Breast Cancer Treatment
Breast Cancer Treatment
Last Section Update: 10/2025
Contributor(s): Sandy Field, PHD; Maureen Williams, ND; Shayna Sandhaus, PhD; Elaine Walsh, MB BCh, PhD
1 Understanding Breast Cancer
Breast cancer is one of the most common cancers among women in the United States, second only to skin cancer.1 It accounts for one in three new cancers in women each year and is the second most common cause of cancer-related death in American women (the most common being lung cancer).2 Men can also get breast cancer and account for about 1% of cases.3 The American Cancer Society estimated that, in women in the United States, about 317,000 invasive breast cancer diagnoses and about 42,000 breast cancer-related deaths would occur in 2025.4
Fortunately, breast cancer treatment has made significant progress over the past several decades. Advances in screening and earlier diagnosis,5 targeted therapies and personalized medicine, as well as improved surgical techniques and greater knowledge about the importance of a healthy diet and lifestyle, mean that women now have a greater likelihood of positive outcomes than in decades past. For instance, the breast cancer death rate has fallen by 44% since 1989. Currently, about 86% of women live for at least 10 years after a breast cancer diagnosis in the United States.6
In this Life Extension Breast Cancer Treatment Protocol, you will learn about current and emerging treatment options, including the latest advances in precision medicine and how treatments can be tailored to your unique characteristics and preferences. You will also find a summary of the evidence on several dietary and lifestyle changes, as well as targeted nutritional interventions, that may have a role in the context of breast cancer treatment. Note that this overview does not cover breast cancer prevention. We at Life Extension encourage you to read and share this information with your healthcare team.
Other Life Extension Protocols that may be relevant for those receiving treatment for breast cancer include:
What Is Breast Cancer?
Breast cancer starts when some breast cells begin to grow out of control and form abnormal growths or lumps. Not all breast lumps are malignant (cancerous), but all lumps should be evaluated by a healthcare professional.1
Types of Breast Cancer
The breast is composed of different types of cells that form ducts, glands, and fatty tissue. The tissue type in which a cancer arises, in addition to individual characteristics of the patient and tumor, determine how breast cancers are classified and how treatment choices are made.1,3
Ductal cancers, which arise from the ductal structures of the breast that carry milk to the nipple, are the most common type of breast cancer, making up about 75% of all cases.1,7 Lobular cancers start in the glandular cells where breast milk is produced and make up about 8% of total breast cancer cases, and mixed ductal and lobular cancers account for another 5%. Cancers that arise in blood vessels or connective tissues of the breast are less common and may require a specialized treatment approach.1,7
Table 1. Breast Cancer Classification by Tissue of Origin
| Type | Characteristics |
|---|---|
| Ductal Breast Cancers | |
| Ductal carcinoma in situ (DCIS)8,9 |
Non-invasive or pre-invasive Does not extend beyond the duct wall |
| Invasive ductal carcinoma (IDC)1 |
Most common type of breast cancer Ductal cancer that has spread beyond the duct wall into the surrounding breast tissue |
| Lobular Breast Cancer | |
| Lobular carcinoma in situ (LCIS)3,10 | Benign; not classified as breast cancer |
| Invasive lobular carcinoma (ILC)1 | Has spread to the surrounding breast tissue Less common than IDC More likely to affect both breasts than other invasive breast cancers |
| Inflammatory Breast Cancer | |
| Inflammatory breast cancer (IBC)1 | Rare and aggressive type of invasive breast cancer Causes edema (swelling) and redness of the skin of the breast, giving the appearance of breast inflammation |
Ductal breast cancers
Ductal breast cancer, the most common type of breast cancer, arises in the ductal structures of the breast, usually from the epithelial cells that line the ducts.1
Ductal carcinoma in situ (DCIS) is a type of non-invasive or pre-invasive breast cancer that does not extend beyond the duct wall. DCIS tumors are usually found with a screening mammogram, though ultrasound, magnetic resonance imaging (MRI), and biopsy can also play a role in diagnosis.8,9 The diagnosis of DCIS has increased more than 10-fold since the widespread adoption of mammography screening. Approximately 20% of new breast cancer diagnoses are DCIS.1,8 Nearly all women with DCIS can be cured, but, if left untreated, about 40% of cases progress to invasive breast cancer.1,11
Invasive ductal carcinoma (IDC) is the most common type of breast cancer, making up approximately 70–80% of cases, and is characterized as ductal cancer that has spread beyond the duct wall into the surrounding breast tissue.1
Lobular breast cancer
Lobular breast cancer arises in a milk-producing gland, or lobule.1
Lobular carcinoma in situ (LCIS) is now considered a benign entity and is no longer classified as breast cancer in the most recent American Joint Committee on Cancer (AJCC) Cancer Staging Manual.3,10
Invasive lobular carcinoma (ILC) has spread to the surrounding breast tissue. It is less common than IDC, making up approximately 10% of total breast cancer cases, and is more likely to affect both breasts than other invasive breast cancers.1
Inflammatory breast cancer
Inflammatory breast cancer (IBC) is a rare and aggressive type of invasive breast cancer in which cancer cells block lymph vessels in the skin of the breast.1 This causes edema (swelling) and redness of the skin of the breast, giving the appearance of breast inflammation. About 1–5% of all breast cancers are IBC.1
Inflammatory breast cancer may not cause a discreet lump or mass and is sometimes not detectable on a mammogram. It is always invasive, at least locally, at the time of diagnosis since it has already spread into the skin. Compared with other types of breast cancer, IBC is more common in younger women, grows and spreads more rapidly, and has a worse prognosis.1
Classification by receptor subtypes
Receptors on the surfaces of breast cancer cells have become an important means of classifying tumors into subtypes to provide more personalized treatment strategies. Testing for the presence of these markers is done in all patients with invasive breast cancer who undergo biopsy or surgery.12
Table 2. Breast Cancer Classification by Receptor Subtype
| Receptor Status | Characteristics |
|---|---|
| Hormone receptor (HR)-positive or HR-negative12,13 | Breast cancers that express estrogen receptors (ERs) and/or progesterone receptors (PRs) in at least 1% of tested cells are HR-positive, while those that do not are HR-negative |
| Human epidermal growth factor receptor 2 (HER2)-positive or HER2-negative12,14 | Express high levels of HER2 (sometimes called ERBB2) or have an abnormally high number of the HER2 gene copies |
| HER2-low15,16 | HER2 expression just below the threshold for HER2-positivity and without an excess of the HER2 gene |
| Triple-negative1 | Breast cancers that have neither ERs, PRs, nor high levels of HER2 are called triple-negative breast cancer (TNBC) |
The breast cancer receptor subtypes are:
- Hormone receptor (HR)-positive or HR-negative. Breast cancers that express estrogen receptors (ERs) and/or progesterone receptors (PRs) in at least 1% of tested cells are HR-positive, while those that do not are HR-negative. They may be further classified as ER-positive or ER-negative and PR-positive or PR-negative. About 75% of women with breast cancer have tumors that are ER-positive, and more than 50% of these are also PR-positive, while very few cases are solely PR-positive.12,13 Hormone receptor status determines which treatments will be effective. ER-positive breast cancers are likely to respond to endocrine therapies that lower estrogen levels or block ERs.12,14 PR-positivity is associated with lower risk of recurrence and longer survival, while PR-negativity is usually an indicator of more aggressive cancer with poorer outcomes.13
- Human epidermal growth factor receptor 2 (HER2)-positive or HER2-negative. Breast cancers that express high levels of HER2 (sometimes called ERBB2) or have an abnormally high number of the HER2 gene copies are HER2-positive, while those that do not are HER2-negative. About 15–20% of breast cancers are HER2-positive. They are typically fast-growing but are likely to respond to HER2-targeted therapies, such as the monoclonal antibody trastuzumab (Herceptin).12 If the level of receptor expression is uncertain, a test that counts copies of the HER2 gene in cancer cells can be done. Tumors that have too many copies of HER2 are also classified as HER2-positive.12,14
- HER2-low. A new category of HER2 expression has recently emerged: HER2-low. Tumors with HER2 expression just below the threshold for HER2-positivity and without an excess of the HER2 gene can now be classified as HER2-low and may be susceptible to certain HER2-targeting therapies.15 Recent estimates suggest 45–55% of breast cancers are HER2-low, and most of these are HR-positive.16
- Triple-negative. Breast cancers that have neither ERs, PRs, nor high levels of HER2 are called triple-negative breast cancer (TNBC). Triple-negative breast cancer grows and spreads faster than other types of breast cancer. It does not respond to receptor-targeting therapies so is typically treated with chemotherapy, alone or in combination with immunotherapy. In general, TNBC is difficult to treat and has a worse prognosis than receptor-positive breast cancers. About 15% of breast cancers are TNBC.1
2 Diagnosis and Staging
Symptoms and Early Detection
Early-stage breast cancer can sometimes cause symptoms, but many cases are identified through screening in people who do not have symptoms.17
Signs and symptoms
The most common finding related to breast cancer is a new irregularly shaped lump or change that can be felt in the breast. These lumps are often painless and hard but can also be soft, painful, tender, or more rounded, and all new breast lumps should be evaluated by a healthcare professional. Other symptoms may include swelling, skin dimpling (feels like the skin of an orange), skin reddening, changes in the size or shape of the breast, pain in the breast or nipple, other nipple changes (eg, nipple turning inward, flaking and/or red skin, discharge), and swollen lymph nodes under the arm or above the collar bone.14,17
Breast self-examination
There is little evidence that regular breast self-examination decreases breast cancer mortality, but it is generally recommended that women be familiar with the way their breasts look and feel in order to detect any changes that might arise.8,17 Many women like to keep track of their breast health by doing breast self-exams on a regular basis; for example, while showering or dressing. However, there is no firm recommendation on when and how to do this.17
Screening methods
Breast cancer is often easier to treat successfully when it is detected at an early stage, before the tumor has grown and spread. Regular screening can help identify breast cancer early.17
Screening recommendations vary depending on age and the number of risk factors. Women who have a strong family history of breast cancer, a genetic mutation known to increase breast cancer risk (eg, BRCA1 or BRCA2), or received chest radiation before the age of 30 are considered to be at high risk, while women without any of these are considered to be at average risk.17
Most breast cancer screening recommendations in the United States agree that women with average risk should have screening mammograms every one to two years from the age of 40–44 years until age 69.18 Many recommendations also suggest healthcare providers perform annual breast exams for women under 40.18 Generally, mammograms are suggested every one to two years after age 70 for women who have at least 10 years of remaining life expectancy.18 In women with high breast cancer risk, a screening mammogram and breast MRI are recommended annually starting at age 30–35, depending on family and patient history.17,18
Mammograms
A mammogram is a low-dose X-ray of the breast that can detect abnormalities in breast tissue.8,17 In the United States, about 10% of women who undergo mammography are asked to return for additional tests, such as a biopsy to further evaluate an abnormality seen on the mammogram.8 During a mammogram, the breasts are compressed, one at a time, between two plates in order to reduce the thickness of the tissue and, thus, the amount of radiation needed to get a good image of the breast.8 Two images of each breast are generally taken.17
A three-dimensional (3D) mammography technique called breast tomosynthesis or digital breast tomosynthesis (DBT) is increasingly being used as an alternative to standard two-dimensional (2D) mammography screening. In DBT, a number of 2D images are taken at various angles and then reconstructed by a computer to provide a 3D image.8,17 This technique may reduce the number of women who are called back for further evaluation and may be more helpful for women with dense breast tissue.17 Several large studies are being conducted to test the value of this new technique compared with conventional 2D imaging.8,17
Ultrasound
Ultrasound is a non-invasive imaging technique that uses sound waves generated by a handheld instrument that is moved over the skin to detect differences in tissue densities. Ultrasound is not typically used for screening but can be helpful if a woman feels a change in her breast that is not detected by mammography. Ultrasound can also be used in addition to standard mammography in women with dense breast tissue. Mammography is less sensitive at detecting small masses in dense breast tissue, which is present in 43% of women age 40–74.8,19 Follow-up ultrasound may also be recommended if a suspicious area is detected on a mammogram, since ultrasound is better at distinguishing benign changes, such as fluid-filled cysts, from potentially cancerous solid masses.19
Magnetic resonance imaging
Magnetic resonance imaging (MRI) is an imaging tool that uses radio waves and strong magnets to detect tissue differences.20 Its use, in conjunction with mammography, is recommended as a screening method for women at high risk for breast cancer, since these techniques complement each other.17,18 MRI can detect some cancers that cannot be identified by mammography but is also more likely to generate false positive findings that result in patients being subject to unnecessary testing, such as biopsy.17
Diagnostic Procedures
Breast cancer diagnosis involves a series of tests to confirm that a symptom or suspicious area detected in the breast during screening is breast cancer. Once the diagnosis is confirmed, additional tests are done to classify the tumor and determine whether it has spread within the breast or to other parts of the body. These are all important considerations in determining the stage of the cancer, the patient’s prognosis, and the treatment approach.14,17
Clinical examination
Clinical examination for the possibility of breast cancer involves the healthcare provider gathering a medical history, performing a thorough physical examination, including a breast exam, and ordering imaging tests to get a complete picture of the patient’s disease. In addition, a biopsy provides critical information about the tumor, and blood tests may be ordered to assess a patient’s general health.14
Imaging techniques
The imaging techniques used in diagnosis of breast cancer are the same as those used in screening, including mammogram, ultrasound, and MRI, and their use may vary depending on the type of breast cancer suspected and features of the area, such as size and depth within the breast.21 Ultrasound and MRI may also be used to guide biopsy procedures.17
Biopsy techniques
Biopsy is an important aspect of cancer diagnosis and involves removing a small sample of the suspicious breast area in order to analyze it in the laboratory to confirm whether or not cancer is present.17 There are different types of biopsy, depending on where the suspected area is, its size, whether there is more than one suspicious area, and the patient’s health and preferences.17
Most biopsies are done with a needle in the office rather than as surgical procedures. Fine needle aspiration biopsy involves the insertion of a very fine needle into the breast to remove a liquid or small tissue sample. Core needle biopsy uses a larger needle and removes a larger sample than fine needle aspiration.17 Ultrasound or MRI may be used to guide these procedures. Sometimes, surgical biopsy is required to remove the lump or area of concern from the breast. A biopsy sample of lymph nodes under the arm may also be taken, as the spread of cancer to other parts of the body often occurs via the lymph system.14,17
Evaluation for metastatic disease
When a cancer spreads from the place it originally developed, such as the breast, to another part of the body, this is termed metastatic disease.14 If metastatic disease is suspected, whole-body imaging and possibly biopsy procedures may be performed, depending on the results from the original diagnostic testing, patient symptoms, and physical exam findings.22
Genetic Testing
About 5–10% of breast cancers are related to inherited (germline) gene mutations. Genetic testing can be helpful for understanding how breast cancer might respond to treatment, whether targeted therapies might be appropriate, and the risk of other cancers.23
Variants (or mutations) of two BRCA genes, BRCA1 and BRCA2, are important inherited contributors to breast cancer risk in women and men. Because the proteins encoded by BRCA1 and BRCA2 are involved in DNA repair and tumor suppression, mutations can lead to abnormal cell growth and increase the likelihood of cancer. In fact, the presence of a BRCA mutation raises a woman’s risk of breast cancer to as high as 70% by the age of 80 years. It also increases the risk of ovarian and other cancers.23 Other germline mutations associated with increased breast cancer risk include ATM, PTEN, CHEK2, CDH1, and PALB2.23
Genetic testing can help identify people with a familial risk of breast cancer. While some experts recommend genetic testing be offered to all patients with breast cancer, the National Comprehensive Cancer Network suggests genetic testing be reserved for patients who have or have had breast cancer and meet any of the following criteria22:
- Breast cancer diagnosed at age ≤50 years old
- Triple-negative breast cancer
- Multiple primary breast cancers
- Lobular breast cancer in addition to a personal or family history of gastric cancer
- Male breast cancer
- Ashkenazi Jewish ancestry
- Family history (ie, first-, second-, and third-degree relatives on the same side of the family) of breast cancer at ≤50 years old, male breast cancer, ovarian cancer, pancreatic cancer, or metastatic or high-risk prostate cancer
- Family history of at least three relatives with either breast or prostate cancer
Testing is done using a blood or saliva sample and usually screens for a panel of different gene mutations. The results provide complex information that should be reviewed with an experienced professional. Importantly, this information may also be relevant to other family members, as they may have inherited the same gene variants.23
Women without breast cancer who are found to have BRCA1, BRCA2, or some other germline mutations, have prevention options, including estrogen-blocking therapy (typically with tamoxifen or a related drug) or prophylactic surgery to remove the breasts (mastectomy). However, in a meta-analysis of data from 21 studies involving BRCA mutation carriers, risk-reducing mastectomy was found to lower breast cancer risk by 87%, but tamoxifen therapy did not significantly reduce risk.24
Staging
Prognostic staging has an important role in treatment decision-making. The American Joint Committee on Cancer is responsible for developing cancer staging standards. Their most recent prognostic staging system, updated in 2018, takes into account the tumor’s anatomical status (TNM classification), cellular features (histological grade), molecular biomarker (ER, PR, and HER2) status, and genomic profile.
TNM classification system
The tumor, node, metastasis (TNM) system is an anatomic staging system used to classify many types of cancer, including breast cancer. Combining the tumor’s gross physical characteristics with data on outcomes allows for categorization into groups based on the predicted course of the disease and probable outcomes.10
TNM staging is based on information about tumor size and degree of invasion, whether the cancer has spread to lymph nodes on the same side of the body, and whether distant metastases are present.10,25
Table 3: Tumor, Node, Metastasis (TNM) Classification System10,26
| Tumor Classification | Node Classification | Metastasis | |
|---|---|---|---|
|
Tx – Primary tumor cannot be assessed |
Nx – Regional lymph nodes cannot be assessed |
M0 – no clinical or radiographic evidence of distant metastases |
|
|
T0 – No evidence of primary tumor |
N0 – No lymph node metastases |
M1 – distant metastases confirmed by clinical and radiographic means or histology |
|
|
Tis – Carcinoma in situ (non- or pre-invasive) |
N1 – Metastasis or micrometastasis involving 1–3 axillary (under the arm) lymph node(s) |
||
|
T1 – Tumor ≤20 mm in greatest dimension |
N2 – Metastasis involving 4–9 axillary lymph nodes or any internal mammary (along the sternum) lymph nodes |
||
|
T2 – Tumor >20 mm but ≤50 mm in greatest dimension |
N3 – Metastasis involving 10 or more axillary lymph nodes (or any in the group closest to the clavicle), a combination of axillary and internal mammary lymph nodes, or any supraclavicular (above the clavicle) lymph nodes |
||
|
T3 – Tumor >50 mm in greatest dimension |
|||
|
T4 – Tumor of any size with direct extension to the chest wall and/or the skin |
Table 4: Breast Cancer Anatomic Stages
| Stage | Definition |
|---|---|
| Stage 0 |
Tis, N0, M0 |
| Stage I |
IA: T1, N0, M0 IB: T0, N1(micro), M0 IB: T1, N1(micro), M0 |
| Stage II |
IIA: T0, N1, M0 IIA: T1, N0, M0 IIA: T2, N0, M0 IIB: T2, N1, M0 IIB: T3, N0, M0 |
| Stage III |
IIIA: T0, N2, M0 IIIA: T1, N2, M0 IIIA: T2, N2, M0 IIIA: T3, N1, M0 IIIA: T3, N2, M0 IIIB: T4, N0, M0 IIIB: T4, N1, M0 IIIB: T4, N2, M0 IIIC: any T, N3, M0 |
| Stage IV |
any T, any N, M1 |
In prognostic staging, these anatomic groupings are modified by information about grade, receptor status, and genomic profile when appropriate and available, which may result in upstaging (for example, in the case of TNBC) or downstaging (for example, in the case of triple receptor positivity: ER+, PR+, and HER2+).25 For women who have not undergone breast cancer surgery, clinical prognostic staging is performed based on results of physical exam, imaging, and biopsy. Pathologic prognostic staging is based on findings from surgery, which provide a more accurate assessment of cancer status.27
Histologic grading
Histological grading is a standardized system for classifying tumors based on cellular characteristics. Tumor cells from biopsy samples are evaluated microscopically and scored based on the degree to which they have changed from normal functional cells. Those scores are then translated into three grades: grade 1 describes a tumor that is well-differentiated (has a high degree of normalcy) with little sign of cancerous change; grade 2 describes a moderately differentiated tumor with a moderate degree of abnormality or cancerous change; and grade 3 describes a poorly differentiated tumor with aggressive cancerous change.22,25
Molecular biomarkers and genomic profile
The presence or absence of the molecular biomarkers ER, PR, and HER2 have well-established implications for treatment responsiveness and breast cancer prognosis. Another protein biomarker called Ki-67 is an indicator of cell proliferation. High expression of Ki-67 has been correlated with more aggressive cancer; however, accurate methods for assessing Ki-67 expression have yet to be standardized.28
Breast cancer can be categorized according to its molecular and genetic characteristics to better understand prognosis and guide treatment. Table 5 describes the main characteristics of the four molecular subtypes of breast cancer: luminal A, luminal B, HER2-positive, and triple negative. In addition, patterns of non-coding RNA (RNA fragments, such as microRNAs, that are not used for synthesizing proteins) have been associated with these subtypes. MicroRNAs are known to have a significant role in cancer cell function and may provide information about future therapeutic targets.29
Table 5: Characteristics of Molecular Subtypes of Breast Cancer13
| Luminal A | Luminal B | HER2-positive | Triple Negative | |
|---|---|---|---|---|
| % of cases | ~50 | ~15 | ~20 | ~15 |
| ER-positive | yes | yes | sometimes | no |
| PR-positive | yes | sometimes | sometimes | no |
| HER2-positive | no | no | yes | no |
| Germline Mutations | — | BRCA2 |
— |
BRCA1 |
| Ki-67 expression | variable | variable | high | high |
| Prognosis | good | medium | medium to poor | poor |
| Systemic therapy options | endocrine therapy | endocrine therapy plus chemotherapy | endocrine, chemotherapy, and Herceptin | chemotherapy or immunotherapy |
Genomic testing is used to detect gene mutations in tumor DNA. These mutations are not inherited but have been acquired since birth or arisen spontaneously in tumor cells. Tumor cells develop many gene mutations because they divide rapidly, and the tumor genomic profile is likely to change over time. Some of these mutations contribute to cancer cell aggressiveness and treatment resistance.30,31
Genomic profiling panels have been developed that can further individualize treatment decisions based on tumor gene patterns. For example, Oncotype DX is a multigene profiling test specifically used to predict risk of recurrence and the potential benefit of chemotherapy in patients with early-stage, ER-positive, HER2-negative (luminal A and luminal B subtypes) breast cancer. For patients with these breast cancer subtypes, the Oncotype DX score, if available, can be factored into prognostic staging and may be helpful for preventing over- and undertreatment.25,32 Other genomic profiling panels include MammaPrint, Prosigna, and EndoPredict.33
Prognostic tools
A tool known as the Clinical Treatment Score post-5 years (CTS5) is used to evaluate the risk of distant recurrence in women with ER+ breast cancer who have completed five years of endocrine therapy. It is calculated using clinical information including patient age, tumor size, quadratic tumor size (tumor size squared [multiplied by itself]), node status, and histologic grade.34,35 Another tool called the Breast Cancer Index (BCI) has been shown to predict which early-stage ER+ patients are likely to benefit from long-term (>5 years) endocrine therapy. It is calculated using genetic information from two tests.33,36 CTS5 and BCI may be especially helpful for deciding whether to prolong endocrine therapy beyond five years in low- and intermediate-risk patients.34,37,38
3 Treatment
Overview
Breast cancer is a challenging diagnosis with treatment protocols that frequently involve multiple modalities, each with potential risks and possible benefits to be thoroughly considered and weighed by the patient and their care team. Good treatment decision-making takes into account a wide range of factors, such as stage and prognosis, previous treatments, possible side effects, length of treatment, and the patient’s health.14 In addition, it is vital to integrate patient values, preferences, and goals into treatment decision-making.39
Standard treatment options usually include some combination of surgery, radiation therapy, and systemic therapy (drugs that enter the bloodstream). Systemic therapy may consist of conventional chemotherapy with drugs that act generally to block cell replication, affecting cancer cells as well as other rapidly dividing cells in the body, or more targeted therapies, like endocrine therapy or immunotherapy (eg, anti-HER2 therapy), that take advantage of specific biomarkers and features of breast cancer cells to target and neutralize them. Patients may also be referred to participate in clinical trials of new treatments.14
Surgical Options
Most breast cancer patients undergo surgery to remove the tumor. The goals of surgery may include removal of the cancer, finding out whether the cancer has spread to lymph nodes or other tissues, restoring the shape of the breast after surgery, and relieving the symptoms of the cancer.40 In some patients with small, low-grade DCIS tumors with a low risk of recurrence, surgery may be the only treatment needed.11
Lumpectomy vs. mastectomy
Depending on the characteristics of the tumor (eg, size, invasiveness, location), the patient may undergo removal of the tumor in a breast-conserving procedure called lumpectomy. This is also known as a partial mastectomy, segmental mastectomy, quadrantectomy, or breast-sparing surgery.14,22 Lumpectomy also involves removal of some normal tissue around the tumor to provide a margin of confidence that all of the cancer has been removed. Narrow surgical margins are associated with increased risk of cancer recurrence, and a wider excision may be indicated for cases with aggressive features.21,22
Lumpectomy is often done in combination with radiation to reduce the risk of cancer recurrence. Studies have demonstrated this practice results in similar survival outcomes as removal of the whole breast (mastectomy) in women who have early-stage cancer and are candidates for either type of surgery.40
In some cases, it is necessary or preferable to remove the entire breast in a procedure called mastectomy. There are several types of mastectomy22:
- Simple mastectomy, in which the breast is removed but the chest muscles and axillary lymph nodes are spared
- Skin-sparing mastectomy, a variation of a simple mastectomy in which enough skin is left to cover the wound and potentially facilitate breast reconstruction
- Nipple-sparing mastectomy, like skin-sparing mastectomy, but the nipple and areola are spared for cosmetic reasons
- Modified radical mastectomy, in which some axillary lymph nodes are removed but the chest muscles are spared
- Radical mastectomy, in which the breast, chest muscles, and axillary lymph nodes are removed; used only in cases with invasion of the chest wall
Double or bilateral mastectomy, in which both breasts are removed, is sometimes performed when the risk of developing a second breast cancer is very high. Women with very high risk, such as those with BRCA gene variants, may opt for prophylactic bilateral mastectomy before a breast cancer diagnosis as a preventive measure.22,40 Prophylactic mastectomy has been shown to reduce breast cancer risk by 87% in BRCA mutation carriers.24
Sentinel lymph node biopsy vs. axillary lymph node dissection
Lymphatic fluid leaving the breast tissue is mainly filtered by a large group of lymph nodes under the arm and below the clavicle known as axillary lymph nodes. Because axillary lymph nodes are often the first site of breast cancer metastasis, their evaluation and monitoring are a critical part of breast cancer management.3,26
In most cases, ultrasound with biopsy is the first step in evaluating axillary lymph nodes. If spread to lymph nodes is absent or limited, this may be followed by sentinel node biopsy, in which a few critical nodes are removed and examined.26,41 Sentinel lymph nodes are the first few lymph nodes to filter fluid from the tumor environment and are generally identified by injecting tracers near the tumor and seeing which lymph node or nodes receive them first.41 In sentinel node biopsy, one or a few (usually two to four) sentinel nodes are identified and removed, often during partial or full mastectomy, and examined for the presence of cancer cells.42 If no cancer is detected, no further axillary surgery is needed.41
Axillary lymph node dissection involves surgical removal of a large number of axillary lymph nodes. It may be offered if initial evaluation shows extensive node involvement or if cancer is found on sentinel node biopsy.26,41 However, axillary lymph node dissection can cause side effects, such as edema (swelling due to fluid retention), limited arm movement, and nerve pain, which may have a substantial negative impact on quality of life. Radiation treatment to the axilla is an alternative to axillary lymph node dissection for some early breast cancer patients found to have cancer in one to two sentinel nodes and has been shown to result in similar survival outcomes.41,42
Reconstructive surgery options
Patients may choose to undergo breast reconstructive surgery at the time of initial breast surgery or later. Reconstructive surgery helps restore the breast’s shape using the patient’s own tissue (muscle, fat, and skin) or a breast implant. Reconstruction has been associated with better mental health and quality of life in patients who have undergone mastectomy. It is best to discuss reconstruction preferences early in treatment planning since timing may need to be coordinated around cancer treatments.22,40,43
A variety of factors affect whether breast reconstruction is appropriate, including patient preference, smoking, other medical conditions, and plans for radiation therapy. For example, smoking and obesity increase the risk of complications after breast reconstruction.21 Women who do not choose reconstruction due to personal preferences, medical issues, or other factors can opt for a surgical closure that leaves a flattened chest wall, and may or may not want to use cosmetic external breast prostheses.44
Radiation Therapy
Indications and types
Radiation therapy involves using high-energy X-rays or other types of radiation to kill cancer cells.14 In external beam radiation therapy, radiation is delivered by a machine from outside the body, while in brachytherapy or internal radiation, it is delivered under the skin by a needle, seed, wire, or catheter. The type and focus of the radiation therapy depends on the type of surgery (breast-conserving or mastectomy), involvement of lymph nodes, and individual patient and tumor factors, such as age, hormone receptor status, and preference.40
Radiation therapy is an important part of breast cancer treatment for many patients. It can reduce the chance of recurrence after breast-conserving surgery and may extend survival. Radiation is indicated after mastectomy in patients when the breast tumor is 5 cm or larger, when the cancer has spread to axillary lymph nodes, or when it is found to extend into surgical margins.22 For patients whose breast cancer does not have these features, decisions around radiation therapy are made on a case-by-case basis.
