Background:

Breast cancers in recently postpartum women may have worse outcomes, but studies examining tumor molecular features by pregnancy recency have shown conflicting results.

Methods:

This analysis used Carolina Breast Cancer Study data to examine clinical and molecular tumor features among women less than 50 years of age who were recently (≤10 years prior) or remotely (>10 years prior) postpartum, or nulliparous. Prevalence odds ratios (POR) and 95% confidence intervals (CI) were estimated using multivariable models.

Results:

Recently postpartum women (N = 618) were more frequently lymph node–positive [POR (95% CI): 1.66 (1.26–2.19)], estrogen receptor (ER)-negative [1.37 (1.02–1.83)], and IHC-based triple negative [1.57 (1.00–2.47)] compared with nulliparous (N = 360) women. Some differences were identified between recent versus remotely postpartum; smaller tumor size [0.67 (0.52–0.86)], p53 wildtype [0.53 (0.36–0.77)], and non–basal-like phenotype [0.53 (0.33–0.84)] were more common among recently postpartum. Recently postpartum (vs. nulliparous) had significant enrichment for adaptive immunity, T cells, B cells, CD8 T cells, activated CD8 T cells/natural killer (NK) cells, and T follicular helper (Tfh) cells and higher overall immune cell scores. These differences were attenuated in remotely (compared with recently) postpartum women.

Conclusions:

These results suggest a dominant effect of parity (vs. nulliparity) and a lesser effect of pregnancy recency on tumor molecular features, although tumor immune microenvironments were altered in association with pregnancy recency.

Impact:

Our study is unique in examining tumor immune microenvironment and RNA-based markers according to time since last childbirth. Future studies should examine the interplay between tumor features, postdiagnostic treatment, and outcomes among recently postpartum women.

See related commentary by McDonald et al., p. 518

This article is featured in Highlights of This Issue, p. 493

Breast cancer is the most commonly diagnosed cancer among women aged 20 to 49 years (1) and the cancer most frequently temporally-related to pregnancy (2). Several studies have found that breast cancer diagnosed during pregnancy or the postpartum years is associated with worse overall and disease-free survival, and plausible mechanisms have been proposed (3–9). Elevated concentrations of estrogen, progesterone, and prolactin during pregnancy may promote the growth and progression of breast tumors (10–12). Postpartum involution, a breast remodeling process that generates a wound-healing/inflammatory microenvironment (13–16), promotes tumor growth in animal studies (17, 18). However, despite the strength of the preclinical and mechanistic data, epidemiologic and clinical studies of breast tumors diagnosed during pregnancy or the postpartum years have reported incongruent results with respect to molecular characteristics (3–8, 19–24).

Inconsistencies in the molecular phenotypes of postpartum-associated breast cancer (PPBC) may be in part due to small sample size, varying definitions that combine breast cancers diagnosed during pregnancy (PrBC) with PPBC (25), and differences in the menopausal status of the study participants. Use of different reference groups has also complicated comparisons across studies. For instance, previous studies have either used only nulliparous women as the reference group (4, 21–24) or combined nulliparous women and parous women diagnosed with breast cancer remotely (after a given number of postpartum years) in one reference group (5–8, 19, 20). To identify the effects of recent pregnancy, an ideal referent group would restrict to premenopausal women and account for age differences. Furthermore, adequate sample size, clear definitions of recently postpartum women (i.e., ≤10 years prior to diagnosis), and distinct comparison groups, including remotely postpartum (>10 years prior to diagnosis) and nulliparous women, would improve inferences and comparisons across studies.

We examined the distribution of a wide array of biological features including ER, progesterone receptor (PR), HER2, p53, IHC- and RNA-based subtypes, risk of recurrence multigene (ROR-P) scores, and multi-gene immune scores among premenopausal women <50 years of age according to parity and recency of pregnancy using data from a large population-based study. Based on prior literature, we used 10 years as the cutoff for our recently postpartum group (3, 13, 25), and we conducted sensitivity analyses and explored smaller windows of time since last childbirth.

Study population

The Carolina Breast Cancer Study (CBCS) is a population-based study of breast cancer in North Carolina (phase I: 1993–1996; phase II: 1996–2001; and phase III: 2008–2013; N = 4,806); details of the study have been described previously (26, 27). Briefly, the primary study enrolled women age 20 to 74 years diagnosed with first primary invasive breast cancer using rapid case ascertainment, with randomized recruitment to oversample Black and younger (age <50 years) women (28). This analysis examined tumor characteristics of premenopausal women under 50 years of age, who contributed tumor tissue (N = 2,081; Supplementary Fig. S1). Written informed consent was obtained from each participant prior to data collection, and the study was conducted in accordance with U.S. Common Rule. The study was approved by the Office of Human Research Ethics/Institutional Review Board at the University of North Carolina at Chapel Hill.

In-person interviews were conducted by trained nurses to collect medical history including detailed information on pregnancy history. Date of breast cancer diagnosis was collected using medical record abstraction. Time since last full-term birth (≥7 months of pregnancy or live birth) was calculated by subtracting date of last full-term birth from date of breast cancer diagnosis. Cases with missing date of last full-term birth were excluded (N = 2). Breast cancers diagnosed during pregnancy were excluded (N = 15). Women diagnosed within 10 years of their last childbirth were included in the “recent” group (N = 618). Women diagnosed more than 10 years after their last childbirth were assigned to the “remote” group (N = 1,086). Women who never had a full-term birth prior to their diagnosis were assigned to the “nulliparous” group (N = 360).

Clinical tumor markers and IHC-based subtyping

Tumor size, grade, node status, and stage were abstracted from medical records and pathology reports. The multiple imputation plus outcome approach was used to impute missing tumor grade values for CBCS phase II participants (n = 578; ref. 29). Estrogen receptor (ER), progesterone receptor (PR), and HER2 status were abstracted from medical records or determined from formalin-fixed, paraffin-embedded (FFPE) tumor tissue blocks, which were previously sectioned and stained for a panel of IHC markers at the Translational Pathology Laboratory at the University of North Carolina, Chapel Hill; details have been described previously (30). Briefly, ER and PR were considered positive if the percentage of positive cells was ≥10%, based on previous work showing that 10% positivity was most closely aligned with RNA-based subtypes (30), and HER2 positivity was defined as IHC 3+ (31). Details of IHC-based breast cancer subtyping have been published previously (26, 32). Briefly, IHC-based subtype was defined as luminal A-like (ER+ or PR+ and HER2), luminal B-like (ER+ or PR+ and HER2+), HER2-type (ER and PR and HER2+), or triple-negative (ER and PR and HER2).

