Next Article in Journal
Anticancer Drug Discovery Based on Natural Products: From Computational Approaches to Clinical Studies
Previous Article in Journal
Surgical, Histopathological, and Quality of Life Outcomes Following Neoadjuvant Chemotherapy and Pancreatectomy for Borderline Resectable and Locally Advanced Pancreatic Cancer
Previous Article in Special Issue
The Effects of Concurrent Training on Molecular, Functional, and Clinical Outcomes in Breast Cancer Survivors: A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Long-Term Adverse Events Following Early Breast Cancer Treatment with a Focus on the BRCA-Mutated Population

1
Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
2
Centre Antoine Lacassagne, 06100 Nice, France
3
Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain
4
Hospital Universitario La Paz, 28046 Madrid, Spain
5
Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
6
The Stefanie Spielman Comprehensive Breast Center, Columbus, OH 43212, USA
7
The Breast Unit, UK Royal Free London NHS Trust, London NW3 2QG, UK
8
Savana Research S.L., 28004 Madrid, Spain
9
Oncology Outcomes Research, AstraZeneca, Cambridge CB2 0AA, UK
10
Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD 20878, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(15), 2506; https://doi.org/10.3390/cancers17152506
Submission received: 10 June 2025 / Revised: 15 July 2025 / Accepted: 21 July 2025 / Published: 30 July 2025

Simple Summary

Assessing long-term adverse events (harmful or unexpected occurrences) in patients with early breast cancer is complex because of differences in characteristics across patient populations. This can be further complicated by variation in how ‘long-term’ adverse events are defined. Patients with inherited (germline) mutations in the BRCA1 or BRCA2 genes (gBRCAm) exhibit distinct types of adverse events, and specific assessment and management strategies may be required for these patients. New treatments present additional challenges, which may include new types of long-term adverse events. Doctors and carers should be aware of these potential events so that they can be effectively managed.

Abstract

Breast cancer (BC) is the most prevalent malignancy in women worldwide. Despite most cases being diagnosed in the early stages, patients typically require a multimodal treatment approach. This typically involves a combination of surgery, radiotherapy, systemic treatments (including chemotherapy or immunotherapy), targeted therapy, and endocrine therapy, depending on the disease subtype and the risk of recurrence. Moreover, patients with BC and germline mutations in the breast cancer genes 1 or 2 (BRCA1/BRCA2), (gBRCAm), who are typically young women, often require more aggressive therapeutic interventions. These mutations present unique characteristics that necessitate a distinct treatment approach, potentially influencing the side effect profiles of patients with BC. Regardless of the clear benefit observed with these treatments in terms of reduced recurrence and mortality rates, long-term, treatment-related adverse events occur that negatively affect the health-related quality of life (HRQoL) of BC survivors. Thus, long-term adverse events need to be factored into the treatment decision algorithm of patients with early BC (eBC). Physical, functional, emotional, and psychosocial adverse events can occur and represent a significant concern and a challenge for clinicians, patients, and their families. This review article provides an overview of the various long-term adverse events that patients with eBC may experience, including their associated risk factors, as well as management and prevention strategies. We also explore the evidence of the long-term impact of treatment on the HRQoL of patients with gBRCAm. By providing a comprehensive overview of current evidence and recommendations regarding patients’ HRQoL, we aim to equip clinicians with scientific and clinical knowledge and provide guidance to optimize care and improve long-term outcomes.

1. Introduction

Breast cancer (BC) is the most commonly diagnosed malignancy and cause of death in women worldwide, accounting for 2.3 million new cases (11.6% of all cancer cases) and 832,000 deaths in 2022. Biological factors, such as age, family history, and reproductive history, along with environmental factors like physical activity, alcohol consumption, weight, and hormonal supplements, have been recognized for their potential role in BC development [1]. Approximately 5–10% of BC cases have a family history, with 30% of these linked to mutations in genes predisposed to BC [2]. Approximately 10% of BC cases harbor a germline mutation in the breast cancer genes 1 (BRCA1) or 2 (BRCA2), (gBRCAm), which affects the homologous repair mechanism of DNA double-strand breaks. BRCA mutation carriers are diagnosed with BC at an early age (<40 years), and recent studies indicate that 55–65% of BRCA1 and 45% of BRCA2 mutation carriers will develop BC by age 70 [3,4]. However, most BC cases are sporadic, resulting from somatic mutations [5].
Standard treatments for early-stage BC (eBC), where the disease is confined to the breast or the axillary lymph nodes, may include surgery, chemotherapy, anti-HER2 therapy, and endocrine therapy (ET) [6,7]. Despite treatment, up to 20% of patients may experience recurrence within the first 10 years [8,9], particularly those with high-risk hormone receptor-positive (HR+) BC or with triple-negative BC (TNBC) [10,11,12,13].
Over 90% of BC cases are diagnosed early, and new therapies have significantly improved patients’ outcomes. Immunotherapy is effective for TNBC, while cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors have demonstrated benefits in HR+/HER2 BC. Furthermore, pembrolizumab, a programmed cell death protein 1 checkpoint inhibitor, has shown significant benefits in overall survival [14]. Doxorubicin plus cyclophosphamide enhances survival and response rates in patients with TNBC [15], while ribociclib and abemaciclib have improved disease-free survival rates [16,17]. Lastly, the poly(ADP)-ribose polymerase (PARP) inhibitor olaparib has improved survival in patients with gBRCAm/HER2-negative BC [18,19,20,21].
Updated guidelines for the diagnosis, treatment, and follow-up of eBC highlight that advancements in screening and therapeutic strategies have enhanced prognosis [22]. However, with 10-year disease-free survival rates ranging from 80 to 95% depending on subtype and stage, preserving patients’ health-related quality of life (HRQoL) post-treatment remains crucial [23,24]. While (neo)adjuvant therapies have been effective in reducing recurrence, they may cause long-term adverse events impacting physical, emotional, and psychosocial well-being [25,26].
Long-term adverse events are not well characterized, as there are no standardized definitions or timelines due to factors such as variability in regimens, patient physiology, and inconsistencies across registries. Consequently, there is no global consensus on the time frame defining an adverse event as early or long-term. In the literature, the time window for describing an adverse event as long-term varies widely, ranging from a few months to several years post-therapy. In addition, the definition of ‘long-term adverse event’ depends on factors such as treatment regimen, patient age and general health, and the specific outcome assessed.
Adverse events occurring from months to years post-therapy significantly impact HRQoL. Common complications include chronic pain (up to 72% of patients) [27], lymphoedema (27–40%) [28], peripheral neuropathy (23–80%), and/or gastrointestinal symptoms (42.8%) arising from chemotherapy treatment [29,30]. Additionally, conservative surgery combined with radiotherapy, while critical in adjuvant care, can lead to late cardiac effects and cosmetic challenges, despite advances aimed at reducing side effects [31].
Patients with eBC may also experience psychological alterations [32], fatigue [33], hormonal and endocrine disturbances [34,35], and sexual dysfunction and infertility [36]. These long-term adverse events also affect patients’ families, with financial strain and emotional stress often leading to depression and impacting recovery [37].
Long-term adverse events pose significant challenges for clinicians, patients, and families. This review examines these events, their risk factors, and management strategies, with a focus on patients with gBRCAm BC, aiming to equip clinicians and patients with the scientific knowledge and tools to optimize care and improve outcomes. Although long-term adverse events can be classified in various ways (e.g., by life impact, treatment modality, severity, or patient-reported outcomes), we have chosen to organize them according to symptoms grouped by affected body systems. This classification reflects how clinical care is typically structured, enabling more effective referral to relevant specialists, enhancing interdisciplinary collaboration, and supporting the development of targeted follow-up strategies. Our approach offers a practical lens for addressing the complex and multifaceted needs of survivors of BC. Despite its clinical relevance, this perspective is underrepresented in the literature, and this review aims to contribute to filling that gap.

2. Methodology

A systematic literature search was conducted in academic databases including PubMed, Embase, and the Cochrane Library, using the keywords ‘breast cancer’, ‘early breast cancer’, ‘long-term toxicity’, ‘adjuvant treatment’, ‘survivorship’, ‘quality of life’, and ‘BRCA mutation’. Articles published between January 1990 and May 2024, in English or Spanish, were included if they assessed, either retrospectively or prospectively, long-term adverse events in patients with eBC.
The search identified 335 articles, of which 243 were screened, and 130 were fully read (Figure 1). The findings of the review were synthesized and summarized in a narrative format.

3. Long-Term Adverse Events in Patients with eBC

Assessing long-term adverse events in eBC is challenging due to patient heterogeneity, varying treatments, comorbidities, and external factors such as lifestyle, genetic predisposition, or healthcare resources, which influence outcomes. The lack of standardized definitions and reporting methods further complicates comparisons across studies [38,39].
Establishing a consensus for a definition of ‘long-term’ adverse events is crucial, as it varies by symptom type (Table 1). For instance, ‘long-term’ hormonal therapy (HT) effects are monitored starting 1 month post-treatment [40], chronic pain after 3 months [41], and fatigue, memory, or psychological issues between 5 and 10 years [42,43]. Cardiac events may appear within a year or up to 8 years post-treatment [44,45,46].
To address these discrepancies, we reviewed studies on ‘long-term’ adverse events by symptom. The following sections outline key adverse events, their associated risk factors, and clinical management recommendations (see Table 2 for an overview).

3.1. Adverse Events Affecting the Chest Wall and Breast

Breast surgery and radiotherapy have the potential to cause various long-term adverse events in the chest wall and breast. The most commonly known are chronic pain (known as post-mastectomy pain syndrome or PMPS), lymphoedema, or skin and soft tissue fibrosis/necrosis related to radiotherapy [38].

3.1.1. Post-Mastectomy Pain Syndrome

PMPS is a chronic pain condition caused by nerve fiber damage, resulting in a type of neuropathic pain. This ache is commonly felt in the anterior/lateral chest wall, axilla, and/or medial upper arm for a period exceeding 3 months post-surgery. Symptoms include neuropathic pain sensations such as burning, tingling, shooting, stinging, or stabbing pain, as well as hyperesthesia [62]. The reported incidence of nerve fiber damage and chronic pain following BC surgery ranges from 20% to 72% depending on the definition and duration of symptoms. Risk factors for persistent pain include postoperative pain, younger age, high body mass index (BMI), axillary radiation, and axillary lymph node dissection [27]. It is important to note that in recent years, surgical de-escalation strategies, such as an increased use of breast-conserving surgery and conservative mastectomies, have been widely adopted, which may influence the prevalence and severity of these long-term adverse events [75,76,77]. PMPS can have a significant negative impact on patients’ HRQoL, particularly their mental health, as indicated by retrospective studies [47,78]. Treatment options for PMPS include analgesics for neuropathic pain, surgical interventions such as neuroma excision or scar release, and complementary therapies like acupuncture or hypnosis [61].

3.1.2. Lymphoedema

Lymphoedema is a chronic and often progressive condition characterized by the accumulation of lymphatic fluid in the interstitial tissues, and most commonly affects the arm, breast, chest wall, and shoulder on the treated side. It remains one of the most significant and burdensome long-term adverse effects of treatment for BC survivors, with a reported prevalence ranging from 27% to 40% depending on treatment type, time since surgery, and diagnostic criteria.
Current clinical practice emphasizes early risk stratification, patient education, and prompt initiation of conservative, non-surgical management. Axillary lymph node dissection (ALND) is the most consistently identified risk factor, followed by regional radiotherapy, higher BMI, postoperative infections, and trauma to the affected limb. Sentinel lymph node biopsy has largely replaced ALND in many early-stage cases, significantly reducing lymphoedema risk, though it does not eliminate it entirely.
Early detection of lymphoedema is essential and supported by routine surveillance methods and patient education on early symptoms. First-line treatment involves complex decongestive therapy, which includes manual lymphatic drainage, compression, exercise, and skin care. Supervised resistance training is now recognized as safe and effective [63,79].
Patient education is vital for prevention and long-term management, emphasizing self-care practices and the use of compression. Contemporary lymphoedema care is proactive, multidisciplinary, and patient-centered, aiming to reduce complications and improve HRQoL for BC survivors [48].

3.1.3. Skin and Soft Tissue Affection

Radiotherapy can induce skin and soft tissue fibrosis and rarely necrosis, which could lead to poor cosmesis. In this regard, the most frequently observed complication following radiotherapy (up to 43% of patients) is radiation-induced fibrosis of the skin and subcutaneous tissue [50,80]. This type of fibrosis is typically observed in areas with overlapping treatment fields after breast-conserving surgery with postoperative radiotherapy or after mastectomy and radiotherapy, particularly in patients with breast implants [31,81].

