Clinical and Sociodemographic Risk Factors Associated With the Development of Second Primary Cancers Among Postmenopausal Breast Cancer Survivors

DOI: https://doi.org/10.21203/rs.3.rs-1028396/v1

Abstract

Introduction: Advances in breast cancer (BC) diagnosis and treatment have increased the number of long-term survivors. Consequently, survivors of primary BC are at a greater risk of developing second primary cancers (SPCs). The risk factors for SPCs among BC survivors including sociodemographic, cancer treatment, comorbidities, and other medications have not been comprehensively examined. The purpose of this study is to assess the incidence and clinicopathologic factors associated with risk of SPCs.

Methods: We analyzed 170, 639 women with early-stage primary BC diagnosed between January 2000-December 2015 from the Medicare-linked Surveillance Epidemiology and End Results (SEER-Medicare) database. SPC was defined as any diagnosis of malignancy occurring within the study period and at least two months after primary BC diagnosis. Univariate analyses compared baseline characteristics between those who developed a SPC and those who did not. We evaluated the cause-specific hazard of developing a SPC in the presence of death as a competing risk.

Results: Of the study cohort, 20,838 (12%) of BC survivors developed a SPC and BC was the most common SPC type (32%). The median time to SPC was 42 months. Women who were white, older, and with fewer comorbidities were more likely to develop a SPC. While statins [hazard ratio (HR) 1.060 (1.016 - 1.106)] and anti-hypertensives [HR 1.517 (1.461 – 1.575)] increased the hazard of developing a SPC, aromatase inhibitor therapy [HR 0.588 (0.542 – 0.638)] and bisphosphonates [HR 0.897 (0.848 – 0.949)] were associated with a decreased hazard of developing any SPC, including non-breast SPCs.

Conclusion: Our study shows that specific clinical factors including type of cancer treatment, medications, and comorbidities are associated with increased risk for the development of SPCs among older BC survivors. These results can increase patient and clinician awareness, target cancer screening among BC survivors, as well as developing risk-adapted management strategies. 

Introduction

Breast cancer (BC) accounts for nearly one in three cancer cases diagnosed in women.1 Early detection as well as improved management have contributed to the increase in survival rates of BC patients. As a result, longer life expectancy is accompanied by increased risk of developing a second primary cancer (SPC), defined as malignant tumors with differing pathologic morphologies and are not metastases from the initial primary cancer.1

The incidence of SPCs among women with primary BC have been inconclusive, ranging from 4–17%, with varying follow up times.1 Though prior studies show that BC survivors are more susceptible to developing a SPC, it has been difficult to quantify that risk and identify specific factors that may predispose some BC survivors to developing SPCs more than others. The American Cancer Society recommends annual mammography screenings for the general population as well as specific guidelines for those with a family history of BC or women with genetic mutations. However, there is currently not enough evidence for specific BC screening recommendations based on having a personal history of BC. There are also no comprehensive screening guidelines for other types of cancer in these cancer survivors.

The purpose of this study is to (1) assess rate and types of SPCs among an extensive cohort of older BC survivors and (2) identify clinical and sociodemographic factors associated with the development of SPCs. Our study will aid in improving clinician and patient awareness of SPCs in BC survivors and can serve as an informative model for developing risk-adapted management strategies.

Materials And Methods

Data Sources

Study participants were selected from the Surveillance, Epidemiology and End Results registry linked to Medicare claims record (SEER-Medicare). We excluded individuals in healthcare maintenance organizations and those without Medicare Parts A and B insurance, due to incomplete claims which are needed to assess comorbidities and cancer treatment (e.g., surgery, chemotherapy, radiation therapy). We excluded individuals without Medicare Part D claims data which were needed to ascertain prescription medication use.

Study Population

We identified patients >66 years of age who were diagnosed with primary BC between January 1, 2000 and December 31, 2015, in the SEER-Medicare database. We only included patients with a pathologically confirmed diagnosis. Those with diagnoses made at autopsy and death certificate were excluded. We further limited our cohort to include only female patients, with early stage (0 to III) BC.

Sociodemographic variables were extracted from linked Medicare claims and included age at diagnosis, race/ethnicity, income quartile, county/residency type, marital status, first treatment regimen, comorbidities, and medications. Comorbidity burden was assessed using the recommended NCI comorbidity index with weights specific for breast cancer. BC treatment (surgery, chemotherapy, radiotherapy) was extracted using International Classification of Diseases, Ninth Revision (ICD-9) and Health Care Procedure Coding System (HCPCS) procedure codes. Patients were considered to use medications of interest if they had used the drug within the year prior to cancer diagnosis until 6 months after diagnosis. Similarly, patients were considered to have specific comorbidities if there was at least one inpatient or two outpatient Medicare claims code for the diagnosis within 1 year prior to cancer diagnosis up until 6 months after primary BC diagnosis. This study was deemed exempt by the Mount Sinai Institutional Review Board.

Study Outcome

The primary outcome was development of SPC. SPCs diagnosed within two months of the primary cancer were excluded to remove what were likely multiple primary tumors.

