (a))
2 (9, 7)
Cohort
N BKM120
1A DL2b 2
100 mg
intermittent
2 (6, 6)
1B
10
100 mg
intermittent
2 (15+, 12)
5 (16+, 9+, 9, 6, 6)
3 (a, a, b)
10 100 mg daily
2 (1+, 2+)
16
13
Total (N) 31
*All occurred in C2+ except the diarrhea, CBR: 63%, NE: Not evaluable, a: off due to AE, b: withdrew consent, # NE for CBR
Conclusion
BKM120 100mg, administered daily or intermittently, plus F was tolerable without DLT during C1. Grade 2/3 AST/ALT and rash
were common in both dosing schedules in subsequent cycles resulting in BKM120 interruption/reduction. Promising activity
observed in this trial warrants further development of this combination in ER+ BC. Phase III studies are ongoing in pts with ER+
MBC progressed on aromoatase inhibitor.
OlympiA
Neo-Olympia
OlympiAD
Setting
Metastatic BC (mBC)
Design
Randomized (1:1),
double-blind,
parallel-group
Olaparib
monotherapy
arm
Comparator
arm(s)
Primary endpoint
Placebo
IDFS
pCR rate
PFS (BICR)
HRQoL
HRQoL
HRQoL
Other objectives
Safety, tolerability
Safety, tolerability
Safety, tolerability
Target
recruitment (pts)
1320
300
310
Secondary
endpoints
*Curative-intent surgery to be performed after 12 wks; pts will then receive olaparib 300 mg bid (Arm A), placebo (Arm B), or
either weekly paclitaxel 80 mg/m2 for 12 wks (then olaparib 300 mg bid) or olaparib 300 mg bid (Arm C). BICR, blinded
independent central review; d, days; DDFS, distant disease-free survival; EFS, event-free survival; IDFS, invasive disease-free
survival; ORR, objective response rate; pCR, pathological complete response; q, every; PFS2, time to second disease
progression or death; TNBC, triple-negative BC
For each trial, eligible pts will have a BRCAm and will undergo gBRCAm testing (Myriad Integrated BRACAnalysis) as part of the
trial. For OlympiA, pts must be at high risk of recurrence and have completed local treatment and either neoadjuvant (without
pCR) or adjuvant chemotherapy. Neo-Olympia pts can have operable, locally advanced or inflammatory BC, must have a tumor
>2cm by clinical exam (or >1cm by radiological exam) and have completed four cycles of anthracycline plus carboplatin without
progression. OlympiAD pts can have TNBC or HER2 BC, and must have received prior anthracycline and taxane in the adjuvant
or metastatic setting, and 2 chemotherapy lines for mBC. OlympiA pts will be treated for up to 12 months (m); efficacy will be
assessed q3m up to 24m, then q6m up to 60m, then q12m. Neo-Olympia pts will be treated for 12 wks (w) pre-surgery, then for
40w post-surgery. In OlympiAD, PFS will be assessed by RECIST v1.1; radiologic exams will be performed at baseline, q6w up to
6m, then q12w until progression. In OlympiA and OlympiAD, IDFS and PFS will be analyzed using stratified log-rank tests; for
Neo-Olympia, pCR rate will be analyzed with an adjusted logistic regression model. Primary analyses will be undertaken after 330
IDFS events (OlympiA), surgery (Neo-Olympia) and 230 PFS events (OlympiAD). Enrollment began in Mar 2014 for OlympiAD,
Apr 2014 for OlympiA and is expected to begin in Q3 2014 for Neo-Olympia.
Phase II
Phase III
n = 240
n = 550
1:1:1
1:1
Randomization
Stratification
DFI: 1 y vs > 1 y
Prior neo/adjuvant taxane (yes vs no)
Key Endpoints
Primary
Secondary
ORR
OS
OS
Safety
Duration of response
Safety
Exploratory
Quality of life
Tumor biomarkers