Tumor-infiltrating lymphocytes (TIL) scores were calculated by adapting a digital algorithm similar to that previously described by Sandhu and colleagues (33) and optimized based on standardized international guidelines on TIL assessment in breast cancer by the International TILs Working Group (34). Briefly, composition algorithms were trained on several annotated representative hematoxylin and eosin (H&E)-stained tumor slides for each tissue feature (immune, epithelial, stromal, adipose) with accuracy optimized to match visual annotation by a trained pathologist's assistant. Percentage area and nuclei per unit area (nuclear density) were calculated for each region.

Genomic assessment

Details of RNA isolation and quantification have been described previously (35). Briefly, RNA was isolated and quantified using Qiagen FFPE RNeasy kit and NanoString assay, respectively (36). Data that passed the NanoString nSolver software quality control were normalized using methods previously described by Bhattacharya and Hamilton and colleagues (37), including reference gene normalization, upper-quartile normalization (38), and Remove Unwanted Variation (RUV) using the RUVg function from the RUVSeq Bioconductor package (39). Only FFPE tumor blocks with adequate tumor cellularity by manual review were included in RNA analyses. Relative to participants without RNA data, those with RNA data had larger tumors, higher tumor stage, and higher grade and were more likely to be lymph node positive, but age at diagnosis or time since last childbirth were not significantly different.

P53 status was determined using a previously validated TP53-dependent signature (40). Tumors were classified as p53 mutant-like versus wild-type–like based on a similarity-to-centroid approach (Pearson coefficient) using distance-weighted discrimination (41). RNA-based intrinsic subtype was determined using the PAM50 gene signature to classify tumors as luminal A, luminal B, HER2-enriched, basal-like, and normal-like, as previously described (35). Normal-like samples were excluded because this subtype is believed to arise due to insufficient tumor cellularity. The PAM50 predictor was used to calculate the risk of recurrence (ROR) score, which incorporates subtype and additional weighting by the proliferation signature (ROR-P) (35). To identify the most aggressive tumors, ROR-P was categorized as low/medium versus high.

We curated a 48-gene panel of immune markers that is representative of at least 10 individual immune cell types based on work from Bindea (42) and Danaher and colleagues (43). As described in Hamilton and colleagues (in preparation), cell type scores for 10 immune cell types, including B Cells, T cells, Th cells, T regulatory cells (Treg), T follicular helper cells (Tfh), CD8 T cells, Neutrophils, Eosinophils, natural killer (NK) cells, macrophages, and the immune checkpoint marker, PDL1, were calculated by computing the median of the log2-transformed expression from each cell's marker genes. For CD8 T cells specifically, we elected to include genes that were both specific and sensitive. These less specific markers, such as GZMM and PRF1, have been shown to be expressed on both NK cells and CD8 T cells; thus, we refer to these cytotoxic cells as CD8 T cells/NK cells in downstream analysis. In addition to cell type scores, overall adaptive and innate immunity scores were calculated, where the median log2-transformed expression for all B-cell, T-cell, CD8 T-cell, Th cell, Treg, and Tfh markers were computed for the adaptive scores, and the median of all eosinophil, neutrophil, NK cell, and macrophage markers were calculated for the innate scores.

Covariate assessment

Race was determined by self-report and categorized as non-Black or Black. Less than 2% of non-Black participants self-identified as multiracial, Hispanic, or other race/ethnicities. Age at diagnosis was obtained from the baseline survey and used as a continuous variable in models. Age at last full-term pregnancy was obtained from baseline survey and categorized as <30 years and ≥ 30years. Information on parity was obtained from baseline survey and categories as nulliparous, or 1, 2, and ≥3 full-term births.

Statistical analyses

Descriptive statistics for patient sociodemographic and tumor characteristics of recently postpartum women were compared with distally postpartum and nulliparous women using χ2 tests (Fisher exact test when cell count <5). Binary logistic regression models were used to compute prevalence odds ratios (POR) and 95% confidence intervals (95% CI) as the measure of association between tumor characteristics and recency of pregnancy. The following tumor characteristics were studied in association with recency of pregnancy: tumor grade (low-intermediate, high), stage of disease (I/II, III/IV), lymph node status (positive, negative), tumor size (≤2 cm, >2 cm), ER (negative, positive), PR status (negative, positive), HER2 status (negative, positive), IHC-based subtype (luminal A, luminal B, HER2-type, triple negative), p53 status (wild-type, mutant), PAM50 intrinsic breast cancer subtype (luminal A, luminal B, HER2-enriched, basal-like), and ROR-P score (low, medium, high). Models were adjusted for study phase (I, II, III), age at diagnosis (continuous), and race (Black, non-Black). Sensitivity analyses included using smaller intervals of time since last full-term birth (≤ 2, >2–5, >5–10 years postpartum) and stratifying by age at last full-term pregnancy (<30 years, ≥30 years). We performed multiple sensitivity analyses, including adjusting for body mass index (BMI) at diagnosis (<25, 25-<30, ≥30), parity (1, 2, ≥3), lactation history (ever, never), and oral contraceptive use (never, current, former). All analyses were conducted in SAS version 9.4 (SAS Institute, Cary, NC). P values were two-sided with an α of 0.05 for statistical significance.

Data availability

For participant confidentiality and due to ethical restrictions, data are available upon request and are subject to data use agreements and other stipulations. Permission to access data from the Carolina Breast Cancer Study may be obtained online (https://unclineberger.org/cbcs/) or by contacting the authors.

Sociodemographic characteristics

Among premenopausal breast cancer cases <50 years at diagnosis in the CBCS (phase I–III, 1993–2013), Table 1 presents the distribution of age at diagnosis, age at last full-term pregnancy, race, parity, and study phase for recently postpartum (diagnosed ≤10 years postpartum), remotely postpartum (diagnosed >10 years postpartum), and nulliparous women diagnosed with breast cancer. Recently postpartum women had a significantly younger age at diagnosis (median = 39 years) compared with remotely postpartum (median = 45 years) and nulliparous (median = 42 years). Compared with remotely postpartum, recently postpartum also had an older age at last full-term pregnancy (median = 33 years vs. 26 years) and a higher frequency of Black race. There was no significant difference in multiparity or study phase between groups.