3.2. Cardiotoxicity

Treatment for eBC, including radiotherapy, chemotherapeutic and biologic agents, and ET, may result in late-onset cardiotoxicity, increasing cardiovascular morbidity and mortality risk [82,83]. In addition, many chemotherapeutic agents used in the neoadjuvant setting are known for their potential to induce cardiac dysfunction [84]. Cancer survivors are 2–7 times more likely to die from cardiovascular diseases than the general population [85], with cardiac effects emerging as early as 6 months post-surgery or up to 8 years after diagnosis [44,45,46]. Long-term cardiac monitoring is essential, as subclinical damage can progress to heart failure years after treatment.
Anthracyclines, common chemotherapeutic agents, can cause cardiotoxicity, including acute or chronic cardiomyopathy, heart failure, arrhythmias, and even death. The risk increases with higher cumulative doses, affecting 1–11% of patients treated with epirubicin at 900 mg/m2 and 7–25% of patients treated with doxorubicin at 500–550 mg/m2, and requires close monitoring [86,87].
Patients with HER2+ eBC face additional cardiotoxicity risks from anti-HER2 drugs, including heart failure (2.5–4%) and decline in left ventricular ejection fraction (LVEF) (3–27%), meaning that cardiac monitoring is required before, during (every 3 months), and after (every 6 months for 2 years) treatment [88,89]. In patients with triple-negative eBC, immune checkpoint inhibitors (ICIs) rarely cause cardiotoxicity, but the appearance of severe cases of ICI-associated myocarditis and pericarditis highlights the need for thorough cardiac evaluation and continuous surveillance post-treatment [90].
The cardiovascular effects of tamoxifen and aromatase inhibitors (AIs) are still under investigation [91,92,93,94]. Tamoxifen, while potentially increasing the risk of stroke, has also demonstrated cardiovascular benefits, such as a lipid-lowering effect [92]. Current evidence suggests that tamoxifen may provide modest cardiovascular protection, particularly in postmenopausal women, regardless of pre-existing coronary heart disease [95]. In contrast, AIs have been associated with a higher risk of heart failure and cardiovascular mortality [51,96]. For example, a UK study of 17,922 patients reported that, compared to tamoxifen, AIs were linked to a 1.86-fold higher risk of heart failure and a 1.50-fold increase in cardiovascular mortality [97].
Anthracycline and taxane-based chemotherapy (ATAX) remains the standard of care for early-stage TNBC, with a lower heart failure risk compared to taxane-only regimens [98]. Radiotherapy, particularly for the left breast, can cause late cardiac toxicities, including coronary artery disease, acute myocardial infarction, cardiomyopathy, or valvular heart disease. However, modern radiotherapy techniques offer better cardiac protection [65,99].
Regular cardiac screening is essential for BC survivors who are at high risk of cardiotoxicity, as shown by the Cardiac-Related Oncologic Late Effects (CAROLE) study, where 77.6% of participants exhibited cardiovascular disease [66]. The study highlights the importance of adopting a multidisciplinary approach involving medical oncologists, radiation oncologists, cardiologists, and other healthcare providers to prevent and manage the late cardiotoxic effects associated with BC treatment. Strategies to reduce anthracycline cardiotoxicity include cardioprotective agents, modified protocols, and alternative therapies [64]. Personalizing treatment helps balance benefits and minimize risks.

3.3. Neurotoxicity

3.3.1. Chemotherapy-Induced Peripheral Neuropathy (CIPN)

Adjuvant chemotherapy, especially with taxanes, can cause chemotherapy-induced peripheral neuropathy (CIPN), a common and potentially severe adverse effect of neoadjuvant chemotherapy (NACT). It is characterized by distal paresthesia (tingling and numbness), pain, and muscle weakness. CIPN prevalence ranges from 23 to 80% of patients [29,52,53], with a higher risk linked to cumulative doses (e.g., paclitaxel > 1000 mg/m2, docetaxel > 400 mg/m2), age, baseline neuropathy, smoking, and diabetes [67].
Persistent CIPN can lead to psychological distress, including depression and anxiety, and significantly affect HRQoL [100]. Regular assessment is crucial for the early detection of any issues. While there is no effective drug to prevent CIPN, cryotherapy, compression therapy, and medical exercise are options to consider. For chronic CIPN, duloxetine is the only treatment supported by level I evidence; other options include venlafaxine, pregabalin, amitriptyline, tramadol, or opioids. Acupuncture may also be considered in selected cases [101]. Currently, there are limited effective treatments for CIPN; once established, prevention and early detection are crucial for effective management. Emerging strategies, such as neuroprotective agents and prehabilitation programs, aim to reduce the incidence and impact of CIPN in NACT BC patients by optimizing their functional status before and during treatment [102].

3.3.2. Cognitive Dysfunction

Cognitive dysfunction, or ‘chemobrain’, affects patients with BC who have undergone chemotherapy or ET, leading to issues with memory, attention, and executive function [54,68]. It impacts up to one in three patients and can persist for 2–3 years post-treatment [103].
The exact cause of chemobrain is unclear, but it may involve neurotoxic effects [104], inflammation, oxidative stress, and hormonal changes, with age and ET being risk factors [105,106]. Symptoms include trouble in multitasking, organizing, concentrating, forgetfulness, and reduced mental speed, sometimes impairing daily life and work [54]. Emotional distress, anxiety, and a reduction in overall HRQoL often accompany this condition. Depression, due to cognitive impairment, can also lead to decreased productivity and financial difficulties.
Interventions like cognitive rehabilitation, exercise, and low-evidence pharmacological treatments are being explored, but more research is needed to develop effective solutions [54].

3.4. Psychological Alterations

A diagnosis of BC is highly distressing for patients and their families, contributing to anxiety and depression due to the challenges of treatment, potential side effects, understanding the prognosis, and an uncertain future. This emotional distress can lead to a reduction in HRQoL and in treatment compliance while increasing mortality risk. Depression prevalence ranges from 9.4 to 66.1% among BC survivors, while anxiety affects 17.9–33.3% of patients [32].
Risk factors for anxiety include unemployment, younger age, physical symptoms, chemotherapy, poor social and cognitive functioning, and poor healthcare communication [55]. Depression is more strongly associated with a younger age at diagnosis, history of psychological disorder, substance abuse, poor social support, and lower socioeconomic status [69,107,108]. Fear of recurrence, affecting up to 71% of patients with BC, is particularly common in younger individuals [56] and in those undergoing breast-conserving surgery, compared to mastectomy [71].
To mitigate these effects, psychological support, psychiatric care, and cognitive–behavioral therapy are crucial for improving mental health outcomes during and after BC treatment [70,71].

3.5. Fatigue

Fatigue is a common symptom in patients with eBC, which can persist even after the completion of cancer treatment. The prevalence of fatigue as a long-term complication in these patients varies, but it is estimated to affect up to 30–50% of patients [33,43,56,109]. The cause of fatigue in BC survivors is multifactorial and can be related to either treatment or to other long-term adverse events such as cardiac, menopause, or psychological causes. Fatigue can have a significant negative impact on the HRQoL of BC survivors, affecting their ability to perform daily activities, work, and socialize. According to a meta-analysis of 12,327 patients, severe fatigue is more likely to occur in those at advanced stages of the disease, receiving chemotherapy, or undergoing a combination of surgery, RT, and chemotherapy with or without ET. Conversely, having family support or not receiving systemic treatment could reduce the risk of severe fatigue [72].
Management of fatigue can involve a combination of lifestyle modifications, such as regular exercise, adequate sleep, stress reduction techniques, and treatment of other comorbidities or late adverse events [110].

3.6. Hormonal Alterations

Patients on ET often experience persistent side effects, including genital atrophy, dizziness, weight gain, hot flashes, cognitive dysfunction, fatigue, and musculoskeletal impairment. These adverse effects can persist over a long period and have a negative impact on a patient’s HRQoL, leading to poor adherence to therapy [111]. For instance, hot flashes within 6 months of starting AIs increase the 5-year discontinuation rate by 14.2% [34]. Weight gain, affecting 50–96% of women with BC, is more pronounced in premenopausal women, those undergoing chemotherapy, and in those overweight at the time of diagnosis, potentially increasing mortality risk [112,113,114,115].
Studies have shown that ET has a significant negative impact on the HRQoL of patients with BC over time; symptoms such as joint pain, lack of energy, weight gain, and vaginal dryness persist in 33–48.7% of patients 5–10 years after diagnosis [40,57].
Nonhormonal pharmacotherapy, such as gabapentin or selective serotonin reuptake inhibitors/serotonin–norepinephrine reuptake inhibitors, may help women experiencing hot flashes. Though evidence is limited, homeopathy or herbal products are commonly used to mitigate hot flashes due to BC treatment [116]. Physical exercise, cognitive–behavioral therapy, and mindfulness have demonstrated efficacy in reducing menopausal symptoms in clinical trials [117,118,119].

3.7. Sexual Disorders and Fertility

Patients with BC have a high prevalence of sexual disorders (affecting up to 90% of patients) [58,120]. The main causes are associated with body image alterations, ET, and psychological impairment, such as depression or anxiety [36]. Treatment focuses on identifying and addressing the physical and/or psychological causes, rather than merely coping with the primary affliction. For example, in cases of vaginal dryness or dyspareunia, advising on the use of lubricants, or providing psychological support and sexual counseling in cases with depression or anxiety [73].
Young BC survivors may experience infertility due to chemotherapy-related gonadotoxicity and the delay in childbearing that is required when women are taking ET. This is an important concern for women, with one study showing that 36% of patients with eBC reported an interest in having children in the future [74]. In a meta-analysis of 112,840 patients with BC, only 6.5% became pregnant after diagnosis, and survivors had a 60% reduction in the likelihood of having a subsequent pregnancy compared with the general population [59]. Encouraging results were provided from the POSITIVE trial, involving 516 women aged 42 or younger and designed to evaluate the safety of temporarily pausing ET for up to 2 years, for women with HR+ eBC who wished to conceive. The study concluded that temporarily stopping ET for pregnancy did not significantly increase the short-term risk of BC recurrence. However, a longer follow-up is needed to confirm the long-term safety of this approach [121]. Oncofertility counseling should be offered to all women of reproductive age. Oocyte/embryo or ovarian tissue cryopreservation is the primary approach available for preserving fertility in patients with BC [122].

3.8. Gastrointestinal Symptoms

Gastrointestinal symptoms are not commonly reported as a long-term adverse effect, but rather as an early-onset event related to chemotherapy. This may change with the recent approval of new drugs to treat eBC, such as abemaciclib, which can cause diarrhea, or immunotherapies, like pembrolizumab. In the KEYNOTE-522 trial, 29.4% of patients treated with pembrolizumab reported some degree of diarrhea, and 62.7% reported nausea [123]. Published follow-up data are limited, and a longer follow-up period is needed to determine whether these symptoms persist in the long term.
In the MonarchE trial, in which patients received 2 years of adjuvant treatment with abemaciclib plus ET [30], 83% of patients experienced diarrhea that improved over time. Nonetheless, between 18 and 24 months during treatment, around 50% of patients still reported some degree of diarrhea. Nausea and vomiting were also reported (in 29.5% and 17.6% of patients, respectively). Further studies are needed to determine whether these symptoms persist post-treatment completion (2 years). In addition, the incorporation of immunotherapy in the early stages of BC treatment has made gastrointestinal symptoms more relevant. Nausea (28–77%), vomiting (40%), constipation (28–33%), and diarrhea (25–34%) are also frequently reported in patients with BC receiving olaparib treatment [124]. Most recently, in the NATALEE trial reporting the benefits of ribociclib in patients with eBC, 23% patients experienced some grade of nausea [17]. Overall, more follow-up is needed to determine whether these symptoms persist in the long term.

3.9. Endocrine Toxicity

ICIs, HT, and radiotherapy lead to dysregulation of immune homeostasis and endocrine toxicity, mainly as thyroid dysfunction, and diabetes mellitus [125].
Radiotherapy is associated with an increased risk of hypothyroidism [126]. In eBC, hypothyroidism was registered in 5.7% of patients at a median time of 3.45 years from the index date [35], with no association with the systemic oncological treatment. Digkas et al. identified a 68% higher risk for hypothyroidism in patients treated with radiotherapy of the regional lymph nodes, with no association observed between hypothyroidism and chemotherapy, ET, or radiotherapy to the breast/chest wall, irrespective of the use of adjuvant chemotherapy treatment. As for endocrine toxicity due to HT, several studies have suggested an association between eBC treatment and diabetes mellitus. Ye et al. recently reported a 30% higher risk of diabetes for patients with primary BC after HT (hazard ratio 1.30), compared to non-HT treated patients, and identified a 15% incidence rate of diabetes in HT treated patients with BC [127].
Endocrine dysfunction appears as one of the most common adverse events after ICI across multiple solid tumors [128] and an evaluation of immune-related adverse events after pembrolizumab treatment reported hypothyroidism and hyperthyroidism in 20% and 2.9% of patients, respectively [129], revealing a relatively high incidence of thyroid dysfunction during eBC treatment.

3.10. Osteomuscular Alterations

Patients with eBC treated with adjuvant therapy face an increased risk of bone loss and osteoporosis. HT significantly disrupts skeletal metabolism, making bone integrity a major concern [130]. AIs reduce estrogenic levels, critical for maintaining bone strength, leading to bone density loss and fracture risk, particularly in postmenopausal women [131,132,133]. Tamoxifen protects bones in postmenopausal women but may cause bone loss in premenopausal women once discontinued [134].
Muscle alterations, including sarcopenia, are another concern. Chemotherapy disrupts muscle protein balance, reducing muscle mass and strength, affecting their overall HRQoL and increasing the risk of other toxicities [135]. Extended HT use further increases the risk of fractures, osteoporosis, bone pain, myalgias, and treatment discontinuation. Physical inactivity, often driven by treatment fatigue, worsens muscle health [136].
Addressing these issues requires interventions such as weight-bearing exercises, calcium and vitamin D supplementation, and pharmacological treatments (e.g., bisphosphonates, denosumab), along with regular monitoring, to support long-term recovery.

