1
|
Maleki R, Ghith A, Heydarlou H, Grzeskowiak LE, Ingman WV. The Role of Breastmilk in Macrophage-Tumour Cell Interactions in Postpartum Breast Cancer. FRONT BIOSCI-LANDMRK 2024; 29:328. [PMID: 39344339 DOI: 10.31083/j.fbl2909328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Lactation is associated with long-term reduced risk of breast cancer. However, there is a transient increased risk of breast cancer in the 5 to 10 years postpartum and this is associated with a high incidence of metastasis and mortality. Breastmilk is a physiological fluid secreted by the mammary glands intimately connected with breast cells and the microenvironment that may affect postpartum breast cancer development and progression. This study aims to investigate the effect of breastmilk on interactions between breast cancer cells and macrophages in vitro. METHODS Human breastmilk from healthy donors (n = 7) was pooled and incubated with breast cancer (MCF-7 and MDA-MB-231) and macrophage (RAW264.7) cell lines to assess cell proliferation, viability, migration, and expression of key genes associated with epithelial-mesenchymal transition (EMT) and macrophage phenotype. Indirect co-culture studies assessed the effect of breastmilk on interactions between breast cancer cells and macrophages. RESULTS Breastmilk increased the proliferation and viability of breast cancer cells, reduced EMT markers, and reduced cell migration in MDA-MB-231 cells. Breastmilk decreased mRNA expression of interleukin 1B (IL1B) and interleukin 10 (IL10) in macrophages. Reduced EMT marker expression was observed in breast cancer cells co-cultured with macrophages pre-treated with breastmilk. Macrophages co-cultured with breast cancer cells pre-treated with breastmilk exhibited increased expression of a pro-inflammatory cytokine tumor necrosis factor A (TNFA) and pro-inflammatory nitric oxide synthase 2 (NOS2), and reduced expression of cytokines IL10 and transforming growth factor B1 (TGFB1) which are associated with the alternatively-activated macrophage phenotype. CONCLUSIONS Breastmilk has the potential to promote breast cancer proliferation, however, it can also reduce breast cancer progression through inhibition of breast cancer cell migration and regulation of macrophage polarisation. These findings suggest that breastmilk has potential to shape the tumour microenvironment in postpartum breast cancer.
Collapse
Affiliation(s)
- Reza Maleki
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Hanieh Heydarlou
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Luke E Grzeskowiak
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Wendy V Ingman
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| |
Collapse
|
2
|
Vickers R, Porter W. Immune Cell Contribution to Mammary Gland Development. J Mammary Gland Biol Neoplasia 2024; 29:16. [PMID: 39177859 PMCID: PMC11343902 DOI: 10.1007/s10911-024-09568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 06/27/2024] [Indexed: 08/24/2024] Open
Abstract
Postpartum breast cancer (PPBC) is a unique subset of breast cancer, accounting for nearly half of the women diagnosed during their postpartum years. Mammary gland involution is widely regarded as being a key orchestrator in the initiation and progression of PPBC due to its unique wound-healing inflammatory signature. Here, we provide dialogue suggestive that lactation may also facilitate neoplastic development as a result of sterile inflammation. Immune cells are involved in all stages of postnatal mammary development. It has been proposed that the functions of these immune cells are partially directed by mammary epithelial cells (MECs) and the cytokines they produce. This suggests that a more niche area of exploration aimed at assessing activation of innate immune pathways within MECs could provide insight into immune cell contributions to the developing mammary gland. Immune cell contribution to pubertal development and mammary gland involution has been extensively studied; however, investigations into pregnancy and lactation remain limited. During pregnancy, the mammary gland undergoes dramatic expansion to prepare for lactation. As a result, MECs are susceptible to replicative stress. During lactation, mitochondria are pushed to capacity to fulfill the high energetic demands of producing milk. This replicative and metabolic stress, if unresolved, can elicit activation of innate immune pathways within differentiating MECs. In this review, we broadly discuss postnatal mammary development and current knowledge of immune cell contribution to each developmental stage, while also emphasizing a more unique area of study that will be beneficial in the discovery of novel therapeutic biomarkers of PPBC.
Collapse
Affiliation(s)
- Ramiah Vickers
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Weston Porter
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
3
|
Fu S, Ke H, Yuan H, Xu H, Chen W, Zhao L. Dual role of pregnancy in breast cancer risk. Gen Comp Endocrinol 2024; 352:114501. [PMID: 38527592 DOI: 10.1016/j.ygcen.2024.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Reproductive history is one of the strongest risk factors for breast cancer in women. Pregnancy can promote short-term breast cancer risk, but also reduce a woman's lifetime risk of breast cancer. Changes in hormone levels before and after pregnancy are one of the key factors in breast cancer risk. This article summarizes the changes in hormone levels before and after pregnancy, and the roles of hormones in mammary gland development and breast cancer progression. Other factors, such as changes in breast morphology and mammary gland differentiation, changes in the proportion of mammary stem cells (MaSCs), changes in the immune and inflammatory environment, and changes in lactation before and after pregnancy, also play key roles in the occurrence and development of breast cancer. This review discusses the dual effects and the potential mechanisms of pregnancy on breast cancer risk from the above aspects, which is helpful to understand the complexity of female breast cancer occurrence.