Table 1.

Distribution of select characteristics by recency of pregnancy among premenopausal women less than 50 years of age in the CBCS, 1999 to 2013 (N = 2,064).

Nulliparous, N = 360Remote, N = 1,086Recent, N = 618P value
Age at diagnosis (years)    <0.0001 
 23–30 23 (6.4) 2 (0.2) 38 (6.2)  
 30–39 104 (28.9) 135 (12.4) 315 (51.0)  
 40–49 233 (64.7) 949 (87.4) 265 (42.9)  
 Median (range) 42 (23, 49) 45 (29, 49) 39 (23, 49)  
Age at last full-term pregnancy (years)    <0.0001 
 13–30 — 755 (69.5) 153 (24.8)  
 30–39 — 331 (30.5) 429 (69.4)  
 40–46 — 0 (0.0) 36 (5.8)  
 Median (range) — 26 (13, 38) 33 (17, 46)  
Race    0.003 
 Black 145 (40.3) 535 (49.3) 267 (43.2)  
 Non-Black 215 (59.7) 551 (50.7) 351 (56.8)  
Parity    0.41 
 1 — 287 (26.4) 157 (25.4)  
 2 — 469 (43.2) 254 (41.1)  
 3–8 — 330 (30.4) 207 (33.5)  
Study phase    0.40 
 1 75 (20.8) 234 (21.6) 115 (18.6)  
 2 66 (18.3) 221 (20.4) 115 (18.6)  
 3 219 (60.8) 631 (58.1) 388 (62.8)  
Nulliparous, N = 360Remote, N = 1,086Recent, N = 618P value
Age at diagnosis (years)    <0.0001 
 23–30 23 (6.4) 2 (0.2) 38 (6.2)  
 30–39 104 (28.9) 135 (12.4) 315 (51.0)  
 40–49 233 (64.7) 949 (87.4) 265 (42.9)  
 Median (range) 42 (23, 49) 45 (29, 49) 39 (23, 49)  
Age at last full-term pregnancy (years)    <0.0001 
 13–30 — 755 (69.5) 153 (24.8)  
 30–39 — 331 (30.5) 429 (69.4)  
 40–46 — 0 (0.0) 36 (5.8)  
 Median (range) — 26 (13, 38) 33 (17, 46)  
Race    0.003 
 Black 145 (40.3) 535 (49.3) 267 (43.2)  
 Non-Black 215 (59.7) 551 (50.7) 351 (56.8)  
Parity    0.41 
 1 — 287 (26.4) 157 (25.4)  
 2 — 469 (43.2) 254 (41.1)  
 3–8 — 330 (30.4) 207 (33.5)  
Study phase    0.40 
 1 75 (20.8) 234 (21.6) 115 (18.6)  
 2 66 (18.3) 221 (20.4) 115 (18.6)  
 3 219 (60.8) 631 (58.1) 388 (62.8)  

Note: Nulliparous, no full-term birth prior to diagnosis. P values generated by χ2 test, except when expected cell count <5, they were calculated by Fisher exact test. Missing values were excluded from percentage calculations. Percentages may not add up to 100 due to rounding.

Clinical and IHC-based tumor features

Although differences for several factors were observed when comparing recently postpartum with nulliparous women, there were few differences between recently and distally postpartum women (Table 2). Compared with nulliparous women, tumors among recently postpartum were more likely to be lymph node–positive, ER/PR-negative, and IHC-based triple negative subtype, after adjusting for age at diagnosis, race, and study phase. Although not statistically significant, recently postpartum women tended to be diagnosed at a later stage and were more frequently IHC-based HER2-type subtype compared with nulliparous women. Sensitivity analysis considering smaller windows of time since last childbirth showed strongest associations with respect to lymph node positivity and later stage for those 0 to 2 years and 2.1 to 5 years postpartum versus nulliparous women (Supplementary Tables S1 and S2). No substantial difference in grade, size, and HER2 status were observed between the recent and nulliparous groups.

Table 2.

Association between recent pregnancy (<10 years) and clinical and IHC-based tumor characteristics among premenopausal women less than 50 years of age in the CBCS, 1999 to 2013 (N = 2,064).