4. Patients with BRCA-Mutated Tumors

Approximately 10% of all BC cases are due to germline mutations in the BRCA1 and BRCA2 genes (gBRCAm), and these patients are at an increased risk of developing a second BC. The toxicity profile of anticancer drugs in these patients may differ from non-gBRCAm patients, as they are on average younger at diagnosis and usually undergo more aggressive treatment approaches, including bilateral mastectomy as the first local treatment recommendation [137] and the addition of bilateral oophorectomy (inducing early menopause) to decrease the risk of a second BC (or primary ovarian cancer). Importantly, recent large-scale international data have confirmed a survival benefit associated with these risk-reducing surgeries in young BRCA patients [138]
Despite these features, treatment consequences in this subgroup of patients are not well studied, as most studies focus on early adverse events, and data for late-onset effects are scarce [139]. While our understanding of long-term adverse events in gBRCAm patients is still evolving, it is crucial to define and characterize the potential impact of early-onset adverse events on patients’ long-term health outcomes.
It is not well established if patients with gBRCAm BC are more likely to develop adverse events post-treatment than those with non-gBRCAm BC. Some studies have shown that patients harboring a gBRCAm BC tumor could present with hematological adverse effects earlier. In a retrospective analysis that included 270 patients, those harboring a gBRCAm appeared to have a higher risk of developing neutropenia after initial anthracycline-based chemotherapy [140], while initial trials with PARP inhibitors suggest higher rates of anemia, lower white-cells counts, fatigue, and lymphopenia post-treatment [18]. Interestingly, recent evidence indicates that there might be a slightly higher risk of contralateral BC in irradiated gBRCA1/2m patients [141]. This study included 3602 patients from the International BRCA1/2 Carrier Cohort Study (IBCSS) and explored the relationship between radiation therapy for primary BC and the risk of developing contralateral BC in gBRCA1/2m patients. Conversely, non-gBRCAm patients showed a significant increase in nausea and vomiting.
In another retrospective analysis, the rate of severe hematological toxicities in patients treated with taxanes was higher in those with gBRCAm compared with the non-gBRCAm control group (59.5% vs. 43.1%; p < 0.001) [142]. In contrast, a larger retrospective analysis showed that anemia and leukopenia were more frequently seen in patients with non-gBRCAm who were receiving taxane-containing chemotherapy compared with patients with gBRCAm [143,144]. The effect of radiotherapy toxicity has been examined in several studies, including patients with gBRCAm, and it was found that rates of radiation-induced adverse events were similar to those in women with sporadic BC. In their matched case–control study, Shanley et al. found no significant differences in acute and late radiation effects between patients with gBRCAm (n = 55) and those with sporadic BC (n = 55) [145]. Similarly, Pierce et al. compared the rates of chronic skin, subcutaneous tissue, lung, and bone adverse events in patients with stage I or II BC treated with breast-conserving surgery and radiotherapy. They found no significant differences between the genetic (n = 71) and sporadic cohorts (n = 213) [146]. Moreover, Park et al. observed no increased risk of acute skin toxicity in non-Caucasian patients with gBRCAm (n = 46) who underwent breast-conserving therapy using radiotherapy compared to women with sporadic BC [147]. More recently, Vliek et al. identified fatigue, nausea, and infections as the most common side effects after high-dose alkylating chemotherapy vs. standard neoadjuvant therapy in TNBC gBRCAm patients, as reported in the randomized phase 3 NeoTN trial [148].
Most studies on long-term adverse events in gBRCAm patients have focused particularly on cardiotoxicity, gonadotoxicity, and the psychological impact of the disease. A retrospective study of 898 patients (167 gBRCA1m, 91 gBRCA2m, and 640 non-gBRCAm) found no difference in hematological events, cardiac alterations, or neuropathy [149]. Also, gBRCAm was not associated with an increased risk of peripheral neuropathy [143]. In terms of cardiotoxicity, it seems that gBRCAm does not confer a higher risk of toxicity. In a single exploratory study involving 401 patients, comprising 232 BRCA1 and 159 BRCA2 mutation carriers, the authors reported a higher likelihood of heart failure based on self-reported symptoms obtained from an anonymous survey compared to historical controls from the general population. However, this study lacked objective confirmatory data, such as echocardiogram reports for most participants, and did not have a direct comparator control group [150]. Furthermore, two other studies found no significant differences in the rates of cardiomyopathy between gBRCAm carriers and non-carrier controls who received anthracyclines [151,152]. In a separate cross-sectional study, 67 patients with an average time of 6 years since BC diagnosis were enrolled. This study showed that women with gBRCAm did not have an increased risk of anthracycline-induced cardiotoxicity compared to those with sporadic BC [153].
Despite the increasing amount of available data on the safety and feasibility of preserving fertility and achieving pregnancy after BC diagnosis in the general population, there are still significant challenges facing patients with gBRCAm. Limited research has been conducted in this group, making it particularly difficult to provide accurate counseling on the risk of chemotherapy-induced gonadotoxicity [154]. In a multicenter survey conducted by Valentini et al., 1954 premenopausal gBRCA patients were studied, 1426 of whom received chemotherapy. Notably, the risk of premature ovarian failure was significantly higher in BRCA2 carriers than in BRCA1 carriers (46.8% vs. 32.7%; p < 0.001), but there was no significant difference in risk between non-carrier controls and either BRCA1 carriers or BRCA2 carriers [155]. Further research is needed to investigate the impact of treatment on the fertility of these patients and the effectiveness of fertility preservation techniques in this population.
Psychological impairment in patients with gBRCAm is also a crucial topic that deserves attention. BC diagnosis is a significant stressor for patients that can be exacerbated after gBRCAm diagnosis. A systematic review of eight studies that investigated the psychological impact of gBRCAm testing found that there was a negative effect on psychological well-being in the first months after positive test disclosure, with increased symptoms of distress, anxiety, and depression. In contrast, no significant clinical symptoms were observed in the intermediate and long-term periods. However, the lack of specific measurements that can accurately identify the psychological burdens of cancer-affected mutation carriers highlights the need for more research in this area [156]. Thus, further studies should focus on developing more precise and reliable screening tools to identify the long-term psychological impact of a positive gBRCAm diagnosis disclosure, allowing for better support and care for those affected women. Understanding the psychological impact of gBRCAm on patients can help healthcare providers to provide better support, reduce distress, and improve the overall HRQoL.
In summary, the available evidence suggests that patients with gBRCAm may have different patterns of toxicity during BC treatment, but the overall incidence is not significantly increased in these patients (Figure 2). However, further research is needed to clarify the association between gBRCAm and long-term adverse events, particularly in the context of novel targeted therapies and immunotherapies.

5. Current Research Needs in Breast Cancer Survivorship

Increasing BC survival rates due to advances in diagnosis and treatment have led to a growing population of BC survivors experiencing the physical and psychological side effects of treatment. Current European Society For Medical Oncology guidelines emphasize monitoring BC survivors for long-term psychological and neurobiological disturbances, including anxiety, depression, sleep disturbances, chronic fatigue, and neurocognitive dysfunction, among others [22].
In 2016, experts identified key priorities to improve BC management, including therapy de-escalation, optimizing the duration of adjuvant treatment, improving care for young patients, and focusing on survivorship and HRQoL [39]. These efforts underscore the growing emphasis on both the physical and psychological aspects of BC patient care.
Emerging treatments, such as immunotherapy, antibody–drug conjugates, and CDK4/6 inhibitors, present new opportunities but also challenges, including the potential for long-term adverse effects (e.g., endocrinopathies, arthritis, xerostomia, neurotoxicity, neutropenia, and gastrointestinal symptoms). While these toxicities are well-studied in metastatic disease, their long-term impact in eBC remains unclear [125,157,158]. Recent trials, like PEONY (phase 3) and coopERA (phase 2), highlight adverse events such as neutropenia, leukopenia, and low white blood cell numbers as emerging adverse factors that require further evaluation [159,160]. Clinical data from the KEYNOTE-522 trial include immune-related adverse events (including hypothyroidism, arthritis, hepatitis, and dermatitis, among others) in a significant subset of patients, linked to treatment response [161], underscoring the need for monitoring as these therapies gain wider use. Additionally, patients with gBRCAm eBC treated with a PARPi, such as olaparib, in the OlympiA clinical trial [18] may face unique adverse effects, including pneumonitis and new primary cancers, with further research needed to understand and manage the long-term impact.

6. Gaps and Future Directions in the Study of Long-Term Adverse Events in BC Survivors

Our review of the current literature reveals variations and gaps in the current understanding of long-term adverse events in BC (see Supplementary Table S1 for a summary of the studies cited). Despite significant advances in documenting treatment-related complications, several limitations persist that warrant further exploration.
  • Limited integration across affected systems: most studies examine individual domains, such as cardiotoxicity [44,66], CIPN [29,67], or psychological distress [32,55], in isolation. There is a lack of comprehensive frameworks to assess how these symptoms may interact over time and to determine their combined impact on the overall quality of life of BC survivors.
  • Inadequate stratification by patient subgroups: while gBRCA1/2 carriers and younger women have been studied in specific contexts [143,156], many studies fail to stratify findings by age, menopausal status, race/ethnicity, or comorbidities. This limits the ability to deliver tailored survivorship care that addresses the distinct risks and experiences of diverse BC patient populations.
  • Understudied and emerging adverse events: several important domains remain under-researched in long-term BC survivorship. These include endocrine dysfunction [35,126], sexual health and intimacy issues [36,58], weight gain, and metabolic syndrome [112,113]. These outcomes are rarely included in clinical trials and are inconsistently addressed in follow-up care plans, limiting comprehensive survivorship planning.
  • Inconsistency in the definition of long-term adverse events, including time frames: there is considerable variability in how studies define long-term and late adverse effects. Some studies classify them as events persisting beyond 1 year post-treatment, while others only focus on those emerging after 5 years [38,42]. Additionally, the heterogeneity in study endpoints, from clinical indicators to patient-reported outcomes, hampers cross-study comparisons and limits the ability to synthesize findings effectively.
  • Need for longitudinal, survivorship-focused research: some clinical trials emphasize efficacy (e.g., disease-free survival) over survivorship quality metrics [16,123]. Long-term safety and well-being outcomes require more deliberate inclusion in research study designs.
  • Opportunity for retrospective studies using electronic health records (EHRs): retrospective analysis of large EHR databases offers a promising avenue to study the impact of long-term adverse events on BC survivorship at scale in real-world clinical practice, beyond a controlled clinical trial setting. Several studies have demonstrated the utility of advanced natural language processing and machine learning to extract and analyze longitudinal data from unstructured clinical notes, enhancing the detection of late toxicities and comorbidities [162,163,164,165]. Leveraging such EHR-based platforms can fill critical gaps, especially in understudied patient subpopulations and rare adverse events, while providing insights into BC survivorship care patterns and outcomes across diverse healthcare systems.

7. Conclusions

As advances in early detection and treatment have improved survival rates for eBC, it has become increasingly essential to prioritize the management of long-term adverse events associated with treatment. These cumulative adverse effects can profoundly impact patients’ HRQoL, overall health, and sustained well-being beyond the initial treatment period.
This review highlights that while improving survival remains a primary goal, equal attention must be given to developing less toxic therapeutic options and employing personalized medicine approaches that tailor treatment to the individual patient’s risk profile. This personalized approach is crucial for mitigating long-term toxicities and optimizing patient outcomes. These treatment plans will provide a framework for continuous monitoring and supportive care, addressing patients’ evolving needs throughout their survivorship journey.
Although the subpopulation of patients with gBRCAm may experience added psychological stress and distinct clinical challenges during treatment, the current evidence does not conclusively demonstrate that their long-term adverse events differ substantially from those experienced by patients with non-gBRCAm BC. Nevertheless, these individuals require particular attention in survivorship programs to support their unique psychosocial and medical needs.
Looking ahead, future research must focus on integrated patient-centered survivorship frameworks with standardized definitions of adverse events, the inclusion of underrepresented BC patient populations, and extended follow-up periods. Such efforts will be vital to understanding and managing the long-term sequelae of BC treatment fully. Moreover, evidence derived from real-world clinical practice and EHRs will offer valuable insights that can inform individualized care strategies. By leveraging these data, healthcare providers can better align interventions with patient-specific needs, ultimately enhancing patient satisfaction and quality of care.
In summary, this review highlights the need to shift the focus from survival alone to a more comprehensive approach to BC care that incorporates the long-term management of adverse events and prioritizes HRQoL. This paradigm shift is essential to ensure that extended survival is matched by a life of sustained health, dignity, and well-being for BC survivors.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers17152506/s1, Table S1. Summary of key clinical studies reporting on long-term adverse events in patients with BC.

Author Contributions

C.B., B.C., B.O., S.R.J., J.V. and P.Z.: writing—review and editing. P.L. and L.C.-H.: methodology, writing—original draft, writing—review and editing. L.B.-A. and X.X.: conceptualization, project administration, supervision, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by AstraZeneca and Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA, who are codeveloping olaparib.

Acknowledgments

The authors thank Carlos Arias, David Casadevall, Ana Luiza Gomez, Judith Marín-Corral, Natalia Polo, Cristina Prieto, and Miren Taberna from Savana Research S.L for their editorial input during manuscript review.

Conflicts of Interest

B.O. acts as a speaker for AstraZeneca. C.B. receives consulting fees from Novartis, AstraZeneca, Daiichi-Sankyo, and Lilly, acts as a speaker for Viatris, Novartis, and Lilly, and receives support from attending meetings and/or travel from MS, AstraZeneca, Daiichi-Sankyo, and Novartis. B.C. is a grant and/or contract receiver from Roche and Pfizer, acts as a consultant for Daiichi-Sankyo, AstraZeneca, MSD, Lilly, Novartis, Pfizer, and Gilead, and receives support for attending meetings and/or travel from Novartis, Pfizer, and Daiichi-Sankyo. P.Z. receives support from attending meetings and/or travel from Pfizer, Novartis, and Daiichi-Sankyo. S.R.J. is a member of the advisory board of Enlace Health and a grant holder from different public and private entities. J.V. has no conflicts of interest. L.C.-H is a full-time employee at Savana Research, Madrid, Spain. P.L. was also a full-time employee at Savana Research during manuscript development. L.B.-A. and X.X. are full-time employees at AstraZeneca.