Collapse
Affiliation(s)
- Shiting Fu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Hao Ke
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | | | - Huaimeng Xu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Wenyan Chen
- Department of Medical Oncology, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China.
| |
Collapse
|
4
|
Dahms P, Lyons TR. Toward Characterizing Lymphatic Vasculature in the Mammary Gland During Normal Development and Tumor-Associated Remodeling. J Mammary Gland Biol Neoplasia 2024; 29:1. [PMID: 38218743 PMCID: PMC10787674 DOI: 10.1007/s10911-023-09554-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/24/2023] [Indexed: 01/15/2024] Open
Abstract
Lymphatic vasculature has been shown to promote metastatic spread of breast cancer. Lymphatic vasculature, which is made up of larger collecting vessels and smaller capillaries, has specialized cell junctions that facilitate cell intravasation. Normally, these junctions are designed to collect immune cells and other cellular components for immune surveillance by lymph nodes, but they are also utilized by cancer cells to facilitate metastasis. Although lymphatic development overall in the body has been well-characterized, there has been little focus on how the lymphatic network changes in the mammary gland during stages of remodeling such as pregnancy, lactation, and postpartum involution. In this review, we aim to define the currently known lymphangiogenic factors and lymphatic remodeling events during mammary gland morphogenesis. Furthermore, we juxtapose mammary gland pubertal development and postpartum involution to show similarities of pro-lymphangiogenic signaling as well as other molecular signals for epithelial cell survival that are critical in these morphogenic stages. The similar mechanisms include involvement of M2-polarized macrophages that contribute to matrix remodeling and vasculogenesis; signal transducer and activator of transcription (STAT) survival and proliferation signaling; and cyclooxygenase 2 (COX2)/Prostaglandin E2 (PGE2) signaling to promote ductal and lymphatic expansion. Investigation and characterization of lymphangiogenesis in the normal mammary gland can provide insight to targetable mechanisms for lymphangiogenesis and lymphatic spread of tumor cells in breast cancer.
Collapse
Affiliation(s)
- Petra Dahms
- Division of Medical Oncology Senior Scientist, Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, 80045, Aurora, CO, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
- Anschutz Medical Campus Graduate Program in Cancer Biology, University of Colorado, Aurora, USA
| | - Traci R Lyons
- Division of Medical Oncology Senior Scientist, Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, 80045, Aurora, CO, USA.
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA.
- Anschutz Medical Campus Graduate Program in Cancer Biology, University of Colorado, Aurora, USA.
| |
Collapse
|
5
|
Updates in the pathology of Pregnancy Associated Breast Cancer (PABC). Pathol Res Pract 2023; 244:154413. [PMID: 36921545 DOI: 10.1016/j.prp.2023.154413] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
Pregnancy associated breast cancer (PABC) is defined as a breast cancer diagnosed during gestation, lactation or within 5 years postpartum. While the development of malignancy during pregnancy is rare, the incidence is increasing. Breast cancer is one of the most common cancers diagnosed during pregnancy, affecting up to 1 in 3000 deliveries. New understanding of the pathophysiology of PABC recently resulted in updated definitions distinguishing breast cancer diagnosed during pregnancy (PrBC) from cancer diagnosed during the postpartum period (PPBC) due to distinct biology and prognosis. Pregnancy has a dual effect on breast cancer development- both protective against cancer and promoting tumor growth. While several hypotheses have been proposed over the years to explain these effects, the most likely hypothesis for the development of PABC is the involution hypothesis, proposing that remodeling programs activated in the immediate postpartum period are similar to wound healing and inflammation that may be associated with tumor development and progression. Although PABCs reflect all subtypes of breast carcinomas, they are most commonly invasive ductal carcinomas of high tumor grade and large tumor size, with more advanced stage at presentation and higher rates of lymph node involvement. Most PABCs are hormone negative tumors (triple negative or HER2 amplified tumors) with high Ki-67 proliferation rates. Several studies have shown that PABCs have different genomic signatures than non-PABC tumors, showing increased expression of immune response mediators. Better understanding of the molecular pathways of tumor initiation and progression, along with prompt diagnosis and novel treatment protocols in the care of PrBC and PPBC are needed to improve outcomes for these young, high-risk breast cancer patients.