Nulliparous, N = 360Remote, N = 1,086Recent, N = 618Adjusted POR (95% CI)a
n%bn%bn%bReferent = remoteReferent = nulliparous
Grade 
 Low-Intermediate 193 53.6 561 51.7 290 46.9 Ref. Ref. 
 High 167 46.4 525 48.3 328 53.1 0.94 (0.73–1.20) 1.13 (0.86–1.48) 
 Missing — — —   
Stage 
 I–II 297 85.3 859 81.4 490 81.3 Ref. Ref. 
 III–IV 51 14.7 196 18.6 113 18.7 0.88 (0.64–1.21) 1.28 (0.88–1.85) 
 Missing 12 — 31 — 15 —   
Node 
 Negative 228 64.4 613 57.1 311 50.9 Ref. Ref. 
 Positive 126 35.6 460 42.9 300 49.1 1.13 (0.89–1.45) 1.66 (1.26–2.19) 
 Missing — 13 — —   
Size 
 ≤2 cm 155 45.1 466 44.4 281 46.8 Ref. Ref. 
 >2 cm 189 54.9 584 55.6 320 53.2 0.67 (0.52–0.86) 0.81 (0.61–1.07) 
 Missing 16 — 36 — 17 —   
ER status 
 Positive 231 67.0 620 59.3 352 58.6 Ref. Ref. 
 Negative 114 33.0 426 40.7 249 41.4 0.95 (0.73–1.23) 1.37 (1.02–1.83) 
 Missing 15 — 40 — 17 —   
PR status 
 Positive 216 62.6 568 54.5 299 50.3 Ref. Ref. 
 Negative 129 37.4 474 45.5 296 49.8 0.98 (0.76–1.26) 1.48 (1.11–1.97) 
 Missing 15 — 44 — 23 —   
HER2 status 
 Negative 253 76.9 820 83.1 449 79.2 Ref. Ref. 
 Positive 76 23.1 167 16.9 118 20.8 1.20 (0.87–1.66) 0.87 (0.62–1.21) 
 Missing 31 — 99 — 51 —   
IHC-based subtype 
 Luminal A 114 45.6 319 41.4 160 36.2 Ref. Ref. 
 Luminal B 80 32.0 177 23.0 122 27.6 1.06 (0.73–1.54) 0.98 (0.66–1.46) 
 HER2-type 14 5.6 58 7.5 41 9.3 1.16 (0.67–2.01) 1.86 (0.95–3.64) 
 Triple negative 42 16.8 216 28.1 119 26.9 0.75 (0.51–1.08) 1.57 (1.00–2.47) 
 Missing 110 — 316 — 176 —   
Nulliparous, N = 360Remote, N = 1,086Recent, N = 618Adjusted POR (95% CI)a
n%bn%bn%bReferent = remoteReferent = nulliparous
Grade 
 Low-Intermediate 193 53.6 561 51.7 290 46.9 Ref. Ref. 
 High 167 46.4 525 48.3 328 53.1 0.94 (0.73–1.20) 1.13 (0.86–1.48) 
 Missing — — —   
Stage 
 I–II 297 85.3 859 81.4 490 81.3 Ref. Ref. 
 III–IV 51 14.7 196 18.6 113 18.7 0.88 (0.64–1.21) 1.28 (0.88–1.85) 
 Missing 12 — 31 — 15 —   
Node 
 Negative 228 64.4 613 57.1 311 50.9 Ref. Ref. 
 Positive 126 35.6 460 42.9 300 49.1 1.13 (0.89–1.45) 1.66 (1.26–2.19) 
 Missing — 13 — —   
Size 
 ≤2 cm 155 45.1 466 44.4 281 46.8 Ref. Ref. 
 >2 cm 189 54.9 584 55.6 320 53.2 0.67 (0.52–0.86) 0.81 (0.61–1.07) 
 Missing 16 — 36 — 17 —   
ER status 
 Positive 231 67.0 620 59.3 352 58.6 Ref. Ref. 
 Negative 114 33.0 426 40.7 249 41.4 0.95 (0.73–1.23) 1.37 (1.02–1.83) 
 Missing 15 — 40 — 17 —   
PR status 
 Positive 216 62.6 568 54.5 299 50.3 Ref. Ref. 
 Negative 129 37.4 474 45.5 296 49.8 0.98 (0.76–1.26) 1.48 (1.11–1.97) 
 Missing 15 — 44 — 23 —   
HER2 status 
 Negative 253 76.9 820 83.1 449 79.2 Ref. Ref. 
 Positive 76 23.1 167 16.9 118 20.8 1.20 (0.87–1.66) 0.87 (0.62–1.21) 
 Missing 31 — 99 — 51 —   
IHC-based subtype 
 Luminal A 114 45.6 319 41.4 160 36.2 Ref. Ref. 
 Luminal B 80 32.0 177 23.0 122 27.6 1.06 (0.73–1.54) 0.98 (0.66–1.46) 
 HER2-type 14 5.6 58 7.5 41 9.3 1.16 (0.67–2.01) 1.86 (0.95–3.64) 
 Triple negative 42 16.8 216 28.1 119 26.9 0.75 (0.51–1.08) 1.57 (1.00–2.47) 
 Missing 110 — 316 — 176 —   

Note: Nulliparous, no full-term birth prior to diagnosis.

aAdjusted for age at diagnosis, race, and study phase.

bPercentages may not add up to 100 due to rounding.

Compared with remotely postpartum women, recently postpartum women did not have more aggressive clinical and molecular tumor characteristics (Table 2). After adjusting for age at diagnosis, race, and study phase, no significant differences in tumor grade, stage, lymph node status, and ER/PR/HER2 status were observed between recent and remote groups and recently postpartum women had significantly smaller tumors compared with remotely postpartum women.

RNA-based tumor characteristics

No significant differences in RNA-based intrinsic subtypes, TP53 status, and ROR-P scores were observed between the recent and nulliparous groups (Table 3). However, compared with remotely postpartum, recently postpartum women had significantly more frequently wild-type TP53 status, and more frequent non–basal-like subtype. No substantial or significant differences in ROR-P scores were observed between the recent and remote groups. Sensitivity analyses conducted with time since pregnancy as a continuous variable showed similar results. Adjusting for BMI at diagnosis, parity, lactation history, and oral contraceptive use did not substantially alter POR estimates. Sensitivity analysis stratifying on age at last full-term pregnancy or including smaller time intervals of recency did not alter the relationships between parity, recency, and RNA-based features (Supplementary Tables S3 and S4). Similarly, stratifying on prior parity status or restricting to only CBCS phase III participants did not change these results.

Table 3.

Association between recent pregnancy (<10 years) and RNA-based tumor characteristics among premenopausal women more than 50 years of age in the CBCS, 1999 to 2013 (N = 966).

Nulliparous, N = 154Remote, N = 520Recent, N = 289Adjusted POR (95% CI)a
n%bn%bn%bReferent = remoteReferent = nulliparous
RNA-based P53 status 
 Wildtype 74 48.0 243 46.7 137 47.4 Ref. Ref. 
 Mutant-like 80 52.0 277 53.3 152 52.6 0.53 (0.36–0.77) 0.80 (0.52–1.24) 
RNA-based subtype (PAM50) 
 Luminal A 69 45.7 203 40.6 108 38.7 Ref. Ref. 
 Luminal B 25 16.6 76 15.2 46 16.5 0.67 (0.39–1.16) 1.00 (0.54–1.84) 
 HER2-enriched 16 10.6 49 9.8 35 12.5 0.64 (0.34–1.23) 1.14 (0.56–2.34) 
 Basal-like 41 27.2 172 34.4 90 32.3 0.53 (0.33–0.84) 1.04 (0.61–1.79) 
 Unknown – 20 – 10 –   
ROR-P 
 Low 33 21.4 135 26.0 64 22.2 Ref. Ref. 
 Medium 71 46.1 231 44.4 124 42.9 0.85 (0.54–1.34) 0.89 (0.51–1.54) 
 High 50 32.5 154 29.6 101 35.0 0.67 (0.39–1.14) 0.74 (0.40–1.38) 
Nulliparous, N = 154Remote, N = 520Recent, N = 289Adjusted POR (95% CI)a
n%bn%bn%bReferent = remoteReferent = nulliparous
RNA-based P53 status 
 Wildtype 74 48.0 243 46.7 137 47.4 Ref. Ref. 
 Mutant-like 80 52.0 277 53.3 152 52.6 0.53 (0.36–0.77) 0.80 (0.52–1.24) 
RNA-based subtype (PAM50) 
 Luminal A 69 45.7 203 40.6 108 38.7 Ref. Ref. 
 Luminal B 25 16.6 76 15.2 46 16.5 0.67 (0.39–1.16) 1.00 (0.54–1.84) 
 HER2-enriched 16 10.6 49 9.8 35 12.5 0.64 (0.34–1.23) 1.14 (0.56–2.34) 
 Basal-like 41 27.2 172 34.4 90 32.3 0.53 (0.33–0.84) 1.04 (0.61–1.79) 
 Unknown – 20 – 10 –   
ROR-P 
 Low 33 21.4 135 26.0 64 22.2 Ref. Ref. 
 Medium 71 46.1 231 44.4 124 42.9 0.85 (0.54–1.34) 0.89 (0.51–1.54) 
 High 50 32.5 154 29.6 101 35.0 0.67 (0.39–1.14) 0.74 (0.40–1.38) 