References

  1. Cancer Research UK. Risk Factors for Breast Cancer. 2023. Available online: https://www.cancerresearchuk.org/about-cancer/breast-cancer/risks-causes/risk-factors (accessed on 28 February 2025).
  2. Wooster, R.; Weber, B.L. Breast and ovarian cancer. N. Engl. J. Med. 2003, 348, 2339–2347. [Google Scholar] [CrossRef] [PubMed]
  3. Antoniou, A.; Pharoah, P.D.; Narod, S.; Risch, H.A.; Eyfjord, J.E.; Hopper, J.L.; Loman, N.; Olsson, H.; Johannsson, O.; Borg, A.; et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: A combined analysis of 22 studies. Am. J. Hum. Genet. 2003, 72, 1117–1130. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, S.; Parmigiani, G. Meta-analysis of BRCA1 and BRCA2 penetrance. J. Clin. Oncol. 2007, 25, 1329–1333. [Google Scholar] [CrossRef] [PubMed]
  5. Malone, K.E.; Daling, J.R.; Doody, D.R.; Hsu, L.; Bernstein, L.; Coates, R.J.; Marchbanks, P.A.; Simon, M.S.; McDonald, J.A.; Norman, S.A.; et al. Prevalence and predictors of BRCA1 and BRCA2 mutations in a population-based study of breast cancer in white and black American women ages 35 to 64 years. Cancer Res. 2006, 66, 8297–8308. [Google Scholar] [CrossRef] [PubMed]
  6. Howlader, N.; Altekruse, S.F.; Li, C.I.; Chen, V.W.; Clarke, C.A.; Ries, L.A.; Cronin, K.A. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J. Natl. Cancer Inst. 2014, 106, dju055. [Google Scholar] [CrossRef] [PubMed]
  7. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  8. Early Breast Cancer Trialists’ Collaborative Group. Aromatase inhibitors versus tamoxifen in early breast cancer: Patient-level meta-analysis of the randomised trials. Lancet 2015, 386, 1341–1352. [Google Scholar] [CrossRef] [PubMed]
  9. Pedersen, R.N.; Esen, B.; Mellemkjær, L.; Christiansen, P.; Ejlertsen, B.; Lash, T.L.; Nørgaard, M.; Cronin-Fenton, D. The incidence of breast cancer recurrence 10-32 years after primary diagnosis. J. Natl. Cancer Inst. 2022, 114, 391–399. [Google Scholar] [CrossRef] [PubMed]
  10. Mamounas, E.P.; Tang, G.; Paik, S.; Baehner, F.L.; Liu, Q.; Jeong, J.H.; Kim, S.R.; Butler, S.M.; Jamshidian, F.; Cherbavaz, D.B.; et al. 21-Gene Recurrence Score for prognosis and prediction of taxane benefit after adjuvant chemotherapy plus endocrine therapy: Results from NSABP B-28/NRG Oncology. Breast Cancer Res. Treat. 2018, 168, 69–77. [Google Scholar] [CrossRef] [PubMed]
  11. Pagani, O.; Francis, P.A.; Fleming, G.F.; Walley, B.A.; Viale, G.; Colleoni, M.; Láng, I.; Gómez, H.L.; Tondini, C.; Pinotti, G.; et al. Absolute improvements in freedom from distant recurrence to tailor adjuvant endocrine therapies for premenopausal women: Results from TEXT and SOFT. J. Clin. Oncol. 2020, 38, 1293–1303. [Google Scholar] [CrossRef] [PubMed]
  12. Cai, S.L.; Liu, J.J.; Liu, Y.X.; Yu, S.H.; Liu, X.; Lin, X.Q.; Chen, H.D.; Fang, X.; Ma, T.; Li, Y.Q.; et al. Characteristics of recurrence, predictors for relapse and prognosis of rapid relapse triple-negative breast cancer. Front. Oncol. 2023, 13, 1119611. [Google Scholar] [CrossRef] [PubMed]
  13. O’Shaughnessy, J.; Tolaney, S.M.; Yardley, D.A.; Hart, L.; Razavi, P.; Fasching, P.A.; Janni, W.; Schwartzberg, L.; Kim, J.; Akdere, M.; et al. Real-world risk of recurrence and treatment outcomes with adjuvant endocrine therapy in patients with stage II-III HR+/HER2- early breast cancer. Breast 2025, 81, 104437. [Google Scholar] [CrossRef] [PubMed]
  14. Schmid, P.; Cortes, J.; Dent, R.; McArthur, H.; Pusztai, L.; Kummel, S.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Overall survival with pembrolizumab in early-stage triple-negative breast cancer. N. Engl. J. Med. 2024, 391, 1981–1991. [Google Scholar] [CrossRef] [PubMed]
  15. Mai, N.; Myers, S.; Shen, S.; Downs-Canner, S.; Robson, M.; Norton, L.; Chen, Y.; Traina, T.; Abuhadra, N. Dose dense doxorubicin plus cyclophosphamide in a modified KEYNOTE522 regimen for triple negative breast cancer. npj Breast Cancer 2024, 10, 39. [Google Scholar] [CrossRef] [PubMed]
  16. Johnston, S.R.D.; Harbeck, N.; Hegg, R.; Toi, M.; Martin, M.; Shao, Z.M.; Zhang, Q.Y.; Martinez Rodriguez, J.L.; Campone, M.; Hamilton, E.; et al. Abemaciclib combined with endocrine therapy for the adjuvant treatment of HR+, HER2-, node-positive, high-risk, early breast cancer (monarchE). J. Clin. Oncol. 2020, 38, 3987–3998. [Google Scholar] [CrossRef] [PubMed]
  17. Slamon, D.; Lipatov, O.; Nowecki, Z.; McAndrew, N.; Kukielka-Budny, B.; Stroyakovskiy, D.; Yardley, D.A.; Huang, C.S.; Fasching, P.A.; Crown, J.; et al. Ribociclib plus endocrine therapy in early breast cancer. N. Engl. J. Med. 2024, 390, 1080–1091. [Google Scholar] [CrossRef] [PubMed]
  18. Tutt, A.N.J.; Garber, J.E.; Kaufman, B.; Viale, G.; Fumagalli, D.; Rastogi, P.; Gelber, R.D.; de Azambuja, E.; Fielding, A.; Balmana, J.; et al. Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef] [PubMed]
  19. Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Goncalves, A.; Lee, K.H.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M.; et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N. Engl. J. Med. 2018, 379, 753–763. [Google Scholar] [CrossRef] [PubMed]
  20. Robson, M.; Im, S.A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A.; et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 2017, 377, 523–533. [Google Scholar] [CrossRef] [PubMed]
  21. Geyer, C.E., Jr.; Garber, J.E.; Gelber, R.D.; Yothers, G.; Taboada, M.; Ross, L.; Rastogi, P.; Cui, K.; Arahmani, A.; Aktan, G.; et al. Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer. Ann. Oncol. 2022, 33, 1250–1268. [Google Scholar] [CrossRef] [PubMed]
  22. Loibl, S.; Andre, F.; Bachelot, T.; Barrios, C.H.; Bergh, J.; Burstein, H.J.; Cardoso, M.J.; Carey, L.A.; Dawood, S.; Del Mastro, L.; et al. Early breast cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2024, 35, 159–182. [Google Scholar] [CrossRef] [PubMed]
  23. Mougalian, S.S.; Hernandez, M.; Lei, X.; Lynch, S.; Kuerer, H.M.; Symmans, W.F.; Theriault, R.L.; Fornage, B.D.; Hsu, L.; Buchholz, T.A.; et al. Ten-year outcomes of patients with breast cancer with cytologically confirmed axillary lymph node metastases and pathologic complete response after primary systemic chemotherapy. JAMA Oncol. 2016, 2, 508–516. [Google Scholar] [CrossRef] [PubMed]
  24. El Saghir, N.S.; Khalil, L.E.; El Dick, J.; Atwani, R.W.; Safi, N.; Charafeddine, M.; Al-Masri, A.; El Saghir, B.N.; Chaccour, M.; Tfayli, A.; et al. Improved survival of young patients with breast cancer 40 years and younger at diagnosis. JCO Glob. Oncol. 2023, 9, e2200354. [Google Scholar] [CrossRef] [PubMed]
  25. Nussbaumer, R.L.; Maggi, N.; Castrezana, L.; Zehnpfennig, L.; Schwab, F.D.; Krol, J.; Oberhauser, I.; Weber, W.P.; Kurzeder, C.; Haug, M.D.; et al. The impact of neoadjuvant systemic treatment on postoperative complications in breast cancer surgery. Breast Cancer Res. Treat. 2023, 197, 333–341. [Google Scholar] [CrossRef] [PubMed]
  26. Stein, K.D.; Syrjala, K.L.; Andrykowski, M.A. Physical and psychological long-term and late effects of cancer. Cancer 2008, 112, 2577–2592. [Google Scholar] [CrossRef] [PubMed]
  27. Andersen, K.G.; Kehlet, H. Persistent pain after breast cancer treatment: A critical review of risk factors and strategies for prevention. J. Pain 2011, 12, 725–746. [Google Scholar] [CrossRef] [PubMed]
  28. Hayes, S.C.; Janda, M.; Cornish, B.; Battistutta, D.; Newman, B. Lymphedema after breast cancer: Incidence, risk factors, and effect on upper body function. J. Clin. Oncol. 2008, 26, 3536–3542. [Google Scholar] [CrossRef] [PubMed]
  29. Seretny, M.; Currie, G.L.; Sena, E.S.; Ramnarine, S.; Grant, R.; MacLeod, M.R.; Colvin, L.A.; Fallon, M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain 2014, 155, 2461–2470. [Google Scholar] [CrossRef] [PubMed]
  30. Rugo, H.S.; O’Shaughnessy, J.; Boyle, F.; Toi, M.; Broom, R.; Blancas, I.; Gumus, M.; Yamashita, T.; Im, Y.H.; Rastogi, P.; et al. Adjuvant abemaciclib combined with endocrine therapy for high-risk early breast cancer: Safety and patient-reported outcomes from the monarchE study. Ann. Oncol. 2022, 33, 616–627. [Google Scholar] [CrossRef] [PubMed]
  31. Brownlee, Z.; Garg, R.; Listo, M.; Zavitsanos, P.; Wazer, D.E.; Huber, K.E. Late complications of radiation therapy for breast cancer: Evolution in techniques and risk over time. Gland. Surg. 2018, 7, 371–378. [Google Scholar] [CrossRef] [PubMed]
  32. Maass, S.W.; Roorda, C.; Berendsen, A.J.; Verhaak, P.F.; de Bock, G.H. The prevalence of long-term symptoms of depression and anxiety after breast cancer treatment: A systematic review. Maturitas 2015, 82, 100–108. [Google Scholar] [CrossRef] [PubMed]
  33. Alvarez-Bustos, A.; de Pedro, C.G.; Romero-Elias, M.; Ramos, J.; Osorio, P.; Cantos, B.; Maximiano, C.; Mendez, M.; Fiuza-Luces, C.; Mendez-Otero, M.; et al. Prevalence and correlates of cancer-related fatigue in breast cancer survivors. Support. Care Cancer 2021, 29, 6523–6534. [Google Scholar] [CrossRef] [PubMed]
  34. Zeng, E.; He, W.; Smedby, K.E.; Czene, K. Adjuvant hormone therapy-related hot flashes predict treatment discontinuation and worse breast cancer prognosis. J. Natl. Compr. Cancer Netw. 2022, 20, 683–689 e682. [Google Scholar] [CrossRef] [PubMed]
  35. Digkas, E.; Smith, D.R.; Wennstig, A.K.; Matikas, A.; Tegnelius, E.; Valachis, A. Incidence and risk factors of hypothyroidism after treatment for early breast cancer: A population-based cohort study. Breast Cancer Res. Treat. 2024, 204, 79–87. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, M.; Kim, Y.H.; Jeon, M.J. Risk factors for negative impacts on sexual activity and function in younger breast cancer survivors. Psychooncology 2015, 24, 1097–1103. [Google Scholar] [CrossRef] [PubMed]
  37. Izci, F.; Ilgun, A.S.; Findikli, E.; Ozmen, V. Psychiatric symptoms and psychosocial problems in patients with breast cancer. J. Breast Health 2016, 12, 94–101. [Google Scholar] [CrossRef] [PubMed]
  38. Lovelace, D.L.; McDaniel, L.R.; Golden, D. Long-term effects of breast cancer surgery, treatment, and survivor care. J. Midwifery Womens Health 2019, 64, 713–724. [Google Scholar] [CrossRef] [PubMed]
  39. Cardoso, F.; Harbeck, N.; Barrios, C.H.; Bergh, J.; Cortes, J.; El Saghir, N.; Francis, P.A.; Hudis, C.A.; Ohno, S.; Partridge, A.H.; et al. Research needs in breast cancer. Ann. Oncol. 2017, 28, 208–217. [Google Scholar] [CrossRef] [PubMed]
  40. Andreu, Y.; Soto-Rubio, A.; Ramos-Campos, M.; Escriche-Saura, A.; Martinez, M.; Gavila, J. Impact of hormone therapy side effects on health-related quality of life, distress, and well-being of breast cancer survivors. Sci. Rep. 2022, 12, 18673. [Google Scholar] [CrossRef] [PubMed]
  41. Hamood, R.; Hamood, H.; Merhasin, I.; Keinan-Boker, L. Chronic pain and other symptoms among breast cancer survivors: Prevalence, predictors, and effects on quality of life. Breast Cancer Res. Treat. 2018, 167, 157–169. [Google Scholar] [CrossRef] [PubMed]
  42. Schmidt, M.E.; Wiskemann, J.; Steindorf, K. Quality of life, problems, and needs of disease-free breast cancer survivors 5 years after diagnosis. Qual. Life Res. 2018, 27, 2077–2086. [Google Scholar] [CrossRef] [PubMed]
  43. Maass, S.; Brandenbarg, D.; Boerman, L.M.; Verhaak, P.F.M.; de Bock, G.H.; Berendsen, A.J. Fatigue among long-term breast cancer survivors: A controlled cross-sectional study. Cancers 2021, 13, 1301. [Google Scholar] [CrossRef] [PubMed]
  44. Zambetti, M.; Moliterni, A.; Materazzo, C.; Stefanelli, M.; Cipriani, S.; Valagussa, P.; Bonadonna, G.; Gianni, L. Long-term cardiac sequelae in operable breast cancer patients given adjuvant chemotherapy with or without doxorubicin and breast irradiation. J. Clin. Oncol. 2001, 19, 37–43. [Google Scholar] [CrossRef] [PubMed]
  45. Bonneterre, J.; Roche, H.; Kerbrat, P.; Fumoleau, P.; Goudier, M.J.; Fargeot, P.; Montcuquet, P.; Clavere, P.; Barats, J.C.; Monnier, A.; et al. Long-term cardiac follow-up in relapse-free patients after six courses of fluorouracil, epirubicin, and cyclophosphamide, with either 50 or 100 mg of epirubicin, as adjuvant therapy for node-positive breast cancer: French adjuvant study group. J. Clin. Oncol. 2004, 22, 3070–3079. [Google Scholar] [CrossRef] [PubMed]
  46. Ganz, P.A.; Hussey, M.A.; Moinpour, C.M.; Unger, J.M.; Hutchins, L.F.; Dakhil, S.R.; Giguere, J.K.; Goodwin, J.W.; Martino, S.; Albain, K.S. Late cardiac effects of adjuvant chemotherapy in breast cancer survivors treated on Southwest Oncology Group protocol s8897. J. Clin. Oncol. 2008, 26, 1223–1230. [Google Scholar] [CrossRef] [PubMed]
  47. Macdonald, L.; Bruce, J.; Scott, N.W.; Smith, W.C.; Chambers, W.A. Long-term follow-up of breast cancer survivors with post-mastectomy pain syndrome. Br. J. Cancer 2005, 92, 225–230. [Google Scholar] [CrossRef] [PubMed]
  48. Davies, C.; Levenhagen, K.; Ryans, K.; Perdomo, M.; Gilchrist, L. Interventions for breast cancer-related lymphedema: Clinical practice guideline from the Academy of Oncologic Physical Therapy of APTA. Phys. Ther. 2020, 100, 1163–1179. [Google Scholar] [CrossRef] [PubMed]
  49. Torgbenu, E.; Luckett, T.; Buhagiar, M.A.; Chang, S.; Phillips, J.L. Prevalence and incidence of cancer related lymphedema in low and middle-income countries: A systematic review and meta-analysis. BMC Cancer 2020, 20, 604. [Google Scholar] [CrossRef] [PubMed]