A growing body of evidence indicates radiation therapy may be unnecessary for some women with early-stage ER-positive tumors who receive surgery followed by five years of endocrine (estrogen-blocking) therapy.45,46
There are many different dosing schedules and strategies for delivery of radiation therapy designed for different situations.40 External beam radiation therapy is the more common mode of delivery and may be administered to the whole breast or the part of the breast closest to the tumor. It may also be applied to lymph nodes.40 Brachytherapy may be an alternative to whole breast irradiation for some women who received breast-conserving surgery. The most common brachytherapy strategy in breast cancer is called intracavitary brachytherapy, in which a catheter is used to connect the outside of the breast to the cavity where the tumor was removed. The catheter is left in place, allowing radiation to be applied directly to the tumor area (usually twice per day for five days) until treatment is complete and the catheter is removed.40
Side effects
Radiation can cause a range of dose-related side effects by damaging healthy tissues near the tumor. The most common side effects of external beam radiation therapy are short-term and include breast swelling, skin changes similar to a sunburn, and fatigue. Long-term changes in breast texture and appearance and an inability to breastfeed may also occur. Axillary lymph node irradiation can cause chronic difficulty with edema in the arm or chest. In rare cases, radiation can damage ribs leading to weakness and fracture.40
Some women experience chronic numbness, pain, and weakness in the shoulder, arm, and hand due to radiation-induced nerve damage. The onset of these nerve problems can be many years after the end of treatment and may persist indefinitely.47 Radiation can also cause scarring in the chest muscles, pleura (lining of the chest cavity), lung tissue, or heart muscle. With modern radiation techniques, these effects are less likely and less severe than in the past, but long-term health implications, such as increased cardiovascular risk in women treated for right-side breast cancer, have been noted.48-50
Brachytherapy is more invasive, but historically more targeted, than external beam radiation therapy and is associated with a somewhat different set of side effects. These include redness or bruising near the site of treatment, breast pain, infection, loss of fatty tissue in the breast, and pockets of fluid accumulation (seromas).40 One advantage of brachytherapy is lower radiation exposure in nearby tissues and organs.51 Nevertheless, brachytherapy has been found to cause rib weakness and fracture slightly more often than external beam radiation.40,51,52
Systemic Therapies
Systemic therapies encompass a wide variety of cancer therapies that are administered directly into the bloodstream intravenously or taken orally. They are called systemic because they reach the whole body, not just the breast. Systemic therapies used in breast cancer include general cancer-killing chemotherapies, endocrine therapies that target ERs, and other targeted therapies designed to take advantage of breast cancer-specific markers, such as the HER2 receptor.40
Systemic chemotherapy that is administered before surgery is called neoadjuvant chemotherapy. Neoadjuvant chemotherapy may be recommended for large tumors to reduce their size prior to surgery, for breast cancer involving many lymph nodes, and for inflammatory breast cancer. Systemic therapies administered after surgery to target residual cancer cells and reduce the risk of recurrence and spread are called adjuvant therapies.40 In advanced and metastatic breast cancer, systemic therapies may be used to reduce further spread, relieve pain and other symptoms, extend life, and improve quality of life.11 Decisions about which systemic therapies to use depend on the patient’s cancer subtype and stage, genomic profile, and individual factors, including reproductive stage, health status, and personal preferences.53-56
Chemotherapy
Chemotherapy is indicated for many patients with invasive breast cancer that has spread to lymph nodes or other parts of the body. In patients with HR-positive, HER2-negative breast cancer, molecular testing may be used to decide whether chemotherapy is likely to be helpful. Chemotherapy is sometimes used prior to hormone therapy or with targeted therapy.11
Many chemotherapy agents used in breast cancer are the same as those used in other cancers. Agents used for neoadjuvant or adjuvant therapy include11,40:
- Anthracyclines, such as doxorubicin (Adriamycin) and epirubicin (Ellence), which inhibit DNA replication and generate free radicals that damage cancer cells
- Taxanes, such as paclitaxel (Taxol) and docetaxel (Taxotere), which interfere with cell division
- Alkylating agents, such as cyclophosphamide (Cytoxan) and carboplatin (Paraplatin), which interfere with normal DNA function
- Fluoropyrimidines, such as 5-fluorouracil (5-FU) and capecitabine (Xeloda), which interfere with DNA replication and repair
A number of other chemotherapy drugs may be used to treat breast cancer that has metastasized.40
By disrupting cell replication, these drugs exert their main toxic effects on cancer cells, which rely on rapid cell division. However, they can also harm healthy cells that divide rapidly, such as skin, hair, and bone marrow cells and the cells lining the digestive tract. This is why these medications cause side effects like hair loss, rashes, mouth sores, nausea and vomiting, diarrhea, and low white blood cell numbers.11,14,40 Other possible side effects of chemotherapy include nail changes, loss of appetite, weight changes, fatigue, hot flashes, and nerve damage.40 Some side effects are severe enough to limit treatment.
Although most side effects resolve after treatment is finished, serious long-term effects can occur following chemotherapy, including infertility, cardiomyopathy due to heart damage, neuropathy due to nerve damage, and diseases of the bone marrow, like leukemia. In some cases, these effects occur many years after the end of chemotherapy.40
Chemotherapy drugs are often used in combinations that take advantage of their different mechanisms of action. A variety of drug combinations are used, depending on tumor characteristics and individual patient factors.33 They are administered in cycles separated by rest periods. The length of treatment is usually three to six months for neoadjuvant or adjuvant treatment, but can vary for metastatic disease.40
Endocrine therapy
Breast cancers that express high levels of estrogen and/or progesterone receptors depend on estrogen signaling to drive their growth. Endocrine (hormonal) therapies targeting estrogen signaling by blocking or breaking down ERs or by reducing synthesis of estrogen are widely used to interrupt cancer cell growth in ER-positive breast cancers.40 Endocrine therapy is usually recommended as an adjuvant (post-surgery) therapy to prevent recurrence in patients with luminal-A or luminal-B breast cancer subtypes, as well as those with HER2-positive tumors that are ER-positive.54 Emerging research suggests endocrine therapy targeting androgen receptors may improve outcomes in triple-negative breast cancer cases with high expression of androgen receptors.57
It is important to recognize that expression of progesterone receptors depends on estrogen signaling, and progesterone receptor activity modulates the activity of ERs. While progesterone receptors are an important, generally favorable, biomarker of prognosis in breast cancer, they are not directly targeted by endocrine therapies.58 Endocrine therapy is sometimes recommended for those rare patients with ER-negative/PR-positive breast cancer, but its efficacy is uncertain.54
Endocrine therapies are associated with a host of side effects related to changes in estrogen signaling. These can include hot flashes, vaginal dryness, altered menstrual cycles, sexual dysfunction, mood difficulties, cognitive dysfunction, insomnia, fatigue, weight gain, and pain.40,59 A substantial proportion of women report profound deterioration of quality of life during long-term endocrine therapy. Although these types of side effects diminish over time for some patients, they are a major reason for stopping therapy. Better support for patients taking long-term endocrine therapy may improve adherence, resulting in greater treatment benefits.59,60
Selective estrogen receptor modulators (SERMs)
Selective estrogen receptor modulators, or SERMs, block estrogen from binding to ERs. Tamoxifen (Nolvadex, Soltamox, et al.) is the most commonly used SERM and is used in both pre- and postmenopausal women with ER-positive breast cancer. Toremifene (Fareston) is also a SERM but is only approved for use in postmenopausal women with metastatic disease.40
The use of tamoxifen has been found to reduce risk of death by 25–31% and risk of recurrence by 41% annually. Five years of treatment has been shown to be more beneficial than one or two years, and 10 years of treatment has been found to improve outcomes even more for certain patients at high risk of recurrence.22,54 Tamoxifen therapy is recommended for five years for patients with ER-positive DCIS and up to 10 years for patients with invasive ER-positive breast cancers. Considerations such as menopausal status, risk of recurrence, and adverse side effects are key to deciding how long to extend therapy for each individual patient.11
Common side effects of SERMs are related to reduced estrogen signaling and are listed above. In addition, SERMs affect estrogen signaling differently in different parts of the body, so can also cause certain pro-estrogenic side effects.61 These include increased risk of endometrial cancer and uterine sarcoma in postmenopausal women as well as blood clots, such as deep vein thrombosis in the leg, which can progress to a life-threatening pulmonary embolism. In rare cases, SERMs are associated with stroke. Their use has also been linked to eye problems, such as cataracts. SERMs may increase bone density in postmenopausal women but may decrease bone density in premenopausal women. It is important to note that the benefits of taking these drugs almost always outweigh the potential risks.40
Aromatase inhibitors
Aromatase is a key enzyme involved in the production of estrogen by cells outside of the ovaries, particularly fat cells. It facilitates the conversion of testosterone into estrogen, a pathway that accounts for a substantial proportion of estrogen synthesis in postmenopausal women. Aromatase inhibitors stop most non-ovarian estrogen production in the body and are an important option for long-term therapy in many cases of ER-positive breast cancer.22,40
In postmenopausal women, five to 10 years of aromatase inhibitor therapy can be used instead of tamoxifen. Alternatively, an aromatase inhibitor may be used before or following tamoxifen to achieve a total of up to 10 years of endocrine therapy. Aromatase inhibitors have demonstrated some outcome-related advantages over tamoxifen, but treatment decisions are sometimes influenced by side effects or treatment resistance. Because they do not affect ovarian estrogen synthesis, aromatase inhibitors are not effective in premenopausal breast cancer patients except in conjunction with ovarian suppression therapy.11,54 Examples of aromatase inhibitors include letrozole (Femara), anastrozole (Arimidex), and exemestane (Aromasin).
Side effects of aromatase inhibitors include the estrogen-depletion symptoms listed above, as well as muscle, bone, and joint pain that sometimes causes patients to discontinue treatment early. In such cases, switching to tamoxifen may be an option. Aromatase inhibitors also contribute to bone loss and increased fracture risk. This is sometimes managed with bone-protective medication.40
The optimal duration of aromatase inhibitor therapy has been the subject of several studies. In most cases, aromatase inhibitor therapy is recommended for at least five years and may be extended for as many as five more years. Prognostic tools, such as CTS5 or BCI, are useful for predicting the likely benefit of extending treatment.34,36 Because the risk of side effects may outweigh additional benefits beyond years seven to eight, an individualized approach is important, weighing the potential benefit a patient may derive from extended aromatase inhibitor therapy against the potential for side effects, such as fractures.62-66
Selective estrogen receptor degraders (SERDs)
The selective estrogen receptor degraders (SERDs) fulvestrant (Faslodex) and elacestrant (Orserdu) are a newer class of drugs that interrupt estrogen signaling by binding to ERs and causing them to break down. These medications are used primarily in postmenopausal women but may be recommended in premenopausal women undergoing ovarian suppression therapy to medically induce menopause. Fulvestrant may be recommended instead of a SERM or aromatase inhibitor, or for patients whose ER-positive tumors have become resistant to another type of endocrine therapy.40 It may also be used in combination with other systemic therapies for metastatic breast cancer. On the other hand, elacestrant is only approved for select patients with ER-positive/HER2-negative tumors that display a specific gene mutation (ESR1) associated with endocrine therapy resistance and have demonstrated resistance to one or more other endocrine therapies.40,61
In addition to hormonal side effects listed above, SERDs can be associated with nausea and pain in the muscles, joints, and bones. Elacestrant use has also been associated with increased cholesterol levels. Patients using fulvestrant, which is given by intramuscular injection, may experience pain at the injection site.40
Ovarian suppression
Ovarian suppression is used in conjunction with an aromatase inhibitor, or sometimes tamoxifen, to treat premenopausal women with ER-positive breast cancer and a high recurrence risk, such as due to young age, high-grade cancer, or lymph node involvement. Ovarian suppression plus tamoxifen or an aromatase inhibitor is generally recommended for five years, followed by up to five years of only tamoxifen, or an aromatase inhibitor if menopause has occurred during treatment.54
Luteinizing hormone-releasing hormone (LHRH) agonists such as goserelin (Zoladex) and leuprolide (Lupron) disrupt normal feedback signaling between the ovaries and the brain, dramatically decreasing ovarian estrogen production and inducing temporary menopause. Alternatively, some women opt for surgical removal of the ovaries, leading to permanent menopause. In some cases, chemotherapy damages ovarian function and induces menopause as a side effect, which may be temporary or permanent. Ovarian suppression results in the estrogen-depletion symptoms common to all endocrine therapies.40
Targeted therapy
The promise of personalized cancer care depends largely on targeted treatment options. This is an area of active research as molecular targets are discovered and new agents directed at them are developed.
HER2-targeting antibodies
Monoclonal antibodies that target HER2, including trastuzumab (Herceptin), pertuzumab (Perjeta), and margetuximab (Margenza), are used to treat patients with HER2-positive breast cancer. These drugs are administered intravenously and may be used alone or in combination with chemotherapy, as well as endocrine therapy when indicated, for early or advanced-stage breast cancer.40
HER2-positive breast cancer patients undergoing adjuvant (post-surgery) chemotherapy are generally co-treated with trastuzumab. This is followed by trastuzumab alone, once every three weeks, for up to one year. Trastuzumab can also be part of neoadjuvant (pre-surgery) treatment for large tumors or those with substantial node involvement.11,40,54 Combining trastuzumab with pertuzumab may slightly improve outcomes in high-risk cases.11,22,54 Margetuximab use is typically limited to advanced breast cancer cases that have not responded to other HER2-targeted therapies.40
The most serious side effect of trastuzumab is cardiac damage. While cardiotoxic effects of trastuzumab usually cause no symptoms and are reversible after therapy ends, it has been associated with life-threatening congestive heart failure in rare cases.67 The risk of lasting heart damage is compounded in patients who also receive cardiotoxic chemotherapy, such as anthracycline, paclitaxel, or cyclophosphamide.68,69 Trastuzumab is not safe in pregnancy.40 Other potential side effects include respiratory problems, diarrhea, indigestion, sleep difficulty, hot flashes, peripheral neuropathy (numbness, tingling, or burning sensation in the hands, feet, arms, or legs), and pain in the joints, muscles, or bones.69
Antibody-drug conjugates are another HER2-targeting treatment option. They are made by attaching HER2-targeting monoclonal antibodies, such as trastuzumab, to chemotherapy drugs. This allows the HER2-targeting antibody to deliver the chemotherapy drug directly to cancer cells. Antibody-drug conjugates include trastuzumab-emtansine (Kadcyla) and trastuzumab-deruxtecan (Enhertu). They are generally reserved for use in cases of advanced metastatic breast cancer that have progressed despite first-line treatment with trastuzumab plus chemotherapy,40 although trastuzumab-emtansine is also used in certain cases of early-stage breast cancer.70
In an important clinical trial, trastuzumab-deruxtecan improved treatment outcomes in breast cancer patients whose tumor expression of HER2 was just below the threshold needed to be classified as HER2-positive and were therefore classified as HER2-low.71 This led to the 2024 Food and Drug Administration (FDA) approval of trastuzumab-deruxtecan for advanced HER2-low breast cancer, regardless of hormone receptor status.61
Common side effects of trastuzumab-drug conjugates include nausea, decreased appetite, fatigue, and headache. These medications can also cause lung, liver, and bone marrow toxicity.72
Kinase inhibitors
Kinase inhibitors are a broad class of drugs that inhibit enzymes called kinases. Kinases activate a range of cellular processes, including metabolism, signaling, growth, and division. Kinase inhibitors are widely used in cancer treatment, including breast cancer.