Note: Nulliparous, no full-term birth prior to diagnosis.

aAdjusted for age at diagnosis, race, and study phase.

bPercentages may not add up to 100 due to rounding.

Tumor microenvironment and immune markers

Despite few changes in epithelial tumor features, we observed an enrichment for adaptive immune cells and higher immune-cell composition among recently postpartum women compared with nulliparous women (Fig. 1A). After adjusting for intrinsic subtype, race, age, and study phase, we saw an enrichment for overall adaptive immunity and adaptive immune cells, such as T cells, B cells, CD8 T cells, activated CD8 T cells/NK cells, and Tfh cells in recent versus nulliparous women (Fig. 1A). Consistent with these findings, tumors from recently and remotely postpartum participants had higher immune-cell composition than tumors from nulliparous participants (Fig. 1B). When comparing remote versus nulliparous women, CD8 T cells were also significantly enriched in remote versus nulliparous women (Fig. 1C); however, many fewer immune cell scores were significant. Nonetheless, no significant differences in immune cell scores were observed between recently versus remotely postpartum women (Fig. 1D). Box plots for signatures that were significant in Fig. 1A showed that the level of each immune cell score was elevated in both recently and remotely postpartum compared with nulliparous, but were modestly attenuated in remotely postpartum (Fig. 1E).

Figure 1.

Tumor immune microenvironment and adaptive immune cell types by recency of pregnancy. A, Volcano plot of immune cell score, adjusted for intrinsic subtype, race, age, and study phase, showing comparison by Recent versus Nulliparous. B, Boxplot of log2 transformed immune-cell composition scores, or TIL scores, from H&E analysis by Recent, Remote, and Nulliparous groups. C and D, Volcano plots of immune cell score, adjusted for intrinsic subtype, race, age, and study phase, showing comparisons by Remote versus Nulliparous, and Recent versus Remote, respectively. E, Boxplot of immune-cell composition scores by Recent, Remote, and Nulliparous groups (P = Kruskal–Wallis). FC, fold change.

Figure 1.

Tumor immune microenvironment and adaptive immune cell types by recency of pregnancy. A, Volcano plot of immune cell score, adjusted for intrinsic subtype, race, age, and study phase, showing comparison by Recent versus Nulliparous. B, Boxplot of log2 transformed immune-cell composition scores, or TIL scores, from H&E analysis by Recent, Remote, and Nulliparous groups. C and D, Volcano plots of immune cell score, adjusted for intrinsic subtype, race, age, and study phase, showing comparisons by Remote versus Nulliparous, and Recent versus Remote, respectively. E, Boxplot of immune-cell composition scores by Recent, Remote, and Nulliparous groups (P = Kruskal–Wallis). FC, fold change.

Close modal

Among premenopausal women, we observed few differences between the tumors of recently and remotely postpartum women in the CBCS. While parity status seems to be associated with breast cancer clinical and molecular tumor characteristics, recent pregnancy among parous women had weak or null associations with clinical and molecular characteristics, except when considering the most recent postpartum (≤5 years) and lymph node status and stage. However, consistent with the postpregnancy wound healing microenvironment hypothesis (13), we observed an enrichment for adaptive immune cells and higher immune-cell composition among recently postpartum women compared with nulliparous women. CD8 immune changes appeared to persist long term, but other immune response changes were attenuated in remotely postpartum versus nulliparous women.

Three previous studies have reported significant differences in some key tumor characteristics between recently postpartum compared with nulliparous women (3, 21, 22). Consistent with our findings, Goddard and colleagues observed an increased lymph node involvement among PPBC women compared with nulliparous women (3). Pilewskie and colleagues found breast cancers diagnosed within 2 years of pregnancy were more likely to be lymph node–positive, hormone receptor—negative, and triple negative compared with nulliparous women, after conditioning multivariate models on categorical age at diagnosis (21). Similarly, Nagatsuma and colleagues reported ER/PR-negative and triple negative tumors to be more common among recently postpartum (≤2 years before diagnosis) women compared with nulliparous women, in a Japanese hospital-based study (22). While these latter studies used a narrower postpartum window than we did to define recency, we observed that several of the differences between recently postpartum and nulliparous women may continue to persist for many years postpartum similar to Goddard and colleagues (3).

Some previous literature seems to contradict the findings of our research. Specifically, we found ER/PR-negative and IHC-based triple negative subtype tumors to be more common among recently postpartum (≤10 years postpartum) compared with nulliparous women, but two other groups found no difference in ER/PR status or IHC-based subtype between recently postpartum (<5 years postpartum) compared with nulliparous young women (4, 24). However, compared with Callihan and colleagues we had several underlying methodologic differences. For example, we adjusted for age, while Callihan and colleagues did not (4). The population studied in Callihan and colleagues has been expanded and the analysis has been updated by Goddard and colleagues in a recent publication corroborating our findings (3). The study by Collins and colleagues also found no significant differences in ER/PR status and IHC-based subtype distribution among women diagnosed within 5 years of their last pregnancy compared with nulliparous women. However, patient ER status definition varied within their study population as their enrollment period began in 2006 and lasted beyond 2010, when ER positivity classification changed from ≥10% to ≥1% of tumor cells; thus, women who would have been classified as ER-negative before 2010 were classified as ER-positive if enrolled after 2010, leading to a slight reduction in ER-negative and triple negative cases (24). Additionally, the Collins and colleagues study population largely consisted of white women and the distribution of ER status has been shown to vary by race (24). An advantage of the current analysis is that it included a significant representation of Black women, a group that has been underrepresented in previous analyses. Some findings of Collins and colleagues were consistent with ours, notably, neither study observed significant difference in tumor grade or stage comparing recently postpartum women with nulliparous women (24).