  50. Classen, J.; Nitzsche, S.; Wallwiener, D.; Kristen, P.; Souchon, R.; Bamberg, M.; Brucker, S. Fibrotic changes after postmastectomy radiotherapy and reconstructive surgery in breast cancer. A retrospective analysis in 109 patients. Strahlenther. Onkol. 2010, 186, 630–636. [Google Scholar] [CrossRef] [PubMed]
  51. Khosrow-Khavar, F.; Filion, K.B.; Al-Qurashi, S.; Torabi, N.; Bouganim, N.; Suissa, S.; Azoulay, L. Cardiotoxicity of aromatase inhibitors and tamoxifen in postmenopausal women with breast cancer: A systematic review and meta-analysis of randomized controlled trials. Ann. Oncol. 2017, 28, 487–496. [Google Scholar] [CrossRef] [PubMed]
  52. Eckhoff, L.; Knoop, A.; Jensen, M.B.; Ewertz, M. Persistence of docetaxel-induced neuropathy and impact on quality of life among breast cancer survivors. Eur. J. Cancer 2015, 51, 292–300. [Google Scholar] [CrossRef] [PubMed]
  53. Tanabe, Y.; Hashimoto, K.; Shimizu, C.; Hirakawa, A.; Harano, K.; Yunokawa, M.; Yonemori, K.; Katsumata, N.; Tamura, K.; Ando, M.; et al. Paclitaxel-induced peripheral neuropathy in patients receiving adjuvant chemotherapy for breast cancer. Int. J. Clin. Oncol. 2013, 18, 132–138. [Google Scholar] [CrossRef] [PubMed]
  54. Onzi, G.R.; D’Agustini, N.; Garcia, S.C.; Guterres, S.S.; Pohlmann, P.R.; Rosa, D.D.; Pohlmann, A.R. Chemobrain in breast cancer: Mechanisms, clinical manifestations, and potential interventions. Drug Saf. 2022, 45, 601–621. [Google Scholar] [CrossRef] [PubMed]
  55. Mitchell, A.J.; Ferguson, D.W.; Gill, J.; Paul, J.; Symonds, P. Depression and anxiety in long-term cancer survivors compared with spouses and healthy controls: A systematic review and meta-analysis. Lancet Oncol. 2013, 14, 721–732. [Google Scholar] [CrossRef] [PubMed]
  56. Luz, P.; Carvalho, A.N.; Oliveira, A.; Menezes, M.; Dinis, R.; Gosalbez, B. Prevalence of fear of death among young breast cancer patients during adjuvant endocrine therapy: Results from a portuguese cohort. Acta Medica Port. 2021, 34, 400–401. [Google Scholar] [CrossRef] [PubMed]
  57. Carmen, A.; Anne, O.; Monika, S.; Daniel, E.; Johannes, G.; Verena, M.; Michael, H.; Christine, B. Does the toxicity of endocrine therapy persist into long-term survivorship?: Patient-reported outcome results from a follow-up study beyond a 10-year-survival. Breast Cancer Res. Treat. 2022, 198, 475–485. [Google Scholar] [CrossRef] [PubMed]
  58. Raggio, G.A.; Butryn, M.L.; Arigo, D.; Mikorski, R.; Palmer, S.C. Prevalence and correlates of sexual morbidity in long-term breast cancer survivors. Psychol. Health 2014, 29, 632–650. [Google Scholar] [CrossRef] [PubMed]
  59. Lambertini, M.; Blondeaux, E.; Bruzzone, M.; Perachino, M.; Anderson, R.A.; de Azambuja, E.; Poorvu, P.D.; Kim, H.J.; Villarreal-Garza, C.; Pistilli, B.; et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J. Clin. Oncol. 2021, 39, 3293–3305. [Google Scholar] [CrossRef] [PubMed]
  60. Shapiro, C.L. Osteoporosis: A long-term and late-effect of breast cancer treatments. Cancers 2020, 12, 3094. [Google Scholar] [CrossRef] [PubMed]
  61. Chang, P.J.; Asher, A.; Smith, S.R. A targeted approach to post-mastectomy pain and persistent pain following breast cancer treatment. Cancers 2021, 13, 5191. [Google Scholar] [CrossRef] [PubMed]
  62. Gartner, R.; Jensen, M.B.; Nielsen, J.; Ewertz, M.; Kroman, N.; Kehlet, H. Prevalence of and factors associated with persistent pain following breast cancer surgery. JAMA 2009, 302, 1985–1992. [Google Scholar] [CrossRef] [PubMed]
  63. Gao, Y.; Rosas, J.C.; Fink, H.; Behrens, S.; Chang-Claude, J.; Seibold, P. Longitudinal changes of health-related quality of life over 10 years in breast cancer patients treated with radiotherapy following breast-conserving surgery. Qual. Life Res. 2023, 32, 2639–2652. [Google Scholar] [CrossRef] [PubMed]
  64. Curigliano, G.; Lenihan, D.; Fradley, M.; Ganatra, S.; Barac, A.; Blaes, A.; Herrmann, J.; Porter, C.; Lyon, A.R.; Lancellotti, P.; et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann. Oncol. 2020, 31, 171–190. [Google Scholar] [CrossRef] [PubMed]
  65. Piroth, M.D.; Baumann, R.; Budach, W.; Dunst, J.; Feyer, P.; Fietkau, R.; Haase, W.; Harms, W.; Hehr, T.; Krug, D.; et al. Heart toxicity from breast cancer radiotherapy: Current findings, assessment, and prevention. Strahlenther. Onkol. 2019, 195, 1–12. [Google Scholar] [CrossRef] [PubMed]
  66. Puckett, L.L.; Saba, S.G.; Henry, S.; Rosen, S.; Rooney, E.; Filosa, S.L.; Gilbo, P.; Pappas, K.; Laxer, A.; Eacobacci, K.; et al. Cardiotoxicity screening of long-term, breast cancer survivors—The CAROLE (Cardiac-Related Oncologic Late Effects) study. Cancer Med. 2021, 10, 5051–5061. [Google Scholar] [CrossRef] [PubMed]
  67. Bao, T.; Basal, C.; Seluzicki, C.; Li, S.Q.; Seidman, A.D.; Mao, J.J. Long-term chemotherapy-induced peripheral neuropathy among breast cancer survivors: Prevalence, risk factors, and fall risk. Breast Cancer Res. Treat. 2016, 159, 327–333. [Google Scholar] [CrossRef] [PubMed]
  68. Lange, M.; Hardy-Leger, I.; Licaj, I.; Pistilli, B.; Rigal, O.; Le Fel, J.; Levy, C.; Capel, A.; Coutant, C.; Meyer, J.; et al. Cognitive impairment in patients with breast cancer before surgery: Results from a CANTO cohort subgroup. Cancer Epidemiol. Biomark. Prev. 2020, 29, 1759–1766. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, X.; Wang, N.; Zhong, L.; Wang, S.; Zheng, Y.; Yang, B.; Zhang, J.; Lin, Y.; Wang, Z. Prognostic value of depression and anxiety on breast cancer recurrence and mortality: A systematic review and meta-analysis of 282,203 patients. Mol. Psychiatry 2020, 25, 3186–3197. [Google Scholar] [CrossRef] [PubMed]
  70. Grassi, L.; Caruso, R.; Riba, M.B.; Lloyd-Williams, M.; Kissane, D.; Rodin, G.; McFarland, D.; Campos-Ródenas, R.; Zachariae, R.; Santini, D.; et al. Anxiety and depression in adult cancer patients: ESMO Clinical Practice Guideline. ESMO Open 2023, 8, 101155. [Google Scholar] [CrossRef] [PubMed]
  71. Vickberg, S.M. The Concerns About Recurrence Scale (CARS): A systematic measure of women’s fears about the possibility of breast cancer recurrence. Ann. Behav. Med. 2003, 25, 16–24. [Google Scholar] [CrossRef] [PubMed]
  72. Abrahams, H.J.G.; Gielissen, M.F.M.; Schmits, I.C.; Verhagen, C.; Rovers, M.M.; Knoop, H. Risk factors, prevalence, and course of severe fatigue after breast cancer treatment: A meta-analysis involving 12,327 breast cancer survivors. Ann. Oncol. 2016, 27, 965–974. [Google Scholar] [CrossRef] [PubMed]
  73. Seav, S.M.; Dominick, S.A.; Stepanyuk, B.; Gorman, J.R.; Chingos, D.T.; Ehren, J.L.; Krychman, M.L.; Su, H.I. Management of sexual dysfunction in breast cancer survivors: A systematic review. Women’s Midlife Health 2015, 1, 9. [Google Scholar] [CrossRef] [PubMed]
  74. Poorvu, P.D.; Gelber, S.I.; Zheng, Y.; Ruddy, K.J.; Tamimi, R.M.; Peppercorn, J.; Schapira, L.; Borges, V.F.; Come, S.E.; Lambertini, M.; et al. Pregnancy after breast cancer: Results from a prospective cohort of young women with breast cancer. Cancer 2021, 127, 1021–1028. [Google Scholar] [CrossRef] [PubMed]
  75. Blair, S.L. De-escalation of breast cancer surgery. Surgery 2023, 174, 123–124. [Google Scholar] [CrossRef] [PubMed]
  76. Soni, A.; Morgan, J.; Wyld, L. A qualitative study exploring the views of global healthcare professionals towards de-escalation of axillary surgery in early breast cancer. Eur. J. Surg. Oncol. 2025, 51, 110079. [Google Scholar] [CrossRef] [PubMed]
  77. De La Cruz, L.; Blankenship, S.A.; Chatterjee, A.; Geha, R.; Nocera, N.; Czerniecki, B.J.; Tchou, J.; Fisher, C.S. Outcomes after oncoplastic breast-conserving surgery in breast cancer patients: A systematic literature review. Ann. Surg. Oncol. 2016, 23, 3247–3258. [Google Scholar] [CrossRef] [PubMed]
  78. Caffo, O.; Amichetti, M.; Ferro, A.; Lucenti, A.; Valduga, F.; Galligioni, E. Pain and quality of life after surgery for breast cancer. Breast Cancer Res. Treat. 2003, 80, 39–48. [Google Scholar] [CrossRef] [PubMed]
  79. Shamsesfandabadi, P.; Shams Esfand Abadi, M.; Yin, Y.; Carpenter, D.J.; Peluso, C.; Hilton, C.; Coopey, S.B.; Gomez, J.; Beriwal, S.; Champ, C.E. Resistance training and lymphedema in breast cancer survivors. JAMA Netw. Open 2025, 8, e2514765. [Google Scholar] [CrossRef] [PubMed]
  80. Jhaveri, J.D.; Rush, S.C.; Kostroff, K.; Derisi, D.; Farber, L.A.; Maurer, V.E.; Bosworth, J.L. Clinical outcomes of postmastectomy radiation therapy after immediate breast reconstruction. Int. J. Radiat. Oncol. Biol. Phys. 2008, 72, 859–865. [Google Scholar] [CrossRef] [PubMed]
  81. Batenburg, M.C.T.; Bartels, M.; Maarse, W.; Witkamp, A.; Verkooijen, H.M.; van den Bongard, H.J.G.D. Factors associated with late local radiation toxicity after post-operative breast irradiation. Breast J. 2022, 2022, 6745954. [Google Scholar] [CrossRef] [PubMed]
  82. Jabbari, A.; Mousavi, E.; Nikoubin-Boroujeni, M.; Tabasi, S.; Arjmandi, N.; Ghelishli, N.; Arab-Bafrani, Z. Cardiac toxicity under concurrent administration of trastuzumab (anti-HER2 therapy) and radiotherapy: Systematic review and meta-analysis. Health Sci. Rep. 2025, 8, e70966. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, X.; Xue, Y.; Hao, M. Cardiotoxicity induced by chemotherapy and immunotherapy in cancer treatment: A bibliometric analysis. Discov. Oncol. 2025, 16, 376. [Google Scholar] [CrossRef] [PubMed]
  84. Zagami, P.; Trapani, D.; Nicolò, E.; Corti, C.; Valenza, C.; Criscitiello, C.; Curigliano, G.; Carey, L.A. Cardiotoxicity of agents used in patients with breast cancer. JCO Oncol. Pract. 2024, 20, 38–46. [Google Scholar] [CrossRef] [PubMed]
  85. Strongman, H.; Gadd, S.; Matthews, A.; Mansfield, K.E.; Stanway, S.; Lyon, A.R.; Dos-Santos-Silva, I.; Smeeth, L.; Bhaskaran, K. Medium and long-term risks of specific cardiovascular diseases in survivors of 20 adult cancers: A population-based cohort study using multiple linked UK electronic health records databases. Lancet 2019, 394, 1041–1054. [Google Scholar] [CrossRef] [PubMed]
  86. Fu, Z.; Lin, Z.; Yang, M.; Li, C. Cardiac toxicity from adjuvant targeting treatment for breast cancer post-surgery. Front. Oncol. 2022, 12, 706861. [Google Scholar] [CrossRef] [PubMed]
  87. Mata Caballero, R.; Serrano Antolin, J.M.; Jimenez Hernandez, R.M.; Talavera Calle, P.; Curcio Ruigomez, A.; Del Castillo Arrojo, S.; Graupner Abad, C.; Cristobal Varela, C.; Alonso Martin, J.J. Incidence of long-term cardiotoxicity and evolution of the systolic function in patients with breast cancer treated with anthracyclines. Cardiol. J. 2022, 29, 228–234. [Google Scholar] [CrossRef] [PubMed]
  88. Bouwer, N.I.; Jager, A.; Liesting, C.; Kofflard, M.J.M.; Brugts, J.J.; Kitzen, J.; Boersma, E.; Levin, M.D. Cardiac monitoring in HER2-positive patients on trastuzumab treatment: A review and implications for clinical practice. Breast 2020, 52, 33–44. [Google Scholar] [CrossRef] [PubMed]
  89. Dent, S.F.; Moore, H.; Raval, P.; Alder, L.; Guha, A. How to manage and monitor cardiac dysfunction in patients with metastatic HER2-positive breast cancer. JACC CardioOncol. 2022, 4, 404–408. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, C.; Fan, P.; Wang, Q. Evolving therapeutics and ensuing cardiotoxicities in triple-negative breast cancer. Cancer Treat. Rev. 2024, 130, 102819. [Google Scholar] [CrossRef] [PubMed]
  91. Sund, M.; Garcia-Argibay, M.; Garmo, H.; Ahlgren, J.; Wennstig, A.K.; Fredriksson, I.; Lindman, H.; Valachis, A. Aromatase inhibitors use and risk for cardiovascular disease in breast cancer patients: A population-based cohort study. Breast 2021, 59, 157–164. [Google Scholar] [CrossRef] [PubMed]
  92. Yoo, J.J.; Jung, E.A.; Kim, Z.; Kim, B.Y. Risk of cardiovascular events and lipid profile change in patients with breast cancer taking aromatase inhibitor: A systematic review and meta-analysis. Curr. Oncol. 2023, 30, 1831–1843. [Google Scholar] [CrossRef] [PubMed]
  93. Cheung, Y.M.; Ramchand, S.K.; Yeo, B.; Grossmann, M. Cardiometabolic effects of endocrine treatment of estrogen receptor-positive early breast cancer. J. Endocr. Soc. 2019, 3, 1283–1301. [Google Scholar] [CrossRef] [PubMed]
  94. Cheang, I.; Gue, Y.; Wu, M.Z.; Huang, J.Y.; Ren, Q.W.; Chen, Z.; Tse, Y.K.; Li, H.L.; Chan, Y.H.; Tse, H.F.; et al. Cardiovascular risks associated with adjuvant endocrine therapy in women with breast cancer: A population-based cohort study. BMC Cancer 2025, 25, 1063. [Google Scholar] [CrossRef] [PubMed]
  95. Forbes, J.F.; Sestak, I.; Howell, A.; Bonanni, B.; Bundred, N.; Levy, C.; von Minckwitz, G.; Eiermann, W.; Neven, P.; Stierer, M.; et al. Anastrozole versus tamoxifen for the prevention of locoregional and contralateral breast cancer in postmenopausal women with locally excised ductal carcinoma in situ (IBIS-II DCIS): A double-blind, randomised controlled trial. Lancet 2016, 387, 866–873. [Google Scholar] [CrossRef] [PubMed]
  96. Cuppone, F.; Bria, E.; Verma, S.; Pritchard, K.I.; Gandhi, S.; Carlini, P.; Milella, M.; Nistico, C.; Terzoli, E.; Cognetti, F.; et al. Do adjuvant aromatase inhibitors increase the cardiovascular risk in postmenopausal women with early breast cancer? Meta-analysis of randomized trials. Cancer 2008, 112, 260–267. [Google Scholar] [CrossRef] [PubMed]
  97. Khosrow-Khavar, F.; Filion, K.B.; Bouganim, N.; Suissa, S.; Azoulay, L. Aromatase inhibitors and the risk of cardiovascular outcomes in women with breast cancer: A population-based cohort study. Circulation 2020, 141, 549–559. [Google Scholar] [CrossRef] [PubMed]