Statistical Analysis

We conducted chi square tests to evaluate differences in sociodemographic and clinical characteristics of patients with and without SPCs. To estimate the effects of covariates on the rate of SPC occurrence, we used a cause-specific hazard model in the presence of competing risk. The time to development of SPCs was defined as the time since the initial BC diagnosis, with a delayed entry of 2 months after the primary BC diagnosis, until SPC occurrence, end of follow-up, or death. The 2-month latency period is in line with SEER Solid Tumor Rules which aims to distinguish single from multiple primary tumors. Patients who experienced death as a competing risk and those who did not develop a SPC were censored. Three cause-specific models were developed: all SPCs, breast only as a SPC, and non-breast SPCs. The results are presented as hazard ratios (HR) with corresponding two-sided 95% confidence intervals (CI). In all models, Her2/neu data was excluded because it was not recorded by the SEER registry for all years evaluated in this study. All statistical analyses were performed using SAS (version 9.4), with the two-sided significance level set at p<0.05.

Results

Patient Characteristics

The final cohort of primary early-stage BC survivors included 170,639 women, with a mean age at diagnosis of 75 years. SPC development occurred in 12% (n=20838) of patients. The total study population consisted of 84% non-Hispanic white, 8% non-Hispanic Black, and 4% Hispanic women. Most of the population lived in urban counties (98%) and 45% were married. The distribution of primary BC stages was as follows: 17% stage 0, 46% stage I, 28% stage II, and 8% stage III. Sixty-eight percent of primary breast tumors were classified as ductal carcinoma. Most tumors were hormone receptor positive (79%), and only 9% were triple negative tumors. Surgery occurred in 94% of the population. Sixteen percent of the study population underwent chemotherapy, and 40% underwent radiotherapy. Most of the women had a Comorbidity Index <1 (74%). Antihypertensives and statins were the most prescribed medications used by 32% and 20% of the cohort, respectively.

Types of Second Primary Cancers

Of the 20,838 patients who developed a SPC, 32% had a second primary BC (Figure 2). Lung, colorectal, and urinary cancers comprised 14%, 11%, and 10% of SPC’s, respectively. Skin and gynecologic cancers accounted for 4% and 6% of SPCs, respectively. Gallbladder/pancreatic and hematologic cancers each occurred in 3% of the patient population. Other SPCs included liver, gastrointestinal, soft tissue and bone, endocrine, and brain, and collectively accounted for 14% of the SPCs. The median time between primary BC and SPC diagnosis was 42 months (IQR: 62).

Factors associated with the development of SPCs

Unadjusted analysis (Table 1) revealed SPC cases were more often diagnosed in white patients (12.5% vs. 11.8% among black, 10.8% among Hispanic and 9.9% among other races), somewhat older (70-74 years 13.5% vs 13.0% for 66-69 years), and those with fewer comorbidities (12.5% NCI comorbidity index <1 vs 11.9% for NCI comorbidity index 1-2 and 9.8% for NCI comorbidity index >2). Additionally, married BC survivors (12.7% vs 11.8%) and those in the highest income quartile (12.8 vs. 11.9%) were more likely due to develop SPCs. SPCs also occurred more often with stage 0 vs. stage I disease (15.1% vs. 12.4%) and those who underwent surgery (12.4% vs 9.8%) or radiation therapy (13.1% vs 11.6%). Chemotherapy was not significantly associated with development of SPC. Additionally, various medications, including statins, antihypertensives, bisphosphonates, SGLT2 inhibitors, and aromatase inhibitors, were significantly associated with SPC development (p<0.001, Table 2).

Table 1

Demographic and Clinical Characteristics associated with development of Second Primary Cancer

Patient Characteristics

(N, %)

Second Primary Cancer Development

N=20838

No Second Primary Cancer Development

N=149801

P-value

Age

     

66-69

5171 (13.0)

34634 (87.0)

<0.001

70-74

6130 (13.5)

39224 (86.5)

75-79

5147 (13.3)

33690 (86.8)

>80

4390 (9.4)

42263 (90.6)

Race/Ethnicity

     

White

17752 (12.5)

124813 (87.6)

<0.001

Black

1539 (11.8)

11545 (88.2)

Hispanic

766 (10.8)

6319 (89.2)

Other

781 (9.9)

7124 (90.1)

Geographic status

     

Urban

20444 (12.2)

146827 (87.8)

0.259

Rural

363 (11.6)

2778 (88.4)

Income quartile

     

First quartile

4990 (11.9)

37016 (88.1)

<0.001

Second quartile

5129 (12.0)

37515 (88.0)

Third quartile

5267 (12.3)

37381 (88.0)

Fourth quartile

5419 (12.8)

36980 (87.2)

Marital Status

     

Married

9668 (12.7)

66642 (87.3)

<0.001

Not married

11170 (11.8)

83159 (88.2)

Tumor Stage

     

Stage 0

4556 (15.1)

25601 (84.9)

<0.001

Stage I

9717 (12.4)

68494 (87.6)

Stage II

5117 (11.0)

41539 (89.0)

Stage III

30 (6.0)

469 (94.0)

Stage IIIA

720 (10.0)

6479 (90.0)

Stage IIIB

331 (8.2)

3728 (92)

Stage IIIIC

367 (9.5)

3491 (91.9)

Tumor Histology

     

Ductal

14061 (12.1)

102546 (87.9)