Only two previous studies have reported on the distribution of tumor characteristics among women >10 years postpartum (remotely postpartum; refs. 3, 4). Similar to our results, both of these studies have reported no significant difference in breast cancer grade, stage, nodal status, ER/PR status, HER2 status, and IHC-based subtypes between the recently and remotely postpartum groups.

To our knowledge, no other studies have examined RNA-based p53 signature, RNA-based intrinsic subtype, or ROR-P scores with respect to recently postpartum and nulliparous premenopausal women at breast cancer diagnosis. However, one previous study did evaluate p53 status by IHC. Our p53 findings corroborate those reported by Pilewskie and colleagues, who found no significant difference in IHC-based p53 status between recently postpartum and nulliparous women at breast cancer diagnosis (21).

Our study is unique among the aforementioned studies in that we examined the immune microenvironment of tumors diagnosed according to time since last childbirth (recently vs. remotely postpartum women). Our results showed an enrichment in adaptive immune cells and a higher immune-cell composition among recently postpartum women (≤10 years postpartum), which is attenuated among remotely postpartum women, compared with nulliparous women. Our findings are consistent with previous literature, which has noted an upregulation of immune-related genes in tumor-associated stroma of animal models (44, 45) or pregnant women (46). Within normal breast tissue, this enrichment of inflammatory and immune response genes may persist many years after the completion of a pregnancy (47, 48). In animal models, tumors during pregnancy or within the postpartum had an enrichment of immune responsiveness genes and immune-cell infiltration (44, 45). Harvell and colleagues noted that compared with nulliparous or women diagnosed >5 years postpartum, tumors diagnosed during pregnancy overexpressed immune response genes (46). Lastly, in normal breast tissue, genes in the immune response and inflammation pathways have been observed to be upregulated among parous compared with nulliparous women (47, 48).

Previously in the CBCS population, we observed that breast cancer–specific and overall survival were worse in recently postpartum women compared with both remotely postpartum and nulliparous women (49). Several previous studies have also reported worse overall and disease-free survival among recently postpartum women (3–9). Therefore, we hypothesized that tumor characteristics might explain the poor survival observed within this population. However, defining the ideal comparison group for recently postpartum women is complex because age, menopausal status and parity are strongly associated with tumor characteristics. Addressing this complexity was a strength of our analyses as we showed comparisons to two different populations (nulliparous women and remotely postpartum parous women), restricted our analysis to premenopausal women less <50 years of age, and controlled for the variation in age between comparison groups. To fully understand how recency of pregnancy affects outcomes, analyses should examine breast cancer recurrence or disease-specific mortality and incorporate both molecular data and treatment information, but our study population (CBCS phase III) has too few recurrences among recently postpartum women (n = 56) to address these subgroups.

Results suggesting a distinct immune microenvironment in recently postpartum women is an intriguing finding. It is increasingly recognized that immune microenvironment plays a key role in response to treatment, and as the science of immune response and chemotherapy response is better defined, there may be implications for understanding PPBC outcomes. Future studies should examine the interplay between tumor microenvironment, postdiagnostic treatment, and breast cancer recurrence/progression among recently postpartum women.

S.N. Vohra reports grants from NIH and Susan G. Komen for the Cure during the conduct of the study. K.E. Reeder-Hayes reports grants from Pfizer Foundation outside the submitted work. No disclosures were reported by the other authors.

S.N. Vohra: Conceptualization, data curation, formal analysis, investigation, visualization, methodology, writing–original draft, writing–review and editing. A. Walens: Formal analysis, visualization, writing–original draft, writing–review and editing. A.M. Hamilton: Data curation, methodology, writing–original draft, writing–review and editing. M.E. Sherman: Conceptualization, writing–review and editing. P. Schedin: Conceptualization, writing–review and editing. H.B. Nichols: Conceptualization, writing–review and editing. K.E. Reeder-Hayes: Conceptualization, writing–review and editing. A.F. Olshan: Conceptualization, funding acquisition, writing–review and editing. M.I. Love: Conceptualization, methodology, writing–review and editing. M.A. Troester: Conceptualization, funding acquisition, investigation, methodology, writing–original draft, project administration, writing–review and editing.

S.N. Vohra was supported by the University Cancer Research Fund of North Carolina, the Komen Graduate Training in Disparities Research (grant no. GTDR16381071), the Doctoral Degree Advancement Award by the University of North Carolina (UNC) – Initiative for Minority Excellence, and the UNC Cancer Control Education Program (T32CA057726). M.A. Troester was supported by the NCI of the NIH (P01CA151135) and the NCI Specialized Program of Research Excellence (SPORE) in Breast Cancer (NIH/NCI P50-CA58223). The Carolina Breast Cancer Study was funded by the University Cancer Research Fund of North Carolina and Susan G. Komen for the Cure (OGUNC1202).