Tyrosine kinase inhibitors. HER2 is a type of enzyme called a tyrosine kinase, and some tyrosine kinase inhibitors (TKIs) have been shown to inhibit HER2. These include lapatinib (Tykerb), neratinib (Nerlynx), and tucatinib (Tukysa).40,68 Tyrosine kinase inhibitors have been shown to improve outcomes in advanced or metastatic HER2-positive breast cancer patients who have stopped responding to trastuzumab. Clinical trials have indicated these TKIs help restore sensitivity to trastuzumab; therefore, TKIs and trastuzumab are often used together even after resistance to trastuzumab has been displayed. They are generally used along with chemotherapy, as well as endocrine therapy when appropriate. In some cases of early-stage breast cancer, neratinib may also be recommended. Common side effects of TKIs include diarrhea, nausea, vomiting, and rash.68 Some TKIs can also cause hand-foot syndrome, marked by soreness, redness, blistering, and peeling of the hands and feet.40
CDK4/6 inhibitors. These oral medications target cyclin-dependent kinases (CDKs) 4 and 6, which, like other CDKs, play a critical role in controlling the cell division cycle. Rapidly dividing breast cancer cells are particularly vulnerable to CDK4/6 inhibitors.73
Palbociclib (Ibrance), ribociclib (Kisqali), and abemaciclib (Verzenio) are CDK4/6 inhibitors used in breast cancer treatment. Multiple clinical trials over the past decade have found that these drugs slow progression and promote stability when added to endocrine therapy in patients with advanced or metastatic HR-positive/HER2-negative breast cancer.74 More recently, several important trials have investigated their potential benefits when used as part of adjuvant therapy in patients with early breast cancer deemed high-risk due to large size, high-grade, or significant lymph node involvement.54,75
A randomized controlled trial in 5,637 high-risk early breast cancer patients found abemaciclib combined with endocrine therapy increased the likelihood of invasive disease-free survival from 76% to 83.6% and distant relapse-free survival from 79.2% to 86% after a median of 54 months of monitoring.76 Another randomized controlled trial compared ribociclib plus endocrine therapy to endocrine therapy alone in 5,101 patients with HR-positive/HER2-negative stage II or III breast cancer. After a median of 34 months, invasive disease-free survival was higher in the ribociclib group (90.4%) than no-ribociclib group (87.1%).77 Although monitoring is ongoing, as of late 2025, neither drug has demonstrated a significant benefit to overall survival in this patient group.75 These findings prompted a change to treatment guidelines in 2024 recommending the use of abemaciclib or ribociclib as part of adjuvant therapy in patients with early but high-risk HR-positive/HER2-negative breast cancer.33,78
On the other hand, two large trials were unable to show adding palbociclib to adjuvant therapy affected survival outcomes in high-risk HR-positive/HER2-negative breast cancer patients without metastatic disease.79,80
CDK4/6 inhibitors can cause low blood cell numbers, leading to increased risk of serious infections. They are also associated with rare but life-threatening inflammation of the lungs. Other possible side effects include fatigue, diarrhea, hair loss, mouth sores, nausea and vomiting, and headache.40
PI3K/AKT/mTOR inhibitors. The phosphatidyl-inositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway controls cell metabolism, growth, proliferation, and survival. Cancer growth depends on dysregulated activation of the PI3K/AKT/mTOR pathway, and therapies that target inhibition may be helpful in cancer treatment.81
Everolimus (Afinitor) is an mTOR inhibitor that was shown to improve the effectiveness of the aromatase inhibitor exemestane in patients with HR-positive/HER2-negative advanced breast cancer that progressed or recurred despite aromatase inhibitor therapy. It was the first PI3K/AKT/mTOR inhibitor to be approved by the FDA for breast cancer treatment in 2012. Several severe adverse events have been associated with everolimus, including mouth sores, anemia, shortness of breath, high blood glucose levels, fatigue, and lung inflammation.81
About 30–40% of breast cancer patients with HR-positive tumors have a mutation in the PIK3CA gene that makes PI3K overactive, promoting proliferation and contributing to treatment resistance. The PI3K inhibitor alpelisib (Piqray), in combination with the SERD fulvestrant, was shown to improve outcomes in patients with breast cancer that was HR-positive/HER2-negative and carried the PIK3CA mutation. This finding led to the 2019 FDA approval of alpelisib, in combination with fulvestrant, for treating breast cancer with these characteristics that has progressed or recurred despite endocrine therapy.40,81,82 The most common severe adverse effects associated with alpelisib were high blood glucose levels, rash, and diarrhea.81
Some breast cancer patients have an AKT1 mutation that dysregulates AKT activity and is associated with endocrine therapy resistance. The AKT inhibitor capivasertib (Truqap) was found to improve the response to fulvestrant in select breast cancer patients.83 Based on this evidence, in 2023, the FDA approved capivasertib for use in combination with fulvestrant to treat patients with HR-positive/HER2-negative advanced or metastatic breast cancer that displays one or more AKT1, PIK3CA, or PTEN mutations and has stopped responding to endocrine therapy.84 Severe side effects include high blood pressure, diarrhea, and rash. There may also be an increased risk of severe respiratory infection in patients taking capivasertib.81
A number of novel PI3K/AKT/mTOR inhibitors, including dual inhibitors, are currently under investigation for their potential role in treating advanced or metastatic, HR-positive, HER2-negative breast cancer. In addition, new methods of testing for PI3K/AKT/mTOR abnormalities are being explored.81
PARP inhibitors
Poly (ADP ribose) polymerase (PARP) inhibitors suppress the activity of a family of DNA repair enzymes called PARP proteins. Cancer cells divide rapidly and generate many DNA errors, making them especially vulnerable to PARP inhibitors. PARP inhibitors appear to be more effective in breast cancer patients who have inherited BRCA mutations, which also compromise DNA repair. Olaparib (Lynparza) and talazoparib (Talzenna) are PARP inhibitors that are approved for use as stand-alone treatment in patients with advanced or metastatic breast cancer who carry an inherited BRCA mutation.40,61
Olaparib is also approved for treatment of BRCA-associated HER2-negative breast cancer with a high risk of recurrence. This is based on evidence showing one year of adjuvant olaparib increased distant disease-free survival and invasive disease-free survival after four years in patients with BRCA mutations and high-risk HR-positive/HER2-negative breast cancer. In addition, four-year overall survival was increased from 86.4% with placebo to 89.8% with olaparib.85 It can be used alongside endocrine therapy and before or after abemaciclib if appropriate.33
These medications are associated with side effects, including nausea and vomiting, diarrhea, loss of appetite, weight loss, fatigue, bone marrow suppression resulting in low blood cell and platelet numbers, and, in rare cases, bone marrow cancer.40
Immunotherapy
Immunotherapies enhance the immune system’s ability to detect and kill cancer cells. Monoclonal antibodies that target HER2 are an example of an immunotherapy (and a targeted therapy). Checkpoint inhibitors, another class of immunotherapy drugs used in cancer treatment, prevent cancer cells from “turning off” immune T cells and evading recognition.40 While breast cancer has historically been thought to be unresponsive to checkpoint inhibitor therapy, new research has indicated these drugs may be an effective option when combined with chemotherapy in select patients.86
Pembrolizumab (Keytruda) is currently the only checkpoint inhibitor approved for treating triple-negative breast cancer. It has been shown to improve outcomes when used with neoadjuvant (pre-surgery) and adjuvant (post-surgery) chemotherapy in patients with early-stage, high-risk, triple-negative breast cancer and when used with chemotherapy in previously untreated patients with advanced or metastatic triple-negative breast cancer whose tumors express high levels of the checkpoint protein PD-L1.86-88
In a randomized controlled trial involving 1,174 patients with stage II or III triple-negative breast cancer, the rate of event-free survival (defined as survival without disease progression, recurrence, occurrence of a second primary cancer, or death from any cause) was higher in those treated before and after surgery with combinations of chemotherapy plus pembrolizumab (84.5%) compared with chemotherapy plus placebo (76.8%).87 In other words, pembrolizumab lowered the relative risk of recurrence, progression, or death by 37%.86 Further analysis of the data indicated pembrolizumab was effective in patients with tumors that were PD-L1-positive or PD-L1-negative.87
Another trial compared pembrolizumab plus chemotherapy to placebo plus chemotherapy in 847 patients with untreated advanced triple-negative breast cancer expressing PD-L1. After a median follow-up of 44.1 months in patients with the highest levels of PD-L1 expression, overall survival was 23.0 months with pembrolizumab and 16.1 months with placebo. However, in those with lower PD-L1 expression, overall survival was not improved with pembrolizumab compared with placebo (17.6 vs. 16.0 months).88
Side effects of pembrolizumab include fatigue, cough, nausea, skin rash, poor appetite, constipation, and diarrhea. Some people experience allergy-like infusion reactions, which can be severe, while receiving pembrolizumab, which is administered intravenously.40 By disabling the immune system’s ability to regulate T cells, checkpoint inhibitors can also cause autoimmunity that may be severe or, in rare cases, life-threatening.86
Emerging Research and Clinical Trials
Novel treatments
New treatments are constantly being developed and tested in all stages of breast cancer. Treatment paradigms are changing as new targets are discovered and existing treatments are shown to work in new settings, combinations, and schedules. For example, agents that target androgen receptors, which may be overexpressed by triple-negative, and possibly ER-negative/HER2-positive, breast cancer cells, are an active area of endocrine therapy research.128 Novel antibody-drug conjugates, new drugs targeting established markers like ERs, HER2, and checkpoint proteins, and drugs with new targets like HER3, other kinases, and other immune-regulating molecules are among the many approaches under investigation.129-131
The wide field of immunotherapy is one of the most promising areas of breast cancer treatment research. For example, ongoing research is investigating new ways of combining checkpoint inhibitors with other therapies, such as antibody-drug conjugates, chemotherapy, or endocrine therapy in breast cancer patients.132-134 Immunotherapy drugs targeting two or more tumor molecules (bispecific antibodies) are a field of emerging clinical research.131,132 Another intriguing potential therapy is adoptive cell therapy, in which a patient’s T cells are collected, modified in the laboratory to combat cancer cells more aggressively, and returned to the bloodstream.131,132 Challenges associated with these approaches include immune dysregulation and the development of serious autoimmune side effects, as well as tumor cell adaptation and treatment resistance.132
Cancer vaccines, which activate immune cells to recognize and kill cancer cells, are another field of active research with potential for use in breast cancer treatment and recurrence prevention. Cancer vaccines have a better side-effect profile than other immunotherapies, but so far have not shown strong benefits.131,132 Some research indicates a role for anti-cancer vaccines in augmenting the effects of other targeted therapies.131
Oncolytic (cancer-killing) viruses, which preferentially infect cancer cells, can directly kill tumor cells, as well as make them easier for immune cells to detect and kill. Clinical trials are being designed to investigate the effects of adding oncolytic virus therapy to various breast cancer treatment protocols.131,132 Cytokines like interferon are up-regulators of immune function and are being explored for their potential role in directing immune activity against breast cancer cells.131
Optimizing radiation therapy is another area of emerging research. Studies are focusing on: establishing markers to identify women who will not be harmed by omitting radiation therapy; finding optimal strategies for minimizing damage to healthy tissue; and developing safer ways of delivering more potent radiation therapy when needed or re-irradiating a breast in cases of recurrence.135
How to participate in clinical trials
Clinical trials are an essential part of improving cancer care and depend on the willingness of individuals to participate. Clinical trials test the safety and efficacy of new treatments or treatment combinations in breast cancer or may be designed to test questions regarding other aspects of care. By supporting research, volunteer participants help further the evidence base and may improve breast cancer treatment for others. They may also benefit personally from test treatments.136 Breast cancer trials generally test new treatments against standard treatments, so all participants receive appropriate care.137
As of early 2025, there were more than 900 clinical trials in breast cancer listed on the National Cancer Institute’s clinical trials website. Each trial is testing a specific question in a certain well-defined category of breast cancer patients. If you are interested in being a part of a clinical trial, find out if you are eligible and explore the possibilities with your provider. More information about clinical trials in breast cancer can be found at the National Cancer Institute’s Participate in Cancer Research website.
4 Dietary and Lifestyle Considerations
Being physically active, adhering to a healthy diet, and maintaining a healthy body weight are important parts of a holistic approach to breast cancer treatment. These factors influence treatment tolerance, recovery during and after cancer treatment, and outcomes. Therefore, diet and exercise during and after cancer treatment should be discussed between patients and their care team as soon as possible after breast cancer diagnosis.147
Adherence to the recommendations encompassed in the American Heart Association’s Life’s Essential 8 could be beneficial for breast cancer survivors.148 Life’s Essential 8 are as follows:
- Eat better
- Be more active
- Quit tobacco
- Get healthy sleep
- Manage weight
- Control cholesterol
- Manage blood sugar
- Manage blood pressure
The following are general recommendations from the American Cancer Society147,149:
- Practice healthy weight management strategies and maintain or increase muscle mass through diet and physical activity.
- Engage in physical activity for a minimum of 10 minutes per day several times per week if possible.
- Build up to 150–300 minutes of moderate-intensity or 75–150 minutes of vigorous activity each week and include resistance training and stretching exercises at least twice per week.
- Follow a healthy eating pattern that includes nutrient-dense foods, such as vegetables, legumes, whole grains, and nuts and seeds, and limits red and processed meats, sugar-sweetened beverages, highly processed foods, refined grains, and alcohol.
Healthy Diet
A healthy diet that aligns with such dietary patterns as the Mediterranean diet has been shown to reduce the risk of developing breast cancer and may improve breast cancer-related outcomes. Such a diet is high in fruits and vegetables, healthy fats, unrefined grains, beans, nuts and seeds, and fish, and low in red and processed meat, refined grains, and added sugars.150 A meta-analysis of findings from 35 observational studies and 14 clinical trials showed healthy high-quality diets reduced overall mortality by 23% compared with unhealthy low-quality diets in breast cancer patients.151 Another meta-analysis pooled findings from four observational studies and found strong adherence to a Mediterranean diet was correlated with a 22% lower risk of all-cause mortality in breast cancer survivors.152 The positive effects of healthy eating on breast cancer outcomes may be due to beneficial modulation of the gut microbiome, inflammatory signaling, and anti-proliferative pathways, as well as other mechanisms that affect cancer cell growth.150
Other dietary regimens, such as modified fasting, calorie restriction, vegan/plant-based diets, and ketogenic diets, are being investigated in breast cancer patients. For example, a systematic review found intermittent fasting (the practice of not eating for long periods, often roughly 16–48 hours, followed by regular eating) may reduce chemotherapy-induced toxicity, and some research suggests it may lower the recurrence rate in breast cancer patients.153,154 A ketogenic diet, characterized by very low carbohydrate and high fat intake, has attracted interest for its potential role in cancer treatment. It is thought to target cancer cells due to their metabolic dependence on glucose, whereas healthy cells can utilize fat metabolites (ketones) as a primary energy source. Some evidence suggests a ketogenic diet may improve treatment responsiveness in breast cancer patients. However, clarity around its role in treatment and concerns about long-term safety remain unresolved.155 Studies examining the benefits of plant foods and plant-based diets have indicated higher intakes of fruits, vegetables, fiber, and soy foods (such as tofu, tempeh, miso, and edamame) may improve breast cancer outcomes.156 Importantly, following a healthy plant-based diet (high in fruits, vegetables, and whole grains) has been linked to lower overall mortality, while an unhealthy plant-based diet (low in fruits and vegetables and high in refined grains) has been correlated with increased mortality in breast cancer patients.156
Physical Activity
Exercise during cancer treatment is safe and there is strong evidence that physical activity before and after diagnosis can reduce the risk of mortality for breast cancer patients. It is also clear that physical activity may reduce the risk of developing cancer in both pre- and postmenopausal women, may reduce the risk of cancer recurrence, and can help cancer survivors cope with and recover from therapy. Exercise programs should be personalized for each patient and tailored to their clinical situation.150,157
A meta-analysis of 24 studies including more than 140,000 participants analyzed the effects of physical activity on breast cancer outcomes and found between 300 and 500 minutes (5 to 8.3 hours) per week of moderate-intensity physical activity improved prognosis and reduced mortality.158 Another meta-analysis of 23 observational studies concluded that about five hours per week of moderate recreational physical activity was associated with a 48% reduction in all-cause mortality and 38% lower breast cancer specific mortality, with minimal additional risk reduction associated with higher levels of physical activity.157
Two Cochrane analyses concluded that structured exercise is safe and beneficial for women with breast cancer, but the two studies looked at different periods during breast cancer treatment.159,160 During adjuvant therapy (chemotherapy and/or radiation), programs typically ran 6–32 weeks (some up to 52), often 2–5 sessions per week for about 30–60 minutes, and produced a moderate boost in physical fitness and a small-to-moderate reduction in fatigue; effects on overall cancer-specific quality of life and depression were minimal.159 After adjuvant therapy, aerobic training was most often 3–5 days per week for 30–90 minutes at about 40–80% of maximal heart rate, and resistance training approximately 2–5 days per week for 30–60 minutes at roughly 65–85% of one-repetition maximum over about 8–24 weeks; these regimens yielded small improvements in fatigue, moderate gains in cardiorespiratory fitness, and modest, variable benefits across quality-of-life domains.160 Across both reviews, the most consistent outcome was improved physical fitness, with fatigue reduction as a close second, while quality-of-life changes were smaller.
Managing fatigue and enhancing recovery
Physical activity may help a patient feel better before treatment and recover more quickly after treatment both physically and mentally. It can also help with sleep, depression, and other mental health challenges that are common with cancer treatment.149,161
Cancer-related fatigue—a feeling of physical, mental, and emotional exhaustion from cancer therapy, the cancer itself, and other cancer-related issues, such as sleep problems—may be alleviated in part by physical activity.161,162 In coordination with the patient’s care team, a physical activity program for overcoming cancer-related fatigue might start out with 10 minutes at a time of exercise or stretching and gradually increase as the patient is able.149,161 Authors of a 2025 meta-analysis on the effects of exercise during and after breast cancer treatment concluded that low-to-moderate-intensity exercise is recommended during treatment, and that moderate-to-high-intensity exercise can be beneficial after treatment.162
Stress Management and Mental Health
Breast cancer and its treatment can be emotionally and physically demanding, and most patients experience some feelings of anxiety, depression, and distress. Mental health support can contribute to better quality of life for both patients and their loved ones. Mental health support may include stress reduction practices, counseling, education, spiritual support, and group support from a variety of health and counseling professionals.163
Managing stress
Breast cancer patients can experience multiple sources of stress, including the seriousness of the condition, treatments and decision-making, fear of recurrence, economic concerns, changes in body image and sexuality, and concerns for family and friends.164
A meta-analysis that evaluated data from 24 randomized controlled trials with a total of 1,187 participants found non-drug interventions, such as yoga, self-regulation, mindfulness, and physical activity, can reduce feelings of stress, anxiety, and depression in breast cancer patients.164 Physical exercise, including aerobic activities and resistance training, has been shown to help patients manage treatment-related anxiety, fatigue, depression, and sleep difficulties.147,149 In addition, mindfulness and relaxation techniques can improve a patient’s ability to adhere to healthy eating regimens while also reducing stress, anxiety, and depression. Mindfulness refers to a practice of slowing down and focusing on the present using breath awareness, guided imagery, body relaxation, and other techniques. Examples of some of these techniques can be found here.165
Support groups and counseling
Peer support groups bring together people with similarities to share experiences, fears, and concerns. These may be people who have the same type of cancer, are getting the same type of treatment, or have other circumstances in common. They may be led by a facilitator who is a healthcare professional or a cancer survivor. Peer support can create a mutual therapeutic and emotional connection and facilitate an educational and supportive patient-centered journey.166 Being comfortable with the approach and structure of the group and feeling safe and respected are key to benefitting from peer support groups.163
Individual counseling can also be helpful for those who have been diagnosed with breast cancer. This allows the patient to focus on their personal concerns and situation with a mental health professional. Individual counseling may be useful for managing fears, coping with life changes, handling family issues, and discussing concerns about body image and sexuality in a private setting. Counseling may also be useful for couples or families as they navigate cancer care together.163
Cognitive behavioral therapy (CBT) in particular has been shown to improve mental health in breast cancer patients. Cognitive behavioral therapy is a technique for identifying and changing unhelpful thoughts and behaviors to improve emotional well-being. One meta-analysis that pooled findings from 16 interventional studies investigating CBT as individual or group therapy found it reduced anxiety and depression in both breast cancer patients and survivors.167 A growing body of evidence indicates internet-based CBT can also improve depression, anxiety, and quality of life in breast cancer patients.168
5 Nutrients
Specific nutrients and dietary supplement ingredients have been investigated for their possible role as part of breast cancer treatment, either to reduce risk of recurrence, prolong survival, or reduce toxic side effects from other treatments. While a number of dietary supplements have shown potential to modify breast cancer risk, general prevention is outside the scope of this protocol. We have limited inclusion of nutrients in this section to those for which there is some clinical evidence in the context of breast cancer treatment (including post-treatment).
Antioxidant Combinations
Antioxidants are members of a broad category of nutrients and other compounds that can donate electrons, effectively neutralizing free radicals and protecting against oxidative damage. Dietary antioxidants are likely to be an important reason some foods, like fruits and vegetables, are linked to lower cancer risk. However, the use of antioxidant supplements during cancer treatment is controversial. Because chemotherapy and radiation kill cancer cells largely by inducing oxidative damage, concerns about antioxidants reducing their effectiveness have been raised. On the other hand, antioxidants protect and repair non-cancerous cells and tissues, possibly making cytotoxic treatments more tolerable and augmenting their effectiveness.169
A meta-analysis of eight observational studies that included a total of 17,062 breast cancer patients found post-diagnosis use of antioxidant supplements, defined as supplements containing vitamins A, C, and/or E, sometimes in combination with other nutrients, was not associated with any significant change in survival outcomes. However, the analysis noted older studies were more likely to find benefits associated with antioxidant use, while more recent studies were more likely to find harms.170 For example, one observational study in which 1,134 breast cancer patients answered questions about supplement use before and during chemotherapy found the use of any antioxidants, specifically vitamins A, C, or E, carotenoids, or CoQ10, before or during treatment was associated with higher recurrence rate and slightly lower survival rate six months after starting treatment.171 Nevertheless, another meta-analysis of observational studies and clinical trials found the use of certain antioxidant supplements (vitamins C and E) or multi-nutrient supplements after diagnosis was associated with lower risk of recurrence and mortality.172
Melatonin
Reported dosage: 3–20 mg nightly
Melatonin, a hormone produced mainly in the pineal gland in the brain,173 is a regulator of circadian signaling throughout the body. Melatonin has demonstrated a range of positive effects on broad health indicators that may be beneficial in cancer therapy: it reduces oxidative stress, suppresses inflammatory processes, preserves mitochondrial function, improves metabolism, and supports normal cell function.174,175 Furthermore, preclinical studies in breast cancer models suggest melatonin reduces tumor growth factor production, modulates expression of estrogen and progesterone receptors as well as HER2, and suppresses tumor kinase signalling.176
Research from the 1990s reported melatonin had positive effects on breast cancer treatment. A controlled clinical trial in 250 patients with metastatic solid tumors, including 77 with breast cancer, reported supplementing with 20 mg melatonin nightly reduced chemotherapy toxicity and improved one-year survival rate (51% with melatonin vs. 23% without melatonin).177 The same research team conducted a trial that had no control group and found melatonin, at 20 mg nightly, improved responsiveness to endocrine therapy in 14 metastatic breast cancer patients.178
More recent trials have shown melatonin may have benefits for specific parameters of sleep, cognition, and mood during breast cancer treatment. A randomized placebo-controlled trial involving 48 women undergoing surgery for breast cancer found 6 mg of melatonin nightly, beginning three days before and continuing for one to two weeks after surgery, increased sleep efficiency (the proportion of time in bed spent sleeping) and reduced wakefulness after sleep onset.179 In another placebo-controlled trial with 43 participants, 6 mg of melatonin nightly beginning one week before breast cancer surgery and continuing for three months reduced the risk of depressive symptoms.180 A randomized controlled trial in 36 breast cancer patients found 20 mg of melatonin nightly, beginning three days before and for one week during the first cycle of adjuvant chemotherapy, improved cognitive performance compared with placebo, and the effect was associated with increased sleep quality and decreased depressive symptoms.181
A randomized placebo-controlled trial in 74 breast cancer patients found 18 mg of melatonin nightly, taken from one week before until one month after completing adjuvant chemotherapy and radiation, improved fatigue scores and reduced the frequency of severe fatigue.182 A longer trial conducted by the same research group included 92 breast cancer patients who received either 18 mg of melatonin per day or placebo for two years following adjuvant therapies. At the end of the trial, those given melatonin were still experiencing less fatigue than those given placebo.183 A trial with no control group that involved patients with metastatic breast cancer undergoing endocrine or trastuzumab therapy found 5 mg of melatonin nightly for two months improved sleep quality and quantity, as well as fatigue, quality of life, and social and cognitive functioning.184 A randomized placebo-controlled trial in 86 postmenopausal breast cancer survivors no longer in active treatment found 3 mg of melatonin nightly for four months improved sleep quality.185 On the other hand, a trial in 78 early-stage breast cancer patients compared 20 mg of melatonin per day to placebo, taken from one day before until two weeks after completing radiation therapy, and found melatonin had no effect on fatigue or other parameters.186 Similarly, a retrospective registry study involving analysis of data from over 37,000 breast cancer patients showed that having been prescribed melatonin for about three months or more was not associated with improved survival.187
Omega-3 Polyunsaturated Fatty Acids
Reported dosage: About 1–1.8 grams omega-3 fatty acids (EPA and/or DHA) daily; this generally was obtained from about 2–5.8 grams of fish oil or 4.5 grams of algae oil daily
The long-chain omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are abundant in oily fish and often obtained in the form of fish oil supplements. They have well-established anti-inflammatory and immune-modulating effects, and higher intake of these fatty acids has been linked to lower breast cancer risk in observational research.191-193
One clinical trial with no control group examined the effect of 1.8 grams of DHA (from 4.5 grams of algae oil) daily on outcomes during an average of 31 months of monitoring in patients who received anthracycline-based chemotherapy for rapidly progressing metastatic breast cancer. The researchers found patients who attained the highest DHA plasma levels progressed more slowly and had longer survival times than those who attained lower levels. Those who achieved high plasma DHA levels had a median time to progression of 8.7 months and overall survival of 34 months, while those who did not had a median time to progression of 3.5 months and overall survival of 18 months. DHA supplementation was not associated with any adverse effects. This finding suggests DHA may sensitize malignant tumors to chemotherapy.194
In a randomized controlled trial involving 48 patients with locally advanced breast cancer, 1 gram of omega-3 fatty acids from fish oil daily during 51 days of neoadjuvant chemotherapy reduced tumor tissue levels of the proliferation marker Ki-67 and vascular endothelial growth factor (VEGF, a protein involved in tumor blood vessel formation). In addition, fish oil extended cancer-free and overall survival compared with placebo: average survival was 30.9 weeks with fish oil and 25.9 weeks with placebo.195
Supplementing with omega-3 fatty acids can improve general health, including cardiovascular and metabolic status, and this may improve overall outcomes in breast cancer. A controlled trial in 32 breast cancer patients found 4 grams of fish oil daily (providing 800 mg EPA and 400 mg DHA) for 60 days beginning at the start of chemotherapy increased chest muscle strength and improved cardiorespiratory fitness.196 Another trial in 51 subjects with HR-negative breast cancer found omega-3 fatty acid supplementation for one year reduced triglyceride levels and led to epigenetic changes favoring healthy lipid metabolism. The trial also showed a roughly 5.8-gram (3.7 gram EPA and 1.6 gram DHA) daily dose had stronger effects than a 1-gram (670 mg EPA and 240 mg DHA) daily dose.197 A randomized controlled trial in 88 newly diagnosed breast cancer patients found 600 mg of omega-3 fatty acids (providing 360 mg EPA and 240 mg DHA) daily plus 1,250 mcg (50,000 IU) of vitamin D weekly for nine weeks during the initiation of chemotherapy improved global health and functioning scores and reduced markers of inflammation compared to vitamin D or fish oil alone or to no supplements.198 However, a trial in 53 women with non-metastatic advanced breast cancer found no cardiometabolic benefit from supplementing with 2.4 grams of omega-3 fatty acids (providing 1,600 mg EPA and 800 mg DHA) daily during neoadjuvant chemotherapy.199
Some evidence suggests omega-3 fatty acids may improve immune function and tumor-suppressive immune activity in breast cancer patients. In a randomized controlled trial, breast cancer patients received either 2 grams of fish oil (providing 1.8 grams of omega-3 fatty acids) daily or a mineral oil placebo for 30 days after diagnosis and before surgery. At the end of the trial, those given fish oil had reduced levels of high-sensitivity C-reactive protein (hsCRP, a marker of inflammation) and higher levels of immune-activating CD4+ T cells (T4 cells), while those given mineral oil had increased hsCRP and decreased T4 cell levels.200
Musculoskeletal pain is a common side effect of aromatase inhibitors, affecting approximately 45–60% of patients, particularly those with obesity. In fact, pain is a major cause of treatment discontinuation.94
The chemotherapy agent paclitaxel frequently causes peripheral neuropathy (ie, numbness in the hands and feet). In a randomized controlled trial, 57 breast cancer patients being treated with paclitaxel took either 640 mg of omega-3 fatty acids (including 346 mg DHA and 64 mg EPA) three times daily or placebo during and for one month following paclitaxel therapy. Those treated with omega-3 fatty acids were about half as likely to experience peripheral neuropathy.201 However, in a randomized placebo-controlled trial in 49 patients, 4 grams of a pharmaceutical formulation providing ethyl esters of EPA (about 1,900 mg) and DHA (about 1,500 mg) per day from one week before beginning paclitaxel, until the end of paclitaxel treatment, did not reduce treatment-induced neuropathy.202
Vitamin D
Reported dosage: Approximately 50 mcg–175 mcg (2,000 IU–7,000 IU) daily
Vitamin D is a steroid hormone that regulates calcium metabolism, modulates immune activity, and promotes normal cell replication. It is synthesized in the body in response to sunlight, is found in foods such as fatty fish, eggs, mushrooms, and fortified dairy products, and is often taken as a supplement. Some studies, though not all, have found a high prevalence of vitamin D deficiency (generally defined as serum 25-hydroxyvitamin D levels < 20 ng/mL) and insufficiency (generally defined as 25-hydroxyvitamin D levels below 30 ng/mL) among newly diagnosed breast cancer patients, particularly those with triple negative breast cancer, suggesting poor vitamin D status may be a contributing factor in its onset.191,203,204
Vitamin D deficiency in breast cancer patients has been correlated with poorer response to neoadjuvant chemotherapy and a higher risk of breast cancer mortality.205,206 One meta-analysis of 12 studies with a total of 8,574 participants found those with the highest vitamin D levels, compared to those with the lowest vitamin D levels, had a 26% relative increase in overall survival, 50% increase in disease-free survival, and 31% increase in breast cancer-specific survival.206 Another meta-analysis included six studies in which surgical samples from a total of 1,291 breast cancer patients treated with neoadjuvant chemotherapy were analyzed for treatment responsiveness. The pooled results showed that pretreatment 25-hydroxyvitamin D levels < 20 ng/mL were associated with a 50% increased risk of a pathologic non-complete response (meaning residual invasive cancer cells were detected) and a 33% increase in risk of no response (meaning invasive disease persisted after treatment).205
In a randomized controlled trial conducted in Brazil, 75 women undergoing neoadjuvant chemotherapy for breast cancer received 50 mcg (2,000 IU) of supplemental vitamin D3 daily or placebo for six months. Both groups were found to have low blood vitamin D levels at the beginning of the trial. Vitamin D levels improved in the vitamin D3 group and 43% achieved a pathologic complete response (defined as the absence of invasive disease in breast and lymph nodes), whereas vitamin D levels were stable in the placebo group and only 24% achieved a pathologic complete response.207 Another randomized clinical trial examined outcomes in 227 women with breast cancer, age 18 to 80 years, undergoing neoadjuvant chemotherapy. Half the women received 1,250 mcg (50,000 IU) of oral vitamin D once per week during neoadjuvant chemotherapy while the other half did not. Over 70% of trial participants, in both groups, had either deficient (<20 ng/mL) or insufficient (20–30 ng/mL) vitamin D levels at baseline, which improved in the supplementation group. In addition, the pathologic complete response rate was 2.3 times higher in those who received vitamin D.208
Vitamin D has demonstrated other effects that suggest it may be helpful in breast cancer treatment. In a randomized placebo-controlled trial in 44 breast cancer patients being treated with tamoxifen, 1,250 mcg (50,000 IU) of vitamin D weekly for eight weeks reduced levels of an indicator of blood vessel formation and tumor growth (angiopoietin-2) more than placebo in a subgroup composed of premenopausal participants whose cancer had not infiltrated blood or lymph vessels.209 Findings from a nine-week, randomized, controlled trial in 88 newly diagnosed breast cancer patients indicated a combination of vitamin D (1,250 mcg [50,000 IU weekly]) plus omega-3 fatty acids (600 mg daily) may be more effective than either alone for lowering inflammation, reducing treatment side effects, and improving nutritional status and overall health and functioning during chemotherapy.198,210
Vitamin C
Reported dosage: 500 mg orally per day. Note: ≥ 1 gram per kg of body weight of intravenously administered vitamin C two to four times per week has also been reported, but this dosage approach requires medical supervision.