  98. Roy, S.; Lakritz, S.; Schreiber, A.R.; Kuna, E.M.; Bradley, C.J.; Kondapalli, L.; Diamond, J.R. Major cardiovascular adverse events in older adults with early-stage triple-negative breast cancer treated with adjuvant taxane + anthracycline versus taxane-based chemotherapy regimens: A SEER-medicare study. Eur. J. Cancer 2024, 196, 113426. [Google Scholar] [CrossRef] [PubMed]
  99. Nitsche, M.; Pahl, R.; Huber, K.; Eilf, K.; Dunst, J. Cardiac toxicity after radiotherapy for breast cancer: Myths and facts. Breast Care 2015, 10, 131–135. [Google Scholar] [CrossRef] [PubMed]
  100. Schwab, L.; Visovsky, C. Psychological distress and quality of life in breast cancer survivors with taxane-induced peripheral neuropathy: A scoping review. Front. Oncol. 2022, 12, 1005083. [Google Scholar] [CrossRef] [PubMed]
  101. Jordan, B.; Margulies, A.; Cardoso, F.; Cavaletti, G.; Haugnes, H.S.; Jahn, P.; Le Rhun, E.; Preusser, M.; Scotte, F.; Taphoorn, M.J.B.; et al. Systemic anticancer therapy-induced peripheral and central neurotoxicity: ESMO-EONS-EANO Clinical Practice Guidelines for diagnosis, prevention, treatment and follow-up. Ann. Oncol. 2020, 31, 1306–1319. [Google Scholar] [CrossRef] [PubMed]
  102. Di Leone, A.; Terribile, D.; Magno, S.; Sanchez, A.M.; Scardina, L.; Mason, E.J.; D’Archi, S.; Maggiore, C.; Rossi, C.; Di Micco, A.; et al. Neoadjuvant chemotherapy in breast cancer: An advanced personalized multidisciplinary prehabilitation model (APMP-M) to optimize outcomes. J. Pers. Med. 2021, 11, 324. [Google Scholar] [CrossRef] [PubMed]
  103. Whittaker, A.L.; George, R.P.; O’Malley, L. Prevalence of cognitive impairment following chemotherapy treatment for breast cancer: A systematic review and meta-analysis. Sci. Rep. 2022, 12, 2135. [Google Scholar] [CrossRef] [PubMed]
  104. Dietrich, J. Chemotherapy associated central nervous system damage. Adv. Exp. Med. Biol. 2010, 678, 77–85. [Google Scholar] [CrossRef] [PubMed]
  105. Arrillaga-Romany, I.C.; Dietrich, J. Imaging findings in cancer therapy-associated neurotoxicity. Semin. Neurol. 2012, 32, 476–486. [Google Scholar] [CrossRef] [PubMed]
  106. Meyers, C.A. How chemotherapy damages the central nervous system. J. Biol. 2008, 7, 11. [Google Scholar] [CrossRef] [PubMed]
  107. Le, N.K.; Gabrick, K.S.; Chouairi, F.; Mets, E.J.; Avraham, T.; Alperovich, M. Impact of socioeconomic status on psychological functioning in survivorship following breast cancer and reconstruction. Breast J. 2020, 26, 1695–1701. [Google Scholar] [CrossRef] [PubMed]
  108. Bickel, E.A.; Fleer, J.; Ranchor, A.V.; Schroevers, M.J. Are cancer patients with high depressive symptom levels able to manage these symptoms without professional care? The role of coping and social support. Psychooncology 2022, 31, 1102–1109. [Google Scholar] [CrossRef] [PubMed]
  109. Di Meglio, A.; Havas, J.; Martin, E.; Pistilli, B.; Menvielle, G.; Dumas, A.; Charles, C.; Everhard, S.; Martin, A.-L.; Coutant, C.; et al. Assessing the risk of severe post-treatment (tx) cancer-related fatigue (CRF) among breast cancer survivors (BCS) in the CANcer TOxicity (CANTO) cohort. J. Clin. Oncol. 2021, 39, 12022. [Google Scholar] [CrossRef]
  110. Roila, F.; Fumi, G.; Fatigoni, S. Management of fatigue following breast cancer treatment. Breast Cancer Manag. 2016, 5, 79–87. [Google Scholar] [CrossRef]
  111. Haidinger, R.; Bauerfeind, I. Long-term side effects of adjuvant therapy in primary breast cancer patients: Results of a web-based survey. Breast Care 2019, 14, 111–116. [Google Scholar] [CrossRef] [PubMed]
  112. Demark-Wahnefried, W.; Campbell, K.L.; Hayes, S.C. Weight management and its role in breast cancer rehabilitation. Cancer 2012, 118, 2277–2287. [Google Scholar] [CrossRef] [PubMed]
  113. Kroenke, C.H.; Chen, W.Y.; Rosner, B.; Holmes, M.D. Weight, weight gain, and survival after breast cancer diagnosis. J. Clin. Oncol. 2005, 23, 1370–1378. [Google Scholar] [CrossRef] [PubMed]
  114. Nichols, H.B.; Trentham-Dietz, A.; Egan, K.M.; Titus-Ernstoff, L.; Holmes, M.D.; Bersch, A.J.; Holick, C.N.; Hampton, J.M.; Stampfer, M.J.; Willett, W.C.; et al. Body mass index before and after breast cancer diagnosis: Associations with all-cause, breast cancer, and cardiovascular disease mortality. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1403–1409. [Google Scholar] [CrossRef] [PubMed]
  115. Vance, V.; Mourtzakis, M.; McCargar, L.; Hanning, R. Weight gain in breast cancer survivors: Prevalence, pattern and health consequences. Obes. Rev. 2011, 12, 282–294. [Google Scholar] [CrossRef] [PubMed]
  116. Heudel, P.E.; Van Praagh-Doreau, I.; Duvert, B.; Cauvin, I.; Hardy-Bessard, A.C.; Jacquin, J.P.; Stefani, L.; Vincent, L.; Dramais, D.; Guastalla, J.P.; et al. Does a homeopathic medicine reduce hot flushes induced by adjuvant endocrine therapy in localized breast cancer patients? A multicenter randomized placebo-controlled phase III trial. Support. Care Cancer 2019, 27, 1879–1889. [Google Scholar] [CrossRef] [PubMed]
  117. Ayers, B.; Smith, M.; Hellier, J.; Mann, E.; Hunter, M.S. Effectiveness of group and self-help cognitive behavior therapy in reducing problematic menopausal hot flushes and night sweats (MENOS 2): A randomized controlled trial. Menopause 2012, 19, 749–759. [Google Scholar] [CrossRef] [PubMed]
  118. Carmody, J.F.; Crawford, S.; Salmoirago-Blotcher, E.; Leung, K.; Churchill, L.; Olendzki, N. Mindfulness training for coping with hot flashes: Results of a randomized trial. Menopause 2011, 18, 611–620. [Google Scholar] [CrossRef] [PubMed]
  119. McCurry, S.M.; Guthrie, K.A.; Morin, C.M.; Woods, N.F.; Landis, C.A.; Ensrud, K.E.; Larson, J.C.; Joffe, H.; Cohen, L.S.; Hunt, J.R.; et al. Telephone-based cognitive behavioral therapy for insomnia in perimenopausal and postmenopausal women with vasomotor symptoms: A MsFLASH randomized clinical trial. JAMA Intern. Med. 2016, 176, 913–920. [Google Scholar] [CrossRef] [PubMed]
  120. Aerts, L.; Christiaens, M.R.; Enzlin, P.; Neven, P.; Amant, F. Sexual functioning in women after mastectomy versus breast conserving therapy for early-stage breast cancer: A prospective controlled study. Breast 2014, 23, 629–636. [Google Scholar] [CrossRef] [PubMed]
  121. Partridge, A.H.; Niman, S.M.; Ruggeri, M.; Peccatori, F.A.; Azim, H.A., Jr.; Colleoni, M.; Saura, C.; Shimizu, C.; Saetersdal, A.B.; Kroep, J.R.; et al. Who are the women who enrolled in the POSITIVE trial: A global study to support young hormone receptor positive breast cancer survivors desiring pregnancy. Breast 2021, 59, 327–338. [Google Scholar] [CrossRef] [PubMed]
  122. Lambertini, M.; Peccatori, F.A.; Demeestere, I.; Amant, F.; Wyns, C.; Stukenborg, J.B.; Paluch-Shimon, S.; Halaska, M.J.; Uzan, C.; Meissner, J.; et al. Fertility preservation and post-treatment pregnancies in post-pubertal cancer patients: ESMO Clinical Practice Guidelines. Ann. Oncol. 2020, 31, 1664–1678. [Google Scholar] [CrossRef] [PubMed]
  123. Schmid, P.; Cortes, J.; Pusztai, L.; McArthur, H.; Kummel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Pembrolizumab for early triple-negative breast cancer. N. Engl. J. Med. 2020, 382, 810–821. [Google Scholar] [CrossRef] [PubMed]
  124. Friedlander, M.; Lee, Y.C.; Tew, W.P. Managing adverse effects associated with poly (ADP-ribose) polymerase inhibitors in ovarian cancer: A synthesis of clinical trial and real-world data. Soc. Clin. Oncol. Educ. Book 2023, 43, e390876. [Google Scholar] [CrossRef] [PubMed]
  125. Johnson, D.B.; Nebhan, C.A.; Moslehi, J.J.; Balko, J.M. Immune-checkpoint inhibitors: Long-term implications of toxicity. Nat. Rev. Clin. Oncol. 2022, 19, 254–267. [Google Scholar] [CrossRef] [PubMed]
  126. Solmunde, E.; Falstie-Jensen, A.M.; Lorenzen, E.L.; Ewertz, M.; Reinertsen, K.V.; Dekkers, O.M.; Cronin-Fenton, D.P. Breast cancer, breast cancer-directed radiation therapy and risk of hypothyroidism: A systematic review and meta-analysis. Breast 2023, 68, 216–224. [Google Scholar] [CrossRef] [PubMed]
  127. Ye, F.; Wen, J.; Yang, A.; Wang, Y.; Li, N.; Yu, P.; Wei, W.; Tang, J. The influence of hormone therapy on secondary diabetes mellitus in breast cancer: A meta-analysis. Clin. Breast Cancer 2022, 22, e48–e58. [Google Scholar] [CrossRef] [PubMed]
  128. Barroso-Sousa, R.; Barry, W.T.; Garrido-Castro, A.C.; Hodi, F.S.; Min, L.; Krop, I.E.; Tolaney, S.M. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: A systematic review and meta-analysis. JAMA Oncol. 2018, 4, 173–182. [Google Scholar] [CrossRef] [PubMed]
  129. Marhold, M.; Udovica, S.; Halstead, A.; Hirdler, M.; Ferner, M.; Wimmer, K.; Bago-Horvath, Z.; Exner, R.; Fitzal, F.; Strasser-Weippl, K.; et al. Emergence of immune-related adverse events correlates with pathological complete response in patients receiving pembrolizumab for early triple-negative breast cancer. Oncoimmunology 2023, 12, 2275846. [Google Scholar] [CrossRef] [PubMed]
  130. Colleoni, M.; Giobbie-Hurder, A. Benefits and adverse effects of endocrine therapy. Ann. Oncol. 2010, 21, vii107–vii111. [Google Scholar] [CrossRef] [PubMed]
  131. Coleman, R.E.; Banks, L.M.; Girgis, S.I.; Vrdoljak, E.; Fox, J.; Cawthorn, S.J.; Patel, A.; Bliss, J.M.; Coombes, R.C.; Kilburn, L.S. Reversal of skeletal effects of endocrine treatments in the Intergroup Exemestane Study. Breast Cancer Res. Treat. 2010, 124, 153–161. [Google Scholar] [CrossRef] [PubMed]
  132. Cuzick, J.; Sestak, I.; Baum, M.; Buzdar, A.; Howell, A.; Dowsett, M.; Forbes, J.F.; on behalf of the ATAC/LATTE investigators. Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 10-year analysis of the ATAC trial. Lancet Oncol. 2010, 11, 1135–1141. [Google Scholar] [CrossRef] [PubMed]
  133. Cohen, A.; Fleischer, J.B.; Johnson, M.K.; Brown, I.N.; Joe, A.K.; Hershman, D.L.; McMahon, D.J.; Silverberg, S.J. Prevention of bone loss after withdrawal of tamoxifen. Endocr. Pract. 2008, 14, 162–167. [Google Scholar] [CrossRef] [PubMed]
  134. Rabaglio, M.; Sun, Z.; Price, K.N.; Castiglione-Gertsch, M.; Hawle, H.; Thurlimann, B.; Mouridsen, H.; Campone, M.; Forbes, J.F.; Paridaens, R.J.; et al. Bone fractures among postmenopausal patients with endocrine-responsive early breast cancer treated with 5 years of letrozole or tamoxifen in the BIG 1-98 trial. Ann. Oncol. 2009, 20, 1489–1498. [Google Scholar] [CrossRef] [PubMed]
  135. Roberto, M.; Barchiesi, G.; Resuli, B.; Verrico, M.; Speranza, I.; Cristofani, L.; Pediconi, F.; Tomao, F.; Botticelli, A.; Santini, D. Sarcopenia in breast cancer patients: A systematic review and meta-analysis. Cancers 2024, 16, 596. [Google Scholar] [CrossRef] [PubMed]
  136. Qian, X.; Li, Z.; Ruan, G.; Tu, C.; Ding, W. Efficacy and toxicity of extended aromatase inhibitors after adjuvant aromatase inhibitors-containing therapy for hormone-receptor-positive breast cancer: A literature-based meta-analysis of randomized trials. Breast Cancer Res. Treat. 2020, 179, 275–285. [Google Scholar] [CrossRef] [PubMed]
  137. Godet, I.; Gilkes, D.M. BRCA1 and BRCA2 mutations and treatment strategies for breast cancer. Integr. Cancer Sci. Ther. 2017, 4, 10.15761. [Google Scholar] [CrossRef] [PubMed]
  138. Blondeaux, E.; Sonnenblick, A.; Agostinetto, E.; Bas, R.; Kim, H.J.; Franzoi, M.A.; Bernstein-Molho, R.; Linn, S.; Kwong, A.; Pogoda, K.; et al. Association between risk-reducing surgeries and survival in young BRCA carriers with breast cancer: An international cohort study. Lancet Oncol. 2025, 26, 759–770. [Google Scholar] [CrossRef] [PubMed]
  139. Tung, N.M.; Garber, J.E. BRCA1/2 testing: Therapeutic implications for breast cancer management. Br. J. Cancer 2018, 119, 141–152. [Google Scholar] [CrossRef] [PubMed]
  140. Huszno, J.; Budryk, M.; Kolosza, Z.; Nowara, E. The influence of BRCA1/BRCA2 mutations on toxicity related to chemotherapy and radiotherapy in early breast cancer patients. Oncology 2013, 85, 278–282. [Google Scholar] [CrossRef] [PubMed]
  141. van Barele, M.; Akdeniz, D.; Heemskerk-Gerritsen, B.A.M.; Genepso; Andrieu, N.; Nogues, C.; Hebon; van Asperen, C.J.; Wevers, M.; Ausems, M.; et al. Contralateral breast cancer risk in patients with breast cancer and a germline-BRCA1/2 pathogenic variant undergoing radiation. J. Natl. Cancer Inst. 2023, 115, 1318–1328. [Google Scholar] [CrossRef] [PubMed]
  142. Furlanetto, J.; Mobus, V.; Schneeweiss, A.; Rhiem, K.; Tesch, H.; Blohmer, J.U.; Lubbe, K.; Untch, M.; Salat, C.; Huober, J.; et al. Germline BRCA1/2 mutations and severe haematological toxicities in patients with breast cancer treated with neoadjuvant chemotherapy. Eur. J. Cancer 2021, 145, 44–52. [Google Scholar] [CrossRef] [PubMed]
  143. Bayraktar, S.; Zhou, J.Z.; Bassett, R.; Gutierrez Barrera, A.M.; Layman, R.M.; Valero, V.; Arun, B. Clinical outcome and toxicity from taxanes in breast cancer patients with BRCA1 and BRCA2 pathogenic germline mutations. Breast J. 2020, 26, 1572–1582. [Google Scholar] [CrossRef] [PubMed]
  144. Drooger, J.C.; Heemskerk-Gerritsen, B.A.M.; Smallenbroek, N.; Epskamp, C.; Seynaeve, C.M.; Jager, A. Toxicity of (neo)adjuvant chemotherapy for BRCA1- and BRCA2-associated breast cancer. Breast Cancer Res. Treat. 2016, 156, 557–566. [Google Scholar] [CrossRef] [PubMed]