Collapse
|
6
|
Han L, Wang N, Li Y, Jiang S, Gu Y. A rapid reduction in tumor size by cyclin-dependent kinase inhibition in hormone receptor-positive postpartum breast cancer: a case report of two patients and a review of the literature. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1413. [PMID: 36660646 PMCID: PMC9843329 DOI: 10.21037/atm-22-5201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022]
Abstract
Background Postpartum breast cancer (PPBC) as an independent entity different from PABC. PPBC is defined as breast cancer (BC) diagnosed within 5 years after delivery in many relevant literatures and is associated with a poor prognosis and a decrease in overall survival. PPBC patients commonly present with inflammatory breast cancer (IBC) phenotype, multifocal lesions, and lymph node metastasis. Hormone receptor-positive (HR+) PPBC is an under-investigated subtype. In PPBC, the risk of death of HR+ subtype significantly increased two-fold, while that was only modestly increased for triple-negative breast cancer (TNBC) subtype, and was not significant in human epidermal growth factor receptor 2-positive (HER2+) subtype. HR+ PPBC is a subtype associating with enhanced signatures of cell cycle control, T-cell activation and exhaustion, decreased HR signaling, and altered P53 signaling. The recommended treatment for HR+ PPBC patients is still lacking. Cyclin-dependent kinase (CDK) 4/6 inhibitors are used as a novel treatment standard not only in pretreated patients but also in the first-line setting of HR+ metastatic breast cancer (MBC). However, there is no clinical case report on the application and efficacy of CDK4/6 inhibitors in HR+ PPBC patients. Case Description This article describes the clinical cases of two patients with advanced HR+ PPBC who were rapidly relieved after receiving leuprorelin combined with letrozole combined with dalpiciclib. We reviewed the related literature of PPBC, and found that HR+ PPBC has not been clinically classified as a BC subtype, and only some basic studies suggested that HR+ PPBC may be sensitive to CDK4/6 inhibitors. The purpose of this study is to provide the basis for the related research on the therapeutic effect of CDK4/6 inhibitors in HR+ PPBC through the report of clinical cases. Conclusions This article reports for the first time the good therapeutic effects of CDK4/6 inhibitors on HR+ PPBC patients. Based on our findings, we suggest that dalpiciclib combined with endocrine therapy can be considered as the first-line treatment for patients with advanced HR+ PPBC. Our case report provides new clinical evidence for the related research on the role of CDK4/6 inhibitors in HR+ PPBC therapy.
Collapse
Affiliation(s)
- Luhong Han
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Li
- Department of Reproductive Medicine, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shan Jiang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Bajpai J, Pathak R, Shylasree TS, Rugo HS. Management of breast cancer diagnosed during pregnancy: global perspectives. Expert Rev Anticancer Ther 2022; 22:1301-1308. [PMID: 36480337 DOI: 10.1080/14737140.2022.2150167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Pregnancy-associated breast cancer (PABC) encompasses breast cancer diagnosed during pregnancy (BCP) or postpartum (PPBC). BCP is especially challenging with concerns regarding maternal and fetal safety synchronously. This review provides a comprehensive global view to optimize care of this unique entity. Areas covered Published literature and practices across the globe including real world published data from the first Indian registry are thoroughly reviewed to derive inferences. Diagnostic delays are common with resultant upstaging and inferior outcomes. Sonography-mammography and a biopsy with immunohistochemistry for estrogen, progesterone and HER-2neu receptors is mandatory. Multidisciplinary specialist teams are critical for trimester dependent management. Stage-wise surgical and systemic treatment remains largely similar to that of the nonpregnant women. Anthracyclines- and taxane-based chemotherapy is found to be safe after the 1st trimester. Frequent fetal and maternal monitoring is required to minimize complications. Chemotherapy should stop three weeks prior to the delivery to prevent peripartum infection/bleeding. Anti- Her-2 targeted therapy, endocrine therapy and radiation therapy are administered post-delivery. Iatrogenic premature delivery leads to poor neurocognition and should be avoided. Expert opinion Stage-wise outcomes are similar to that of non-pregnant patients with breast cancer, and underscores the importance of early detection especially in low- and middle-income countries. Global collaborations are warranted. AREAS COVERED Published literature and practices across the globe including real world published data from the first Indian registry are thoroughly reviewed to derive inferences. Diagnostic delays are common with resultant upstaging and inferior outcomes. Sonography-mammography and a biopsy with immunohistochemistry for estrogen, progesterone and HER-2neu receptors is mandatory. Multidisciplinary specialist teams are critical for trimester dependent management. Stage-wise surgical and systemic treatment remains largely similar to that of the nonpregnant women. Anthracyclines- and taxane-based chemotherapy is found to be safe after the 1st trimester. Frequent fetal and maternal monitoring is required to minimize complications. Chemotherapy should stop three weeks prior to the delivery to prevent peripartum infection/bleeding. Anti- Her-2 targeted therapy, endocrine therapy and radiation therapy are administered post-delivery. Iatrogenic premature delivery leads to poor neurocognition and should be avoided. EXPERT OPINION Stage-wise outcomes are similar to that of non-pregnant patients with breast cancer, and underscores the importance of early detection especially in low- and middle-income countries. Global collaborations are warranted.
Collapse
Affiliation(s)
- Jyoti Bajpai
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - Rima Pathak
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, India
| | - T S Shylasree
- MD,FRCOG Consultant Gynaecological Oncologist, Aberdeen Royal Infirmary and NHS Grampian North Cancer Alliance United Kingdom, UK
| | - Hope S Rugo
- Professor of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| |
Collapse
|
8
|
Park S, Lee JS, Yoon JS, Kim NH, Park S, Youn HJ, Lee JW, Lee JE, Lee J, Hur H, Jeong J, Kim KC, Bae SY. The Risk Factors, Incidence and Prognosis of Postpartum Breast Cancer: A Nationwide Study by the SMARTSHIP Group. Front Oncol 2022; 12:889433. [PMID: 35847886 PMCID: PMC9283825 DOI: 10.3389/fonc.2022.889433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022] Open
Abstract
The term ‘pregnancy-associated breast cancer’ is no longer used as it has been consistently reported that breast cancer during pregnancy and breast cancer after delivery (postpartum breast cancer) have different characteristics and prognosis. The purpose of this study is to define postpartum breast cancer by analyzing the incidence rate, related factors, and prognosis according to the timing of breast cancer. Data from the Korean National Health Insurance Service were used to analyze 1,292,727 women aged 20-49 years who birthed their first child between 2007 and 2012. The annual incidence rate of breast cancer after delivery increased every year (7.7 per 10,000 person-years after 5 years, 19.36 per 10,000 person-years after 10 years). The risk of breast cancer was significantly higher (hazard ratio 1.15, 95% CI 1.05-1.27, P=0.0037) in women diagnosed with gestational diabetes, but that was not associated with overall survival (OS). Patients diagnosed with breast cancer within 5 years of delivery had a poorer prognosis than those diagnosed later (5-year OS, <5 years: 91.1% vs. 5-10 years: 96.0%). In multivariate analysis of OS, the hazard ratio of patients diagnosed within 5 years after delivery was twice as high as of patients diagnosed between 5 and 10 years. Women diagnosed with gestational diabetes had an increased risk of breast cancer. Breast cancer patients diagnosed within 5 years of delivery had a poorer prognosis than those diagnosed later. In this regard, careful screening for early diagnosis of high-risk patients and intensive research on new treatment strategies are needed.
Collapse
Affiliation(s)
- Sungmin Park
- Department of Surgery, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Ji Sung Lee
- Clinical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Sun Yoon
- Department of Biostatistics, Korea University, Seoul, South Korea
| | - Nam Hyoung Kim
- Advertising and Branding, Kaywon University of Art and Design, Uiwang-si, South Korea
| | - Seho Park
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Jo Youn
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Jong Won Lee
- Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Jung Eun Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, South Korea
| | - Jihyoun Lee
- Department of Surgery, Soonchunhyang University Seoul Hospital, Seoul, South Korea
| | - Ho Hur
- Department of Surgery, National Health Insurance Service Ilsan Hospital, Koyang-si, South Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Kweon-Cheon Kim
- Department of Surgery, Chosun University Medical School, Gwangju, South Korea
| | - Soo Youn Bae
- Department of Surgery, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, South Korea
- *Correspondence: Soo Youn Bae,
| |
Collapse
|
9
|
Puchar A, Despierres M, Boudy AS, Selleret L, Gligorov J, Richard S, Zilberman S, Ferrier C, Dabi Y, Varlas V, Thomassin-Naggara I, Bendifallah S, Touboul C, Darai E. Prognosis of triple-negative breast cancer associated with pregnancy: A propensity score-matched analysis from the French CALG (Cancer Associé à la Grossesse) network. Breast 2022; 61:168-174. [PMID: 35042133 PMCID: PMC8777280 DOI: 10.1016/j.breast.2022.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Triple-negative (TN) breast cancer represents one third of pregnancy-associated breast cancers (PABC). The aims of the current study were to describe oncological and obstetrical outcomes of patients with TN-PABC and to compare their prognosis with TN-non-PABC patients using a propensity score. Materials and methods Between January 2005 and December 2020, data of patients with histologically proven TN-PABC were collected and compared with data of TN-non-PABC patients under the age of 46 years diagnosed during the same period using a propensity score (PS). Results After PS matching (tumor size and lymph node involvement),there were 59 patients in each group. The median follow-up was 14 months (IQR 4.8–40.1) for the TN-PABC group and 60 months (IQR 30.7–101.4) for the TN-non-PABC group. Eight recurrences occurred in the TN-PABC group and 10 in the TN-non-PABC group (adjusted OR (AOR) = 0.60 (0.21–1.60), HR (Cox adjusted model- AHR) = 1.25 (0.53–2.94)). Two patients died in the TN-PABC group, and six in the TN-non-PABC group with an AOR = 0.23 (0.03–1.01) and an AHR = 0.58 (0.12–2.69). All the patients diagnosed during the second (n = 17) and third trimesters (n = 28) continued their pregnancies, with a median term at delivery of 38 WG (IQR 36–39). All patients gave birth to healthy newborns. Conclusion Although the TN subtype is associated with poor prognosis in pregnant patients due to advanced stage at diagnosis and high lymph node involvement, our PS-matched case-control study showed that pregnancy per se does not worsen the prognosis in terms of recurrence-free and overall survival. Triple-negative breast cancer are associated with poor prognosis. Breast cancers occurring during pregnancy differ from those occurring in the post-partum period. Pregnancy per se does not worsen the prognosis in triple-negative breast cancer.
Collapse
Affiliation(s)
- Anne Puchar
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France.
| | - Marie Despierres
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France
| | - Anne-Sophie Boudy
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France
| | - Lise Selleret
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France
| | - Joseph Gligorov
- Cancer Associé à La Grossesse (CALG), French CALG Network, France; UMRS-938 4. Faculté́ de Médecine Sorbonne Université́, France; Department of Oncology, Tenon University Hospital, Assistance Publique des Ho^pitaux de Paris (AP-HP), Sorbonne University, France
| | - Sandrine Richard
- Cancer Associé à La Grossesse (CALG), French CALG Network, France; Department of Oncology, Tenon University Hospital, Assistance Publique des Ho^pitaux de Paris (AP-HP), Sorbonne University, France
| | - Sonia Zilberman
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France
| | - Clément Ferrier
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France
| | - Yohann Dabi
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France
| | - Valentin Varlas
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France
| | - Isabelle Thomassin-Naggara
- Cancer Associé à La Grossesse (CALG), French CALG Network, France; Department of Radiology, Tenon University Hospital, Assistance Publique des Ho^pitaux de Paris (AP-HP), Sorbonne University, France
| | - Sofiane Bendifallah
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France; UMRS-938 4. Faculté́ de Médecine Sorbonne Université́, France
| | - Cyril Touboul
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France; UMRS-938 4. Faculté́ de Médecine Sorbonne Université́, France
| | - Emile Darai
- Department of Gynaecology and Obstetrics, Tenon University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University, France; Cancer Associé à La Grossesse (CALG), French CALG Network, France; UMRS-938 4. Faculté́ de Médecine Sorbonne Université́, France
| |
Collapse
|
10
|
Muñoz-Montaño WR, Cabrera-Galeana P, De la Garza-Ramos C, Azim HA, Tabares A, Perez V, Porras Reyes F, Sanchez Benitez D, Alvarado-Miranda A, Lara-Medina F, Vazquez Romo R, Bargallo-Rocha E, Arrieta O, Villarreal-Garza C. Prognosis of breast cancer diagnosed during pregnancy and early postpartum according to immunohistochemical subtype: A matched case-control study. Breast Cancer Res Treat 2021; 188:489-500. [PMID: 34132938 DOI: 10.1007/s10549-021-06225-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/08/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Pregnancy-associated breast cancer (PABC) poses a clinical challenge and its prognosis remains controversial. During the pregnancy and postpartum periods, the breast undergoes biological events that may uniquely influence disease behavior and treatment response. This study aimed to assess if a PABC diagnosis influences survival compared to non-PABC. METHODS A single-center record review was performed to identify PABC patients diagnosed from January 2007 through June 2018. Two controls were matched to each PABC case by stage, immunohistochemical (IHC) subtype, age (± 3) and year of diagnosis (± 2). Disease-free survival (DFS) and overall survival (OS) were estimated with the Kaplan-Meier method and compared with the log-rank test. Multivariate analysis was used to assess the impact of PABC on outcomes. RESULTS 125 PABC patients (pregnant: 62; postpartum: 63) and 250 controls were included. Median follow-up was 67.7 and 73.4 months, respectively. 4-year DFS was 62% in pregnant vs 78% in controls (p = 0.010), and 63% in postpartum vs 83% in controls (p = 0.034). Subanalysis by IHC subtype revealed a significantly inferior DFS in PABC with hormone receptor-positive/HER2-negative (p = 0.032) and HER2-positive disease (p = 0.005) compared to corresponding non-PABC patients. 4-year OS was similar between case groups and controls. Multivariate analysis supported the independent impact of pregnant and postpartum status on DFS (p < 0.05). CONCLUSION Patients diagnosed during pregnancy and early postpartum are at high risk of recurrence. Further research is warranted to better characterize PABC tumor biology and enable the identification of novel therapeutic interventions to improve treatment outcomes.
Collapse
Affiliation(s)
- Wendy R Muñoz-Montaño
- Breast Medical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Paula Cabrera-Galeana
- Breast Medical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Cynthia De la Garza-Ramos
- Breast Cancer Center, Hospital Zambrano Hellion, Tecnologico de Monterrey, Av. Batallon de San Patricio 112, 66278, Real San Agustin, San Pedro Garza Garcia, NL, Mexico
| | - Hatem A Azim
- Breast Cancer Center, Hospital Zambrano Hellion, Tecnologico de Monterrey, Av. Batallon de San Patricio 112, 66278, Real San Agustin, San Pedro Garza Garcia, NL, Mexico
| | - Ariana Tabares
- Breast Medical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Victor Perez
- Oncological Pathology of Mammary Tumors Unit, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Fanny Porras Reyes
- Oncological Pathology of Mammary Tumors Unit, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | | | - Alberto Alvarado-Miranda
- Breast Medical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Fernando Lara-Medina
- Breast Medical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Rafael Vazquez Romo
- Breast Surgical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Enrique Bargallo-Rocha
- Breast Surgical Oncology, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico
| | - Oscar Arrieta
- Research Unit, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico. .,Head of Thoracic Oncology Unit and Laboratory of Personalized Medicine, National Institute of Cancer, San Fernando #22, Section XVI, 14080, Tlalpan, Mexico City, Mexico.
| | - Cynthia Villarreal-Garza
- Breast Cancer Center, Hospital Zambrano Hellion, Tecnologico de Monterrey, Av. Batallon de San Patricio 112, 66278, Real San Agustin, San Pedro Garza Garcia, NL, Mexico.
| |
Collapse
|
11
|
Amant F, Lefrère H, Borges VF, Cardonick E, Lambertini M, Loibl S, Peccatori F, Partridge A, Schedin P. The definition of pregnancy-associated breast cancer is outdated and should no longer be used. Lancet Oncol 2021; 22:753-754. [PMID: 34087122 PMCID: PMC8868503 DOI: 10.1016/s1470-2045(21)00183-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 11/17/2022]
Affiliation(s)
- Frédéric Amant
- Department of Oncology, KU Leuven, Leuven, Belgium; Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands.
| | - Hanne Lefrère
- Department of Oncology, KU Leuven, Leuven, Belgium; Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Virginia F Borges
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elyce Cardonick
- Cooper Maternal Fetal Medicine, MD Anderson Cancer Center at Cooper, Camden, NJ, USA
| | - Matteo Lambertini
- University of Genova, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sibylle Loibl
- German Breast Group, Neu-Isenburg, Germany; Centre for Haematology and Oncology Bethanien, Frankfurt, Germany
| | - Fedro Peccatori
- Gynecologic Oncology Programme, European Institute of Oncology IRCCS, Milan, Italy
| | - Ann Partridge
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Pepper Schedin
- Knight Cancer Institute, University of Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
12
|
Lefrère H, Lenaerts L, Borges VF, Schedin P, Neven P, Amant F. Postpartum breast cancer: mechanisms underlying its worse prognosis, treatment implications, and fertility preservation. Int J Gynecol Cancer 2021; 31:412-422. [PMID: 33649008 PMCID: PMC7925817 DOI: 10.1136/ijgc-2020-002072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Breast cancers that occur in young women up to 5 to 10 years' postpartum are associated with an increased risk for metastasis and death compared with breast cancers diagnosed in young, premenopausal women during or outside pregnancy. Given the trend to delay childbearing, this frequency is expected to increase. The (immuno)biology of postpartum breast cancer is poorly understood and, hence, it is unknown why postpartum breast cancer has an enhanced risk for metastasis or how it should be effectively targeted for improved survival. The poorer prognosis of women diagnosed within 10 years of a completed pregnancy is most often contributed to the effects of mammary gland involution. We will discuss the most recent data and mechanistic insights of the most important processes associated with involution and their role in the adverse effects of a postpartum diagnosis. We will also look into the effect of lactation on breast cancer outcome after diagnosis. In addition, we will discuss the available treatment strategies that are currently being used to treat postpartum breast cancer, keeping in mind the importance of fertility preservation in this group of young women. These additional insights might offer potential therapeutic options for the improved treatment of women with this specific condition.
Collapse
Affiliation(s)
- Hanne Lefrère
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium.,Department of Gynecology, AVL NKI, Amsterdam, Noord-Holland, The Netherlands
| | - Liesbeth Lenaerts
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium
| | - Virginia F Borges
- Department of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Pepper Schedin
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado, USA.,Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Patrick Neven
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium.,Department of Gynecology and Obstetrics, Katholieke Universiteit Leuven UZ Leuven, Leuven, Flanders, Belgium.,Multidisciplinary Breast Centre, UZ-KU Leuven Cancer Institute (LKI), Katholieke Universiteit Leuven UZ Leuven, Leuven, Flanders, Belgium
| | - Frédéric Amant
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium .,Department of Gynecology, AVL NKI, Amsterdam, Noord-Holland, The Netherlands.,Department of Gynecology and Obstetrics, Katholieke Universiteit Leuven UZ Leuven, Leuven, Flanders, Belgium.,Department of Gynecological Oncology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Elder AM, Stoller AR, Black SA, Lyons TR. Macphatics and PoEMs in Postpartum Mammary Development and Tumor Progression. J Mammary Gland Biol Neoplasia 2020; 25:103-113. [PMID: 32535810 PMCID: PMC7395889 DOI: 10.1007/s10911-020-09451-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Postpartum mammary gland involution is a mammalian tissue remodeling event that occurs after pregnancy and lactation to return the gland to the pre-pregnant state. This event is characterized by apoptosis and lysosomal-mediated cell death of the majority of the lactational mammary epithelium, followed by remodeling of the extracellular matrix, influx of immune cell populations (in particular, T helper cells, monocytes, and macrophages), and neo-lymphangiogenesis. This postpartum environment has been shown to be promotional for tumor growth and metastases and may partially account for why women diagnosed with breast cancer during the postpartum period or within 5 years of last childbirth have an increased risk of developing metastases when compared to their nulliparous counterparts. The lymphatics and macrophages present during mammary gland involution have been implicated in promoting the observed growth and metastasis. Of importance are the macrophages, which are of the "M2" phenotype and are known to create a pro-tumor microenvironment. In this report, we describe a subset of postpartum macrophages that express lymphatic proteins (PoEMs) and directly interact with lymphatic vessels to form chimeric vessels or "macphatics". Additionally, these PoEMs are very similar to tumor-associated macrophages that also express lymphatic proteins and are present at the sites of lymphatic vessels where tumors escape the tissue and enter the lymphatic vasculature. Further characterizing these PoEMs may offer insight in preventing lymphatic metastasis of breast cancer, as well as provide information for how developmental programming of lymphatic endothelial cells and macrophages can contribute to different disease progression.
Collapse
Affiliation(s)
- Alan M Elder
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
- Graduate Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexander R Stoller
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
| | - Sarah A Black
- University of Colorado School of Medicine, Aurora, CO, USA
| | - Traci R Lyons
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA.
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA.
- Graduate Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
14
|
Farmaki E, Kaza V, Chatzistamou I, Kiaris H. CCL8 Promotes Postpartum Breast Cancer by Recruiting M2 Macrophages. iScience 2020; 23:101217. [PMID: 32535027 PMCID: PMC7300153 DOI: 10.1016/j.isci.2020.101217] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 04/27/2020] [Accepted: 05/26/2020] [Indexed: 12/23/2022] Open
Abstract
The microenvironment of postpartum mammary gland promotes tumor growth and metastasis in animal models and is linked to increased risk of breast cancer and poor disease outcome in patients. Our previous studies showed the involvement of the chemokine CCL8 in breast cancer metastasis through modulation of the tumor-promoting activity of the tumor microenvironment. Here we show that CCL8 is highly expressed during mammary gland involution and enhances the infiltration of M2 subtype macrophages at the second phase of involution. Cancer cell inoculation studies in Ccl8-deficient animals indicate that CCL8 accelerates tumor onset during involution but not in nulliparous animals. Depletion of macrophages abolished the tumor-promoting effect of CCL8 in involution suggesting the specific role of CCL8 in promoting tumor growth by recruiting macrophages. These results underscore the role of CCL8 in the development of postpartum breast cancer and suggest the potential value of targeting CCL8 in disease management. CCL8 exhibits increased expression during mammary gland involution CCL8 has tumor promoting activity and promotes postpartum breast cancer Targeting CCL8 could have beneficial value for the management of postpartum breast cancer
Collapse
Affiliation(s)
- Elena Farmaki
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Vimala Kaza
- Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology and Immunology, University of South Carolina, School of Medicine, Columbia, SC 29209, USA
| | - Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA; Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
15
|
Hitchcock JR, Hughes K, Harris OB, Watson CJ. Dynamic architectural interplay between leucocytes and mammary epithelial cells. FEBS J 2019; 287:250-266. [PMID: 31691481 PMCID: PMC7003847 DOI: 10.1111/febs.15126] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/19/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022]
Abstract
The adult mammary gland undergoes dynamic changes during puberty and the postnatal developmental cycle. The mammary epithelium is composed of a bilayer of outer basal, or myoepithelial, cells and inner luminal cells, the latter lineage giving rise to the milk-producing alveolar cells during pregnancy. These luminal alveolar cells undergo Stat3-mediated programmed cell death following the cessation of lactation. It is established that immune cells in the microenvironment of the gland have a role to play both in the ductal outgrowth during puberty and in the removal of dead cells and remodelling of the stroma during the process of postlactational regression. However, most studies have focussed on the role of the stromal immune cell compartment or have quantified immune cell populations in tissue extracts. Our recent development of protocols for deep imaging of the mammary gland in three dimensions (3D) has enabled the architectural relationship between immune cells and the epithelium to be examined in detail, and we have discovered a surprisingly dynamic relationship between the basal epithelium and leucocytes. Furthermore, we have observed morphological changes in the myoepithelial cells, as involution progresses, which were not revealed by previous work in 2D tissue sections and whole tissue. This dynamic architecture suggests a role for myoepithelial cells in the orderly progression of involution. We conclude that deep imaging of mammary gland and other tissues is essential for analysing complex interactions between cellular compartments.
Collapse
|
16
|
Elder AM, Tamburini BAJ, Crump LS, Black SA, Wessells VM, Schedin PJ, Borges VF, Lyons TR. Semaphorin 7A Promotes Macrophage-Mediated Lymphatic Remodeling during Postpartum Mammary Gland Involution and in Breast Cancer. Cancer Res 2018; 78:6473-6485. [PMID: 30254150 PMCID: PMC6239927 DOI: 10.1158/0008-5472.can-18-1642] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/15/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Postpartum mammary gland involution is a tissue remodeling event that occurs in all mammals in the absence of nursing or after weaning to return the gland to the pre-pregnant state. The tissue microenvironment created by involution has proven to be tumor promotional. Here we report that the GPI-linked protein semaphorin 7A (SEMA7A) is expressed on mammary epithelial cells during involution and use preclinical models to demonstrate that tumors induced during involution express high levels of SEMA7A. Overexpression of SEMA7A promoted the presence of myeloid-derived podoplanin (PDPN)-expressing cells in the tumor microenvironment and during involution. SEMA7A drove the expression of PDPN in macrophages, which led to integrin- and PDPN-dependent motility and adherence to lymphatic endothelial cells to promote lymphangiogenesis. In support of this mechanism, mammary tissue from SEMA7A-knockout mice exhibited decreased myeloid-derived PDPN-expressing cells, PDPN-expressing endothelial cells, and lymphatic vessel density. Furthermore, coexpression of SEMA7A, PDPN, and macrophage marker CD68 predicted for decreased distant metastasis-free survival in a cohort of over 600 cases of breast cancer as well as in ovarian, lung, and gastric cancers. Together, our results indicate that SEMA7A may orchestrate macrophage-mediated lymphatic vessel remodeling, which in turn drives metastasis in breast cancer.Signficance: SEMA7A, which is expressed on mammary cells during glandular involution, alters macrophage biology and lymphangiogenesis to drive breast cancer metastasis. Cancer Res; 78(22); 6473-85. ©2018 AACR.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Breast Neoplasms/pathology
- Cell Movement
- Crosses, Genetic
- Endothelial Cells/pathology
- Epithelial Cells/metabolism
- Female
- GPI-Linked Proteins/metabolism
- Humans
- Integrins/metabolism
- Lymphangiogenesis
- Lymphatic Vessels/pathology
- Macrophages/cytology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Human/pathology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Metastasis
- Postpartum Period
- Semaphorins/genetics
- Semaphorins/metabolism
- Tumor Microenvironment
Collapse
Affiliation(s)
- Alan M Elder
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Beth A J Tamburini
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora, Colorado
| | - Lyndsey S Crump
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Sarah A Black
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Veronica M Wessells
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Pepper J Schedin
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
- Department of Cell, Development and Cancer Biology, Oregon Health Sciences University, Oregon
| | - Virginia F Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| |
Collapse
|
17
|
Freedman RA, Partridge AH. Emerging Data and Current Challenges for Young, Old, Obese, or Male Patients with Breast Cancer. Clin Cancer Res 2018; 23:2647-2654. [PMID: 28572259 DOI: 10.1158/1078-0432.ccr-16-2552] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/20/2017] [Accepted: 04/04/2017] [Indexed: 11/16/2022]
Abstract
There are distinct subgroups of patients who have historically been underrepresented in most prospective clinical trials in breast cancer, including the youngest and oldest patients, male patients, and those struggling with obesity. Herein, we review current and future directions in research for each of these special populations with breast cancer, highlighting significant knowledge gaps and priorities in tumor biology and heterogeneity, therapeutic decision making promotion of adherence, supportive care, and psychosocial and functional well-being. In younger women, future study should focus on the biological underpinnings of aggressive disease and optimizing adherence and treatment decision making while addressing their unique survivorship needs. The latter includes generating a scientific basis for interruption of therapy for pregnancy. Among older patients, interventions should focus on increasing clinical trial accrual, predicting and mitigating toxicity so that functional status can be optimized, tailoring needs for dose modification, and anticipating life expectancy in the context of competing causes of death. For men with breast cancer, we need worldwide collaboration to answer even basic questions on optimal treatment, supportive care, and survivorship strategies. Finally, for those struggling with obesity, we need to better understand the biological associations with cancer incidence, prognosis and outcome, and how we can best intervene to assure weight loss at the "right time." It is only through highly collaborative, far-reaching, prospective, multidisciplinary, patient-centered, and patient-engaged efforts that we can optimize the physical and psychologic outcomes for all patients with breast cancer. Clin Cancer Res; 23(11); 2647-54. ©2017 AACRSee all articles in this CCR Focus section, "Breast Cancer Research: From Base Pairs to Populations."
Collapse
|
18
|
Prognosis of pregnancy-associated breast cancer. Breast Cancer Res Treat 2017; 163:417-421. [DOI: 10.1007/s10549-017-4224-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022]
|