Vitamin C, or ascorbic acid, is abundant in fruits and vegetables and is important for immune health and protecting cells from free radicals.191 Vitamin C has well known free radical-scavenging and anti-inflammatory properties. Preclinical research has also demonstrated that it has the ability to inhibit abnormal cell proliferation, suppress expression of proteins involved in cell transformation, and at high concentrations can raise oxidative stress in tumor cells.211 In addition, meta-analyses of observational studies have found post-diagnosis vitamin C supplementation was associated with a 16–21% reduction in relative risk of overall mortality.170,172,212
Some clinical data support a benefit of vitamin C for reducing adverse treatment effects in breast cancer patients. A randomized controlled trial in 40 early-stage breast cancer patients found 500 mg of vitamin C, along with 400 IU of vitamin E (as alpha tocopherol acetate), during chemotherapy mitigated the reduction in antioxidant enzyme levels, rise in oxidative stress, and DNA damage caused by chemotherapy and cancer.213 Whether these effects could translate into protection against side effects or improved outcomes is not yet known.
Because intestinal absorption of vitamin C is limited, intravenous administration can be used to achieve higher blood levels. One observational study examined data from 424 breast cancer patients treated with adjuvant radiation therapy, 70 of whom were co-treated with intravenous vitamin C twice weekly for at least four weeks. Those who received intravenous vitamin C at ≤1 gram/kg body weight or did not receive vitamin C had an increase in the neutrophil-to-lymphocyte ratio—a change in immune cell proportions that reflects inflammation and is correlated with increased overall and breast cancer-specific mortality. However, those who received intravenous vitamin C at >1 gram/kg body weight experienced a decrease in this ratio.214
Another observational study found 35 triple-negative breast cancer patients who received intravenous vitamin C at 1 gram/kg body weight, beginning three days before and continuing every other day during one full cycle of chemotherapy, had fewer adverse side effects and longer progression-free and overall survival than 35 matched patients who received chemotherapy alone. In those who received vitamin C, average progression-free survival was seven months and overall survival was 27 months, while in those who did not receive vitamin C, average progression-free survival was 4.5 months and overall survival was 18 months.215 This equates to a 56% relative increase in progression-free survival and a 50% relative increase in overall survival. An observational study compared data from 53 patients with non-metastasized invasive breast cancer who received 7.5 grams of intravenous vitamin C once weekly for at least four weeks during standard treatment and 72 similar patients who did not receive vitamin C. Those who received vitamin C had fewer chemotherapy and radiation therapy side effects such as nausea, loss of appetite, fatigue, depression, sleep disorders, dizziness, and bruising/bleeding disorders than those who did not.216
Vitamin E
Reported dosage: 400 IU–1,300 IU (approximately 180 mg–600 mg) daily
Vitamin E is a fat-soluble antioxidant nutrient found in fatty foods, such as nuts, seeds, avocados, olives, and plant oils. In breast cancer cell studies and animal models of breast cancer, vitamin E has been shown to suppress tumor growth and proliferation, promote cell death, enhance tumor suppressor gene activity, and improve immune function.217
In an open-label, randomized, controlled trial in 31 cancer patients, 600 mg of vitamin E daily during and for three months after completing chemotherapy resulted in a 66% reduction in neuropathy.218 This dose was equivalent to about 900–1,300 IU per day, depending on the type of vitamin E used (not specified in the publication). In a randomized controlled trial involving 74 breast cancer patients, a combination of 400 IU (180 mg) vitamin E plus carnitine during four cycles of chemotherapy with doxorubicin plus cyclophosphamide resulted in a 24.2% relative reduction in cardiac events compared with chemotherapy alone. The form of vitamin E used in this study was dl-alpha-tocopherol.219 A meta-analysis of findings from nine studies also indicated vitamin E supplementation may reduce the risk of breast cancer recurrence.220 In addition, one randomized controlled trial found combination treatment with 400 IU (about 270 mg) of vitamin E as alpha-tocopherol per day plus pentoxifylline (Trental, a drug that improves blood flow) for six months following radiation therapy reduced radiation-related scarring in the breast and chest wall, though it had no effect on survival during two years of monitoring.221
Green Tea
Reported dosage: 940 mg–1,200 mg oral EGCG daily; 600 micromol/L (about 275 mg/L) EGCG spray used topically during radiation therapy
Consumption of green tea may reduce the risk of developing breast cancer and may improve outcomes after breast cancer diagnosis.191,222 The protective effects of green tea have been attributed to bioactive polyphenolic compounds called catechins, the most abundant of which is epigallocatechin-3-gallate (EGCG). Catechins have numerous modulating effects on cellular pathways known to be involved in cancer.191,223,224
One small study provided preliminary evidence that cancer-related pathways may be affected by consumption of supplemental EGCG in breast cancer patients. Molecular markers on surgical tissue samples from 13 women with early-stage breast cancer waiting for surgery who took 2,175 mg green tea extract, containing about 940 mg EGCG, per day (roughly equivalent to 8‒10 cups of green tea) for an average of 35 days prior to surgery were compared with those from 15 matched breast cancer patients who received no green tea. The women in the green tea extract group had lower levels of the proliferation marker Ki-67 in both benign and malignant cells compared with the control group.225
There is also some evidence that green tea can impact the risk of breast cancer recurrence. An observational study that monitored 518 women with triple-negative breast cancer for a median of 9.1 years found those who reported drinking green tea regularly during the five years after diagnosis were 46% less likely to have a recurrence compared with those who did not drink green tea.226 A meta-analysis of seven observational studies involving a total of 13,956 women with a breast cancer diagnosis found those who drank the most green tea (generally 3–8 cups per day) had a 19% lower likelihood of recurrence.224
Green tea may have particular benefits for breast cancer patients being treated with radiation therapy. In a small, randomized, placebo-controlled trial involving 10 women with metastatic breast cancer undergoing radiotherapy, 400 mg of EGCG three times daily during therapy reduced blood levels of growth factors and enzyme activity related to cancer growth compared with placebo.227 Findings from preliminary clinical trials suggest an EGCG-containing topical formula (600 micromol/L EGCG spray) may alleviate radiation-induced dermatitis when applied to the skin of breast cancer patients undergoing radiation therapy.228,229
Curcumin
Reported dosage: 6,000 mg of standard curcumin orally per day or 300 mg intravenously per week
Dosage note: Some curcumin formulations have been shown to increase absorption and thus may achieve blood levels comparable to higher-dose unformulated curcumin. The studies described here used an unformulated curcumin preparation so this dosage range should not be confused with a dosage range for enhanced-absorption curcumin formulations. Also, curcumin should not be administered intravenously without qualified medical supervision.
Curcumin is a polyphenolic compound extracted from turmeric. In laboratory studies, curcumin has been found to slow proliferation of breast cancer cells, cause tumor cells to die, and prevent tumors from developing blood vessels to supply nutrients.191
A preliminary dose-finding case series included nine patients with advanced metastatic breast cancer receiving chemotherapy with docetaxel once every three weeks for five or six cycles. They received oral curcumin at doses of 500 mg to 8,000 mg per day for seven days during each three-week cycle, timed to include four days before, the day of, and two days after chemotherapy. Three patients achieved stable disease and five had a partial response six or more weeks after their last cycle of docetaxel. However, two participants experienced diarrhea and one experienced low white blood cell numbers, which may have been related to curcumin. The study authors determined 6,000 mg daily was a safe oral dose for future research.230 In a randomized, double-blind, placebo-controlled trial involving 30 breast cancer patients, 2,000 mg of oral curcumin three times daily during radiation treatment reduced the severity of radiation dermatitis: average radiation dermatitis scores, based on a 0–4.0 scale, were 2.6 in the curcumin group and 3.4 in the placebo group at the end of radiation therapy.231 This equates to a 24% relative reduction in radiation dermatitis scores with curcumin. The curcumin preparation used in the study was a branded standardized curcumin extract called Curcumin C3 Complex.
High oral doses of curcumin are sometimes used in trials due to low absorption of standard curcumin preparations. Intravenous curcumin has been explored as an alternative in some trials.232 In a randomized controlled trial, 150 patients with progressive, advanced, or metastatic breast cancer received paclitaxel plus 300 mg of intravenous curcumin or placebo weekly for 12 weeks. The curcumin group had a higher overall response rate than the placebo group (51% vs. 33%) four weeks after the end of treatment, but the difference had diminished (29% vs. 20%) by 12 weeks after the treatment period. The curcumin group also reported less fatigue and better physical performance, suggesting better treatment tolerance.233
Coenzyme Q10
Reported dosage: 100 mg daily
Coenzyme Q10 (CoQ10) is an antioxidant involved in cellular energy production. Muscle tissues, including heart muscle, have a high requirement for CoQ10.
Observational evidence suggests low CoQ10 levels are associated with breast cancer risk and with expression of genetic biomarkers related to tumor growth.234,235 Other observational studies and case descriptions have reported CoQ10, in combination with essential fatty acids and other antioxidants (eg, vitamin C, vitamin E, selenium, folic acid, and beta-carotene), extended survival time beyond median predicted survival (based on patient demographics and tumor characteristics and stage) in advanced and metastatic breast cancer patients.236-238
CoQ10 has been shown to reduce fatigue in healthy people and those with chronic conditions, but to date there is little evidence of benefit for cancer-related fatigue.239 A randomized controlled trial in 57 breast cancer patients with cancer-related fatigue undergoing chemotherapy found a combination supplement providing 30 mg of CoQ10 plus amino acids, minerals, and B vitamins once daily for 21 days reduced the most severe fatigue levels and global fatigue scores, but did not change average fatigue levels or quality of life scores.240 Another randomized placebo-controlled trial in 236 breast cancer patients found no improvement in fatigue, mood, or quality of life after 24 weeks of supplementing with 100 mg of CoQ10 plus 100 IU of vitamin E daily.241
In a meta-analysis of findings from nine randomized controlled trials in breast cancer survivors who had received tamoxifen for at least six months, 100 mg of CoQ10 per day for 45–90 days decreased levels of VEGF (needed for tumor blood vessel formation), interleukin-8 (IL-8, an inflammatory cytokine), and matrix metalloproteinases (proteins involved in tumor cell growth and proliferation), but did not alter markers of inflammation and oxidative stress.242 Whether or not these effects of CoQ10 could lead to better treatment outcomes is not known.
Sulforaphane and Indole-3-carbinol
Reported dosage: Approximately 35 mg sulforaphane or 300 mg diindolylmethane (DIM) daily
Numerous observational studies have indicated high consumption of cruciferous vegetables (eg, broccoli, kale, cabbage, Brussels sprouts) is associated with a lower risk of a range of cancers, including breast cancer.243 Sulforaphane is a sulfur-containing phytochemical from cruciferous vegetables that has been shown in preclinical research to have anti-inflammatory and anti-cancer properties. Unfortunately, clinical research investigating its potential role in breast cancer treatment is sparse.244
In a preliminary randomized controlled trial, 30 newly diagnosed postmenopausal breast cancer patients received broccoli sprout extract containing 200 micromoles (about 35 mg) of sulfur compounds (including sulforaphane) or placebo daily for two weeks prior to surgery. The dose of broccoli sprout extract was equivalent to about 500 grams (more than one pound) of fresh broccoli. Tissue samples at the beginning and end of the trial showed markers of cancerous activity were not statistically different between the two groups.245 A trial ongoing as of late of 2025 is evaluating the potential for supplemental sulforaphane to reduce chemotherapy-induced cardiac dysfunction.246
Indole-3-carbinol (I3C) and its metabolite diindolylmethane (DIM) are other sulfur compounds found in crucifers. Preclinical and clinical evidence suggests I3C and DIM have multiple anti-cancer actions. Among them, these compounds have been shown to shift estrogen metabolism toward production of non-toxic and potentially anti-cancer byproducts, and may thereby help protect against hormone-dependent breast cancer onset and progression.247-249 In a randomized placebo-controlled trial involving 98 breast cancer patients taking tamoxifen, 150 mg of DIM twice daily for 12 months promoted favorable changes in estrogen metabolism. However, DIM also decreased the amount of tamoxifen-related compounds in circulation compared with placebo, suggesting it could reduce tamoxifen’s effectiveness.250 Until more is known about this interaction, DIM should not be used by breast cancer patients taking tamoxifen.
Flaxseeds
Reported dosage: 25 grams–50 grams daily
Flaxseeds have been studied for their potential role in breast cancer prevention and treatment mainly because of their high lignan content. Lignans are plant fibers that are digested by intestinal bacteria into phytoestrogens—compounds that alter estrogen signaling by binding ERs, blocking estrogen, and exerting weak estrogen-like activity. In addition, phytoestrogens from flaxseeds have antioxidant properties. Flaxseeds are also a rich source of alpha-linolenic acid, an omega-3 fatty acid, which has demonstrated anti-cancer effects in the laboratory and may lower breast cancer risk.251
Higher flaxseed and lignan intakes have been associated with lower breast cancer risk in multiple observational studies. The amount associated with this benefit is 30 grams (about one ounce) per day.251 In a randomized controlled trial, 32 newly diagnosed postmenopausal breast cancer patients were given muffins with either 25 grams (about 2 tablespoons) of flaxseed with 50 mg of lignan or no flaxseed (placebo) to be eaten daily until surgery (an average of 32–39 days). Tumor cell proliferation and a breast tumor cell biomarker, c-erbB2, decreased significantly in the flax muffin group, while programmed tumor cell death increased. None of these changes were seen in the placebo group.252 A trial lacking a control group investigated the effects of 50 mg of flaxseed lignan per day for 12 months in 45 premenopausal women with a high breast cancer risk. The participants had high Ki-67 expression levels in biopsy samples and either a history of prior breast cancer or precancerous breast changes. Lignan supplementation led to a reduction in Ki-67 in 36 (80%) participants and the percentage of women with precancerous cellular changes decreased.253
In one animal study, flaxseed consumption enhanced the anti-tumor effect of tamoxifen.251 However, it is still not known whether flaxseed use can improve breast cancer survival outcomes.
Soy
Reported dosage: Approximately 60 mg of soy isoflavones or 2–3 servings of soy food daily
Dietary intake of soy-containing foods may improve outcomes in breast cancer.147,254 Soybeans are an important source of phytoestrogens called isoflavones. Phytoestrogens, including soy isoflavones, have a structure that allows them to bind to ERs and mildly stimulate estrogenic signaling. This leads to downregulation of estrogen signaling in conditions of high estrogen levels (such as pre-menstrually) or upregulation in conditions of low estrogen levels (such as after menopause).255
Isoflavones bind more strongly to ER-beta than ER-alpha. ER-alpha is the main ER type in breast and uterine tissue and contributes to estrogen-related cell proliferation.256 Soy isoflavones also have epigenetic effects leading to reduced ER-alpha expression, making them potentially protective against breast cancer.257 Finally, isoflavones have demonstrated a range of health benefits that may be related to their ability to reduce oxidative stress and inflammation and promote healthy metabolism.258
A recent systematic review and meta-analysis that included 11 observational studies evaluating the impacts of soy intake on breast cancer outcomes found greater intake of soy isoflavones (a type of phytoestrogen) was associated with a 26% reduced risk of breast cancer recurrence. Subgroup analysis showed high soy isoflavone intake was associated with a 28% lower recurrence risk in postmenopausal women and an 18% lower recurrence risk among patients with ER-positive cancer. A soy isoflavone intake of 60 mg per day, equivalent to about 2–3 servings of soy food (eg, soy milk, tofu, or cooked soybeans) per day, was linked to the greatest risk reduction. In addition, the analysis found higher soy protein or product intake was associated with a 25% reduction in cancer-related mortality in those with ER-positive breast cancer.254
Boswellia
Reported dosage: 2,400 mg daily; 2% Boswellia cream topically twice daily during radiation therapy
Boswellia (frankincense, Boswellia serrata) and its active compounds, boswellic acids, have demonstrated anti-cancer effects in a variety of cancer cell types and animal models. Boswellia has well-known anti-inflammatory activity and is widely used to treat such conditions as arthritis and asthma. In a small pilot trial, 18 breast cancer patients were treated with 2,400 mg of boswellia extract per day for a median of 11 days between diagnosis and surgery. Surgical specimens showed levels of Ki-67, a marker of tumor cell proliferation, were reduced by an average of 13.8% compared with biopsy samples. Similar assessments in 18 matched patients who did not receive boswellia showed Ki-67 levels increased by an average of 54.6% between biopsy and surgery.263 In a randomized controlled trial,114 women undergoing radiation treatment for breast cancer received either a topical 2% boswellia cream or a placebo cream to use twice daily on irradiated skin throughout radiation therapy (generally five sessions per week for five weeks). The boswellia cream reduced the severity of acute radiation-related skin damage (assessed as redness) compared with a placebo cream: 49% of those treated with placebo and 22% of those treated with boswellia had the highest intensity of redness following radiation.264
Crocin
Reported dosage: 30 mg daily
Crocin is a carotenoid and the main active compound found in the yellow spice saffron (Crocus sativus). Like other carotenoids, crocin has anti-inflammatory and antioxidant properties.265 Saffron and its carotenoids have demonstrated neuroprotective effects and have shown promise in the treatment of neurological disorders, including depression and anxiety.266 In pre-clinical experiments, it has also demonstrated a number of anti-cancer properties.265
In a randomized controlled trial, 72 patients with non-metastatic HER2-positive or triple-negative breast cancer were given either 30 mg of crocin or placebo daily during doxorubicin-based chemotherapy. At the end of treatment, depression and anxiety scores dropped by approximately 30–40% in the crocin group but had increased by approximately 30% in the placebo group. The crocin group also had longer survival, but this effect may have been due to chance as the study was small.267 Longer and larger trials are needed to determine whether crocin can be beneficial as part of breast cancer treatment.
6 Repurposed Drugs in Breast Cancer
Metformin
Metformin is used to treat type 2 diabetes, though ongoing research has provided some preliminary evidence that it may provide some clinical benefit in certain types of breast cancer when combined with standard therapies.268 Analysis of gene expression profiles in postmenopausal breast cancer survivors showed that metformin treatment induced gene expression patterns that might be protective against new tumor development.269 Preclinical research suggests metformin can directly impair the growth and spread of breast tumors, as well as sensitize breast cancer stem cells to chemotherapy drugs.270 However, various pharmacokinetic characteristics of metformin present a challenge for its use in cancer, and metformin’s preclinically demonstrated anti-cancer effects require concentrations of the drug that are much higher than those achieved in treating type 2 diabetes.271 Nevertheless, clinical data on the benefits of metformin for breast cancer patients are mixed, and benefits may be restricted to patients with specific disease characteristics or genomic profiles.271,272
Intriguingly, one study that examined data from 44,541 women who were monitored for approximately eight to 14 years found those with type 2 diabetes treated with metformin had a 14% lower incidence of estrogen receptor-positive breast cancer than those without type 2 diabetes, and the risk decreased with longer use of metformin. However, they also had a 25% higher incidence of estrogen receptor-negative and 74% higher incidence of triple-negative breast cancer than those who did not have diabetes.273 Having diabetes worsens survival outcomes in women with hormone receptor-positive breast cancer, but treatment with metformin appears to reverse the negative impact of diabetes on disease-free survival, distant disease-free survival, and overall survival.271,274,275
Clinical trials have found metformin is not beneficial in breast cancer patients without diabetes. A large randomized controlled trial that included 3,649 women with high-risk non-metastatic breast cancer and without diabetes compared metformin to placebo after standard anti-cancer treatment, monitoring participants for up to 10 years. The trial found metformin was no better than placebo at improving survival, and this lack of effect was observed in women with both HR-positive and HR-negative cancers.276 A meta-analysis of five smaller randomized controlled trials that included a combined total of 396 non-diabetic breast cancer patients found adding metformin to standard anti-cancer treatment did not slow progression or improve overall survival.277 Another meta-analysis that included data from five randomized controlled trials with a total of 412 participants found metformin also had no effect on outcomes in non-diabetic women with metastatic or recurrent breast cancer.278
Statins
There is some limited evidence that statins (a family of drugs used to reduce high cholesterol levels) may improve mortality outcomes in breast cancer patients.279 Large meta-analyses of observational data involving women with breast cancer have found statin use to be associated with a 25–27% lower risk of breast cancer recurrence, as well as an 18–20% lower risk of breast cancer-specific and 18% lower risk of all-cause mortality.279-281 Although statins do not appear to reduce the risk of developing breast cancer, the associations between statins and breast cancer outcomes appear to be independent of timing of initiation of statin use (before or after breast cancer diagnosis) and choice of statin drug.280 Another meta-analysis found statin use after breast cancer diagnosis was only linked to better cancer-specific survival in those with HR-positive tumors.282 In addition, there is some limited evidence that combining statin use with cancer therapies may extend disease-free survival in triple-negative breast cancer patients.283 One large observational study in Finland found statin use after breast cancer diagnosis reduced the relative risk of breast cancer death by 51% in those whose cholesterol levels decreased with statin therapy, but was unchanged in those whose cholesterol levels did not decrease.284
Statins may also play a role in reducing adverse side effects of cancer treatment. In a randomized placebo-controlled trial that included 89 women recently diagnosed with breast cancer, rosuvastatin (Crestor), taken during and after chemotherapy for a total of six months, protected against chemotherapy-induced cardiotoxic effects linked to heart failure.285 Observational studies comparing data from women with breast cancer who were taking statin drugs before and during chemotherapy with data from similar women who did not take statins found statin users had a lower risk of cardiotoxicity related to chemotherapy.286,287 One interesting study compared the use of a 1% atorvastatin (Lipitor) gel on irradiated skin twice daily for six weeks to a placebo gel in 70 breast cancer patients receiving radiation therapy. Those who used topical atorvastatin had decreased severity of radiation-induced skin symptoms, including itching, pain, and edema.288
7 Support and Resources for Patients and Caregivers
If you have any questions on the scientific content of this protocol, please call a Life Extension Oncology Wellness Specialist at 1-866-864-3027.
Navigating the Healthcare System
Understanding insurance and financial assistance
For many people with breast cancer, diagnosis and treatment bring the burden of financial stress to an emotionally challenging time. Dubbed “financial toxicity,” this type of stress is a well-known side effect of cancer care that may persist for years after diagnosis and has been shown to reduce quality of life, lead to treatment disruptions, and contribute to mortality.289,290 It is critical for patients to work with their healthcare providers and insurance companies to make sure they understand the direct and indirect out-of-pocket costs of treatment and know how to access available resources. Measuring and monitoring the financial burden of treatment allows patients and their care team to consider adjustments to protocols that prioritize overall well-being.290
The American Cancer Society offers guidance for cancer patients seeking assistance with treatment expenses, as well as costs of day-to-day life while dealing with cancer.
Finding the right healthcare team
After receiving a breast cancer diagnosis, it is important to find a care team that can provide the best care possible while addressing the patient’s unique condition, needs, and preferences.291 In addition to one’s primary care physician and other personal connections, there are numerous resources available for finding cancer care centers. For example, the National Cancer Institute-designated cancer centers are recognized for their rigorous standards, state-of-the-art research, and commitment to developing new and better approaches to preventing, diagnosing, and treating cancer.292 The Association of Community Cancer Centers and the Association of American Cancer Institutes are other resources that provide profiles of member cancer centers by state. Important considerations for finding the right team and center are their location, facilities, specialties, and availability of multidisciplinary teams, as well as access to clinical trials. The American Cancer Society has a worksheet that can help a patient decide whether a particular doctor or center is a good fit for them.291
Finding a patient advocate
Partnering with a knowledgeable patient advocate can be very helpful in ensuring your interests are guiding decisions in your cancer care journey. There are several patient advocacy resources available to patients. Often, the treatment center at which you are receiving treatment will have patient advocacy resources available. Also, the American Cancer Society can provide access to patient navigators to assist patients. They can be reached at 1-800-227-2345. Local or statewide services may be available as well. For example, the Patient Advocate Foundation assists breast cancer patients in partnership with relevant local organizations. Insurance providers may provide specific resources related to patient advocacy and care navigation as well. Private patient advocacy services are available as well and may be worth pursuing for patients with disposable resources.
Disclaimer and Safety Information
This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.
The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.
- American Cancer Society (ACS). About Breast Cancer. Accessed 02/02/2025, https://www.cancer.org/content/dam/CRC/PDF/Public/8577.00.pdf
- Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA: a cancer journal for clinicians . Jan–Feb 2024;74(1):12–49. doi:10.3322/caac.21820 https://www.ncbi.nlm.nih.gov/pubmed/38230766
- National Cancer Institute. Breast Cancer Treatment (PDQ®)-HealthProfessional Version. Updated 3/14/2025. Accessed 03/28/2025, https://www.cancer.gov/types/breast/hp/breast-treatment-pdq#_298
- American Cancer Society. Key Statistics for Breast Cancer. Accessed Oct. 6,2025, https://www.cancer.org/cancer/types/breast-cancer/about/how-common-is-breast-cancer.html
- Hendrick RE, Baker JA, Helvie MA. Breast cancer deaths averted over 3decades. Cancer. May 1 2019;125(9):1482–1488.doi:10.1002/cncr.31954 https://www.ncbi.nlm.nih.gov/pubmed/30740647
- ACS. American Cancer Society. Breast Cancer Facts and Figures 2024–2025.Available at https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/breast-cancer-facts-and-figures/2024/breast-cancer-facts-and-figures-2024.pdf Accessed 12/30/2024. 2024;
- Clinical Key. Clinical Overview: Breast Cancer in Females. Elsevier BV.Updated Feb. 18, 2025. Accessed Mar. 13, 2025, https://www.clinicalkey.com/
- National Cancer Institute. Breast Cancer Screening (PDQ) - HealthProfessional Version. Updated 4/10/2025. Accessed 02/02/2025, https://www.cancer.gov/types/breast/hp/breast-screening-pdq
- NBCF team. National Breast Cancer Foundation, Inc. Ductal Carcinoma In Situ(DCIS). Updated 3/25/2025. Accessed 4/14/2025, https://www.nationalbreastcancer.org/dcis/
- Burstein HJ. Tumor, node, metastasis (TNM) staging classification for breastcancer. UpToDate. Updated 4/24/2024. Accessed 7/30/2024, https://www.uptodate.com/contents/tumor-node-metastasis-tnm-staging-classification-for-breast-cancer
- Trayes KP, Cokenakes SEH. Breast Cancer Treatment. Am Fam Physician. Aug 1 2021;104(2):171–178. https://www.ncbi.nlm.nih.gov/pubmed/34383430
- American Cancer Society (ACS). Understanding a Breast Cancer Diagnosis.Accessed 02/02/2025, https://www.cancer.org/content/dam/CRC/PDF/Public/8580.00.pdf
- Orrantia-Borunda EA-N, P.; Acuña-Aguilar, L. E.; et al. . Subtypes of BreastCancer. In: Mayrovitz HN, editor. Breast Cancer [Internet]. Brisbane (AU):Exon Publications; 2022 Aug 6. Chapter 3. Available at: https://www.ncbi.nlm.nih.gov/books/NBK583808/ . 2022;
- National Cancer Institute. Breast Cancer Treatment (PDQ) - Patient Version.Updated 12/11/2024. Accessed 02/02/2025, https://www.cancer.gov/types/breast/patient/breast-treatment-pdq
- Kang S, Kim SB. HER2-Low Breast Cancer: Now and in the Future. Cancer research and treatment : official journal of Korean Cancer Association . Jul 2024;56(3):700–720. doi:10.4143/crt.2023.1138 https://www.ncbi.nlm.nih.gov/pubmed/38291745
- Zhang H, Peng Y. Current Biological, Pathological and Clinical Landscape ofHER2-Low Breast Cancer. Cancers (Basel). Dec 252022;15(1)doi:10.3390/cancers15010126 https://www.ncbi.nlm.nih.gov/pubmed/36612123
- American Cancer Society. Breast Cancer Early Detection and Diagnosis.Updated 12/19/2023. Accessed 02/02/2025, https://www.cancer.org/cancer/types/breast-cancer/screening-tests-and-early-detection.html
- Katsika L, Boureka E, Kalogiannidis I, et al. Screening for Breast Cancer: AComparative Review of Guidelines. Life (Basel). Jun 192024;14(6)doi:10.3390/life14060777 https://www.ncbi.nlm.nih.gov/pubmed/38929759
- American Cancer Society. Breast Ultrasound. Updated 12/19/2023. Accessed4/18/2025, https://www.cancer.org/cancer/types/breast-cancer/screening-tests-and-early-detection/breast-ultrasound.html
- American Cancer Society. Breast MRI. Updated 1/14/2022. Accessed 4/14/2025, https://www.cancer.org/cancer/types/breast-cancer/screening-tests-and-early-detection/breast-mri-scans.html
- National Comprehensive Cancer Network (NCCN). Clinical Practice Guidelinesin Oncology: Breast Cancer. Accessed 7/30/2024, https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf
- Choi L. Breast Cancer. Merck Manual: Professional Version. Updated 9/2023.Accessed 8/22/2024, https://www.merckmanuals.com/professional/gynecology-and-obstetrics/breast-cancer/breast-cancer
- American Society of Clinical Oncology. Hereditary breast and ovarian cancer. https://www.cancer.net/cancer-types/hereditary-breast-and-ovarian-cancer
- Liu T, Yu J, Gao Y, et al. Prophylactic Interventions for Hereditary Breastand Ovarian Cancer Risks and Mortality in BRCA1/2 Carriers. Cancers (Basel) . Dec 24 2023;16(1)doi:10.3390/cancers16010103 https://www.ncbi.nlm.nih.gov/pubmed/38201529
- Teichgraeber DC, Guirguis MS, Whitman GJ. Breast Cancer Staging: Updates inthe AJCC Cancer Staging Manual, 8th Edition, and Current Challenges forRadiologists, From the AJR Special Series on Cancer Staging. AJR Am J Roentgenol . Aug 2021;217(2):278–290. doi:10.2214/AJR.20.25223 https://www.ncbi.nlm.nih.gov/pubmed/33594908
- Chung HL, Le-Petross HT, Leung JWT. Imaging Updates to Breast Cancer LymphNode Management. Radiographics. Sep–Oct 2021;41(5):1283–1299.doi:10.1148/rg.2021210053 https://www.ncbi.nlm.nih.gov/pubmed/34469221
- Amin MB, Edge SB, Greene FL, et al. AJCC cancer staging manual. vol1024. Springer.
- Gown AM. The Biomarker Ki-67: Promise, Potential, and Problems in BreastCancer. Applied immunohistochemistry & molecular morphology : AIMM . Aug 1 2023;31(7):478–484. doi:10.1097/PAI.0000000000001087 https://www.ncbi.nlm.nih.gov/pubmed/36730064
- Arun RP, Cahill HF, Marcato P. Breast Cancer Subtype-Specific miRNAs:Networks, Impacts, and the Potential for Intervention. Biomedicines. Mar 11 2022;10(3)doi:10.3390/biomedicines10030651 https://www.ncbi.nlm.nih.gov/pubmed/35327452
- Chakraborty S, Guan Z, Kostrzewa CE, Shen R, Begg CB. Identifying somaticfingerprints of cancers defined by germline and environmental risk factors.Genet Epidemiol. Dec 2024;48(8):455–467. doi:10.1002/gepi.22565 https://www.ncbi.nlm.nih.gov/pubmed/38686586
- Chen C, Lin CJ, Pei YC, et al. Comprehensive genomic profiling of breastcancers characterizes germline-somatic mutation interactions mediatingtherapeutic vulnerabilities. Cell Discov. Dec 19 2023;9(1):125.doi:10.1038/s41421-023-00614-3 https://www.ncbi.nlm.nih.gov/pubmed/38114467
- Zhu H, Dogan BE. American Joint Committee on Cancer's Staging System forBreast Cancer, Eighth Edition: Summary for Clinicians. Eur J Breast Health . Jul 2021;17(3):234–238. doi:10.4274/ejbh.galenos.2021.2021-4-3 https://www.ncbi.nlm.nih.gov/pubmed/34263150
- Gradishar WJ, Moran MS, Abraham J, et al. Breast Cancer, Version 3.2024,NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network . 01 Jul. 2024 2024;22(5):331–357. doi:10.6004/jnccn.2024.0035https://jnccn.org/view/journals/jnccn/22/5/article-p331.xml https://jnccn.org/downloadpdf/view/journals/jnccn/22/5/article-p331.pdf
- Wimmer K, Hlauschek D, Balic M, et al. Is the CTS5 a helpful decision-makingtool in the extended adjuvant therapy setting? Breast Cancer Res Treat . Jun 2024;205(2):227–239. doi:10.1007/s10549-023-07186-6 https://www.ncbi.nlm.nih.gov/pubmed/38273214
- Tajiri W, Ijichi H, Takizawa K, et al. The clinical usefulness of the CTS5in the prediction of late distant recurrence in postmenopausal women withestrogen receptor-positive early breast cancer. Breast Cancer. Jan2021;28(1):67–74. doi:10.1007/s12282-020-01130-y https://www.ncbi.nlm.nih.gov/pubmed/32601800
- Noordhoek I, Treuner K, Putter H, et al. Breast Cancer Index PredictsExtended Endocrine Benefit to Individualize Selection of Patients with HR(+)Early-stage Breast Cancer for 10 Years of Endocrine Therapy. Clin Cancer Res . Jan 1 2021;27(1):311–319. doi:10.1158/1078-0432.CCR-20-2737 https://www.ncbi.nlm.nih.gov/pubmed/33109739
- Bartlett JMS, Xu K, Wong J, et al. Validation of the Prognostic Performanceof Breast Cancer Index in Hormone Receptor-Positive Postmenopausal BreastCancer Patients in the TEAM Trial. Clin Cancer Res. Apr 152024;30(8):1509–1517. doi:10.1158/1078-0432.CCR-23-2436 https://www.ncbi.nlm.nih.gov/pubmed/38345755
- Shah S, Shaing C, Khatib J, et al. The Utility of Breast Cancer Index (BCI)Over Clinical Prognostic Tools for Predicting the Need for ExtendedEndocrine Therapy: A Safety Net Hospital Experience. Clinical breast cancer . Dec 2022;22(8):823–827. doi:10.1016/j.clbc.2022.08.003 https://www.ncbi.nlm.nih.gov/pubmed/36089460
- Yeo HY, Liew AC, Chan SJ, Anwar M, Han CH, Marra CA. Understanding PatientPreferences Regarding the Important Determinants of Breast Cancer Treatment:A Narrative Scoping Review. Patient Prefer Adherence.2023;17:2679–2706. doi:10.2147/PPA.S432821 https://www.ncbi.nlm.nih.gov/pubmed/37927344
- American Cancer Society. Treating Breast Cancer. Updated 1/11/2023. Accessed02/02/2025, https://www.cancer.org/cancer/types/breast-cancer/treatment.html
- Owusu-Brackett N, Facer B, Quiroga D, et al. Axillary Management: How MuchIs Too Much? Curr Oncol Rep. Jul 2024;26(7):735–743.doi:10.1007/s11912-024-01539-0 https://www.ncbi.nlm.nih.gov/pubmed/38748364
- James J, Law M, Sengupta S, Saunders C. Assessment of the axilla in womenwith early-stage breast cancer undergoing primary surgery: a review. World J Surg Oncol . May 9 2024;22(1):127. doi:10.1186/s12957-024-03394-6 https://www.ncbi.nlm.nih.gov/pubmed/38725006
- Tomita K, Kubo T. Recent advances in surgical techniques for breastreconstruction. Int J Clin Oncol. Jul 2023;28(7):841–846.doi:10.1007/s10147-023-02313-1 https://www.ncbi.nlm.nih.gov/pubmed/36848021
- Paganini A, Westesson LM, Hansson E, Karlsson SA. Women's decision processwhen actively choosing to 'go flat' after breast cancer: a constructivistgrounded theory study. BMC Womens Health. Mar 15 2024;24(1):178.doi:10.1186/s12905-024-03015-0 https://www.ncbi.nlm.nih.gov/pubmed/38491353
- Kunkler IH, Williams LJ, Jack WJL, Cameron DA, Dixon JM. Breast-ConservingSurgery with or without Irradiation in Early Breast Cancer. N Engl J Med . Feb 16 2023;388(7):585–594. doi:10.1056/NEJMoa2207586 https://www.ncbi.nlm.nih.gov/pubmed/36791159
- Whelan TJ, Smith S, Parpia S, et al. Omitting Radiotherapy afterBreast-Conserving Surgery in Luminal A Breast Cancer. N Engl J Med.Aug 17 2023;389(7):612–619. doi:10.1056/NEJMoa2302344 https://www.ncbi.nlm.nih.gov/pubmed/37585627
- Harris SR. Brachial plexopathy after breast cancer: A persistent late effectof radiotherapy. PM R. Jan 2024;16(1):85–91. doi:10.1002/pmrj.13007 https://www.ncbi.nlm.nih.gov/pubmed/37272709
- Catalano O, Fusco R, Carriero S, Tamburrini S, Granata V. UltrasoundFindings After Breast Cancer Radiation Therapy: Cutaneous, Pleural,Pulmonary, and Cardiac Changes. Korean journal of radiology. Nov2024;25(11):982–991. doi:10.3348/kjr.2024.0672 https://www.ncbi.nlm.nih.gov/pubmed/39473089
- Sardar P, Kundu A, Chatterjee S, et al. Long-term cardiovascular mortalityafter radiotherapy for breast cancer: A systematic review and meta-analysis.Clin Cardiol. Feb 2017;40(2):73–81. doi:10.1002/clc.22631 https://www.ncbi.nlm.nih.gov/pubmed/28244595
- Jacobs JEJ, L'Hoyes W, Lauwens L, et al. Mortality and Major Adverse CardiacEvents in Patients With Breast Cancer Receiving Radiotherapy: The FirstDecade. J Am Heart Assoc. Apr 18 2023;12(8):e027855.doi:10.1161/JAHA.122.027855 https://www.ncbi.nlm.nih.gov/pubmed/37026536
- Chargari C, Deutsch E, Blanchard P, et al. Brachytherapy: An overview forclinicians. CA: a cancer journal for clinicians. Sep2019;69(5):386–401. doi:10.3322/caac.21578 https://www.ncbi.nlm.nih.gov/pubmed/31361333
- Huo J, Giordano SH, Smith BD, Shaitelman SF, Smith GL. Contemporary ToxicityProfile of Breast Brachytherapy Versus External Beam Radiation AfterLumpectomy for Breast Cancer. Int J Radiat Oncol Biol Phys. Mar 152016;94(4):709–18. doi:10.1016/j.ijrobp.2015.12.013 https://www.ncbi.nlm.nih.gov/pubmed/26972643
- Al Sukhun S, Temin S, Barrios CH, et al. Systemic Treatment of Patients WithMetastatic Breast Cancer: ASCO Resource-Stratified Guideline. JCO Glob Oncol . Jan 2024;10:e2300285. doi:10.1200/GO.23.00285 https://www.ncbi.nlm.nih.gov/pubmed/38206277
- Gradishar WJ, Moran MS, Abraham J, et al. Breast Cancer, Version 3.2024,NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network : JNCCN . Jul 2024;22(5):331–357. doi:10.6004/jnccn.2024.0035 https://www.ncbi.nlm.nih.gov/pubmed/39019058
- Sung WWY, Sharma K, Chan AW, Al Khaifi M, Oldenburger E, Chuk E. A narrativereview of the challenges and impact of breast cancer treatment in olderadults beyond cancer diagnosis. Ann Palliat Med. Nov2024;13(6):1521–1529. doi:10.21037/apm-24-90 https://www.ncbi.nlm.nih.gov/pubmed/39523217
- Hu L, Xu B, Chau PH, et al. Reproductive Concerns Among Young Adult WomenWith Breast Cancer: A Systematic Review and Meta-Analysis. Psycho-oncology . Aug 2024;33(8):e9304. doi:10.1002/pon.9304 https://www.ncbi.nlm.nih.gov/pubmed/39160674
- Choupani E, Mahmoudi Gomari M, Zanganeh S, et al. Newly Developed TargetedTherapies Against the Androgen Receptor in Triple-Negative Breast Cancer: AReview. Pharmacol Rev. Mar 2023;75(2):309–327.doi:10.1124/pharmrev.122.000665 https://www.ncbi.nlm.nih.gov/pubmed/36781219
- Li Z, Wei H, Li S, Wu P, Mao X. The Role of Progesterone Receptors in BreastCancer. Drug Des Devel Ther. 2022;16:305–314.doi:10.2147/DDDT.S336643 https://www.ncbi.nlm.nih.gov/pubmed/35115765
- Long Y, Xie S, Liu Q, et al. The experiences of adjuvant endocrine therapyfor women breast cancer survivors: A literature review. Medicine (Baltimore) . Dec 22 2023;102(51):e36704. doi:10.1097/MD.0000000000036704 https://www.ncbi.nlm.nih.gov/pubmed/38134074
- Todd A, Waldron C, McGeagh L, et al. Identifying determinants of adherenceto adjuvant endocrine therapy following breast cancer: A systematic reviewof reviews. Cancer Med. Feb 2024;13(3):e6937. doi:10.1002/cam4.6937 https://www.ncbi.nlm.nih.gov/pubmed/38240343
- Lee EY, Lee DW, Lee KH, Im SA. Recent Developments in the TherapeuticLandscape of Advanced or Metastatic Hormone Receptor-Positive Breast Cancer. Cancer research and treatment : official journal of Korean Cancer Association . Oct 2023;55(4):1065–1076. doi:10.4143/crt.2023.846 https://www.ncbi.nlm.nih.gov/pubmed/37817306
- Iwase T, Saji S, Iijima K, et al. Postoperative Adjuvant Anastrozole for 10or 5 Years in Patients With Hormone Receptor-Positive Breast Cancer: AERAS,a Randomized Multicenter Open-Label Phase III Trial. J Clin Oncol.Jun 20 2023;41(18):3329–3338. doi:10.1200/JCO.22.00577 https://www.ncbi.nlm.nih.gov/pubmed/37079878
- Gnant M, Fitzal F, Rinnerthaler G, et al. Duration of AdjuvantAromatase-Inhibitor Therapy in Postmenopausal Breast Cancer. N Engl J Med . Jul 29 2021;385(5):395–405. doi:10.1056/NEJMoa2104162 https://www.ncbi.nlm.nih.gov/pubmed/34320285
- Burstein HJ, Lacchetti C, Anderson H, et al. Adjuvant Endocrine Therapy forWomen With Hormone Receptor-Positive Breast Cancer: ASCO Clinical PracticeGuideline Focused Update. J Clin Oncol. Feb 10 2019;37(5):423–438.doi:10.1200/JCO.18.01160 https://www.ncbi.nlm.nih.gov/pubmed/30452337
- Blok EJ, Kroep JR, Meershoek-Klein Kranenbarg E, et al. Optimal Duration ofExtended Adjuvant Endocrine Therapy for Early Breast Cancer; Results of theIDEAL Trial (BOOG 2006-05). J Natl Cancer Inst. Jan 12018;110(1)doi:10.1093/jnci/djx134 https://www.ncbi.nlm.nih.gov/pubmed/28922787
- Yung RL, Davidson NE. Searching for the IDEAL Duration of Adjuvant EndocrineTherapy. J Natl Cancer Inst. Jan 1 2018;110(1):6–8.doi:10.1093/jnci/djx143 https://www.ncbi.nlm.nih.gov/pubmed/28922788
- Khoury K, Lynce F, Barac A, et al. Long-term follow-up assessment of cardiacsafety in SAFE-HEaRt, a clinical trial evaluating the use of HER2-targetedtherapies in patients with breast cancer and compromised heart function.Breast Cancer Res Treat. Feb 2021;185(3):863–868.doi:10.1007/s10549-020-06053-y https://www.ncbi.nlm.nih.gov/pubmed/33400034
- Stanowicka-Grada M, Senkus E. Anti-HER2 Drugs for the Treatment of AdvancedHER2 Positive Breast Cancer. Curr Treat Options Oncol. Nov2023;24(11):1633–1650. doi:10.1007/s11864-023-01137-5 https://www.ncbi.nlm.nih.gov/pubmed/37878202
- Jackson C, Finikarides L, Freeman ALJ. The adverse effects oftrastuzumab-containing regimes as a therapy in breast cancer: A piggy-backsystematic review and meta-analysis. PLoS One.2022;17(12):e0275321. doi:10.1371/journal.pone.0275321 https://www.ncbi.nlm.nih.gov/pubmed/36454979
- Geyer CE, Jr., Untch M, Huang CS, et al. Survival with Trastuzumab Emtansinein Residual HER2-Positive Breast Cancer. N Engl J Med. Jan 162025;392(3):249–257. doi:10.1056/NEJMoa2406070 https://www.ncbi.nlm.nih.gov/pubmed/39813643
- Modi S, Jacot W, Yamashita T, et al. Trastuzumab Deruxtecan in PreviouslyTreated HER2-Low Advanced Breast Cancer. N Engl J Med. Jul 72022;387(1):9–20. doi:10.1056/NEJMoa2203690 https://www.ncbi.nlm.nih.gov/pubmed/35665782
- Fu Z, Liu J, Li S, Shi C, Zhang Y. Treatment-related adverse eventsassociated with HER2-Targeted antibody-drug conjugates in clinical trials: asystematic review and meta-analysis. EClinicalMedicine. Jan2023;55:101795. doi:10.1016/j.eclinm.2022.101795 https://www.ncbi.nlm.nih.gov/pubmed/36712893
- Pellarin I, Dall'Acqua A, Favero A, et al. Cyclin-dependent protein kinasesand cell cycle regulation in biology and disease. Signal Transduct Target Ther . Jan 13 2025;10(1):11. doi:10.1038/s41392-024-02080-z https://www.ncbi.nlm.nih.gov/pubmed/39800748
- Danbala IA, Wang X, Su Y, et al. Efficacy of cyclin-dependent kinases 4 and6 inhibitors in the treatment of HR +/HER2 - advanced breast cancer: ameta-analysis. Eur J Med Res. Jun 24 2025;30(1):519.doi:10.1186/s40001-025-02806-x https://www.ncbi.nlm.nih.gov/pubmed/40551183
- Niraula S. Adjuvant CDK4/6 inhibitors in breast cancer: Interpreting trialdesign, evidence, and uncertainty. Cancer Treat Rev. May2025;136:102944. doi:10.1016/j.ctrv.2025.102944 https://www.ncbi.nlm.nih.gov/pubmed/40262369
- Rastogi P, O'Shaughnessy J, Martin M, et al. Adjuvant Abemaciclib PlusEndocrine Therapy for Hormone Receptor-Positive, Human Epidermal GrowthFactor Receptor 2-Negative, High-Risk Early Breast Cancer: Results From aPreplanned monarchE Overall Survival Interim Analysis, Including 5-YearEfficacy Outcomes. J Clin Oncol. Mar 20 2024;42(9):987–993.doi:10.1200/JCO.23.01994 https://www.ncbi.nlm.nih.gov/pubmed/38194616
- Slamon D, Lipatov O, Nowecki Z, et al. Ribociclib plus Endocrine Therapy inEarly Breast Cancer. N Engl J Med. Mar 21 2024;390(12):1080–1091.doi:10.1056/NEJMoa2305488 https://www.ncbi.nlm.nih.gov/pubmed/38507751
- Caswell-Jin JL, Freedman RA, Hassett MJ, et al. Optimal AdjuvantChemotherapy and Targeted Therapy for Early Breast Cancer-CDK4/6 Inhibitors:ASCO Rapid Guideline Update Clinical Insights. JCO Oncol Pract. Mar2025;21(3):287–291. doi:10.1200/OP-24-00663 https://www.ncbi.nlm.nih.gov/pubmed/39303175
- Loibl S, Martin M, Bonnefoi H, et al. Final survival results from thePENELOPE-B trial investigating palbociclib versus placebo for patients withhigh-risk HR+/HER2- breast cancer and residual disease after neoadjuvantchemotherapy. Ann Oncol. Jul 2025;36(7):832–837.doi:10.1016/j.annonc.2025.03.010 https://www.ncbi.nlm.nih.gov/pubmed/40139460
- Gnant M, Dueck AC, Frantal S, et al. Adjuvant Palbociclib for Early BreastCancer: The PALLAS Trial Results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol . Jan 20 2022;40(3):282–293. doi:10.1200/JCO.21.02554 https://www.ncbi.nlm.nih.gov/pubmed/34874182
- Hao C, Wei Y, Meng W, Zhang J, Yang X. PI3K/AKT/mTOR inhibitors for hormonereceptor-positive advanced breast cancer. Cancer Treat Rev. Jan2025;132:102861. doi:10.1016/j.ctrv.2024.102861 https://www.ncbi.nlm.nih.gov/pubmed/39662202
- Andre F, Ciruelos E, Rubovszky G, et al. Alpelisib for PIK3CA-Mutated,Hormone Receptor-Positive Advanced Breast Cancer. N Engl J Med. May16 2019;380(20):1929–1940. doi:10.1056/NEJMoa1813904 https://www.ncbi.nlm.nih.gov/pubmed/31091374
- Turner NC, Oliveira M, Howell SJ, et al. Capivasertib in HormoneReceptor-Positive Advanced Breast Cancer. N Engl J Med. Jun 12023;388(22):2058–2070. doi:10.1056/NEJMoa2214131 https://www.ncbi.nlm.nih.gov/pubmed/37256976
- Food and Drug Administration. FDA approves capivasertib with fulvestrant forbreast cancer. Updated 11/16/2023. Accessed 9/5/2024, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-capivasertib-fulvestrant-breast-cancer
- Geyer CE, Jr., Garber JE, Gelber RD, et al. Overall survival in the OlympiAphase III trial of adjuvant olaparib in patients with germline pathogenicvariants in BRCA1/2 and high-risk, early breast cancer. Ann Oncol.Dec 2022;33(12):1250–1268. doi:10.1016/j.annonc.2022.09.159 https://www.ncbi.nlm.nih.gov/pubmed/36228963
- Dvir K, Giordano S, Leone JP. Immunotherapy in Breast Cancer. Int J Mol Sci. Jul 9 2024;25(14)doi:10.3390/ijms25147517 https://www.ncbi.nlm.nih.gov/pubmed/39062758
- Schmid P, Cortes J, Dent R, et al. Event-free Survival with Pembrolizumab inEarly Triple-Negative Breast Cancer. N Engl J Med. Feb 102022;386(6):556–567. doi:10.1056/NEJMoa2112651 https://www.ncbi.nlm.nih.gov/pubmed/35139274
- Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus Chemotherapy inAdvanced Triple-Negative Breast Cancer. N Engl J Med. Jul 212022;387(3):217–226. doi:10.1056/NEJMoa2202809 https://www.ncbi.nlm.nih.gov/pubmed/35857659
- Ballinger TJ, Thompson WR, Guise TA. The bone-muscle connection in breastcancer: implications and therapeutic strategies to preserve musculoskeletalhealth. Breast Cancer Res. Nov 23 2022;24(1):84.doi:10.1186/s13058-022-01576-2 https://www.ncbi.nlm.nih.gov/pubmed/36419084
- Stokes G, Herath M, Samad N, Trinh A, Milat F. 'Bone Health-Across a Woman'sLifespan'. Clin Endocrinol (Oxf). Apr 2025;102(4):389–402.doi:10.1111/cen.15203 https://www.ncbi.nlm.nih.gov/pubmed/39871618
- Motlaghzadeh Y, Wu JY. Approach to Bone Health in the Patient With BreastCancer. J Clin Endocrinol Metab. Sep 16 2024;109(10):e1902–e1910.doi:10.1210/clinem/dgae404 https://www.ncbi.nlm.nih.gov/pubmed/38864566
- Goel B, Virmani T, Jain V, Kumar G, Sharma A, Al Noman A. Unveiling the LinkBetween Breast Cancer Treatment and Osteoporosis: Implications forAnticancer Therapy and Bone Health. Biomed Res Int.2024;2024:5594542. doi:10.1155/2024/5594542 https://www.ncbi.nlm.nih.gov/pubmed/39574432
- Teissonniere M, Point M, Biver E, et al. Bone Effects of Anti-CancerTreatments in 2024. Calcified tissue international. Mar 272025;116(1):54. doi:10.1007/s00223-025-01362-0 https://www.ncbi.nlm.nih.gov/pubmed/40146323
- Christensen Holz S. Aromatase Inhibitor Musculoskeletal Syndrome and BoneLoss: a Review of the Current Literature. Curr Oncol Rep. Jul2023;25(7):825–831. doi:10.1007/s11912-023-01413-5 https://www.ncbi.nlm.nih.gov/pubmed/37052869
- Nunes FAP, de Farias MLF, Oliveira FP, et al. Use of aromatase inhibitors inpatients with breast cancer is associated with deterioration of bonemicroarchitecture and density. Arch Endocrinol Metab. Nov 32021;65(4):505–511. doi:10.20945/2359-3997000000385 https://www.ncbi.nlm.nih.gov/pubmed/34283901
- Kuba S, Chiba K, Watanabe K, et al. Aromatase inhibitors, bonemicrostructure, and estimated bone strength in postmenopausal women withbreast cancer: a 5-year prospective study. Journal of bone and mineral metabolism . Mar 2025;43(2):133–140. doi:10.1007/s00774-024-01560-0 https://www.ncbi.nlm.nih.gov/pubmed/39508861
- Kuba S, Watanabe K, Chiba K, et al. Adjuvant endocrine therapy effects onbone mineral density and microstructure in women with breast cancer. Journal of bone and mineral metabolism . Nov 2021;39(6):1031–1040. doi:10.1007/s00774-021-01239-w https://www.ncbi.nlm.nih.gov/pubmed/34191126
- Blomqvist C, Vehmanen L, Kellokumpu-Lehtinen PL, et al. Long-term effects ofaromatase inhibitor withdrawal on bone mineral density in early breastcancer patients: 10-year follow-up results of the BREX study. Breast Cancer Res Treat . Jul 2024;206(1):57–65. doi:10.1007/s10549-024-07252-7 https://www.ncbi.nlm.nih.gov/pubmed/38561578
- Castaneda S, Casas A, Gonzalez-Del-Alba A, et al. Bone loss induced bycancer treatments in breast and prostate cancer patients. Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico . Nov 2022;24(11):2090–2106. doi:10.1007/s12094-022-02872-1 https://www.ncbi.nlm.nih.gov/pubmed/35779210
- Diana A, Carlino F, Giunta EF, et al. Cancer Treatment-Induced Bone Loss(CTIBL): State of the Art and Proper Management in Breast Cancer Patients onEndocrine Therapy. Curr Treat Options Oncol. Apr 16 2021;22(5):45.doi:10.1007/s11864-021-00835-2 https://www.ncbi.nlm.nih.gov/pubmed/33864145
- Shapiro CL, Van Poznak C, Lacchetti C, et al. Management of Osteoporosis inSurvivors of Adult Cancers With Nonmetastatic Disease: ASCO ClinicalPractice Guideline. J Clin Oncol. Nov 1 2019;37(31):2916–2946.doi:10.1200/JCO.19.01696 https://www.ncbi.nlm.nih.gov/pubmed/31532726
- Lu H, Lei X, Zhao H, et al. Bone Mineral Density at the Time of InitiatingAromatase Inhibitor Therapy Is Associated With Decreased Fractures in WomenWith Breast Cancer. J Bone Miner Res. May 2021;36(5):861–871.doi:10.1002/jbmr.4250 https://www.ncbi.nlm.nih.gov/pubmed/33484602
- Bailey S, Mhango G, Lin JJ. The impact of bone mineral density screening onincident fractures and healthcare resource utilization among postmenopausalbreast cancer survivors treated with aromatase inhibitors. Osteoporos Int . Sep 2022;33(9):1989–1997. doi:10.1007/s00198-022-06458-z https://www.ncbi.nlm.nih.gov/pubmed/35697870
- LeBoff MS, Greenspan SL, Insogna KL, et al. The clinician's guide toprevention and treatment of osteoporosis. Osteoporos Int. Oct2022;33(10):2049–2102. doi:10.1007/s00198-021-05900-y https://www.ncbi.nlm.nih.gov/pubmed/35478046
- Kim KY, Kim KM. Similarities and differences between bone qualityparameters, trabecular bone score and femur geometry. PLoS One.2022;17(1):e0260924. doi:10.1371/journal.pone.0260924 https://www.ncbi.nlm.nih.gov/pubmed/35015756
- Catalano A, Gaudio A, Agostino RM, Morabito N, Bellone F, Lasco A.Trabecular bone score and quantitative ultrasound measurements in theassessment of bone health in breast cancer survivors assuming aromataseinhibitors. J Endocrinol Invest. Nov 2019;42(11):1337–1343.doi:10.1007/s40618-019-01063-0 https://www.ncbi.nlm.nih.gov/pubmed/31127591
- Ye C, Leslie WD. Fracture risk and assessment in adults with cancer. Osteoporos Int . Mar 2023;34(3):449–466. doi:10.1007/s00198-022-06631-4 https://www.ncbi.nlm.nih.gov/pubmed/36512057
- Hadji P, Aapro MS, Body JJ, et al. Management of AromataseInhibitor-Associated Bone Loss (AIBL) in postmenopausal women with hormonesensitive breast cancer: Joint position statement of the IOF, CABS, ECTS,IEG, ESCEO IMS, and SIOG. Journal of bone oncology. Jun2017;7:1–12. doi:10.1016/j.jbo.2017.03.001 https://www.ncbi.nlm.nih.gov/pubmed/28413771
- Bassatne A, Bou Khalil A, Chakhtoura M, Arabi A, Van Poznak C, El-HajjFuleihan G. Effect of antiresorptive therapy on aromatase inhibitor inducedbone loss in postmenopausal women with early-stage breast cancer: Asystematic review and meta-analysis of randomized controlled trials. Metabolism . Mar 2022;128:154962. doi:10.1016/j.metabol.2021.154962 https://www.ncbi.nlm.nih.gov/pubmed/34958816
- Waqas K, Lima Ferreira J, Tsourdi E, Body JJ, Hadji P, Zillikens MC. Updatedguidance on the management of cancer treatment-induced bone loss (CTIBL) inpre- and postmenopausal women with early-stage breast cancer. Journal of bone oncology . Jun 2021;28:100355. doi:10.1016/j.jbo.2021.100355 https://www.ncbi.nlm.nih.gov/pubmed/33948427
- Coleman R. Clinical benefits of bone targeted agents in early breast cancer.Breast (Edinburgh, Scotland). Nov 2019;48 Suppl 1:S92–S96.doi:10.1016/S0960-9776(19)31133-6 https://www.ncbi.nlm.nih.gov/pubmed/31839171
- Liu Y, Zhao S, Zhang Y, Onwuka JU, Zhang Q, Liu X. Bisphosphonates andbreast cancer survival: a meta-analysis and trial sequential analysis of81508 participants from 23 prospective epidemiological studies. Aging (Albany NY) . Aug 10 2021;13(15):19835–19866. doi:10.18632/aging.203395 https://www.ncbi.nlm.nih.gov/pubmed/34375305
- Awan AA, Stober C, Pond GR, et al. A randomised trial comparing 6-monthlyadjuvant zoledronate with a single one-time dose in patients with earlybreast cancer. Breast Cancer Res Treat. Dec 2024;208(3):523–533.doi:10.1007/s10549-024-07443-2 https://www.ncbi.nlm.nih.gov/pubmed/39083190
- Amgen. Prolia (denosumab). Full prescribing information. Updated 9/2011.Accessed 6/9/2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/125320s5s6lbl.pdf
- Eastell R. Evaluation and management of aromatase inhibitor-induced boneloss. UpToDate. Updated 7/29/2024. Accessed 6/9/2025, https://www.uptodate.com/contents/evaluation-and-management-of-aromatase-inhibitor-induced-bone-loss#H1674841258
- Coleman R, Zhou Y, Jandial D, Cadieux B, Chan A. Bone Health Outcomes fromthe International, Multicenter, Randomized, Phase 3, Placebo-ControlledD-CARE Study Assessing Adjuvant Denosumab in Early Breast Cancer. Adv Ther . Aug 2021;38(8):4569–4580. doi:10.1007/s12325-021-01812-9 https://www.ncbi.nlm.nih.gov/pubmed/34185259
- Bracchi P, Zecca E, Brunelli C, et al. A real-world study on the prevalenceand risk factors of medication related osteonecrosis of the jaw in cancerpatients with bone metastases treated with Denosumab. Cancer Med.Sep 2023;12(17):18317–18326. doi:10.1002/cam4.6429 https://www.ncbi.nlm.nih.gov/pubmed/37559413
- Moretti L, Richelmi L, Cosentini D, et al. Adjuvant denosumab for earlybreast cancer-Evidence and controversy. Breast (Edinburgh, Scotland) . Dec 2024;78:103826. doi:10.1016/j.breast.2024.103826 https://www.ncbi.nlm.nih.gov/pubmed/39509862
- Gnant M, Frantal S, Pfeiler G, et al. Long-Term Outcomes of AdjuvantDenosumab in Breast Cancer. NEJM Evid. Dec2022;1(12):EVIDoa2200162. doi:10.1056/EVIDoa2200162 https://www.ncbi.nlm.nih.gov/pubmed/38319865
- Coleman R, Finkelstein DM, Barrios C, et al. Adjuvant denosumab in earlybreast cancer (D-CARE): an international, multicentre, randomised,controlled, phase 3 trial. Lancet Oncol. Jan 2020;21(1):60–72.doi:10.1016/S1470-2045(19)30687-4 https://www.ncbi.nlm.nih.gov/pubmed/31806543
- Scafetta R, Donato M, Gullotta C, et al. Comparative analysis of Denosumaband Zoledronic acid in advanced breast cancer patients receiving CDK4/6inhibitors. Breast (Edinburgh, Scotland). Aug 2025;82:104502.doi:10.1016/j.breast.2025.104502 https://www.ncbi.nlm.nih.gov/pubmed/40424680
- White L. Rochester Study: Women with Breast Cancer Have Low Vitamin DLevels. University of Rochester Medical Center. Updated 10/8/2009. Accessed6/9/2025, https://www.urmc.rochester.edu/news/story/rochester-study--women-with-breast-cancer-have-low-vitamin-d-levels
- Wu J, Liang XY, Hu L, et al. Bone health in newly diagnosed female breastcancer patients in China: a cross-sectional study. Sci Rep. Feb 12025;15(1):3982. doi:10.1038/s41598-024-84698-1 https://www.ncbi.nlm.nih.gov/pubmed/39893249
- Abdel-Razeq H, Al-Rasheed U, Mashhadani N, et al. The efficacy of acomprehensive bone health program in maintaining bone mineral density inpostmenopausal women with early-stage breast cancer treated with endocrinetherapy: real-world data. Ir J Med Sci. Dec 2022;191(6):2511–2515.doi:10.1007/s11845-021-02897-5 https://www.ncbi.nlm.nih.gov/pubmed/35088228
- Arul Vijaya Vani S, Ananthanarayanan PH, Kadambari D, Harichandrakumar KT,Niranjjan R, Nandeesha H. Effects of vitamin D and calcium supplementationon side effects profile in patients of breast cancer treated with letrozole.Clin Chim Acta. Aug 1 2016;459:53–56. doi:10.1016/j.cca.2016.05.020 https://www.ncbi.nlm.nih.gov/pubmed/27221206
- Peppone LJ, Ling M, Huston AJ, et al. The effects of high-dose calcitrioland individualized exercise on bone metabolism in breast cancer survivors onhormonal therapy: a phase II feasibility trial. Support Care Cancer. Aug 2018;26(8):2675–2683. doi:10.1007/s00520-018-4094-4 https://www.ncbi.nlm.nih.gov/pubmed/29470705
- Shane EB, J. R. UpToDate: Treatment of hypercalcemia. Available at https://www.uptodate.com/contents/treatment-of-hypercalcemia?topicRef=842&source=see_link Last updated 03/08/2024. Accessed 08/05/2025. 2024;
- Khan AF, Karami S, Peidl AS, Waiters KD, Babajide MF, Bawa-Khalfe T.Androgen Receptor in Hormone Receptor-Positive Breast Cancer. Int J Mol Sci . Dec 29 2023;25(1)doi:10.3390/ijms25010476 https://www.ncbi.nlm.nih.gov/pubmed/38203649
- Agostinetto E, Curigliano G, Piccart M. Emerging treatments in HER2-positiveadvanced breast cancer: Keep raising the bar. Cell Rep Med. Jun 182024;5(6):101575. doi:10.1016/j.xcrm.2024.101575 https://www.ncbi.nlm.nih.gov/pubmed/38759648
- Guglielmi G, Del Re M, Gol LS, Bengala C, Danesi R, Fogli S. Pharmacologicalinsights on novel oral selective estrogen receptor degraders in breastcancer. Eur J Pharmacol. Apr 15 2024;969:176424.doi:10.1016/j.ejphar.2024.176424 https://www.ncbi.nlm.nih.gov/pubmed/38402929
- Alaluf E, Shalamov MM, Sonnenblick A. Update on current and new potentialimmunotherapies in breast cancer, from bench to bedside. Front Immunol . 2024;15:1287824. doi:10.3389/fimmu.2024.1287824 https://www.ncbi.nlm.nih.gov/pubmed/38433837
- Heater NK, Warrior S, Lu J. Current and future immunotherapy for breastcancer. J Hematol Oncol. Dec 25 2024;17(1):131.doi:10.1186/s13045-024-01649-z https://www.ncbi.nlm.nih.gov/pubmed/39722028
- Vasileiou M, Diamantoudis SC, Tsianava C, Nguyen NP. ImmunotherapeuticStrategies Targeting Breast Cancer Stem Cells. Current oncology (Toronto, Ont) . May 29 2024;31(6):3040–3063. doi:10.3390/curroncol31060232 https://www.ncbi.nlm.nih.gov/pubmed/38920716
- Serrano Garcia L, Javega B, Llombart Cussac A, et al. Patterns of immuneevasion in triple-negative breast cancer and new potential therapeutictargets: a review. Front Immunol. 2024;15:1513421.doi:10.3389/fimmu.2024.1513421 https://www.ncbi.nlm.nih.gov/pubmed/39735530
- Rodin D, Glicksman RM, Hepel JT, et al. Early-Stage Breast Cancer: ACritical Review of Current and Emerging Practice. Int J Radiat Oncol Biol Phys . Dec 1 2024;120(5):1260–1272. doi:10.1016/j.ijrobp.2024.08.037 https://www.ncbi.nlm.nih.gov/pubmed/39237044
- Wegge-Larsen C, Mehlsen M, Jensen AB. The motivation of breast cancerpatients to participate in a national randomized control trial. Support Care Cancer . Jul 15 2023;31(8):468. doi:10.1007/s00520-023-07930-0 https://www.ncbi.nlm.nih.gov/pubmed/37452876
- National Cancer Institute. Clinical Trials Information for Patients andCaregivers. Accessed 01/21/2025, https://www.cancer.gov/research/participate/clinical-trials
- Gao Y, Hao J, Zhang Z. Adjuvant Chemotherapy for Breast Cancer in OlderAdult Patients. Clin Interv Aging. 2024;19:1281–1286.doi:10.2147/CIA.S470262 https://www.ncbi.nlm.nih.gov/pubmed/39050516
- Torrisi C, Wareg NK, Anbari AB. Decision-making for bilateral risk-reducingmastectomy for an increased lifetime breast cancer risk: A qualitativemetasynthesis. Psycho-oncology. Mar 2024;33(3):e6311.doi:10.1002/pon.6311 https://www.ncbi.nlm.nih.gov/pubmed/38429973
- Afzal MZ, Vahdat LT. Evolving Management of Breast Cancer in the Era ofPredictive Biomarkers and Precision Medicine. J Pers Med. Jul 32024;14(7):719. doi:10.3390/jpm14070719 https://www.ncbi.nlm.nih.gov/pubmed/39063972
- Iams WT, Mackay M, Ben-Shachar R, et al. Concurrent Tissue and CirculatingTumor DNA Molecular Profiling to Detect Guideline-Based Targeted Mutationsin a Multicancer Cohort. JAMA Netw Open. Jan 2 2024;7(1):e2351700.doi:10.1001/jamanetworkopen.2023.51700 https://www.ncbi.nlm.nih.gov/pubmed/38252441
- Lloyd MR, Jhaveri K, Kalinsky K, Bardia A, Wander SA. Precision therapeuticsand emerging strategies for HR-positive metastatic breast cancer. Nature reviews Clinical oncology . Oct 2024;21(10):743–761. doi:10.1038/s41571-024-00935-6 https://www.ncbi.nlm.nih.gov/pubmed/39179659
- Andre F, Ismaila N, Allison KH, et al. Biomarkers for Adjuvant Endocrine andChemotherapy in Early-Stage Breast Cancer: ASCO Guideline Update. J Clin Oncol . Jun 1 2022;40(16):1816–1837. doi:10.1200/JCO.22.00069 https://www.ncbi.nlm.nih.gov/pubmed/35439025
- Addanki S, Meas S, Sarli VN, Singh B, Lucci A. Applications of CirculatingTumor Cells and Circulating Tumor DNA in Precision Oncology for BreastCancers. Int J Mol Sci. Jul 16 2022;23(14)doi:10.3390/ijms23147843 https://www.ncbi.nlm.nih.gov/pubmed/35887191
- Kotsifaki A, Maroulaki S, Armakolas A. Exploring the Immunological Profilein Breast Cancer: Recent Advances in Diagnosis and Prognosis throughCirculating Tumor Cells. Int J Mol Sci. Apr 292024;25(9)doi:10.3390/ijms25094832 https://www.ncbi.nlm.nih.gov/pubmed/38732051
- Mazzeo R, Sears J, Palmero L, et al. Liquid biopsy in triple-negative breastcancer: unlocking the potential of precision oncology. ESMO open.Oct 2024;9(10):103700. doi:10.1016/j.esmoop.2024.103700 https://www.ncbi.nlm.nih.gov/pubmed/39288656
- Rock CL, Thomson CA, Sullivan KR, et al. American Cancer Society nutritionand physical activity guideline for cancer survivors. CA: a cancer journal for clinicians . May 2022;72(3):230–262. doi:10.3322/caac.21719 https://www.ncbi.nlm.nih.gov/pubmed/35294043
- Ositelu KC, Peesay T, Garcia C, Akhter N. Life's Essential 8 andCardiovascular Disease in Breast Cancer Survivors. Current cardiology reports . Feb 15 2025;27(1):55. doi:10.1007/s11886-025-02216-9 https://www.ncbi.nlm.nih.gov/pubmed/39954113
- American Cancer Society. Physical Activity and the Person with Cancer.Updated 3/16/2022. Accessed 9/9/2024, https://www.cancer.org/cancer/survivorship/be-healthy-after-treatment/physical-activity-and-the-cancer-patient.html
- Khalifa A, Guijarro A, Nencioni A. Advances in Diet and Physical Activity inBreast Cancer Prevention and Treatment. Nutrients. Jul 132024;16(14):2262. doi:10.3390/nu16142262 https://www.ncbi.nlm.nih.gov/pubmed/39064705
- Castro-Espin C, Agudo A. The Role of Diet in Prognosis among CancerSurvivors: A Systematic Review and Meta-Analysis of Dietary Patterns andDiet Interventions. Nutrients. Jan 142022;14(2)doi:10.3390/nu14020348 https://www.ncbi.nlm.nih.gov/pubmed/35057525
- Chen G, Leary S, Niu J, Perry R, Papadaki A. The Role of the MediterraneanDiet in Breast Cancer Survivorship: A Systematic Review and Meta-Analysis ofObservational Studies and Randomised Controlled Trials. Nutrients.Apr 27 2023;15(9)doi:10.3390/nu15092099 https://www.ncbi.nlm.nih.gov/pubmed/37432242
- Anemoulis M, Vlastos A, Kachtsidis V, Karras SN. Intermittent Fasting inBreast Cancer: A Systematic Review and Critical Update of Available Studies.Nutrients. Jan 19 2023;15(3):532. doi:10.3390/nu15030532 https://www.ncbi.nlm.nih.gov/pubmed/36771239
- Marinac CR, Nelson SH, Breen CI, et al. Prolonged Nightly Fasting and BreastCancer Prognosis. JAMA Oncol. Aug 1 2016;2(8):1049–55.doi:10.1001/jamaoncol.2016.0164 https://www.ncbi.nlm.nih.gov/pubmed/27032109
- Urzi AG, Tropea E, Gattuso G, et al. Ketogenic Diet and Breast Cancer:Recent Findings and Therapeutic Approaches. Nutrients. Oct 132023;15(20)doi:10.3390/nu15204357 https://www.ncbi.nlm.nih.gov/pubmed/37892432
- Hardt L, Mahamat-Saleh Y, Aune D, Schlesinger S. Plant-Based Diets andCancer Prognosis: a Review of Recent Research. Current nutrition reports . Dec 2022;11(4):695–716. doi:10.1007/s13668-022-00440-1 https://www.ncbi.nlm.nih.gov/pubmed/36138327
- Cariolou M, Abar L, Aune D, et al. Postdiagnosis recreational physicalactivity and breast cancer prognosis: Global Cancer Update Programme (CUPGlobal) systematic literature review and meta-analysis. Int J Cancer . Feb 15 2023;152(4):600–615. doi:10.1002/ijc.34324 https://www.ncbi.nlm.nih.gov/pubmed/36279903
- Lee J. A Meta-analysis of the Association Between Physical Activity andBreast Cancer Mortality. Cancer nursing. Jul/Aug2019;42(4):271–285. doi:10.1097/NCC.0000000000000580 https://www.ncbi.nlm.nih.gov/pubmed/29601358
- Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvanttherapy for breast cancer. Cochrane Database Syst Rev. Sep 212016;9(9):CD005001. doi:10.1002/14651858.CD005001.pub3 https://www.ncbi.nlm.nih.gov/pubmed/27650122
- Lahart IM, Metsios GS, Nevill AM, Carmichael AR. Physical activity for womenwith breast cancer after adjuvant therapy. Cochrane Database Syst Rev . Jan 29 2018;1(1):CD011292. doi:10.1002/14651858.CD011292.pub2 https://www.ncbi.nlm.nih.gov/pubmed/29376559
- (ACS) ACS. Chemotherapy Side Effects. Updated 05/01/2020. Accessed01/25/2023, https://www.cancer.org/treatment/treatments-and-side-effects/treatment-types/chemotherapy/chemotherapy-side-effects.html
- Lee J, Hwang Y. The effects of exercise interventions on fatigue, bodycomposition, physical fitness, and biomarkers in breast cancer patientsduring and after treatment: a systematic review and meta-analysis ofrandomized controlled trials. Journal of cancer survivorship : research and practice . Mar 8 2025;doi:10.1007/s11764-025-01772-x https://www.ncbi.nlm.nih.gov/pubmed/40056311
- American Cancer Society. Psychosocial Support Options for People withCancer. Updated 6/9/2023. Accessed 9/9/2024, https://www.cancer.org/cancer/survivorship/coping/understanding-psychosocial-support-services.html
- Ding X, Zhao F, Zhu M, et al. A systematic review and meta-analysis ofinterventions to reduce perceived stress in breast cancer patients. Complement Ther Clin Pract . Feb 2024;54:101803. doi:10.1016/j.ctcp.2023.101803 https://www.ncbi.nlm.nih.gov/pubmed/38159534
- American Cancer Society. Practice Mindfulness and Relaxation. Updated12/2/2020. Accessed 9/9/2024, https://www.cancer.org/cancer/survivorship/coping/practice-mindfulness-and-relaxation.html
- Clougher D, Ciria-Suarez L, Medina JC, Anastasiadou D, Racioppi A,Ochoa-Arnedo C. What works in peer support for breast cancer survivors: Aqualitative systematic review and meta-ethnography. Applied psychology Health and well-being . May 2024;16(2):793–815. doi:10.1111/aphw.12473 https://www.ncbi.nlm.nih.gov/pubmed/37493002
- Azizi M, Heshmatnia F, Milani H, Shahhosseini Z, Marvdashti LM, Moghadam ZB.The Effectiveness of Cognitive Behavioral Therapy on Depression and AnxietySymptoms in Breast Cancer Patients and Survivors: A Systematic Review ofInterventional Studies. Brain Behav. Oct 2024;14(10):e70098.doi:10.1002/brb3.70098 https://www.ncbi.nlm.nih.gov/pubmed/39467209
- Li M, Liu F, Han X, Li J, Fan Y. Effect of Internet-Based CognitiveBehavioral Therapy on Psychological Distress and Quality of Life AmongBreast Cancer Survivors: A Meta-Analysis of Randomized Controlled Trials.Psycho-oncology. Nov 2024;33(11):e70014. doi:10.1002/pon.70014 https://www.ncbi.nlm.nih.gov/pubmed/39562510
- Yasueda A, Urushima H, Ito T. Efficacy and Interaction of AntioxidantSupplements as Adjuvant Therapy in Cancer Treatment: A Systematic Review.Integr Cancer Ther. Mar 2016;15(1):17–39.doi:10.1177/1534735415610427 https://www.ncbi.nlm.nih.gov/pubmed/26503419
- Li Y, Lin Q, Lu X, Li W. Post-Diagnosis use of Antioxidant VitaminSupplements and Breast Cancer Prognosis: A Systematic Review andMeta-Analysis. Clinical breast cancer. Dec 2021;21(6):477–485.doi:10.1016/j.clbc.2021.09.001 https://www.ncbi.nlm.nih.gov/pubmed/34635464
- Ambrosone CB, Zirpoli GR, Hutson AD, et al. Dietary Supplement Use DuringChemotherapy and Survival Outcomes of Patients With Breast Cancer Enrolledin a Cooperative Group Clinical Trial (SWOG S0221). J Clin Oncol.Mar 10 2020;38(8):804–814. doi:10.1200/JCO.19.01203 https://www.ncbi.nlm.nih.gov/pubmed/31855498
- Kanellopoulou A, Riza E, Samoli E, Benetou V. Dietary Supplement Use afterCancer Diagnosis in Relation to Total Mortality, Cancer Mortality andRecurrence: A Systematic Review and Meta-Analysis. Nutr Cancer.2021;73(1):16–30. doi:10.1080/01635581.2020.1734215 https://www.ncbi.nlm.nih.gov/pubmed/32148118
- Leelaviwat N, Mekraksakit P, Cross KM, et al. Melatonin: Translation ofOngoing Studies Into Possible Therapeutic Applications Outside SleepDisorders. Clin Ther. May 2022;44(5):783–812.doi:10.1016/j.clinthera.2022.03.008 https://www.ncbi.nlm.nih.gov/pubmed/35400533
- Azzeh FS, Kamfar WW, Ghaith MM, et al. Unlocking the health benefits ofmelatonin supplementation: A promising preventative and therapeuticstrategy. Medicine (Baltimore). Sep 20 2024;103(38):e39657.doi:10.1097/MD.0000000000039657 https://www.ncbi.nlm.nih.gov/pubmed/39312371
- Cardinali DP, Pandi-Perumal SR, Brown GM. Melatonin as a Chronobiotic andCytoprotector in Non-communicable Diseases: More than an Antioxidant. Sub-cellular biochemistry . 2024;107:217–244. doi:10.1007/978-3-031-66768-8_11 https://www.ncbi.nlm.nih.gov/pubmed/39693027
- Pardi PC, Turri JAO, Bayer L, et al. Biological action of melatonin ontarget receptors in breast cancer. Rev Assoc Med Bras (1992).2024;70(3):e20231260. doi:10.1590/1806-9282.20231260 https://www.ncbi.nlm.nih.gov/pubmed/38656007
- Lissoni P, Barni S, Mandala M, et al. Decreased toxicity and increasedefficacy of cancer chemotherapy using the pineal hormone melatonin inmetastatic solid tumour patients with poor clinical status. Eur J Cancer . Nov 1999;35(12):1688–92. doi:10.1016/s0959-8049(99)00159-8 https://www.ncbi.nlm.nih.gov/pubmed/10674014
- Lissoni P, Barni S, Meregalli S, et al. Modulation of cancer endocrinetherapy by melatonin: a phase II study of tamoxifen plus melatonin inmetastatic breast cancer patients progressing under tamoxifen alone. Br J Cancer . Apr 1995;71(4):854–6. doi:10.1038/bjc.1995.164 https://www.ncbi.nlm.nih.gov/pubmed/7710954
- Madsen MT, Hansen MV, Andersen LT, et al. Effect of Melatonin on Sleep inthe Perioperative Period after Breast Cancer Surgery: A Randomized,Double-Blind, Placebo-Controlled Trial. J Clin Sleep Med. Feb2016;12(2):225–33. doi:10.5664/jcsm.5490 https://www.ncbi.nlm.nih.gov/pubmed/26414973
- Hansen MV, Andersen LT, Madsen MT, et al. Effect of melatonin on depressivesymptoms and anxiety in patients undergoing breast cancer surgery: arandomized, double-blind, placebo-controlled trial. Breast Cancer Res Treat . Jun 2014;145(3):683–95. doi:10.1007/s10549-014-2962-2 https://www.ncbi.nlm.nih.gov/pubmed/24756186
- Palmer ACS, Zortea M, Souza A, et al. Clinical impact of melatonin on breastcancer patients undergoing chemotherapy; effects on cognition, sleep anddepressive symptoms: A randomized, double-blind, placebo-controlled trial.PLoS One. 2020;15(4):e0231379. doi:10.1371/journal.pone.0231379 https://www.ncbi.nlm.nih.gov/pubmed/32302347
- Sedighi Pashaki A, Mohammadian K, Afshar S, et al. A Randomized, Controlled,Parallel-Group, Trial on the Effects of Melatonin on Fatigue Associated withBreast Cancer and Its Adjuvant Treatments. Integr Cancer Ther.Jan–Dec 2021;20:1534735420988343. doi:10.1177/1534735420988343 https://www.ncbi.nlm.nih.gov/pubmed/33543655
- Sedighi Pashaki A, Sheida F, Moaddab Shoar L, et al. A Randomized,Controlled, Parallel-Group, Trial on the Long-term Effects of Melatonin onFatigue Associated With Breast Cancer and Its Adjuvant Treatments. Integr Cancer Ther . Jan–Dec 2023;22:15347354231168624. doi:10.1177/15347354231168624 https://www.ncbi.nlm.nih.gov/pubmed/37139718
- Innominato PF, Lim AS, Palesh O, et al. The effect of melatonin on sleep andquality of life in patients with advanced breast cancer. Support Care Cancer . Mar 2016;24(3):1097–105. doi:10.1007/s00520-015-2883-6 https://www.ncbi.nlm.nih.gov/pubmed/26260726
- Chen WY, Giobbie-Hurder A, Gantman K, et al. A randomized,placebo-controlled trial of melatonin on breast cancer survivors: impact onsleep, mood, and hot flashes. Breast Cancer Res Treat. Jun2014;145(2):381–8. doi:10.1007/s10549-014-2944-4 https://www.ncbi.nlm.nih.gov/pubmed/24718775
- Mukhopadhyay ND, Khorasanchi A, Pandey S, et al. Melatonin Supplementationfor Cancer-Related Fatigue in Patients With Early Stage Breast CancerReceiving Radiotherapy: A Double-Blind Placebo-Controlled Trial. The oncologist . Feb 2 2024;29(2):e206–e212. doi:10.1093/oncolo/oyad250 https://www.ncbi.nlm.nih.gov/pubmed/37699115
- Pistiolis L, Khaki D, Kovacs A, Olofsson Bagge R. The Effect of MelatoninIntake on Survival of Patients with Breast Cancer-A Population-BasedRegistry Study. Cancers (Basel). Nov 292022;14(23)doi:10.3390/cancers14235884 https://www.ncbi.nlm.nih.gov/pubmed/36497366
- Zetner D, Kamby C, Christophersen C, et al. Effect of melatonin cream onacute radiation dermatitis in patients with primary breast cancer: Adouble-blind, randomized, placebo-controlled trial. J Pineal Res.Aug 2023;75(1):e12873. doi:10.1111/jpi.12873 https://www.ncbi.nlm.nih.gov/pubmed/37055944
- Zetner D, Kamby C, Gulen S, et al. Quality-of-life outcomes followingtopical melatonin application against acute radiation dermatitis in patientswith early breast cancer: A double-blind, randomized, placebo-controlledtrial. J Pineal Res. Jan 2023;74(1):e12840. doi:10.1111/jpi.12840 https://www.ncbi.nlm.nih.gov/pubmed/36385713
- Ben-David MA, Elkayam R, Gelernter I, Pfeffer RM. Melatonin for Preventionof Breast Radiation Dermatitis: A Phase II, Prospective, Double-BlindRandomized Trial. The Israel Medical Association journal : IMAJ.Mar–Apr 2016;18(3-4):188–92. https://www.ncbi.nlm.nih.gov/pubmed/27228641
- Mokbel K, Mokbel K. Harnessing Micronutrient Power: Vitamins, Antioxidantsand Probiotics in Breast Cancer Prevention. Anticancer Res. Jun2024;44(6):2287–2295. doi:10.21873/anticanres.17036 https://www.ncbi.nlm.nih.gov/pubmed/38821606
- Gutierrez S, Svahn SL, Johansson ME. Effects of Omega-3 Fatty Acids onImmune Cells. Int J Mol Sci. Oct 112019;20(20)doi:10.3390/ijms20205028 https://www.ncbi.nlm.nih.gov/pubmed/31614433
- Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules toman. Biochem Soc Trans. Oct 15 2017;45(5):1105–1115.doi:10.1042/BST20160474 https://www.ncbi.nlm.nih.gov/pubmed/28900017
- Bougnoux P, Hajjaji N, Ferrasson MN, Giraudeau B, Couet C, Le Floch O.Improving outcome of chemotherapy of metastatic breast cancer bydocosahexaenoic acid: a phase II trial. Br J Cancer. Dec 152009;101(12):1978–85. doi:10.1038/sj.bjc.6605441 https://www.ncbi.nlm.nih.gov/pubmed/19920822
- Darwito D, Dharmana E, Riwanto I, et al. Effects of Omega-3 Supplementationon Ki-67 and VEGF Expression Levels and Clinical Outcomes of LocallyAdvanced Breast Cancer Patients Treated with Neoadjuvant CAF Chemotherapy: ARandomized Controlled Trial Report. Asian Pac J Cancer Prev. Mar 262019;20(3):911–916. doi:10.31557/APJCP.2019.20.3.911 https://www.ncbi.nlm.nih.gov/pubmed/30912414
- Suzumura DN, Schleder JC, Appel MH, Naliwaiko K, Tanhoffer R, Fernandes LC.Fish Oil Supplementation Enhances Pulmonary Strength and Endurance in WomenUndergoing Chemotherapy. Nutr Cancer. Aug–Sep 2016;68(6):935–42.doi:10.1080/01635581.2016.1187282 https://www.ncbi.nlm.nih.gov/pubmed/27340931
- Frankhouser DE, DeWees T, Snodgrass IF, et al. Randomized dose-responsetrial of n-3 fatty acids in hormone receptor negative breast cancersurvivors - impact on breast adipose oxylipin and DNA methylation patterns.Am J Clin Nutr. Jul 2025;122(1):70–82.doi:10.1016/j.ajcnut.2025.04.021 https://www.ncbi.nlm.nih.gov/pubmed/40288580
- Almassri HF, Abdul Kadir A, Srour M, Foo LH. The effects of Omega-3 fattyacids and vitamin D supplementation on the quality of life and bloodinflammation markers in newly diagnosed breast cancer women: Anopen-labelled randomised controlled trial. Clin Nutr ESPEN. Feb2025;65:64–75. doi:10.1016/j.clnesp.2024.11.014 https://www.ncbi.nlm.nih.gov/pubmed/39577691
- de la Rosa Oliva F, Meneses Garcia A, Ruiz Calzada H, et al. Effects ofomega-3 fatty acids supplementation on neoadjuvant chemotherapy-inducedtoxicity in patients with locally advanced breast cancer: a randomized,controlled, double-blinded clinical trial. Nutr Hosp. Aug 262019;36(4):769–776. Efecto de la suplementacion con acidos grasos omega -3sobre la toxicidad secundaria a quimioterapia neoadyuvante en pacientes concancer de mama localmente avanzado: ensayo clinico aleatorizado.doi:10.20960/nh.2338 https://www.ncbi.nlm.nih.gov/pubmed/31192682
- Paixao E, Oliveira ACM, Pizato N, et al. The effects of EPA and DHA enrichedfish oil on nutritional and immunological markers of treatment naive breastcancer patients: a randomized double-blind controlled trial. Nutr J. Oct 23 2017;16(1):71. doi:10.1186/s12937-017-0295-9 https://www.ncbi.nlm.nih.gov/pubmed/29061183
- Ghoreishi Z, Esfahani A, Djazayeri A, et al. Omega-3 fatty acids areprotective against paclitaxel-induced peripheral neuropathy: a randomizeddouble-blind placebo controlled trial. BMC Cancer. Aug 152012;12(1):355. doi:10.1186/1471-2407-12-355 https://www.ncbi.nlm.nih.gov/pubmed/22894640
- Tawfik B, Dayao ZR, Brown-Glaberman UA, et al. A pilot randomized,placebo-controlled, double-blind study of omega-3 fatty acids to preventpaclitaxel-associated acute pain syndrome in breast cancer patients:Alliance A22_Pilot2. Support Care Cancer. Oct 17 2023;31(12):637.doi:10.1007/s00520-023-08082-x https://www.ncbi.nlm.nih.gov/pubmed/37847317
- Voutsadakis IA. Vitamin D baseline levels at diagnosis of breast cancer: Asystematic review and meta-analysis. Hematol Oncol Stem Cell Ther.Mar 2021;14(1):16–26. doi:10.1016/j.hemonc.2020.08.005 https://www.ncbi.nlm.nih.gov/pubmed/33002425
- Chang SW, Lee HC. Vitamin D and health - The missing vitamin in humans. Pediatr Neonatol. Jun 2019;60(3):237–244.doi:10.1016/j.pedneo.2019.04.007 https://www.ncbi.nlm.nih.gov/pubmed/31101452
- Shu C, Yang Q, Huang J, et al. Pretreatment plasma vitamin D and response toneoadjuvant chemotherapy in breast cancer: evidence from pooled analysis ofcohort studies. International journal of surgery (London, England).Dec 1 2024;110(12):8126–8135. doi:10.1097/JS9.0000000000002142 https://www.ncbi.nlm.nih.gov/pubmed/39806750
- Li C, Li H, Zhong H, Li X. Association of 25-hydroxyvitamin D level withsurvival outcomes in female breast cancer patients: A meta-analysis. J Steroid Biochem Mol Biol . Sep 2021;212:105947. doi:10.1016/j.jsbmb.2021.105947 https://www.ncbi.nlm.nih.gov/pubmed/34214604
- Omodei MS, Chimicoviaki J, Buttros DAB, et al. Vitamin D SupplementationImproves Pathological Complete Response in Breast Cancer Patients UndergoingNeoadjuvant Chemotherapy: A Randomized Clinical Trial. Nutr Cancer.Mar 17 2025;77(6):648–657. doi:10.1080/01635581.2025.2480854 https://www.ncbi.nlm.nih.gov/pubmed/40098326
- Ozkurt E, Ordu C, Ozmen T, et al. Vitamin D Supplementation DuringNeoadjuvant Chemotherapy for Breast Cancer Improves Pathological CompleteResponse: A Prospective Randomized Clinical Trial. World journal of surgery . Jun 2025;49(6):1396–1405. doi:10.1002/wjs.12587 https://www.ncbi.nlm.nih.gov/pubmed/40229998
- Shahvegharasl Z, Pirouzpanah S, Mahboob SA, et al. Effects ofcholecalciferol supplementation on serum angiogenic biomarkers in breastcancer patients treated with tamoxifen: A controlled randomized clinicaltrial. Nutrition. Apr 2020;72:110656. doi:10.1016/j.nut.2019.110656 https://www.ncbi.nlm.nih.gov/pubmed/31901710
- Almassri HF, Abdul Kadir A, Srour M, Foo LH. The Effects of Omega-3 FattyAcids and Vitamin D Supplementation on the Nutritional Status of Women withBreast Cancer in Palestine: An Open-Label Randomized Controlled Trial. Nutrients . Nov 20 2024;16(22)doi:10.3390/nu16223960 https://www.ncbi.nlm.nih.gov/pubmed/39599746
- Codini M. Why Vitamin C Could Be an Excellent Complementary Remedy toConventional Therapies for Breast Cancer. Int J Mol Sci. Nov 92020;21(21)doi:10.3390/ijms21218397 https://www.ncbi.nlm.nih.gov/pubmed/33182353
- Harris HR, Orsini N, Wolk A. Vitamin C and survival among women with breastcancer: a meta-analysis. Eur J Cancer. May 2014;50(7):1223–31.doi:10.1016/j.ejca.2014.02.013 https://www.ncbi.nlm.nih.gov/pubmed/24613622
- Suhail N, Bilal N, Khan HY, et al. Effect of vitamins C and E on antioxidantstatus of breast-cancer patients undergoing chemotherapy. J Clin Pharm Ther . Feb 2012;37(1):22–6. doi:10.1111/j.1365-2710.2010.01237.x https://www.ncbi.nlm.nih.gov/pubmed/21204889
- Park H, Kang J, Choi J, Heo S, Lee DH. The Effect of High Dose IntravenousVitamin C During Radiotherapy on Breast Cancer Patients'Neutrophil-Lymphocyte Ratio. J Altern Complement Med. Nov2020;26(11):1039–1046. doi:10.1089/acm.2020.0138 https://www.ncbi.nlm.nih.gov/pubmed/32876471
- Ou J, Zhu X, Zhang H, et al. A Retrospective Study of Gemcitabine andCarboplatin With or Without Intravenous Vitamin C on Patients With AdvancedTriple-Negative Breast Cancer. Integr Cancer Ther. Jan–Dec2020;19:1534735419895591. doi:10.1177/1534735419895591 https://www.ncbi.nlm.nih.gov/pubmed/32070148
- Vollbracht C, Schneider B, Leendert V, Weiss G, Auerbach L, Beuth J.Intravenous vitamin C administration improves quality of life in breastcancer patients during chemo-/radiotherapy and aftercare: results of aretrospective, multicentre, epidemiological cohort study in Germany. In Vivo . Nov–Dec 2011;25(6):983–90. https://www.ncbi.nlm.nih.gov/pubmed/22021693
- de Sousa Coelho M, Pereira IC, de Oliveira KGF, et al. Chemopreventive andanti-tumor potential of vitamin E in preclinical breast cancer studies: Asystematic review. Clin Nutr ESPEN. Feb 2023;53:60–73.doi:10.1016/j.clnesp.2022.11.001 https://www.ncbi.nlm.nih.gov/pubmed/36657931
- Argyriou AA, Chroni E, Koutras A, et al. Vitamin E for prophylaxis againstchemotherapy-induced neuropathy: a randomized controlled trial. Neurology. Jan 11 2005;64(1):26–31. doi:10.1212/01.WNL.0000148609.35718.7D https://www.ncbi.nlm.nih.gov/pubmed/15642899
- Moustafa I, Connolly C, Anis M, Mustafa H, Oosthuizen F, Viljoen M. Aprospective study to evaluate the efficacy and safety of vitamin E andlevocarnitine prophylaxis against doxorubicin-induced cardiotoxicity inadult breast cancer patients. J Oncol Pharm Pract. Mar2024;30(2):354–366. doi:10.1177/10781552231171114 https://www.ncbi.nlm.nih.gov/pubmed/37157803
- de Oliveira VA, Oliveira IKF, Pereira IC, et al. Consumption andsupplementation of vitamin E in breast cancer risk, treatment, and outcomes:A systematic review with meta-analysis. Clin Nutr ESPEN. Apr2023;54:215–226. doi:10.1016/j.clnesp.2023.01.032 https://www.ncbi.nlm.nih.gov/pubmed/36963866
- Jacobson G, Bhatia S, Smith BJ, Button AM, Bodeker K, Buatti J. Randomizedtrial of pentoxifylline and vitamin E vs standard follow-up after breastirradiation to prevent breast fibrosis, evaluated by tissue compliancemeter. Int J Radiat Oncol Biol Phys. Mar 1 2013;85(3):604–8.doi:10.1016/j.ijrobp.2012.06.042 https://www.ncbi.nlm.nih.gov/pubmed/22846413
- Wang Y, Zhao Y, Chong F, et al. A dose-response meta-analysis of green teaconsumption and breast cancer risk. Int J Food Sci Nutr. Sep2020;71(6):656–667. doi:10.1080/09637486.2020.1715353 https://www.ncbi.nlm.nih.gov/pubmed/31959020
- Sinha D, Biswas J, Nabavi SM, Bishayee A. Tea phytochemicals for breastcancer prevention and intervention: From bench to bedside and beyond. Semin Cancer Biol. Oct 2017;46:33–54. doi:10.1016/j.semcancer.2017.04.001 https://www.ncbi.nlm.nih.gov/pubmed/28396252
- Gianfredi V, Nucci D, Abalsamo A, et al. Green Tea Consumption and Risk ofBreast Cancer and Recurrence-A Systematic Review and Meta-Analysis ofObservational Studies. Nutrients. Dec 3 2018;10(12):1886.doi:10.3390/nu10121886 https://www.ncbi.nlm.nih.gov/pubmed/30513889
- Yu SS, Spicer DV, Hawes D, et al. Biological effects of green tea capsulesupplementation in pre-surgery postmenopausal breast cancer patients. Frontiers in oncology . 2013;3:298. doi:10.3389/fonc.2013.00298 https://www.ncbi.nlm.nih.gov/pubmed/24380073
- Bao PP, Zhao GM, Shu XO, et al. Modifiable Lifestyle Factors andTriple-negative Breast Cancer Survival: A Population-based ProspectiveStudy. Epidemiology (Cambridge, Mass). Nov 2015;26(6):909–16.doi:10.1097/EDE.0000000000000373 https://www.ncbi.nlm.nih.gov/pubmed/26360370
- Zhang G, Wang Y, Zhang Y, et al. Anti-cancer activities of teaepigallocatechin-3-gallate in breast cancer patients under radiotherapy.Current molecular medicine. Feb 2012;12(2):163–76.doi:10.2174/156652412798889063 https://www.ncbi.nlm.nih.gov/pubmed/22280355
- Zhao H, Zhu W, Jia L, et al. Phase I study of topicalepigallocatechin-3-gallate (EGCG) in patients with breast cancer receivingadjuvant radiotherapy. The British journal of radiology.2016;89(1058):20150665. doi:10.1259/bjr.20150665 https://www.ncbi.nlm.nih.gov/pubmed/26607642
- Zhu W, Jia L, Chen G, et al. Epigallocatechin-3-gallate amelioratesradiation-induced acute skin damage in breast cancer patients undergoingadjuvant radiotherapy. Oncotarget. Jul 26 2016;7(30):48607–48613.doi:10.18632/oncotarget.9495 https://www.ncbi.nlm.nih.gov/pubmed/27224910
- Bayet-Robert M, Kwiatkowski F, Leheurteur M, et al. Phase I dose escalationtrial of docetaxel plus curcumin in patients with advanced and metastaticbreast cancer. Cancer Biol Ther. Jan 2010;9(1):8–14.doi:10.4161/cbt.9.1.10392 https://www.ncbi.nlm.nih.gov/pubmed/19901561
- Ryan JL, Heckler CE, Ling M, et al. Curcumin for radiation dermatitis: a randomized, double-blind, placebo-controlled clinical trial of thirty breastcancer patients. Radiation research. Jul 2013;180(1):34–43.doi:10.1667/RR3255.1 https://www.ncbi.nlm.nih.gov/pubmed/23745991
- Mayo B, Penroz S, Torres K, Simon L. Curcumin Administration Routes inBreast Cancer Treatment. Int J Mol Sci. Oct 262024;25(21)doi:10.3390/ijms252111492 https://www.ncbi.nlm.nih.gov/pubmed/39519045
- Saghatelyan T, Tananyan A, Janoyan N, et al. Efficacy and safety of curcuminin combination with paclitaxel in patients with advanced, metastatic breastcancer: A comparative, randomized, double-blind, placebo-controlled clinicaltrial. Phytomedicine. Apr 15 2020;70:153218.doi:10.1016/j.phymed.2020.153218 https://www.ncbi.nlm.nih.gov/pubmed/32335356
- Chai W, Cooney RV, Franke AA, et al. Plasma coenzyme Q10 levels andpostmenopausal breast cancer risk: the multiethnic cohort study. Cancer Epidemiol Biomarkers Prev . Sep 2010;19(9):2351–6. doi:10.1158/1055-9965.EPI-10-0396 https://www.ncbi.nlm.nih.gov/pubmed/20668119
- Abdi S, Montazeri V, Garjani A, Shayanfar A, Pirouzpanah S. Coenzyme Q10 inassociation with metabolism-related AMPK/PFKFB3 and angiogenic VEGF/VEGFR2genes in breast cancer patients. Molecular biology reports. Apr2020;47(4):2459–2473. doi:10.1007/s11033-020-05310-z https://www.ncbi.nlm.nih.gov/pubmed/32140960
- Hertz N, Lister RE. Improved survival in patients with end-stage cancertreated with coenzyme Q(10) and other antioxidants: a pilot study. J Int Med Res . Nov–Dec 2009;37(6):1961–71. doi:10.1177/147323000903700634 https://www.ncbi.nlm.nih.gov/pubmed/20146896
- Lockwood K, Moesgaard S, Hanioka T, Folkers K. Apparent partial remission ofbreast cancer in 'high risk' patients supplemented with nutritionalantioxidants, essential fatty acids and coenzyme Q10. Mol Aspects Med . 1994;15 Suppl:s231–40. doi:10.1016/0098-2997(94)90033-7 https://www.ncbi.nlm.nih.gov/pubmed/7752835
- Lockwood K, Moesgaard S, Folkers K. Partial and complete regression ofbreast cancer in patients in relation to dosage of coenzyme Q10. Biochem Biophys Res Commun. Mar 30 1994;199(3):1504–8. doi:10.1006/bbrc.1994.1401 https://www.ncbi.nlm.nih.gov/pubmed/7908519
- Barnish M, Sheikh M, Scholey A. Nutrient Therapy for the Improvement ofFatigue Symptoms. Nutrients. Apr 302023;15(9)doi:10.3390/nu15092154 https://www.ncbi.nlm.nih.gov/pubmed/37432282
- Iwase S, Kawaguchi T, Yotsumoto D, et al. Efficacy and safety of an aminoacid jelly containing coenzyme Q10 and L-carnitine in controlling fatigue inbreast cancer patients receiving chemotherapy: a multi-institutional,randomized, exploratory trial (JORTC-CAM01). Support Care Cancer.Feb 2016;24(2):637–646. doi:10.1007/s00520-015-2824-4 https://www.ncbi.nlm.nih.gov/pubmed/26105516
- Lesser GJ, Case D, Stark N, et al. A randomized, double-blind,placebo-controlled study of oral coenzyme Q10 to relieve self-reportedtreatment-related fatigue in newly diagnosed patients with breast cancer.J Support Oncol. Mar 2013;11(1):31–42.doi:10.1016/j.suponc.2012.03.003 https://www.ncbi.nlm.nih.gov/pubmed/22682875
- Alimohammadi M, Rahimi A, Faramarzi F, et al. Effects of coenzyme Q10supplementation on inflammation, angiogenesis, and oxidative stress inbreast cancer patients: a systematic review and meta-analysis of randomizedcontrolled- trials. Inflammopharmacol. Jun 2021;29(3):579–593.doi:10.1007/s10787-021-00817-8 https://www.ncbi.nlm.nih.gov/pubmed/34008150
- Guo C, Liu Y, Fu H, Zhang X, Li M. Effect of cruciferous vegetable intake oncancer: An umbrella review of meta-analysis. J Food Sci. Sep2024;89(9):5230–5244. doi:10.1111/1750-3841.17300 https://www.ncbi.nlm.nih.gov/pubmed/39138635
- Jabbarzadeh Kaboli P, Afzalipour Khoshkbejari M, Mohammadi M, et al. Targetsand mechanisms of sulforaphane derivatives obtained from cruciferous plantswith special focus on breast cancer - contradictory effects and futureperspectives. Biomed Pharmacother. Jan https://www.ncbi.nlm.nih.gov/pubmed/31739165 2020;121:109635.doi:10.1016/j.biopha.2019.109635
- Wang Z, Tu C, Pratt R, et al. A Presurgical-Window Intervention Trial of Isothiocyanate-Rich Broccoli Sprout Extract in Patients with Breast Cancer.Mol Nutr Food Res. Jun 2022;66(12):e2101094.doi:10.1002/mnfr.202101094 https://www.ncbi.nlm.nih.gov/pubmed/35475592
- Singh SP. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library ofMedicine (US). Identifier NCT03934905. Phase II Trial of Effects of theNutritional Supplement Sulforaphane on Doxorubicin-Associated CardiacDysfunction (CRI18-026). Updated 11/20/2024. 4/16/2025. https://clinicaltrials.gov/study/NCT03934905
- Reyes-Hernandez OD, Figueroa-Gonzalez G, Quintas-Granados LI, et al. 3,3'-Diindolylmethane and indole-3-carbinol: potential therapeutic moleculesfor cancer chemoprevention and treatment via regulating cellular signalingpathways. Cancer Cell Int. Aug 26 2023;23(1):180.doi:10.1186/s12935-023-03031-4 https://www.ncbi.nlm.nih.gov/pubmed/37633886
- Reed GA, Peterson KS, Smith HJ, et al. A phase I study of indole-3-carbinolin women: tolerability and effects. Cancer Epidemiol Biomarkers Prev . Aug 2005;14(8):1953–60. doi:10.1158/1055-9965.EPI-05-0121 https://www.ncbi.nlm.nih.gov/pubmed/16103443
- Dalessandri KM, Firestone GL, Fitch MD, Bradlow HL, Bjeldanes LF. Pilotstudy: effect of 3,3'-diindolylmethane supplements on urinary hormonemetabolites in postmenopausal women with a history of early-stage breastcancer. Nutr Cancer. 2004;50(2):161–7.doi:10.1207/s15327914nc5002_5 https://www.ncbi.nlm.nih.gov/pubmed/15623462
- Thomson CA, Chow HHS, Wertheim BC, et al. A randomized, placebo-controlledtrial of diindolylmethane for breast cancer biomarker modulation in patientstaking tamoxifen. Breast Cancer Res Treat. Aug 2017;165(1):97–107.doi:10.1007/s10549-017-4292-7 https://www.ncbi.nlm.nih.gov/pubmed/28560655
- Calado A, Neves PM, Santos T, Ravasco P. The Effect of Flaxseed in BreastCancer: A Literature Review. Front Nutr. 2018;5:4.doi:10.3389/fnut.2018.00004 https://www.ncbi.nlm.nih.gov/pubmed/29468163
- Thompson LU, Chen JM, Li T, Strasser-Weippl K, Goss PE. Dietary flaxseedalters tumor biological markers in postmenopausal breast cancer. Clin Cancer Res . May 15 2005;11(10):3828–35. doi:10.1158/1078-0432.CCR-04-2326 https://www.ncbi.nlm.nih.gov/pubmed/15897583
- Fabian CJ, Kimler BF, Zalles CM, et al. Reduction in Ki-67 in benign breasttissue of high-risk women with the lignan secoisolariciresinol diglycoside.Cancer Prev Res (Phila). Oct 2010;3(10):1342–50.doi:10.1158/1940-6207.CAPR-10-0022 https://www.ncbi.nlm.nih.gov/pubmed/20724470
- van Die MD, Bone KM, Visvanathan K, et al. Phytonutrients and outcomesfollowing breast cancer: a systematic review and meta-analysis ofobservational studies. JNCI Cancer Spectr. Jan 42024;8(1)doi:10.1093/jncics/pkad104 https://www.ncbi.nlm.nih.gov/pubmed/38070485
- Chavda VP, Chaudhari AZ, Balar PC, Gholap A, Vora LK. Phytoestrogens:Chemistry, potential health benefits, and their medicinal importance. Phytother Res . Jun 2024;38(6):3060–3079. doi:10.1002/ptr.8196 https://www.ncbi.nlm.nih.gov/pubmed/38602108
- Messina M, Barnes S, Setchell KD. Perspective: Isoflavones-IntriguingMolecules but Much Remains to Be Learned about These Soybean Constituents.Adv Nutr. May 2025;16(5):100418. doi:10.1016/j.advnut.2025.100418 https://www.ncbi.nlm.nih.gov/pubmed/40157603
- Li J, Yu J, Zou H, Zhang J, Ren L. Estrogen receptor-mediated healthbenefits of phytochemicals: a review. Food Funct. Dec 112023;14(24):10681–10699. doi:10.1039/d3fo04702d https://www.ncbi.nlm.nih.gov/pubmed/38047630
- Rasheed S, Rehman K, Shahid M, Suhail S, Akash MSH. Therapeutic potentialsof genistein: New insights and perspectives. J Food Biochem. Sep2022;46(9):e14228. doi:10.1111/jfbc.14228 https://www.ncbi.nlm.nih.gov/pubmed/35579327
- Messina M, Nechuta S. A Review of the Clinical and Epidemiologic EvidenceRelevant to the Impact of Postdiagnosis Isoflavone Intake on Breast CancerOutcomes. Current nutrition reports. Mar 25 2025;14(1):50.doi:10.1007/s13668-025-00640-5 https://www.ncbi.nlm.nih.gov/pubmed/40131602
- Dong JY, Qin LQ. Soy isoflavones consumption and risk of breast cancerincidence or recurrence: a meta-analysis of prospective studies. Breast Cancer Res Treat . Jan 2011;125(2):315–23. doi:10.1007/s10549-010-1270-8 https://www.ncbi.nlm.nih.gov/pubmed/21113655
- Guha N, Kwan ML, Quesenberry CP, Jr., Weltzien EK, Castillo AL, Caan BJ. Soyisoflavones and risk of cancer recurrence in a cohort of breast cancersurvivors: the Life After Cancer Epidemiology study. Breast Cancer Res Treat . Nov 2009;118(2):395–405. doi:10.1007/s10549-009-0321-5 https://www.ncbi.nlm.nih.gov/pubmed/19221874
- Ho SC, Yeo W, Goggins W, et al. Pre-diagnosis and early post-diagnosisdietary soy isoflavone intake and survival outcomes: A prospective cohortstudy of early stage breast cancer survivors. Cancer Treat Res Commun . 2021;27:100350. doi:10.1016/j.ctarc.2021.100350 https://www.ncbi.nlm.nih.gov/pubmed/33770661
- Valente IVB, Garcia D, Abbott A, et al. The anti-proliferative effects of afrankincense extract in a window of opportunity phase ia clinical trial forpatients with breast cancer. Breast Cancer Res Treat. Apr2024;204(3):521–530. doi:10.1007/s10549-023-07215-4 https://www.ncbi.nlm.nih.gov/pubmed/38194131
- Togni S, Maramaldi G, Bonetta A, Giacomelli L, Di Pierro F. Clinicalevaluation of safety and efficacy of Boswellia-based cream for prevention ofadjuvant radiotherapy skin damage in mammary carcinoma: a randomized placebocontrolled trial. Eur Rev Med Pharmacol Sci. Apr2015;19(8):1338–44. https://www.ncbi.nlm.nih.gov/pubmed/25967706
- Bao X, Hu J, Zhao Y, Jia R, Zhang H, Xia L. Advances on the anti-tumormechanisms of the carotenoid Crocin. PeerJ. 2023;11:e15535.doi:10.7717/peerj.15535 https://www.ncbi.nlm.nih.gov/pubmed/37404473
- Bej E, Volpe AR, Cesare P, Cimini A, d'Angelo M, Castelli V. Therapeuticpotential of saffron in brain disorders: From bench to bedside. Phytother Res . May 2024;38(5):2482–2495. doi:10.1002/ptr.8169 https://www.ncbi.nlm.nih.gov/pubmed/38446350
- Salek R, Dehghani M, Mohajeri SA, Talaei A, Fanipakdel A, Javadinia SA.Amelioration of anxiety, depression, and chemotherapy related toxicity aftercrocin administration during chemotherapy of breast cancer: A double blind,randomized clinical trial. Phytother Res. Sep 2021;35(9):5143–5153.doi:10.1002/ptr.7180 https://www.ncbi.nlm.nih.gov/pubmed/34164855
- Skuli SJ, Alomari S, Gaitsch H, Bakayoko A, Skuli N, Tyler BM. Metformin andCancer, an Ambiguanidous Relationship. Pharmaceuticals (Basel). May19 2022;15(5):626. doi:10.3390/ph15050626 https://www.ncbi.nlm.nih.gov/pubmed/35631452
- Stromland PP, Bertelsen BE, Viste K, et al. Effects of metformin ontranscriptomic and metabolomic profiles in breast cancer survivors enrolledin the randomized placebo-controlled MetBreCS trial. Sci Rep. May15 2025;15(1):16897. doi:10.1038/s41598-025-01705-9 https://www.ncbi.nlm.nih.gov/pubmed/40374694
- Samuel SM, Varghese E, Koklesova L, Liskova A, Kubatka P, Busselberg D.Counteracting Chemoresistance with Metformin in Breast Cancers: TargetingCancer Stem Cells. Cancers (Basel). Sep 12020;12(9)doi:10.3390/cancers12092482 https://www.ncbi.nlm.nih.gov/pubmed/32883003
- Cejuela M, Martin-Castillo B, Menendez JA, Pernas S. RETRACTED: Metforminand Breast Cancer: Where Are We Now? Int J Mol Sci. Feb 282022;23(5):2705. doi:10.3390/ijms23052705 https://www.ncbi.nlm.nih.gov/pubmed/35269852
- Benjamin DJ, Haslam A, Prasad V. Cardiovascular/anti-inflammatory drugsrepurposed for treating or preventing cancer: A systematic review andmeta-analysis of randomized trials. Cancer Med. Mar2024;13(5):e7049. doi:10.1002/cam4.7049 https://www.ncbi.nlm.nih.gov/pubmed/38491813
- Park YM, Bookwalter DB, O'Brien KM, Jackson CL, Weinberg CR, Sandler DP. Aprospective study of type 2 diabetes, metformin use, and risk of breastcancer. Ann Oncol. Mar 2021;32(3):351–359.doi:10.1016/j.annonc.2020.12.008 https://www.ncbi.nlm.nih.gov/pubmed/33516778
- Barakat HE, Hussein RRS, Elberry AA, Zaki MA, Elsherbiny Ramadan M. Factorsinfluencing the anticancer effects of metformin on breast cancer outcomes: asystematic review and meta-analysis. Expert review of anticancer therapy . Apr 2022;22(4):415–436. doi:10.1080/14737140.2022.2051482 https://www.ncbi.nlm.nih.gov/pubmed/35259320
- Sonnenblick A, Agbor-Tarh D, Bradbury I, et al. Impact of Diabetes, Insulin,and Metformin Use on the Outcome of Patients With Human Epidermal GrowthFactor Receptor 2-Positive Primary Breast Cancer: Analysis From the ALTTOPhase III Randomized Trial. J Clin Oncol. May 12017;35(13):1421–1429. doi:10.1200/JCO.2016.69.7722 https://www.ncbi.nlm.nih.gov/pubmed/28375706
- Goodwin PJ, Chen BE, Gelmon KA, et al. Effect of Metformin vs Placebo onInvasive Disease-Free Survival in Patients With Breast Cancer: The MA.32Randomized Clinical Trial. JAMA. May 24 2022;327(20):1963–1973.doi:10.1001/jama.2022.6147 https://www.ncbi.nlm.nih.gov/pubmed/35608580
- Wang Q, Ma X, Long J, Du X, Pan B, Mao H. Metformin and survival of womenwith breast cancer: A meta-analysis of randomized controlled trials. J Clin Pharm Ther . Mar 2022;47(3):263–269. doi:10.1111/jcpt.13500 https://www.ncbi.nlm.nih.gov/pubmed/34397110
- Morio K, Kurata Y, Kawaguchi-Sakita N, Shiroshita A, Kataoka Y. Efficacy ofMetformin in Patients With Breast Cancer Receiving Chemotherapy or EndocrineTherapy: Systematic Review and Meta-analysis. Ann Pharmacother. Mar2022;56(3):245–255. doi:10.1177/10600280211025792 https://www.ncbi.nlm.nih.gov/pubmed/34137294
- Jeong GH, Lee KH, Kim JY, et al. Statin and Cancer Mortality and Survival:An Umbrella Systematic Review and Meta-Analysis. J Clin Med. Jan 232020;9(2):326. doi:10.3390/jcm9020326 https://www.ncbi.nlm.nih.gov/pubmed/31979352
- Lv H, Shi D, Fei M, et al. Association Between Statin Use and Prognosis ofBreast Cancer: A Meta-Analysis of Cohort Studies. Frontiers in oncology . 2020;10:556243. doi:10.3389/fonc.2020.556243 https://www.ncbi.nlm.nih.gov/pubmed/33178584
- Zhao G, Ji Y, Ye Q, et al. Effect of statins use on risk and prognosis ofbreast cancer: a meta-analysis. Anti-cancer drugs. Jan 12022;33(1):e507–e518. doi:10.1097/CAD.0000000000001151 https://www.ncbi.nlm.nih.gov/pubmed/34407042
- Xu WH, Zhou YH. The relationship between post-diagnostic statin usage andbreast cancer prognosis varies by hormone receptor phenotype: a systemicreview and meta-analysis. Archives of gynecology and obstetrics.Nov 2021;304(5):1315–1321. doi:10.1007/s00404-021-06065-z https://www.ncbi.nlm.nih.gov/pubmed/33891208
- McKechnie T, Brown Z, Lovrics O, et al. Concurrent Use of Statins inPatients Undergoing Curative Intent Treatment for Triple Negative BreastCancer: A Systematic Review and Meta-Analysis. Clinical breast cancer . Apr 2024;24(3):e103–e115. doi:10.1016/j.clbc.2023.12.001 https://www.ncbi.nlm.nih.gov/pubmed/38296737
- Murto MO, Simolin N, Arponen O, et al. Statin Use, Cholesterol Level, andMortality Among Females With Breast Cancer. JAMA Netw Open. Nov 12023;6(11):e2343861. doi:10.1001/jamanetworkopen.2023.43861 https://www.ncbi.nlm.nih.gov/pubmed/37976058
- Nabati M, Janbabai G, Esmailian J, Yazdani J. Effect of Rosuvastatin inPreventing Chemotherapy-Induced Cardiotoxicity in Women With Breast Cancer:A Randomized, Single-Blind, Placebo-Controlled Trial. J Cardiovasc Pharmacol Ther . May 2019;24(3):233–241. doi:10.1177/1074248418821721 https://www.ncbi.nlm.nih.gov/pubmed/30599756
- Abdel-Qadir H, Bobrowski D, Zhou L, et al. Statin Exposure and Risk of HeartFailure After Anthracycline- or Trastuzumab-Based Chemotherapy for EarlyBreast Cancer: A Propensity Score‒Matched Cohort Study. J Am Heart Assoc . Jan 19 2021;10(2):e018393. doi:10.1161/JAHA.119.018393 https://www.ncbi.nlm.nih.gov/pubmed/33401953
- Calvillo-Arguelles O, Abdel-Qadir H, Michalowska M, et al. CardioprotectiveEffect of Statins in Patients With HER2-Positive Breast Cancer ReceivingTrastuzumab Therapy. The Canadian journal of cardiology. Feb2019;35(2):153–159. doi:10.1016/j.cjca.2018.11.028 https://www.ncbi.nlm.nih.gov/pubmed/30760421
- Ghasemi A, Ghashghai Z, Akbari J, Yazdani-Charati J, Salehifar E,Hosseinimehr SJ. Topical atorvastatin 1% for prevention of skin toxicity inpatients receiving radiation therapy for breast cancer: a randomized,double-blind, placebo-controlled trial. Eur J Clin Pharmacol. Feb2019;75(2):171–178. doi:10.1007/s00228-018-2570-x https://www.ncbi.nlm.nih.gov/pubmed/30291370
- Celi KY, Celi KS, Sarikose S, Arslan HN. Evaluation of financial toxicityand associated factors in female patients with breast cancer: a systematicreview and meta-analysis. Support Care Cancer. Nov 132023;31(12):691. doi:10.1007/s00520-023-08172-w https://www.ncbi.nlm.nih.gov/pubmed/37953376
- Oshima SM, Tait SD, Rushing C, et al. Patient Perspectives on the FinancialCosts and Burdens of Breast Cancer Surgery. JCO Oncol Pract. Jun2021;17(6):e872–e881. doi:10.1200/OP.20.00780 https://www.ncbi.nlm.nih.gov/pubmed/33566677
- American Cancer Society. Choosing a Cancer Center or Hospital. Updated 9/17/2021. Accessed 9/12/2024, https://www.cancer.org/cancer/managing-cancer/finding-care/where-to-find-cancer-care/choosing-a-cancer-center-or-hospital.html
- NCI. National Cancer Institute. NCI-designated cancer centers. Available at https://www.cancer.gov/research/infrastructure/cancer-centers. Last updated 12/12/2024. Accessed 02/01/2025