<0.001

Lobular

1947 (11.7)

14723 (88.3)

Other

4830 (12.9)

32532 (87.1)

Estrogen Receptor Status

     

ER+

14275 (11.4)

111440 (88.6)

0.004

ER-

2708 (12.0)

19822 (88.0)

Progesterone Receptor Status

     

PR+

12015 (11.3)

93917 (88.7)

0.07

PR-

4710 (11.7)

35604 (88.3)

Her2 Receptor Status

     

Her2+

278 (4.9)

5350 (95.1)

0.10

Her2-

2407 (5.5)

41621 (94.5)

Breast Subtype

     

Her2+/HR+

191 (4.8)

3775 (95.2)

0.37

Her2+/HR-

87 (5.3)

1561 (94.7)

Her2-/HR+

2156 (5.5)

37229 (94.5)

Triple Negative

250 (5.5)

4331 (94.5)

Surgery

     

Yes

19818 (12.4)

140397 (87.6)

<0.001

No

1020 (9.8)

9404 (90.2)

Chemotherapy

     

Yes

3511 (12.1)

25404 (87.9)

0.69

No

17327 (12.2)

124397 (87.8)

Radiotherapy

     

Yes

8987 (13.1)

59824 (86.9)

<0.001

No

11851 (11.6)

89977 (88.4)

NCI Comorbidity Index

     

<1

16884 (12.5)

117801 (87.5)

<0.001

1-2

2472 (11.9)

18366 (88.1)

>2

1482 (9.1)

13634 (90.2)

 

Table 2

Medications associated with the development of second primary cancers among primary breast cancer survivors

Medication

Second Primary Cancer Development, (N= 20, 838)

No Second Primary Cancer Development, (N =149, 801)

P-value

Statins, N (%)

4564 (22)

29349 (20)

<0.001

Antihypertensive, N (%)

7649 (37)

47988 (32)

<0.001

Bisphosphonates, N (%)

1432 (7)

9300 (6)

<0.001

Aromatase Inhibitor, N (%)

631 (3)

10523 (7)

<0.001

Metformin, N (%)

1239 (6)

7636 (5)

<0.001

Insulin, N (%)

584 (3)

3779 (3)

0.02

Thiazolidinediones, N (%)

228 (1)

1337 (1)

0.004

SGLT-2 Inhibitors, N (%)

<11 (0.01)

110 (0.07)

<0.001

DPP-4 Inhibitors, N (%)

269 (1.3)

1793 (1.2)

0.24

GLP-1 Agonists, N (%)

37 (0.18)

236 (0.16)

0.50

 

Table 3

Multivariable Cox proportional cause-specific hazard models to evaluate the effects of covariates on the rate of second primary cancer development among primary breast cancer survivors.

Patient Characteristic

All SPCs

HR (95% CI)

Breast Only SPCs*

HR (95% CI)

Non-Breast SPCs*

HR (95% CI)

Age (yrs) (REF= 66-69)

70 - 74

1.056 (1.017 - 1.096)*

0.962 (0.903 - 1.025)

1.106 (1.056 - 1.158)*

75 - 79

1.072 (1.031 - 1.115)*

0.915 (0.855 - 0.980)*

1.148 (1.094 - 1.205)*

≥ 80

0.956 (0.916 - 0.998)*

0.756 (0.701 - 0.816)*

1.032 (0.979 - 1.087)

Race/Ethnicity

     

White

REF

REF

REF

Black

0.962 (0.911 - 1.016)

1.106 (1.010 - 1.211)*

0.893 (0.834 - 0.955)*

Hispanic

0.831 (0.773 - 0.894)*

0.866 (0.764 - 0.982)*

0.797 (0.728 - 0.872)*

Other

0.794 (0.739 - 0.853)*

0.815 (0.720 - 0.922)*

0.768 (0.702 - 0.840)*

Marital Status

     

Married

0.991 (0.964 - 1.020)*

1.021 (0.971 - 1.073)

0.982 (0.949 - 1.017)

Income Quartile

     

First (Lowest)

REF

REF

REF

Second

1.032 (0.993 - 1.074)

1.052 (0.982 - 1.128)

1.025 (0.977 - 1.075)

Third

1.073 (1.032 - 1.117)*

1.114 (1.039 - 1.195)*

1.058 (1.008 - 1.110)*

Fourth (Highest)

1.112 (1.069 - 1.157)*

1.158 (1.080 - 1.242)*

1.095 (1.044 - 1.149)*

Geographic Residence (REF=urban)

Rural

0.931 (0.839 - 1.034)

0.987 (0.823 - 1.183)

0.906 (0.796 - 1.030)

Breast Cancer Stage

     

Stage I

REF

REF

REF

Stage 0

1.101 (1.058 - 1.146)*

1.384 (1.296 - 1.478)*

0.955 (0.907 - 1.004)*

Stage II

0.984 (0.949 - 1.020)

0.901 (0.843 - 0.964)*

1.013 (0.971 - 1.057)

Stage III

0.804 (0.561 - 1.152)

1.207 (0.698 - 2.088)

0.622 (0.386 - 1.003)*

Stage IIIA

0.989 (0.914 - 1.071)

0.927 (0.797 - 1.078)

1.000 (0.911 - 1.098)

Stage IIIB

1.071 (0.956 - 1.199)

1.181 (0.965 - 1.446)

1.007 (0.879 - 1.154)

Stage IIIC

1.139 (1.022 - 1.269)

1.325 (1.098 - 1.598)*

1.051 (0.920 - 1.200)

Tumor Histology

     

Ductal

REF

REF

REF

Lobular

0.996 (0.949 - 1.045)

1.191 (1.098 - 1.292)*

0.910 (0.857 - 0.966)*

Other

1.026 (0.992 - 1.061)

1.108 (1.046 - 1.173)*

0.992 (0.952 - 1.033)

ER Negative

1.118 (1.059 - 1.181)

1.366 (1.241 - 1.504)*

1.024 (0.959 - 1.094)

PR Negative

0.986 (0.944 - 1.030)

0.954 (0.880 - 1.034)

1.002 (0.951 - 1.056)

Comorbidity Index

     

0.00 - 1.00

REF

REF

REF

1.01 - 2.00

0.785 (0.742 - 0.830)

0.723 (0.653 - 0.800)

0.781 (0.730 - 0.836)

> 2.00

0.731 (0.685 - 0.781)

0.724 (0.642 - 0.816)

0.684 (0.633 - 0.741)

Surgery

0.812 (0.749 - 0.879)*

0.808 (0.722 - 0.903)*

0.786 (0.726 - 0.851)*

Radiation Therapy

1.000 (0.970 - 1.030)

0.881 (0.836 - 0.929)*

1.073 (1.035 - 1.113)*

Chemotherapy

1.016 (0.972 - 1.062)

0.809 (0.744 - 0.879)*

1.132 (1.074 - 1.193)*

Aromatase Inhibitor

0.588 (0.542 - 0.638)

0.417 (0.353 - 0.494)*

0.625 (0.569 - 0.686)*

Statins

1.060 (1.016 - 1.106)*

1.083 (1.006 - 1.166)*

1.081 (1.027 - 1.138)*

Anti-hypertensives

1.517 (1.461 - 1.575)*

1.829 (1.712 - 1.953)*

1.480 (1.413 - 1.549)*

Bisphosphonates

0.897 (0.848 - 0.949)*

0.875 (0.793 - 0.966)*

0.894 (0.835 - 0.958)*

Metformin

1.058 (0.988 - 1.132)

1.039 (0.919 - 1.175)

1.078 (0.993 - 1.170)

Insulin

0.973 (0.888 - 1.065)

1.061 (0.903 - 1.247)

0.937 (0.839 - 1.047)

Thiazolidinediones

0.832 (0.726 - 0.952)*

0.665 (0.512 - 0.862)*

0.900 (0.768 - 1.055)

SGLT2-Inhibitors

0.386 (0.124 - 1.200)

0.458 (0.064 - 3.269)

0.334 (0.083 - 1.338)

DDP4-Inhibitors

1.068 (0.941 - 1.213)

1.149 (0.916 - 1.442)

1.071 (0.918 - 1.248)

GLP1-Agonists

1.260 (0.902 - 1.760)

1.109 (0.606 - 2.031)

1.351 (0.905 - 2.018)

 

In multivariable-adjusted models, the hazard of developing any SPC was greater in patients aged 70-79 [HR 1.072 (1.031-1.115)], highest income quartile [HR 1.112 (1.069 - 1.157)], and with stage 0 BC compared to stage I [HR 1.101 (1.058 - 1.146)]. Those who were married [HR 0.991 (0.964-1.020)] and with greater number of comorbidities [HR 0.731 (0.685 - 0.781)] had a lower hazard of developing a SPC. However, BC survivors with certain comorbidities had increased hazard of developing any SPC: diabetes [HR 1.360 (1.299 - 1.424)], renal disease [HR 1.356 (1.284 - 1.433)], and chronic obstructive pulmonary disease (COPD) [HR 1.726 (1.665 - 1.790)]. We also observed a lower hazard of SPC in patients who were taking bisphosphonates [HR 0.897 (0.848 - 0.949)] and aromatase inhibitors [HR 0.588 (0.542 - 0.638)]. Medications such as anti-hypertensives [HR 1.517 (1.461 - 1.575)] and statins [HR 1.060 (1.016 - 1.106)] increased the hazard of SPCs.

In contrast to any SPC, the hazard of developing breast-only SPC was significantly higher in black BC survivors [HR 1.106 (1.010 - 1.211)] and those diagnosed at more advanced stage (stage IIIC vs. stage I) [HR 1.325 (1.098 - 1.598)]. While patients with estrogen receptor (ER) negative status had an increased hazard for BC only SPC [HR 1.366 (1.241 - 1.504)], patients who received chemotherapy [HR 0.809 (0.744 - 0.879)] and radiation therapy [HR 0.881 (0.836 - 0.929)], as well as those taking thiazolidinediones [HR 0.665 (0.512 - 0.862)] had a significantly lower hazard.

Compared to the breast-only SPC group, the factors associated with greater hazard for non-breast SPC were somewhat different. The hazard for non-breast SPCs was higher in patients 70-74 years old [HR 1.106 (1.056 - 1.158)] and 75-79 years old [HR 1.148 (1.094 - 1.205)] compared to those 66-69 years old. In contrast to the breast-only SPC group, patients who underwent chemotherapy [HR 1.132 (1.074 - 1.193)] or radiation therapy [HR 1.073 (1.035 - 1.113)] for their initial primary BC had an increased risk of developing a non-breast SPC. We observed a similar trend for decreased hazard of developing any SPC, including BC, in patients who were on medications such as aromatase inhibitors and bisphosphonates, whereas an increased hazard of SPC was observed for those with comorbidities such as diabetes, COPD, and renal disease.

Discussion

We found that the incidence of any SPC among older women with primary BC is 12%. Women of white race, who were married, and in a higher income quartile, were more likely to develop SPC. Additionally, initial BC with ductal histology, hormone positivity, treated with surgical intervention, were also associated with SPC. There are many possible explanations for these associations which will be further discussed below.

Sociodemographic factors associated with the development of SPC included older age at diagnosis, white race, being married and higher income quartile. While older age at BC diagnosis was associated with increased development of SPC, we observed a decline in SPC incidence among women ≥80 years after adjusting for competing risk of death. Compared to white women, the incidence of any SPC was lower for women of non-Hispanic black, Hispanic, and other race. The higher incidence of SPC among white women may be due to their overall higher survival rates after BC diagnosis, thus allowing them the time to develop a SPC.2 Conversely, the incidence of a breast SPC specifically was significantly higher in black women which may be attributed to factors such as poorer access to healthcare, barriers to breast screening, and more advanced stage of BC at initial diagnosis.2 These results are consistent with Qian et al., who also investigated risk factors for SPCs in the SEER database population and found black race associated with increased risk of second BC.3

We also observed that non-married women and those who have higher income were more likely to develop a SPC. This increased risk persisted in the adjusted models for any SPC. Patients who are married are more likely to get psychosocial and financial support which may aid in early cancer detection, appropriate treatment, and prolonged survival.4 Prolonged survival is an independent risk factor for the development of SPC. Several studies have reported the incidence of SPC types in primary breast cancer survivors.5 We found that second BCs represented 32% of all SPCs and the incidence was higher in women with ductal carcinoma and hormone receptor positive BCs. In general, these types of BC are more responsive to treatment and result in longer survival times, whereas hormone receptor negative BCs, which comprise 30% of tumors, are known to have a more aggressive clinical course as they can not be treated with hormone therapy.6 They are thus more likely to be poorly differentiated, involve increased recurrence rates as well as increased mortality rates.6

The current treatment for BC includes surgery, chemotherapy, radiotherapy, and more recently immunotherapy. The majority of BC survivors in our cohort received surgery as initial treatment for their primary BC, and surgery was associated with a higher hazard of developing any SPC. This is supported by other studies which showed that patients who did not receive surgery for BC conversely had a higher risk of any SPC including breast.7 Those who received surgery alone (without chemotherapy or radiation therapy) were also at higher risk for developing a contralateral breast cancer.8 This may be due to increased surveillance of the contralateral breast post-BC treatment and suggest that a combination of surgery with or without chemotherapy and radiation therapy is the most effective treatment against BC, improving survival and allowing more time for SPC to develop. We found that chemotherapy was associated with a decreased risk for breast SPC, but increased risk of all other SPCs, which is corroborated by other studies linking certain chemotherapy drugs with different types of cancers. Li et al. reported that SPCs, particularly non-breast cancers such as colon and lung cancer, were higher in patients who received chemotherapy for BC, even after adjusting for known confounders.7 Another SEER-based study determined that chemotherapy for BC patients was associated with increased incidences for all SPC, except for some hematologic malignancies.9 Though the mechanism of how chemotherapy may inadvertently stimulate cancer growth remains largely unclear, it has been shown to be most commonly linked to leukemia and myelodysplastic syndrome.

Research regarding the risks of radiation for BC are also generally inconclusive. Some studies have concluded that radiation therapy for BC increases the risk for SPC in the healthy contralateral breast or ipsilateral lung compared to an unexposed population.10 Other studies have shown that only 8% of SPCs are related to radiotherapy.11 Here, we found that similar to chemotherapy, radiation therapy had a decreased risk of second primary breast cancer, but increased risk of non-breast SPCs. Although the mechanisms underlying radiation-induced tumorigenesis is unclear, irradiation of surrounding tissues may cause secondary malignancies of these tissues particularly the lungs which was the second highest SPC in our cohort. Aromatase inhibitor therapy is the gold standard for the treatment of hormone-receptor positive BC in post-menopausal BC survivors. In our study, women treated with aromatase inhibitors were less likely to develop any SPC, including. non-breast SPCs. Preclinical studies have shown that aromatase inhibitors in combination with standard cisplatin chemotherapy for non-small cell lung cancer (NSCLC) decreases tumor progression.12 Post-menopausal hormone exposure was also associated with a reduced risk for later development of NSCLC in the general population.13

Finally, we observed significant associations between certain medications and the development of SPCs, particularly statins, antihypertensives, and bisphosphonates. Statins and anti-hypertensives were both associated with increased hazard of developing any SPC including breast SPC, whereas bisphosphonates were associated with decreased hazard of developing all SPCs. Bisphosphonates have been shown to decrease risk of both locoregional/distant BC recurrence or second primary BC.14 Its effect on the development of other SPCs is less well understood. However, anti-tumor properties have been shown in preclinical studies15 and it is effective in reducing the risk of bone metastases.14 A large population-based study determined that certain antihypertensives, including loop and thiazide diuretics, were associated with adverse BC outcomes, such as increased risk of breast SPCs, recurrence, and BC mortality,16 although the mechanisms to explain these associations remain unclear. Possible explanations include that patients on these antihypertensives are more unhealthy generally, and thus either did not tolerate BC treatment or are more likely to have captured adverse outcomes due to their more frequent medical visits. Additionally, thiazide diuretics specifically are associated with insulin resistance, which has been found to be an established risk factor for breast cancer, which may also explain the risk associated with antihypertensives.16 While the biological mechanisms are unclear, statins have also been shown to impact cancer outcomes, with varying results for different cancer types. For example, a SEER-based study determined that statin use improved overall and lung cancer specific survival in patients with stage IV NSCLC, citing in vitro studies that have demonstrated reduced proliferation, migration, and increased apoptosis of lung cancer cell lines with simvastatin use.17 Another possible mechanism of statin anticancer effects posited in this study is the inhibition of tumor cell proliferation, angiogenesis, and migration by reduction of systemic cholesterol.17

Limitations

This study has some limitations. First, it is possible that certain SPCs may have been recurrence or metastases of primary BC. However, this was mitigated by excluding patients in whom the SPC was a BC diagnosed within two months of the primary BC diagnosis. In our adjusted analyses we also modeled all SPCs, breast only SPCs, and non-breast SPCs to isolate the effects of treatment and medications on the development of SPCs. Our cohort was limited to postmenopausal women; thus, our findings are not necessarily generalizable to premenopausal women. Postmenopausal women are at an increased risk of SPCs due to increasing age and comorbidities. As such, our results can assist in determining those who may benefit from increased cancer surveillance. We could not adjust for other potential confounders, such as family history of cancer, reproductive or lifestyle factors such as smoking or obesity. Finally, we do not have detailed treatment information such as type of chemotherapy or radiation therapy dose.

Conclusion

In summary, SPCs pose a threat to BC survivors, though the nuances in potential biologic and epidemiologic explanations remain unclear. Our comprehensive exploration uncovered several demographic and treatment factors that may help guide more detailed recommendations for cancer prevention and screening among postmenopausal BC survivors.

Declarations

Acknowledgements

This work was supported by the National Cancer Institute of the National Institutes of Health (T32CA225617 to SB). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Data Availability

The data that support the findings of this study are available from NCI SEER-Medicare, but restrictions apply to the availability of these data, which were used under license for the current study, and so are not publicly available. Data are however available from the authors upon reasonable request and with permission of NCI SEER-Medicare.

Statements and Declarations

Conflict of Interest: The authors declare that they have no conflict of interest.

References

  1. Cheng Y, Huang Z, Liao Q, et al. Risk of second primary breast cancer among cancer survivors: Implications for prevention and screening practice. PLoS One. 2020;15(6):e0232800. doi:10.1371/journal.pone.0232800
  2. Yedjou CG, Tchounwou PB, Payton M, et al. Assessing the Racial and Ethnic Disparities in Breast Cancer Mortality in the United States. Int J Environ Res Public Health. 2017;14(5):E486. doi:10.3390/ijerph14050486
  3. Qian X, Jia H, Zhang Y, Ma B, Qin G, Wu Z. Risk factors and prediction of second primary cancer in primary female non-metastatic breast cancer survivors. Aging (Albany NY). 2020;12(19):19628-19640. doi:10.18632/aging.103939
  4. Goodwin JS, Hunt WC, Key CR, Samet JM. The effect of marital status on stage, treatment, and survival of cancer patients. JAMA. 1987;258(21):3125-3130.
  5. Li Z, Wu Q, Song J, Zhang Y, Zhu S, Sun S. Risk of Second Primary Female Genital Malignancies in Women with Breast Cancer: a SEER Analysis. Horm Cancer. 2018;9(3):197-204. doi:10.1007/s12672-018-0330-0
  6. Nahleh Z. Androgen receptor as a target for the treatment of hormone receptor-negative breast cancer: an unchartered territory. Future Oncol. 2008;4(1):15-21. doi:10.2217/14796694.4.1.15
  7. Li Z, Wang K, Shi Y, Zhang X, Wen J. Incidence of second primary malignancy after breast cancer and related risk factors-Is breast-conserving surgery safe? A nested case-control study. Int J Cancer. 2020;146(2):352-362. doi:10.1002/ijc.32259
  8. Kirova YM, De Rycke Y, Gambotti L, Pierga J-Y, Asselain B, Fourquet A. Second malignancies after breast cancer: the impact of different treatment modalities. Br J Cancer. 2008;98(5):870-874. doi:10.1038/sj.bjc.6604241
  9. Wei J-L, Jiang Y-Z, Shao Z-M. Survival and chemotherapy-related risk of second primary malignancy in breast cancer patients: a SEER-based study. Int J Clin Oncol. 2019;24(8):934-940. doi:10.1007/s10147-019-01430-0
  10. Mazonakis M, Stratakis J, Lyraraki E, Damilakis J. Risk of contralateral breast and ipsilateral lung cancer induction from forward-planned IMRT for breast carcinoma. Physica Medica. 2019;60:44-49. doi:10.1016/j.ejmp.2019.03.021
  11. Berrington de Gonzalez A, Curtis RE, Kry SF, et al. Proportion of second cancers attributable to radiotherapy treatment in adults: a cohort study in the US SEER cancer registries. Lancet Oncol. 2011;12(4):353-360. doi:10.1016/S1470-2045(11)70061-4
  12. Márquez-Garbán DC, Chen H-W, Goodglick L, Fishbein MC, Pietras RJ. Targeting aromatase and estrogen signaling in human non-small cell lung cancer. Ann N Y Acad Sci. 2009;1155:194-205. doi:10.1111/j.1749-6632.2009.04116.x
  13. Schwartz AG, Wenzlaff AS, Prysak GM, et al. Reproductive factors, hormone use, estrogen receptor expression and risk of non small-cell lung cancer in women. J Clin Oncol. 2007;25(36):5785-5792. doi:10.1200/JCO.2007.13.3975
  14. Korde LA, Doody DR, Hsu L, Porter PL, Malone KE. Bisphosphonate Use and Risk of Recurrence, Second Primary Breast Cancer, and Breast Cancer Mortality in a Population-Based Cohort of Breast Cancer Patients. Cancer Epidemiol Biomarkers Prev. 2018;27(2):165-173. doi:10.1158/1055-9965.EPI-17-0556
  15. Croucher P, Jagdev S, Coleman R. The anti-tumor potential of zoledronic acid. The Breast. 2003;12(2):S30-S36. doi:https://doi.org/10.1016/S0960-9776(03)80161-3
  16. Chen L, Chubak J, Boudreau DM, Barlow WE, Weiss NS, Li CI. Use of Antihypertensive Medications and Risk of Adverse Breast Cancer Outcomes in a SEER-Medicare Population. Cancer Epidemiol Biomarkers Prev. 2017;26(11):1603-1610. doi:10.1158/1055-9965.EPI-17-0346
  17. Lin JJ, Ezer N, Sigel K, Mhango G, Wisnivesky JP. The effect of statins on survival in patients with stage IV lung cancer. Lung Cancer. 2016;99:137-142. doi:10.1016/j.lungcan.2016.07.006
  18. Du R, Lin L, Cheng D, et al. Thiazolidinedione therapy and breast cancer risk in diabetic women: A systematic review and meta-analysis. Diabetes Metab Res Rev. 2018;34(2):e2961. doi:10.1002/dmrr.2961
  19. Gnant M. Can Oral Bisphosphonates Really Reduce the Risk of Breast Cancer in Healthy Women? JCO. 2010;28(22):3548-3551. doi:10.1200/JCO.2010.29.6327
  20. Agarwal S, Pappas L, Matsen CB, Agarwal JP. Second primary breast cancer after unilateral mastectomy alone or with contralateral prophylactic mastectomy. Cancer Med. 2020;9(21):8043-8052. doi:10.1002/cam4.3394
  21. Ahmed AT, Welch BT, Brinjikji W, et al. Racial Disparities in Screening Mammography in the United States: A Systematic Review and Meta-analysis. Journal of the American College of Radiology. 2017;14(2):157-165.e9. doi:10.1016/j.jacr.2016.07.034
  22. Berrington de Gonzalez A, Curtis RE, Gilbert E, et al. Second solid cancers after radiotherapy for breast cancer in SEER cancer registries. Br J Cancer. 2010;102(1):220-226. doi:10.1038/sj.bjc.6605435
  23. Chen L, Chubak J, Boudreau DM, Barlow WE, Weiss NS, Li CI. Use of Antihypertensive Medications and Risk of Adverse Breast Cancer Outcomes in a SEER-Medicare Population. Cancer Epidemiol Biomarkers Prev. 2017;26(11):1603-1610. doi:10.1158/1055-9965.EPI-17-0346
  24. Coyle C, Cafferty FH, Vale C, Langley RE. Metformin as an adjuvant treatment for cancer: a systematic review and meta-analysis. Ann Oncol. 2016;27(12):2184-2195. doi:10.1093/annonc/mdw410
  25. Donin N, Filson C, Drakaki A, et al. Risk of second primary malignancies among cancer survivors in the United States, 1992 through 2008. Cancer. 2016;122(19):3075-3086. doi:10.1002/cncr.30164
  26. Donin NM, Kwan L, Lenis AT, Drakaki A, Chamie K. Second primary lung cancer in United States Cancer Survivors, 1992-2008. Cancer Causes Control. 2019;30(5):465-475. doi:10.1007/s10552-019-01161-7
  27. Faria J, Negalha G, Azevedo A, Martel F. Metformin and Breast Cancer: Molecular Targets. J Mammary Gland Biol Neoplasia. 2019;24(2):111-123. doi:10.1007/s10911-019-09429
  28. Keegan THM, Bleyer A, Rosenberg AS, Li Q, Goldfarb M. Second Primary Malignant Neoplasms and Survival in Adolescent and Young Adult Cancer Survivors. JAMA Oncol. 2017;3(11):1554-1557. doi:10.1001/jamaoncol.2017.0465
  29. Kim HY, Jung YJ, Lee SH, Jung HJ, Pak K. Is Beta-Blocker Use Beneficial in Breast Cancer? A Meta-Analysis. Oncology. 2017;92(5):264-268. doi:10.1159/000455143
  30. Kuo JH, Chabot JA, Lee JA. Breast cancer in thyroid cancer survivors: An analysis of the Surveillance, Epidemiology, and End Results-9 database. Surgery. 2016;159(1):23-29. doi:10.1016/j.surg.2015.10.009
  31. Kurian AW, Canchola AJ, Ma CS, Clarke CA, Gomez SL. Magnitude of reduction in risk of second contralateral breast cancer with bilateral mastectomy in patients with breast cancer: Data from California, 1998 through 2015. Cancer. 2020;126(5):958-970. doi:10.1002/cncr.32618
  32. Lee A, Djamgoz MBA. Triple negative breast cancer: Emerging therapeutic modalities and novel combination therapies. Cancer Treat Rev. 2018;62:110-122. doi:10.1016/j.ctrv.2017.11.003
  33. Li D, Weng S, Zhong C, et al. Risk of Second Primary Cancers Among Long-Term Survivors of Breast Cancer. Front Oncol. 2020;9. doi:10.3389/fonc.2019.01426
  34. Li S, Yang J, Shen Y, et al. Clinicopathological features, survival and risk in breast cancer survivors with thyroid cancer: an analysis of the SEER database. BMC Public Health. 2019;19(1):1592. doi:10.1186/s12889-019-7947-y
  35. Lin JJ, Gallagher EJ, Sigel K, et al. Survival of patients with stage IV lung cancer with diabetes treated with metformin. Am J Respir Crit Care Med. 2015;191(4):448-454. doi:10.1164/rccm.201407-1395OC
  36. Liu Y, Dong C, Chen L. The clinicopathological features of second primary cancer in patients with prior breast cancer. Medicine (Baltimore). 2017;96(16):e6675. doi:10.1097/MD.0000000000006675
  37. Marcu LG, Santos A, Bezak E. Risk of second primary cancer after breast cancer treatment. Eur J Cancer Care (Engl). 2014;23(1):51-64. doi:10.1111/ecc.12109
  38. Matsuo K, Mandelbaum RS, Machida H, et al. Decreasing secondary primary uterine cancer after breast cancer: A population-based analysis. Gynecol Oncol. 2019;154(1):169-176. doi:10.1016/j.ygyno.2019.05.014
  39. Nasioudis D, Wilson E, Mastroyannis SA, et al. Increased Risk of Breast and Uterine Cancer Among Women With Ovarian Granulosa Cell Tumors. Anticancer Res. 2019;39(9):4971-4975. doi:10.21873/anticanres.13686
  40. Phadke S, Clamon G. Beta blockade as adjunctive breast cancer therapy: A review. Crit Rev Oncol Hematol. 2019;138:173-177. doi:10.1016/j.critrevonc.2019.04.006
  41. Poleszczuk J, Luddy K, Chen L, et al. Neoadjuvant radiotherapy of early-stage breast cancer and long-term disease-free survival. Breast Cancer Res. 2017;19(1):75. doi:10.1186/s13058017-0870-1
  42. Sakai T, Ozkurt E, DeSantis S, et al. National trends of synchronous bilateral breast cancer incidence in the United States. Breast Cancer Res Treat. 2019;178(1):161-167. doi:10.1007/s10549-019-05363-0
  43. Schonfeld SJ, Berrington de Gonzalez A, Visvanathan K, Pfeiffer RM, Anderson WF. Declining second primary ovarian cancer after first primary breast cancer. J Clin Oncol. 2013;31(6):738-743. doi:10.1200/JCO.2012.43.2757
  44. Sung H, Hyun N, Leach CR, Yabroff KR, Jemal A. Association of First Primary Cancer With Risk of Subsequent Primary Cancer Among Survivors of Adult-Onset Cancers in the United States. JAMA. 2020;324(24):2521-2535. doi:10.1001/jama.2020.23130
  45. Vancura A, Bu P, Bhagwat M, Zeng J, Vancurova I. Metformin as an Anticancer Agent. Trends Pharmacol Sci. 2018;39(10):867-878. doi:10.1016/j.tips.2018.07.006
  46. Wang R, Yin Z, Liu L, et al. Second Primary Lung Cancer After Breast Cancer: A Population-Based Study of 6,269 Women. Front Oncol. 2018;8:427. doi:10.3389/fonc.2018.00427
  47. Yesensky J, Kyrillos A, Kuchta K, Yao K, Wang C-H, Bhayani MK. Risk of Development of Second Primary Head and Neck Cancer following an Index Breast Cancer. Otolaryngoly Head Neck Surg. 2018;158(2):303-308. doi:10.1177/0194599817737271
  48. Zhong M, He X, Lei K. Survival of Patients with First and Metachronous Second Primary Breast Cancer or Lung Cancer Malignancy: Comparisons Using the SEER Database. Adv Ther. 2020;37(5):2236-2245. doi:10.1007/s12325-020-01322-0