  145. Shanley, S.; McReynolds, K.; Ardern-Jones, A.; Ahern, R.; Fernando, I.; Yarnold, J.; Evans, G.; Eccles, D.; Hodgson, S.; Ashley, S.; et al. Late toxicity is not increased in BRCA1/BRCA2 mutation carriers undergoing breast radiotherapy in the United Kingdom. Clin. Cancer Res. 2006, 12, 7025–7032. [Google Scholar] [CrossRef] [PubMed]
  146. Pierce, L.J.; Strawderman, M.; Narod, S.A.; Oliviotto, I.; Eisen, A.; Dawson, L.; Gaffney, D.; Solin, L.J.; Nixon, A.; Garber, J.; et al. Effect of radiotherapy after breast-conserving treatment in women with breast cancer and germline BRCA1/2 mutations. J. Clin. Oncol. 2000, 18, 3360–3369. [Google Scholar] [CrossRef] [PubMed]
  147. Park, H.; Choi, D.H.; Noh, J.M.; Huh, S.J.; Park, W.; Nam, S.J.; Lee, J.E. Acute skin toxicity in Korean breast cancer patients carrying BRCA mutations. Int. J. Radiat. Biol. 2014, 90, 90–94. [Google Scholar] [CrossRef] [PubMed]
  148. Vliek, S.; Hilbers, F.S.; van Werkhoven, E.; Mandjes, I.; Kessels, R.; Kleiterp, S.; Lips, E.H.; Mulder, L.; Kayembe, M.T.; Loo, C.E.; et al. High-dose alkylating chemotherapy in BRCA-altered triple-negative breast cancer: The randomized phase III NeoTN trial. NPJ Breast Cancer 2023, 9, 75. [Google Scholar] [CrossRef] [PubMed]
  149. Friedlaender, A.; Vuilleumier, A.; Viassolo, V.; Ayme, A.; De Talhouet, S.; Combes, J.D.; Peron, J.; Bodmer, A.; Giraud, S.; Buisson, A.; et al. BRCA1/BRCA2 germline mutations and chemotherapy-related hematological toxicity in breast cancer patients. Breast Cancer Res. Treat. 2019, 174, 775–783. [Google Scholar] [CrossRef] [PubMed]
  150. Sajjad, M.; Fradley, M.; Sun, W.; Kim, J.; Zhao, X.; Pal, T.; Ismail-Khan, R. An exploratory study to determine whether BRCA1 and BRCA2 mutation carriers have higher risk of cardiac toxicity. Genes 2017, 8, 59. [Google Scholar] [CrossRef] [PubMed]
  151. Barac, A.; Lynce, F.; Smith, K.L.; Mete, M.; Shara, N.M.; Asch, F.M.; Nardacci, M.P.; Wray, L.; Herbolsheimer, P.; Nunes, R.A.; et al. Cardiac function in BRCA1/2 mutation carriers with history of breast cancer treated with anthracyclines. Breast Cancer Res. Treat. 2016, 155, 285–293. [Google Scholar] [CrossRef] [PubMed]
  152. Pearson, E.J.; Nair, A.; Daoud, Y.; Blum, J.L. The incidence of cardiomyopathy in BRCA1 and BRCA2 mutation carriers after anthracycline-based adjuvant chemotherapy. Breast Cancer Res. Treat. 2017, 162, 59–67. [Google Scholar] [CrossRef] [PubMed]
  153. Demissei, B.G.; Lv, W.; Wilcox, N.S.; Sheline, K.; Smith, A.M.; Sturgeon, K.M.; McDermott-Roe, C.; Musunuru, K.; Lefebvre, B.; Domchek, S.M.; et al. BRCA1/2 mutations and cardiovascular function in breast cancer survivors. Front. Cardiovasc. Med. 2022, 9, 833171. [Google Scholar] [CrossRef] [PubMed]
  154. Lambertini, M.; Goldrat, O.; Toss, A.; Azim, H.A., Jr.; Peccatori, F.A.; Ignatiadis, M.; Del Mastro, L.; Demeestere, I. Fertility and pregnancy issues in BRCA-mutated breast cancer patients. Cancer Treat. Rev. 2017, 59, 61–70. [Google Scholar] [CrossRef] [PubMed]
  155. Valentini, A.; Finch, A.; Lubinski, J.; Byrski, T.; Ghadirian, P.; Kim-Sing, C.; Lynch, H.T.; Ainsworth, P.J.; Neuhausen, S.L.; Greenblatt, E.; et al. Chemotherapy-induced amenorrhea in patients with breast cancer with a BRCA1 or BRCA2 mutation. J. Clin. Oncol. 2013, 31, 3914–3919. [Google Scholar] [CrossRef] [PubMed]
  156. Ringwald, J.; Wochnowski, C.; Bosse, K.; Giel, K.E.; Schaffeler, N.; Zipfel, S.; Teufel, M. Psychological distress, anxiety, and depression of cancer-affected BRCA1/2 mutation carriers: A systematic review. J. Genet. Couns. 2016, 25, 880–891. [Google Scholar] [CrossRef] [PubMed]
  157. Cecco, S.; Puligheddu, S.; Fusaroli, M.; Gerratana, L.; Yan, M.; Zamagni, C.; De Ponti, F.; Raschi, E. Emerging toxicities of antibody-drug conjugates for breast cancer: Clinical prioritization of adverse events from the FDA Adverse Event Reporting System. Target. Oncol. 2024, 19, 435–445. [Google Scholar] [CrossRef] [PubMed]
  158. Ding, H.; Xu, W.; Dai, M.; Li, S.; Xin, W.; Tong, Y.; He, C.; Mi, X.; Zhan, Z.; Fang, L. Hematological toxicity of cyclin-dependent kinase 4/6 inhibitors in patients with breast cancer: A network meta-analysis and pharmacovigilance study. Expert Opin. Drug Saf. 2024, 24, 157–165. [Google Scholar] [CrossRef] [PubMed]
  159. Huang, L.; Pang, D.; Yang, H.; Li, W.; Wang, S.; Cui, S.; Liao, N.; Wang, Y.; Wang, C.; Chang, Y.C.; et al. Neoadjuvant-adjuvant pertuzumab in HER2-positive early breast cancer: Final analysis of the randomized phase III PEONY trial. Nat. Commun. 2024, 15, 2153. [Google Scholar] [CrossRef] [PubMed]
  160. Hurvitz, S.A.; Bardia, A.; Quiroga, V.; Park, Y.H.; Blancas, I.; Alonso-Romero, J.L.; Vasiliev, A.; Adamchuk, H.; Salgado, M.; Yardley, D.A.; et al. Neoadjuvant palbociclib plus either giredestrant or anastrozole in oestrogen receptor-positive, HER2-negative, early breast cancer (coopERA Breast Cancer): An open-label, randomised, controlled, phase 2 study. Lancet Oncol. 2023, 24, 1029–1041. [Google Scholar] [CrossRef] [PubMed]
  161. Connors, C.; Valente, S.A.; ElSherif, A.; Escobar, P.; Chichura, A.; Kopicky, L.; Roesch, E.; Ritner, J.; McIntire, P.; Wu, Y.; et al. Real-world outcomes with the KEYNOTE-522 regimen in early-stage triple-negative breast cancer. Ann. Surg. Oncol. 2025, 32, 912–921. [Google Scholar] [CrossRef] [PubMed]
  162. Segura, T.; Medrano, I.H.; Collazo, S.; Maté, C.; Sguera, C.; Del Rio-Bermudez, C.; Casero, H.; Salcedo, I.; García-García, J.; Alcahut-Rodríguez, C.; et al. Symptoms timeline and outcomes in amyotrophic lateral sclerosis using artificial intelligence. Sci. Rep. 2023, 13, 702. [Google Scholar] [CrossRef] [PubMed]
  163. Calleja-Panero, J.L.; Esteban Mur, R.; Jarque, I.; Romero-Gómez, M.; Group, S.R.; García Labrador, L.; González Calvo, J. Chronic liver disease-associated severe thrombocytopenia in Spain: Results from a retrospective study using machine learning and natural language processing. Gastroenterol. Hepatol. 2024, 47, 236–245. [Google Scholar] [CrossRef] [PubMed]
  164. González-Juanatey, C.; Anguita-Sánchez, M.; Barrios, V.; Núñez-Gil, I.; Gómez-Doblas, J.J.; García-Moll, X.; Lafuente-Gormaz, C.; Rollán-Gómez, M.J.; Peral-Disdier, V.; Martínez-Dolz, L.; et al. Impact of advanced age on the incidence of major adverse cardiovascular events in patients with type 2 diabetes mellitus and stable coronary artery disease in a real-world setting in spain. J. Clin. Med. 2023, 12, 5218. [Google Scholar] [CrossRef] [PubMed]
  165. Abrisqueta-Costa, P.; García-Marco, J.A.; Gutiérrez, A.; Hernández-Rivas, J.; Andreu-Lapiedra, R.; Arguello-Tomas, M.; Leiva-Farré, C.; López-Roda, M.D.; Callejo-Mellén, Á.; Álvarez-García, E.; et al. Real-world evidence on adverse events and healthcare resource utilization in patients with chronic lymphocytic leukaemia in Spain using natural language processing: The SRealCLL study. Cancers 2024, 16, 4004. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Literature search and review approach.
Figure 1. Literature search and review approach.
Cancers 17 02506 g001
Figure 2. Schematic representation of the possible early and later-onset adverse events related to treatment in patients with eBC and gBRCA mutation. AE: adverse event, eBC: early breast cancer, gBRCA: germline BRCA, GI: gastrointestinal. Red dot represents tumor location.
Figure 2. Schematic representation of the possible early and later-onset adverse events related to treatment in patients with eBC and gBRCA mutation. AE: adverse event, eBC: early breast cancer, gBRCA: germline BRCA, GI: gastrointestinal. Red dot represents tumor location.
Cancers 17 02506 g002
Table 1. Time period considerations for BC adverse events.
Table 1. Time period considerations for BC adverse events.
Long-Term Adverse EventsTime Span
Chest wall and breast adverse events
PMPS7–12 years after BC diagnosis [47]
Lymphedema10 years after BC diagnosis [48]
After BC primary treatment [49]
Skin and soft tissue affectionAfter BC primary treatment [50]
Cardiologic
Heart failure>6 months after BC diagnosis [51], 5–8 years after BC diagnosis [45], 8 years after BC diagnosis [45], 10–13 years after BC diagnosis [46], and 11 years after BC diagnosis [44]
Arrhythmia, acute ischemic heart disease, ischemic stroke, or transient ischemic attack>6 months after BC diagnosis [51]
Neurotoxicity
CIPN>3 weeks after BC treatment [52], after the first administration of chemotherapy [53]
Cognitive dysfunctionDuring chemotherapy treatment, after cessation of treatment, >6 months post-treatment cessation, >1 year post-treatment cessation, >3 years post-treatment cessation [54]
Psychological alterations
AnxietyDifferent timepoints ranging from 1.8 to 21 years after BC diagnosis [55]
DepressionDifferent timepoints ranging from 1.8 to 21 years after BC diagnosis [55]
Fear of death>1 year after BC diagnosis [56]
Women’s health
FatigueAfter BC primary treatment, >5 years after BC diagnosis [43]
Hormonal alterationsAfter BC primary treatment [40], >8 years after BC diagnosis [57]
Sexual disorders>3 years after BC diagnosis [58]
Reduced fertility>2 years after BC diagnosis [59]
GI symptoms
DiarrheaAfter BC primary treatment [30]
Endocrine symptoms
Hypothyroidism>3 years after BC diagnosis [35]
Osteomuscular adverse events
OsteoporosisAfter BC primary treatment [60]
BC: breast cancer, CIPN: chemotherapy-induced peripheral neuropathy, GI: gastrointestinal, PMPS: post-mastectomy pain syndrome. The gray background indicates different categories of adverse events.
Table 2. Description of key adverse events in patients with BC: prevalence, associated risk factors, and management.
Table 2. Description of key adverse events in patients with BC: prevalence, associated risk factors, and management.
Type of Adverse EventPrevalenceRisk FactorsManagement
Chest wall and breast
PMPS [61,62]28.2–65%Postoperative pain, younger age, high BMI, axillary radiation, and axillary lymph node dissectionAnalgesics, surgical interventions, acupuncture, or hypnosis
Lymphedema [28,48,49]27–40%ALND, mastectomy, adjuvant therapies, high BMIPhysiotherapy
Skin and soft tissue affections [31,63]Up to 43%RadiotherapyPhysiotherapy, anti-inflammatory drugs
Cardiologic
Cardiac toxicity [64,65,66]1–51.5%Age, history of heart disease, maximum cumulative dose of anthracyclines, endocrine therapy, radiation to the left breastPrevention: use of alternative chemotherapeutic agents, cardioprotective agents
Treatment: same guidelines for heart failure for other causes
Neurologic
CIPN [29,67]23–80%Age, taxane treatment, baseline neuropathy, smoking, diabetesDuloxetine (level I evidence), venlafaxine, pregabalin, amitriptyline, and tramadol. In selected patients, acupuncture can also be an option
Cognitive dysfunction [54,68]28–33%Age, chemotherapy, endocrine therapyCognitive rehabilitation, physical exercise, and low evidence for pharmacological treatment
Psychological alterations
Depression [32,69]9.4–66.1%Younger age at diagnosis, history of psychological disorder, substance abuse, poor social support, and lower socioeconomic status.Psychological/psychiatric support and cognitive–behavioral therapy
Anxiety [32,70]17.9–33.3%Younger age, physical symptoms, chemotherapy, poor social and cognitive functioning, and communication problems with healthcare providersPsychological/psychiatric support and cognitive–behavioral therapy
Fear of death [56,71]71%Uncertain future, young age, breast-conserving surgeryPsychological/psychiatric support and cognitive–behavioral therapy
Women’s health
Fatigue [33,72]30–50%Relation with long-term adverse events such as cardiac, menopause, or psychologicalLifestyle modifications, such as regular exercise, adequate sleep, stress reduction techniques, and treatment of other comorbidities or late adverse events
Hormonal alterations [34,40]33–48.7%Endocrine therapy, chemotherapyGabapentin or SSRIs/SNRIs for hot flashes. Physical exercise, cognitive-behavioral therapy, and mindfulness
Sexual disorders [58,73]90%Body image alterations, endocrine therapy, and psychological impairment, such as depression or anxietyThe treatment of associated factors (vaginal dryness, dyspareunia, depression, or anxiety, etc.)
Sexual counseling
Reduced fertility [59,74]60%Gonadotoxic chemotherapyOncofertility counseling
GI symptoms
Diarrhea [30]29.4–83%Treatment with abemaciclib or immunotherapyDose reduction or interruption according to severity. Loperamide for abemaciclib toxicity and corticosteroids for immunotherapy according to severity
Nausea [30]23.0–77%Treatment with abemaciclib, olaparib, or ribociclibIntegration of strategies to prevent or lessen its impact
Vomiting [30]40%Treatment with olaparibIntegration of strategies to prevent or lessen its impact
Endocrine symptoms
Hypothyroidism [35]5–6%Radiotherapy treatmentHormonal supplementation
ALND: axillary lymph node dissection, BMI: body mass index, CIPN: chemotherapy-induced peripheral neuropathy, GI: gastrointestinal, PMPS: post-mastectomy pain syndrome, SSRIs/SNRIs: selective serotonin reuptake inhibitors/serotonin–norepinephrine reuptake inhibitors. The gray background indicates different categories of adverse events.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Obispo, B.; Bailleux, C.; Cantos, B.; Zamora, P.; Jhawar, S.R.; Varghese, J.; Cabal-Hierro, L.; Luz, P.; Berrocal-Almanza, L.; Xu, X. Long-Term Adverse Events Following Early Breast Cancer Treatment with a Focus on the BRCA-Mutated Population. Cancers 2025, 17, 2506. https://doi.org/10.3390/cancers17152506