This research was supported by a grant from UNC Lineberger Comprehensive Cancer Center, which is funded by the University Cancer Research Fund of North Carolina, the Susan G. Komen Foundation (OGUNC1202), the Komen Graduate Training in Disparities Research Grant (GTDR16381071), the NCI of the NIH (P01CA151135), and the NCI Specialized Program of Research Excellence (SPORE) in Breast Cancer (NIH/NCI P50-CA58223). This research recruited participants and/or obtained data with the assistance of Rapid Case Ascertainment (RCA), a collaboration between the North Carolina Central Cancer Registry and UNC Lineberger. RCA is supported by a grant from the NCI of the NIH (P30CA016086). We are grateful to CBCS participants and study staff. We also acknowledge the late Robert C. Millikan, founder of the CBCS phase III.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Bleyer
A
,
Barr
R
,
Hayes-Lattin
B
,
Thomas
D
,
Ellis
C
,
Anderson
B
, et al
.
The distinctive biology of cancer in adolescents and young adults
.
Nat Rev Cancer
2008
;
8
:
288
98
.
2.
Cardonick
E
,
Dougherty
R
,
Grana
G
,
Gilmandyar
D
,
Ghaffar
S
,
Usmani
A
.
Breast cancer during pregnancy: maternal and fetal outcomes
.
Cancer J
2010
;
16
:
76
82
.
3.
Goddard
ET
,
Bassale
S
,
Schedin
T
,
Jindal
S
,
Johnston
J
,
Cabral
E
, et al
.
Association between postpartum breast cancer diagnosis and metastasis and the clinical features underlying risk
.
JAMA Network Open
2019
;
2
:
e186997
.
4.
Callihan
EB
,
Gao
D
,
Jindal
S
,
Lyons
TR
,
Manthey
E
,
Edgerton
S
, et al
.
Postpartum diagnosis demonstrates a high risk for metastasis and merits an expanded definition of pregnancy-associated breast cancer
.
Breast Cancer Res Treat
2013
;
138
:
549
59
.
5.
Dimitrakakis
C
,
Zagouri
F
,
Tsigginou
A
,
Marinopoulos
S
,
Sergentanis
TN
,
Keramopoulos
A
, et al
.
Does pregnancy-associated breast cancer imply a worse prognosis? A matched case-case study
.
Breast Care (Basel)
2013
;
8
:
203
7
.
6.
Ali
SA
,
Gupta
S
,
Sehgal
R
,
Vogel
V
.
Survival outcomes in pregnancy associated breast cancer: a retrospective case control study
.
Breast J
2012
;
18
:
139
44
.
7.
Azim Jr HA,
Botteri
E
,
Renne
G
,
Dell'orto
P
,
Rotmensz
N
,
Gentilini
O
, et al
.
The biological features and prognosis of breast cancer diagnosed during pregnancy: a case-control study
.
Acta Oncol
2012
;
51
:
653
61
.
8.
Madaras
L
,
Kovács
KA
,
Szász
AM
,
Kenessey
I
,
Tőkés
AM
,
Székely
B
, et al
.
Clinicopathological features and prognosis of pregnancy associated breast cancer - a matched case control study
.
Pathol Oncol Res
2014
;
20
:
581
90
.
9.
Amant
F
,
Minckwitz
GV
,
Han
SN
,
Bontenbal
M
,
Ring
AE
,
Giermek
J
, et al
.
Prognosis of women with primary breast cancer diagnosed during pregnancy: results from an international collaborative study
.
J Clin Oncol
2013
;
31
:
2532
9
.
10.
Pathak
DR
.
Dual effect of first full term pregnancy on breast cancer risk: empirical evidence and postulated underlying biology
.
Cancer Causes Control
2002
;
13
:
295
8
.
11.
Henderson
BE
,
Ross
R
,
Bernstein
L
.
Estrogens as a cause of human cancer: the richard and hinda rosenthal foundation award lecture
.
Cancer Res
1988
;
48
:
246
53
.
12.
Tworoger
SS
,
Hankinson
SE
.
Prolactin and breast cancer risk
.
Cancer Lett
2006
;
243
:
160
9
.
13.
Schedin
P
.
Pregnancy-associated breast cancer and metastasis
.
Nat Rev Cancer
2006
;
6
:
281
91
.
14.
Lyons
TR
,
O'Brien
J
,
Borges
VF
,
Conklin
MW
,
Keely
PJ
,
Eliceiri
KW
, et al
.
Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2
.
Nat Med
2011
;
17
:
1109
15
.
15.
Watson
CJ
.
Involution: apoptosis and tissue remodelling that convert the mammary gland from milk factory to a quiescent organ
.
Breast Cancer Res
2006
;
8
:
203
.
16.
Schedin
P
,
O'Brien
J
,
Rudolph
M
,
Stein
T
,
Borges
V
.
Microenvironment of the involuting mammary gland mediates mammary cancer progression
.
J Mammary Gland Biol Neoplasia
2007
;
12
:
71
82
.
17.
Wallace
TR
,
Tarullo
SE
,
Crump
LS
,
Lyons
TR
.
Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms
.
Journal of Cancer Metastasis and Treatment
2019
;
5
:
9
.
18.
Lyons
TR
,
Borges
VF
,
Betts
CB
,
Guo
Q
,
Kapoor
P
,
Martinson
HA
, et al
.
Cyclooxygenase-2–dependent lymphangiogenesis promotes nodal metastasis of postpartum breast cancer
.
J Clin Invest
2014
;
124
:
3901
12
.
19.
Halaska
MJ
,
Pentheroudakis
G
,
Strnad
P
,
Stankusova
H
,
Robova
Chod J
, et al
.
Presentation, management and outcome of 32 patients with pregnancy-associated breast cancer: a matched controlled study
.
Breast J
2009
;
15
:
461
7
.
20.
Genin
AS
,
Lesieur
B
,
Gligorov
J
,
Antoine
M
,
Selleret
L
,
Rouzier
R
.
Pregnancy-associated breast cancers: Do they differ from other breast cancers in young women?
The Breast
2012
;
21
:
550
5
.
21.
Pilewskie
M
,
Gorodinsky
P
,
Fought
A
,
Hansen
N
,
Bethke
K
,
Jeruss
J
, et al
.
Association between recency of last pregnancy and biologic subtype of breast cancer
.
Ann Surg Oncol
2012
;
19
:
1167
73
.
22.
Nagatsuma
AK
,
Shimizu
C
,
Takahashi
F
,
Tsuda
H
,
Saji
S
,
Hojo
T
, et al
.
Impact of recent parity on histopathological tumor features and breast cancer outcome in premenopausal Japanese women
.
Breast Cancer Res Treat
2013
;
138
:
941
50
.
23.
Asztalos
S
,
Pham
TN
,
Gann
PH
,
Hayes
MK
,
Deaton
R
,
Wiley
EL
, et al
.
High incidence of triple negative breast cancers following pregnancy and an associated gene expression signature
.
Springerplus
2015
;
4
:
710
.
24.
Collins
LC
,
Gelber
S
,
Marotti
JD
,
White
S
,
Ruddy
K
,
Brachtel
EF
, et al
.
Molecular phenotype of breast cancer according to time since last pregnancy in a large cohort of young women
.
Oncologist
2015
;
20
:
713
8
.
25.
Amant
F
,
Lefrére
H
,
Borges
VF
,
Cardonick
E
,
Lambertini
M
,
Loibl
S
, et al
.
The definition of pregnancy-associated breast cancer is outdated and should no longer be used
.
Lancet Oncol
2021
;
22
:
753
4
.
26.
Millikan
RC
,
Newman
B
,
Tse
CK
,
Moorman
P
,
Conway
K
,
Dressler
LG
, et al
.
Epidemiology of basal-like breast cancer
.
Breast Cancer Res Treat
2008
;
109
:
123
39
.
27.
Hair
BY
,
Hayes
S
,
Tse
CK
,
Bell
MB
,
Olshan
AF
.
Racial differences in physical activity among breast cancer survivors: implications for breast cancer care
.
Cancer
2014
;
120
:
2174
82
.
28.
Moorman
PG
,
Newman
B
,
Millikan
RC
,
Tse
CK
,
Sandler
DP
.
Participation rates in a case-control study: the impact of age, race, and race of interviewer
.
Ann Epidemiol
1999
;
9
:
188
95
.
29.
Ali
AM
,
Dawson
SJ
,
Blows
FM
,
Provenzano
E
,
Ellis
IO
,
Baglietto
L
, et al
.
Comparison of methods for handling missing data on immunohistochemical markers in survival analysis of breast cancer
.
Br J Cancer
2011
;
104
:
693
9
.
30.
Allott
EH
,
Cohen
SM
,
Geradts
J
,
Sun
X
,
Khoury
T
,
Bshara
W
, et al
.
Performance of three-biomarker immunohistochemistry for intrinsic breast cancer subtyping in the AMBER consortium
.
Cancer Epidemiol Biomarkers Prev
2016
;
25
:
470
8
.
31.
Wolff
AC
,
Hammond
ME
,
Schwartz
JN
,
Hagerty
KL
,
Allred
DC
,
Cote
RJ
, et al
.
American society of clinical oncology/college of American pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer
.
Arch Pathol Lab Med
2007
;
131
:
18
43
.
32.
Carey
LA
,
Perou
CM
,
Livasy
CA
,
Dressler
LG
,
Cowan
D
,
Conway
K
, et al
. Race,
Breast cancer subtypes, and survival in the Carolina Breast Cancer Study
.
JAMA
2006
;
295
:
2492
502
.
33.
Sandhu
R
,
Chollet-Hinton
L
,
Kirk
EL
,
Midkiff
B
,
Troester
MA
.
Digital histologic analysis reveals morphometric patterns of age-related involution in breast epithelium and stroma
.
Hum Pathol
2016
;
48
:
60
8
.
34.
Salgado
R
,
Denkert
C
,
Demaria
S
,
Sirtaine
N
,
Klauschen
F
,
Pruneri
G
, et al
.
The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an international TILs working group 2014
.
Ann Oncol
2015
;
26
:
259
71
.
35.
Troester
MA
,
Sun
X
,
Allott
EH
,
Geradts
J
,
Cohen
SM
,
Tse
CK
, et al
.
Racial differences in PAM50 subtypes in the Carolina Breast Cancer Study
.
J Natl Cancer Inst
2018
;
110
:
176
82
.
36.
Geiss
GK
,
Bumgarner
RE
,
Birditt
B
,
Dahl
T
,
Dowidar
N
,
Dunaway
DL
, et al
.
Direct multiplexed measurement of gene expression with color-coded probe pairs
.
Nat Biotechnol
2008
;
26
:
317
25
.
37.
Bhattacharya
A
,
Hamilton
AM
,
Furberg
H
,
Pietzak
E
,
Purdue
MP
,
Troester
MA
, et al
.
An approach for normalization and quality control for NanoString RNA expression data
.
Brief Bioinform
2020
;
22
:
bbaa 163
.
38.
Bullard
JH
,
Purdom
E
,
Hansen
KD
.,
Dudoit S. Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments
.
BMC Bioinformatics
2010
;
11
:
94
.
39.
Risso
D
,
Ngai
J
,
Speed
TP
.,
Dudoit S. Normalization of RNA-seq data using factor analysis of control genes or samples
.
Nat Biotechnol
2014
;
32
:
896
902
.
40.
Troester
MA
,
Herschkowitz
JI
,
Oh
DS
,
He
X
,
Hoadley
KA
,
Barbier
CS
, et al
.
Gene expression patterns associated with p53 status in breast cancer
.
BMC Cancer
2006
;
6
:
276
.
41.
Marron
JS
,
Todd
MJ
,
Ahn
J
.,
Distance-Weighted Discrimination
.
J Am Statist Assoc
2007
;
102
:
1267
71
.
42.
Bindea
G
,
Mlecnik
B
,
Tosolini
M
,
Kirilovsky
A
,
Waldner
M
,
Obenauf
AC
, et al
.
Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer
.
Immunity
2013
;
39
:
782
95
.
43.
Danaher
P
,
Warren
S
,
Dennis
L
,
D'Amico
L
,
White
A
,
Disis
ML
, et al
.
Gene expression markers of tumor infiltrating leukocytes
.
J Immunother Cancer
2017
;
5
:
18
.
44.
Fornetti
J
,
Martinson
HA
,
Betts
CB
,
Lyons
TR
,
Jindal
S
,
Guo
Q
, et al
.
Mammary gland involution as an immunotherapeutic target for postpartum breast cancer
.
J Mammary Gland Biol Neoplasia
2014
;
19
:
213
28
.
45.
Martinson
HA
,
Jindal
S
,
Durand-Rougely
C
,
Borges
VF
,
Schedin
P
.
Wound healing-like immune program facilitates postpartum mammary gland involution and tumor progression
.
Int J Cancer
2015
;
136
:
1803
13
.
46.
Harvell
DM
,
Kim
J
,
O'Brien
J
,
Tan
AC
,
Borges
VF
,
Schedin
P
, et al
.
Genomic signatures of pregnancy-associated breast cancer epithelia and stroma and their regulation by estrogens and progesterone
.
Horm Cancer
2013
;
4
:
140
53
.
47.
Rotunno
M
,
Sun
X
,
Figueroa
J
,
Sherman
ME
,
Garcia-Closas
M
,
Meltzer
P
, et al
.
Parity-related molecular signatures and breast cancer subtypes by estrogen receptor status
.
Breast Cancer Res
2014
;
16
:
R74
.
48.
Asztalos
S
,
Gann
PH
,
Hayes
MH
,
Nonn
L
,
Beam
CA
,
Dai
Y
, et al
.
Gene expression patterns in the human breast after pregnancy
.
Cancer Prev Res (Phila)
2010
;
3
:
301
11
.
49.
Sun
X
,
Nichols
HB
,
Tse
CK
,
Bell
MB
,
Robinson
WR
,
Sherman
ME
, et al
.
Association of parity and time since last birth with breast cancer prognosis by intrinsic subtype
.
Cancer Epidemiol Biomarkers Prev
2016
;
25
:
60
7
.

Supplementary data