AMA Style

Obispo B, Bailleux C, Cantos B, Zamora P, Jhawar SR, Varghese J, Cabal-Hierro L, Luz P, Berrocal-Almanza L, Xu X. Long-Term Adverse Events Following Early Breast Cancer Treatment with a Focus on the BRCA-Mutated Population. Cancers. 2025; 17(15):2506. https://doi.org/10.3390/cancers17152506

Chicago/Turabian Style

Obispo, Berta, Caroline Bailleux, Blanca Cantos, Pilar Zamora, Sachin R. Jhawar, Jajini Varghese, Lucia Cabal-Hierro, Paulo Luz, Luis Berrocal-Almanza, and Xiaoqing Xu. 2025. "Long-Term Adverse Events Following Early Breast Cancer Treatment with a Focus on the BRCA-Mutated Population" Cancers 17, no. 15: 2506. https://doi.org/10.3390/cancers17152506

APA Style

Obispo, B., Bailleux, C., Cantos, B., Zamora, P., Jhawar, S. R., Varghese, J., Cabal-Hierro, L., Luz, P., Berrocal-Almanza, L., & Xu, X. (2025). Long-Term Adverse Events Following Early Breast Cancer Treatment with a Focus on the BRCA-Mutated Population. Cancers, 17(15), 2506. https://doi.org/10.3390/cancers17152506

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop