Abstract
Background
Cancer cases diagnosed each year are increasing, mainly because the population is ageing and, in part, due to early detection. This implies that there are more and more persons that receive medical anticancer therapies and that are interested in maintaining their quality of life. Many oncological treatments, including chemotherapy, immunotherapy, surgery, and radiotherapy, and combined therapy are associated with cutaneous toxicity and long-term side effects to different tissues and organs. This is particularly relevant when new therapies are used since these may cause new and unexpected side effects that may be short-lived but, in some cases, may become chronic or permanent. Patients often seek advice with their oncologists on what can be done and what cannot be done. Notably, many of the cutaneous side effects can be prevented or reduced by adequate interventions.
Summary
The aim of this review is to highlight how oncological patients may benefit from a closer collaboration between specialists in different branches. We will focus on women with breast cancer since we think that they may derive a special benefit from this collaboration, but we will analyse other cancers in future papers.
Key Messages
The working group was created to help the medical doctor in the prevention and management of all the adverse effects of the oncological treatments, supporting patients in this phase of their life, including nutritional assessment and dietary support.
Keywords: Breast cancer, Aesthetic medicine, Toxic side effects, Multidisciplinary team
Introduction
Annual incidence of all tumours can be estimated around 190 cases/100,000 with a cure rate of approximately 65%. Breast cancer is the most common cancer in women. About 287,850 new cases of invasive breast cancer will be diagnosed every year in the USA [1]. Thus, the number of cancer survivors has reached considerable numbers, accounting for 9.8 millions and 12 millions individuals in the USA and in Europe, respectively [2]. This marked increase in survival is mainly due to earlier diagnosis and better treatments, but many survivors live with acute and chronic consequences of those treatments, including cardiotoxicity, secondary tumours, and infertility [3, 4]. Moreover, the psychological implications of diagnosis and treatments on patients and their families should not be ignored. Adverse effects (AEs) may include any organ or apparatus, thus more specialists are needed to offer uniform and shared diagnostic and therapeutic care paths [5]. The prevention and management of these side effects represent a crucial point to obtain optimal care that should include every feature of the patient's health and not only the treatment of cancer [6]. In addition to being frequently disabling, skin and annexa damages including alopecia, skin rashes, or skin alterations are certainly the most visible and assessable by the patient [7], producing an important psychological impact, alteration of body-image, cultural and gender identity, sexuality, and health-mind relationship [8, 9]. For these reasons, toxic side effects involving the skin and cutaneous adnexa, given by different pathogenetic mechanisms depending on the drug, are considered more and more relevant [10]. On the other hand, it is widely accepted that the skin signals reveal what happens within the body: the skin can be considered “a great sentinel” [11] and its monitoring during anticancer treatment is therefore very important [12]. The frequency of dermatological AEs depends on the agent administered and less frequently on tumour type, 18–72% of patients show dermatological AEs during cytotoxic chemotherapy, 75–90% [13] during target therapy and >30% of patients in immunotherapies [14]. Radiotherapy causes a variety of side effects, commonly referred to as radiodermatitis that can range from mild erythema to severe confluent moist desquamation. All patients receiving radiotherapy are at risk of developing these reactions [15]. Therefore, early assessment and management promote patient comfort and enhance quality of life (QoL). Combining radiotherapy and systemic therapy may exacerbate cutaneous reactions, resulting in severe xerosis, inflammation, skin thinning, and necrosis of the upper dermis and epidermis. Side effects tend to appear early on, and although mostly mild at onset, can become severe and significantly impair QoL [6]. These reactions may lead to dose reduction or discontinuation of therapy, which could be detrimental to the outcome of treatment. The correct diagnosis and management of dermatological AEs are crucial, both to optimize therapeutic success rates and to reduce the patient's suffering [16]. We are therefore witnessing the evolution of the concept of “cure” into “taking care,” at the basis of the new holistic vision of the patient, which focuses not only on the disease burden, but also on additional therapeutic goals, as (QoL) [17], a previously ignored factor, but which now seems to be crucial for patient's prognosis [7, 18]. The appearance of severe cutaneous AEs can lead to a decrease in drug dosage, and in some cases to discontinuation of therapy [19]. The heterogeneous clinical pictures that can arise during treatment are generally linked to the mechanism of action of the agents, but since drugs are often used simultaneously in polychemotherapy regimens, cumulative toxicities may appear [20, 21]. It is therefore understandable that cutaneous AEs are frequently unpredictable since they can also be worsened by the interactions with supportive treatments including antibiotics, antiemetics, analgesics, or antidepressant drugs [22]. In this complex scenario, a multidisciplinary team (MDT) plays a crucial role in providing optimal care for breast cancer survivors [23, 24]. Professionals, as the oncologist, the surgeon, the radiotherapist, are supported by the nutritionist, the endocrinologist, the gynaecologist/andrologist, the pain therapist, the aesthetic specialist, the dermatologist, and other figures, previously erroneously considered of secondary importance. Here, we propose the onset of a MDT in which all these health care providers cooperate to “take care” of their patients. To simplify, the specific case of patients affected by breast cancer will be used as an example.
Dermatological Toxicity
Concerning the toxic side effects of chemotherapy on the skin and cutaneous annexa, they may comprise hair thinning and hair loss and alterations of oral and any mucosae. Here, we will focus on drugs that are commonly used in the treatment of breast cancer. The most evident example is alopecia (hair loss) which is inevitably caused by anthracyclines and by taxanes [25]. Docetaxel may also damage nails, and this may be a cause of relevant stress for some patients [26]. Fluorouracil and capecitabine may cause reddening and tenderness of the palms and of the soles (hand-foot-syndrome), as also observed with liposomal formulations of doxorubicin [27]. Hyperpigmentation may also be associated with the hand-foot-syndrome, while flagellated dermatitis, a fairly specific form of hyperpigmentation, is reported only with bleomycin. It is important to note that hormones may also be associated with cutaneous damage which is due to the decrease in oestrogen levels caused by luteinising hormone-releasing hormone analogues and by aromatase inhibitors. Xerosis is very common during almost all oncological treatments [28], so the main goal of this phase is to maintain physiological skin homeostasis in the best possible condition. Therefore, the correct preventive measures should be adapted according to the planned oncological regimen and the cutaneous or systemic comorbidities of the patient.
Nails can also be damaged by anticancer agents, and for this reason, preventive measures may be useful. General preventive recommendations should be suggested to all oncological patients, while specific protective measures may be proposed according to the different protocols. The preventive approach can be supported by professional figures to perform, for example, correct nail management and to reduce elements that may predispose to damage [29, 30]. Concerning the effects on hair follicles, there are several types of treatment-induced alopecia, according to the different oncological protocols (classic chemotherapy, target therapies, hormonal therapy, immunotherapy, and radiotherapy) [31]. Notably, alopecia in women almost invariably has a strong emotional impact and represents one of the most common reasons of chemotherapy refusal or scarce compliance [12]. General preventive measures have been suggested such as scalp cooling or local vasoconstrictors, the rationale is the decrease in drug delivered to the follicular unit. Scalp cooling, to date, is the only Food and Drug Administration approved useful intervention able to prevent classic chemotherapy-induced alopecia with an effectiveness of 50–80% [31, 32, 33, 34]. This preventive measure has unsatisfactory results in the other type of chemotherapy-induced alopecia.
Besides the toxic effects on the skin tissue, even mucositis represents one of the main AEs of chemotherapies. Mucositis may be caused by different drugs, and it may be different according to the anticancer treatment administered, as, for example, taxanes, anthracyclines, irinotecan [35]. It is important to underline that the mucosae are not just limited to the oral cavity, but they line the entire gastrointestinal tract, the urogenital tract, and the eyes; therefore, these areas must also be considered [36]. The prevention of oral mucositis is very relevant since a severe involvement can interfere with the patient's ability to eat and more importantly to drink and may contribute to the worsening of the general conditions and therefore of the prognosis [37]. Recent studies focused the attention to the oral and intestinal microbiota that could be relevant to mucositis development and treatment [38], but no clear high-risk pattern has been identified and no single probiotic formulation has emerged from human clinical trials for strong recommendation. Noteworthy, mucositis caused by chemotherapy are due to damage to proliferating cells in mucosal crypts: bacteria or fungal infection of the damaged mucosa can certainly occur but starting antifungal treatment for any oral mucositis is not advisable [39].
Aesthetic Medicine to Support Oncology
The objective of good management of side effects during chemotherapy is to correctly diagnose and treat any possible cutaneous AEs to avoid drug dose reduction or discontinuations, as well as to maintain the patient's QoL [40]. Xerosis, itching, different kinds of rash, bullous lesions, acral erythema, photoreactions, cutaneous and mucosal pigmentations, nail alterations, mucositis, and alopecia are just some of the clinical manifestations during therapy [41]. Some new forms are now emerging during immunostimulating therapies: vitiligo, lichenoid reaction, bullous dermatoses, maculopapular rashes, and life-threatening severe cutaneous adverse reactions [42]. Several publications provide recommendations or algorithms for pharmaceutical treatment, but skincare itself remains unclear and not sufficiently detailed. What is certain is that a close and timely collaboration between the different specialists would ensure that the best decisions are made for patients, with the aim of managing adverse dermatological reactions from the beginning of treatment to ensure continuation of therapies without suspensions, changes, and interruptions of treatments [6, 23]. Therefore, in a multidimensional and multidisciplinary evaluation, aesthetic medicine, which has always been attentive to “skin health,” plays an important role for prevention and cure [18, 43, 44]. In particular, the preventive role of aesthetic medicine focuses on maintaining the integrity of the skin during oncological treatments with basic dermo-cosmetic therapy: this must include cosmetics for correct cleansing, care and photoprotection [45]. The role of the treatment of skin damage is to monitor, evaluate and treat acute and late toxicity from mild skin discomfort to medium-grade toxicity with advanced dermo-cosmetic therapies [46]. A constant dermo-cosmetological vigilance can avoid annoying skin sequelae; therefore, a greater awareness of the oncologists regarding the need for the precious support of specialist dermatologists, aesthetic doctors, and cosmetologists even before the start of oncological therapies becomes fundamental, with a “skin care” protocol that prepares the skin for the extreme stress that awaits it [47, 48]. Indeed, skin compatibility is an essential requirement of any cosmetic prescribed to cancer patients whose skin is often more delicate and may have been weakened by previous lengthy treatments or may in any case be suffering from diseases already present before the start of oncological therapies [49]. Before recommending a cosmetic product or treatment, a careful preliminary medical history must be carried out as it is necessary to respect the skin's hydrolipidic mantle and its sensitivity in the daily routine [6, 46]. It may also be useful to carry out an “aesthetic skin check-up,” characterized by a series of clinical and instrumental evaluations, to help the aesthetic doctor in prescribing the best cosmetic support therapy during therapies and post-treatment [50]. An “aesthetic skin check-up” as recommended in aesthetic medicine aims at clinically evaluating the skin using tools that allow the recording and monitoring of physiological parameters over time, including the use of equipment before, during and after oncological treatments [51]. The parameters that can be evaluated are, among others, corneometry that is an estimate of the water content of the stratum corneum; sebometry which is an accurate and reproducible measurement of the sebum level of the skin surface and Ph. Also measurement of skin elasticity based on the method of suction/elongation and subsequent skin release and measurement of melanin and haemoglobin with instruments, allow the evaluation, through reflectance spectrophotometry, of the two main components responsible for skin colour, melanin and haemoglobin (erythema) [51, 52, 53]. Therefore, aesthetic medicine is crucial for the management of cancer patient, in order to evaluate the skin parameters and set the “tailor-made” support therapy to guarantee a condition of equilibrium and to monitor the variations in an objective way [54].
Nutritional Assessment
Until recently, nutritional care was not considered a critical issue in patients immediately after cancer diagnosis. Indeed, some researchers believed that nutrition could have stimulatory effects on tumour growth [55, 56]. Some of these beliefs were based, in part, on the results of in vitro or in vivo animal research [57]. Recently, DeWys et al. [58] have shown that without adequate nutrition, malnutrition, and weight loss in people with cancer could represent a major cause of mortality. This pioneer study paved the way for new therapeutic approaches to cancer patients, also based on nutritional support. In recent years, the concept that an increase in body weight in breast cancer patients (BCP) represents a risk factor associated with comorbidities clearly emerged [59]. Therefore, it would be appropriate for BCP care that nutrition experts encourage weight management during all phases of cancer care [60, 61, 62, 63]. In women with breast cancer (especially those in the pre-menopausal stage), weight gain usually affects them in the early stages (grade I or II) of the disease [64, 65]. In addition, changes in body composition can negatively affect the nutritional status of patients, increasing the risk of tumour recurrence and death [66, 67, 68]. It should also be kept in mind that the nutritional status of BCP changes during treatments [69] for different reasons, as tumour size, digestive symptoms, increased nutritional needs, and erroneous belief of what a healthy diet represents [70, 71]. Antineoplastic treatment induces a decrease in bone mineral density and an increase in body weight, mainly due to an increase in fat mass and a decrease in muscle mass and strength, also known as sarcopenic obesity [65, 71, 72]. Thus, nutritional therapy could improve and prevent adverse changes in the body composition of BCPs in the early stages of the disease [73, 74, 75, 76, 77]. From the studies cited so far, and according to the clinical practice guidelines for overweight and obesity and with the recommendations for cancer survivors, the following nutritional interventions for BCPs should be addressed: (1) limit weight loss to between 5% and 10% of initial body weight by calorie restriction based on age and initial body mass index, (2) increase diet quality with nutrient-rich foods, and (3) reduce simple sugars and added fats [78, 79, 80, 81]. In addition, even normal-weight BCP patients should still receive appropriate nutritional therapies to prevent weight gain, nutrient deficiencies, and adverse health consequences [65, 82, 83]. Nutritional therapy for weight loss aims at achieving a negative energy balance in an individual's total energy requirements by increasing physical activity and reducing dietary energy intake. Calorie restriction will depend on the patient's nutritional status, but a usual therapy may consider a reduction of 500–1,000 kcal/day. Most behavioural programs are designed to achieve a loss of 0.5–1 kg/week, encouraging patients to be engaged in a healthy nutritional behaviour [79, 84, 85]. Since specific nutritional guidelines for BCPs are still lacking, the American Cancer Society suggests a nutritional approach based on guidelines published in 2000 by the National Institutes of Health in the USA [85]. These guidelines provide more specific nutrient recommendations (fat intake, carbohydrates, fibre, cholesterol, etc.) for BCP, and their strength lies in the consideration of caloric restriction to promote healthy weight loss [86, 87], providing proper macronutrient distribution to customize the patient's eating plan. For body weight control in BCP, the American Cancer Society recommends a healthy low-fat diet (<30% from total energy) with a strong fruit and vegetable base (5–9 servings/day) and encourages physical activity [88]. Within this context, one multicentre study reported that only 30–48% of people newly diagnosed with BCP made positive changes in their eating habits. These changes included increasing consumption of fruits and vegetables and reducing consumption of fats and simple sugars. Nevertheless, nearly half of newly diagnosed BCP patients do not introduce these positive changes into their lifestyle [89]. Furthermore, an evidence-based nutrition intervention program proposed by the Oncology Nursing Society and supported by the National Cancer Institute recommends individualized nutrition therapy as the most effective measure for cancer patients. The nutritionist in such therapy should work together with the patient, his or her family, and a multidisciplinary oncology team to provide appropriate nutritional assessment and follow-up of the intervention [86, 90]. The evidence-based ESPEN guidelines were also drafted to translate the best current evidence and expert opinion into recommendations for MDTs responsible for identifying, preventing, and treating the reversible elements of malnutrition in adult cancer patients [91].
Nutraceuticals and Food Supplements
Several recently published papers show that antioxidants can play an important role in preventing breast cancer and can positively affect the therapeutic interventions [92]. Melatonin [93, 94, 95, 96], resveratrol [97, 98, 99, 100], curcumin [101, 102, 103, 104], vitamins C [105, 106, 107, 108], D [109, 110, 111, 112], and E [113, 114, 115] and even synthetic antioxidants such as N-acetyl cysteine [116, 117, 118] have been studied with the aim of evaluating their real effectiveness. While we have a great deal of in vitro evidence, clinical trials are still lacking, and often not set up in a consistent way. It is essential to appropriately stratify patients in relationship to the tumour isotype, and consider the chemical interactions between pro-oxidants and antioxidants (“redoxidomics”), to define the optimal dosage of these molecules for a personalized medicine [92]. Another issue concerns the study of interactions between different types of antioxidants according to their origin. Noteworthy, exogenous antioxidants carry out their actions in aqueous phase (e.g., vitamin C) or in lipid phase (e.g., vitamin E) and are in redox balance between them and with endogenous antioxidants (e.g., glutathione) in preventing or repairing oxidative damage on biochemicals or cell components (e.g., plasma membranes) [119]. Many studies have also highlighted the “multitarget” role of some antioxidants especially with polyphenolic structure. Molecules as quercetin, for example, in addition to the ability of reactive oxygen species scavenging, possess anti-inflammatory, gene modulation, and anti-proliferative properties [119]. These molecules, found in different foods, behave as real drugs and are often defined as “nutraceuticals.” The term “nutraceutical” combines two words: “nutrient,” a food component, and “pharmaceutical,” a drug. The term was coined in 1989 by Stephen De Felice, chairman of the Foundation for Innovation in Medicine. Unfortunately, while studying the antioxidant effects in vitro is very simple, unravelling the potential anti-proliferative properties towards breast cancer or the dose-response relationships is much more complex. Poschner and collaborators [98] investigated the role of resveratrol and other polyphenols in inhibiting oestrogen metabolism in human mammal cellular lines (ERα+ and ERα‒). Experimental results highlight that only high concentrations of polyphenols can inhibit the proliferation of cancer cells while low concentrations could even stimulate cell growth [98]. A similar behaviour was demonstrated by Patisaul and Jefferson [120] for phytoestrogens, as the genistein, that only at high doses is able to inhibit the proliferation of cells ERα + and ERα‒ while, on the other hand, promoting proliferation at low doses. This evidence, related to the dual action of resveratrol and genistein, appears to be particularly worrying in the light of the current market trend that proposes phytoestrogen supplements as isoflavones, for a preventive and protective role towards breast cancers. These suggestions were not (yet) supported by strong clinical evidence. For this reason, foods such as soy, rich in isoflavones, should be cautiously consumed by women with a high risk of developing breast cancer as they still lack studies on their biological actions at cellular level [121]. These results show how dietary supplementation with polyphenols in the prevention and treatment of breast cancer is currently controversial, once again due to the lack of clinical trials aimed at mainly defining the posology. In vitro research, does not allow to study pharmacokinetics and bioavailability of antitumor nutraceuticals, being unable to dissect the interactions with the microbiota and with the immune system as well. The microbiota, able to produce immunomodulatory and antineoplastic molecules [122], could convert nutraceuticals (prebiotics) into metabolites with anticancer properties. Some vitamin deficiencies can worsen the clinical history of patients with breast cancer. A meta-analysis performed by Harris and collaborators [105] highlighted that a post-diagnosis vitamin C supplementation could be associated with a decreased mortality in patients with breast cancer. Even the presence of adequate quantities of vitamin C in the normal diet of patients before the diagnosis seems to be associated with a decrease in total mortality associated with cancer. Another meta-analysis performed by Hossain and Coworkers [112], demonstrated that vitamin D dietary deficiencies can promote the onset of breast cancer while a supplementation exerts a protective effect. Nevertheless, besides these encouraging results, the evidence to support vitamin supplementations for preventing breast cancer is still limited. Miotti and Gnagnarella [123] and Mokbel [124] highlight that, despite several biological mechanisms have been clarified, there is still no clear evidence concerning the role of vitamin supplementation in decreasing the risk of mammary cancer onset. This limitation could be overcome through well-structured clinical trials. Similar conclusions were reached by Avila-Galvez and co-authors about the preventive role of polyphenols [125] and Gonzalez-Gonzalez et al. [93] about melatonin. Some components of the Mediterranean diet, as resveratrol, retinoids, epigallocatechin 3-gallate, and omega-3 PUFAs could represent innovative anticancer candidates. However, few data are available on the potential role of nutraceuticals and food supplements during anticancer therapy or as potential chemotherapeutic agents [126].
Conclusion
Patients with cancer undergo a variety of oncological treatments, including surgery, radiation, traditional chemotherapy, targeted immune checkpoint inhibitor, and endocrine therapies. Many of the pathways targeted by anticancer therapies are also important for skin, hair, mucosal, and nail homeostasis; therefore, dermatological toxicities are among the most frequently encountered AEs. When severe, these conditions may result in interruption of cancer therapy, and late or persistent complications can arise. According to QoL studies, women are affected to a greater degree than men by the toxic dermatological effects of cancer therapy [4, 7, 9, 10], and these toxicities can affect a woman's self-image, cultural identity, femininity, sexuality, and mental health [127, 128, 129]. Recognition and management of these late effects, focusing on how and when to act, should be better dissected and understood to improve the QoL of the patients. In the present review, we focused on the role of the oncologists, the aesthetic medicine specialists, the dermatologists, radiotherapists, and the nutritionists in supporting the BCP. A MDT assumes a crucial role in the care of and for breast cancer survivors (online suppl. Fig. 1; see www.karger.com/doi/10.1159/000528280 for all online suppl. material). Indeed, “living well means living longer,” so it must be done to improve the prognosis of the cancer patient, allowing us to better investigate the role of the involved specialists in supporting the recovery of the affected patient.
Conflict of Interest Statement
The authors have no conflicts of interest to declare.
Funding Sources
This research was funded by “Fondo di Ateneo per la Ricerca 2021 (Margherita Maioli).”
Author Contributions
All authors were involved in project conception and in writing the article. All authors have read and agreed to the published version of the manuscript.
Supplementary Material
Supplementary data
Funding Statement
This research was funded by “Fondo di Ateneo per la Ricerca 2021 (Margherita Maioli).”
References
- 1.American Cancer Society What are the key statistics about breast cancer? 2014. Cancer.org .
- 2.Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin. 2020;70((1)):7–30. doi: 10.3322/caac.21590. [DOI] [PubMed] [Google Scholar]
- 3.Ferreira MN, Ramseier JY, Leventhal JS. Dermatologic conditions in women receiving systemic cancer therapy. Int J Womens Dermatol. 2019;5((5)):285–307. doi: 10.1016/j.ijwd.2019.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Gandhi M, Oishi K, Zubal B, Lacouture ME. Unanticipated toxicities from anticancer therapies survivors' perspectives. Support Care Cancer. 2010;18((11)):1461–1468. doi: 10.1007/s00520-009-0769-1. [DOI] [PubMed] [Google Scholar]
- 5.Berardi R, Morgese F, Rinaldi S, Torniai M, Mentrasti G, Scortichini L. Benefits and limitations of a multidisciplinary approach in cancer patient management. Cancer Manag Res. 2020;12:9363–9374. doi: 10.2147/CMAR.S220976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bensadoun RJ, Humbert P, Krutman J, Luger T, Triller R, Rougier A, et al. Daily baseline skin care in the prevention and supportive care of skin toxicity in oncology patients recommendations from a multinational expert panel. Cancer Manag Res. 2013;5:401–408. doi: 10.2147/CMAR.S52256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hackbarth M, Haas N, Fotopoulou C, Lichtenegger W, Sehouli J. Chemotherapy-induced dermatological toxicity frequencies and impact on quality of life in women's cancers. Results of a prospective study. Support Care Cancer. 2008;16((3)):267–273. doi: 10.1007/s00520-007-0318-8. [DOI] [PubMed] [Google Scholar]
- 8.Lee J, Lim J, Park JS, Kim M, Kim TY, Kim TM, et al. The impact of skin problems on the quality of life in patients treated with anticancer agents a cross-sectional study. Cancer Res Treat. 2018;50((4)):1186–1193. doi: 10.4143/crt.2017.435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ra HS, Shin SJ, Kim JH, Lim H, Cho BC, Roh MR. The impact of dermatological toxicities of anti-cancer therapy on the dermatological quality of life of cancer patients. J Eur Acad Dermatol Venereol. 2013;27((1)):e53–e59. doi: 10.1111/j.1468-3083.2012.04466.x. [DOI] [PubMed] [Google Scholar]
- 10.Bolomsky A, Schreder M, Meisner T, Hose D, Ludwig H, Pfeifer S. Immunomodulatory drugs thalidomide and lenalidomide affect osteoblast differentiation of human bone marrow stromal cells invitro. Exp Hematol. 2014;42((7)):516–525. doi: 10.1016/j.exphem.2014.03.005. [DOI] [PubMed] [Google Scholar]
- 11.Carlesimo M MFRA, Caro G, Fortuna MC, Muscianese M. Late effects in oncotherapy skin the great sentinel. Arch of Clinical Exper Dermatol. 2020;2:p 3–4. [Google Scholar]
- 12.Frick MA, Vachani CC, Hampshire MK, Bach C, Arnold-Korzeniowski K, Metz JM. Patient-reported survivorship care practices and late effects after treatment of hodgkin and non-hodgkin lymphoma. JCO Clin Cancer Inform. 2018;2:1–10. doi: 10.1200/CCI.18.00015. [DOI] [PubMed] [Google Scholar]
- 13.Rosen AC, Case EC, Dusza SW, Balagula Y, Gordon J, West DP. Impact of dermatologic adverse events on quality of life in 283 cancer patients a questionnaire study in a dermatology referral clinic. Am J Clin Dermatol. 2013;14((4)):327–333. doi: 10.1007/s40257-013-0021-0. [DOI] [PubMed] [Google Scholar]
- 14.Haanen JBAG, Obeid M, Spain L, Carbonnel F, Wang Y, Robert C, et al. Management of toxicities from immunotherapy ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33((12)):1217–1238. doi: 10.1016/j.annonc.2022.10.001. [DOI] [PubMed] [Google Scholar]
- 15.Lynch TJ, Kim ES, Eaby B, Garey J, West DP, Lacouture ME. Epidermal growth factor receptor inhibitor–associated cutaneous toxicities an evolving paradigm in clinical management. Oncologist. 2007;12((5)):610–621. doi: 10.1634/theoncologist.12-5-610. [DOI] [PubMed] [Google Scholar]
- 16.Tattersall IW, Leventhal JS. Cutaneous toxicities of immune checkpoint inhibitors the role of the dermatologist. Yale J Biol Med. 2020;93((1)):123–132. [PMC free article] [PubMed] [Google Scholar]
- 17.van Leeuwen M, Husson O, Alberti P, Arraras JI, Chinot OL, Costantini A, et al. Understanding the quality of life (QOL) issues in survivors of cancer towards the development of an EORTC QOL cancer survivorship questionnaire. Health Qual Life Outcomes. 2018;16((1)):114. doi: 10.1186/s12955-018-0920-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jourdan M, Madfes DC, Lima E, Tian Y, Seité S. Skin care management for medical and aesthetic procedures to prevent scarring. Clin Cosmet Investig Dermatol. 2019;12:799–804. doi: 10.2147/CCID.S218134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Grávalos C, Sanmartin O, Gurpide A, Espana A, Majem M, Suh Oh HJ, et al. Clinical management of cutaneous adverse events in patients on targeted anticancer therapies and immunotherapies a national consensus statement by the spanish academy of dermatology and venereology and the spanish society of medical oncology. Clin Transl Oncol. 2019;21((5)):556–571. doi: 10.1007/s12094-018-1953-x. [DOI] [PubMed] [Google Scholar]
- 20.Masui K, Gini B, Wykosky J, Zanca C, Mischel PS, Furnari FB. A tale of two approaches complementary mechanisms of cytotoxic and targeted therapy resistance may inform next-generation cancer treatments. Carcinogenesis. 2013;34((4)):725–738. doi: 10.1093/carcin/bgt086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Fisusi FA, Akala EO. Drug combinations in breast cancer therapy. Pharm Nanotechnol. 2019;7((1)):3–23. doi: 10.2174/2211738507666190122111224. https://10.2174/2211738507666190122111224 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Barber FD. Adverse events of oncologic immunotherapy and their management. Asia Pac J Oncol Nurs. 2019;6((3)):212–226. doi: 10.4103/apjon.apjon_6_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Oliveri S, Faccio F, Pizzoli S, Monzani D, Redaelli C, Indino M. A pilot study on aesthetic treatments performed by qualified aesthetic practitioners efficacy on health-related quality of life in breast cancer patients. Qual Life Res. 2019;28((6)):1543–1553. doi: 10.1007/s11136-019-02133-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pillay B, Wootten AC, Crowe H, Corcoran N, Tran B, Bowden P, et al. The impact of multidisciplinary team meetings on patient assessment management and outcomes in oncology settings a systematic review of the literature. Cancer Treat Rev. 2016;42:56–72. doi: 10.1016/j.ctrv.2015.11.007. [DOI] [PubMed] [Google Scholar]
- 25.Zheng R, Han S, Duan C, Chen K, You Z, Jia J, et al. Role of taxane and anthracycline combination regimens in the management of advanced breast cancer a meta-analysis of randomized trials. Med. 2015;94((17)):e803. doi: 10.1097/MD.0000000000000803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Poi MJ, Berger M, Lustberg M, Layman R, Shapiro CL, Ramaswamy B, et al. Docetaxel-induced skin toxicities in breast cancer patients subsequent to paclitaxel shortage a case series and literature review. Support Care Cancer. 2013;21((10)):2679–2686. doi: 10.1007/s00520-013-1842-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Stintzing S, Fischer von Weikersthal L, Vehling-Kaiser U, Stauch M, Hass HG, Dietzfelbinger H, et al. Correlation of capecitabine-induced skin toxicity with treatment efficacy in patients with metastatic colorectal cancer results from the German AIO KRK-0104 tria. Br J Cancer. 2011;105((2)):206–211. doi: 10.1038/bjc.2011.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Valentine J, Belum VR, Duran J, Ciccolini K, Schindler K, Wu S. Incidence and risk of xerosis with targeted anticancer therapies. J Am Acad Dermatol. 2015;72((4)):656–667. doi: 10.1016/j.jaad.2014.12.010. [DOI] [PubMed] [Google Scholar]
- 29.Mittal S, Khunger N, Kataria SP. Nail changes with chemotherapeutic agents and targeted therapies. Indian Dermatol Online J. 2022;13((1)):13–22. doi: 10.4103/idoj.IDOJ_801_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Robert C, Sibaud V, Mateus C, Verschoore M, Charles C, Lanoy E. Nail toxicities induced by systemic anticancer treatments. Lancet Oncol. 2015;16((4)):e181–e189. doi: 10.1016/S1470-2045(14)71133-7. [DOI] [PubMed] [Google Scholar]
- 31.Rossi A, Caro G, Fortuna MC, Pigliacelli F, D'Arino A, Carlesimo M. Prevention and treatment of chemotherapy-induced alopecia. Dermatol Pract Concept. 2020;10((3)):e2020074. doi: 10.5826/dpc.1003a74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Breed WPM, Van Den Hurk CJG, Peerbooms M. Presentation impact and prevention of chemotherapy-induced hair loss scalp cooling potentials and limitations. Expert Rev Dermatol. 2011;6((1)):109–125. [Google Scholar]
- 33.Macduff C, Mackenzie T, Hutcheon A, Melville L, Archibald H. The effectiveness of scalp cooling in preventing alopecia for patients receiving epirubicin and docetaxel. Eur J Cancer Care. 2003;12((2)):154–161. doi: 10.1046/j.1365-2354.2003.00382.x. [DOI] [PubMed] [Google Scholar]
- 34.Rossi A, Fortuna MC, Caro G, Pranteda G, Garelli V, Pompili U. Chemotherapy-induced alopecia management clinical experience and practical advice. J Cosmet Dermatol. 2017;16((4)):537–541. doi: 10.1111/jocd.12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Pulito C, Cristaudo A, Porta C, Zapperi S, Blandino G, Morrone A, et al. Oral mucositis the hidden side of cancer therapy. J Exp Clin Cancer Res. 2020;39((1)):210. doi: 10.1186/s13046-020-01715-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Migliorati C, Hewson I, Lalla RV, Antunes HS, Estilo CL, Hodgson B, et al. Systematic review of laser and other light therapy for the management of oral mucositis in cancer patients. Support Care Cancer. 2013;21((1)):333–341. doi: 10.1007/s00520-012-1605-6. [DOI] [PubMed] [Google Scholar]
- 37.Rastogi M, Khurana R, Revannasiddaiah S, Jaiswal I, Nanda SS, Gupta P, et al. Role of benzydamine hydrochloride in the prevention of oral mucositis in head and neck cancer patients treated with radiotherapy (>50 Gy) with or without chemotherapy. Support Care Cancer. 2017;25((5)):1439–1443. doi: 10.1007/s00520-016-3548-9. [DOI] [PubMed] [Google Scholar]
- 38.Donnelly JP, Bellm LA, Epstein JB, Sonis ST, Symonds RP. Antimicrobial therapy to prevent or treat oral mucositis. Lancet Infect Dis. 2003;3((7)):405–412. doi: 10.1016/s1473-3099(03)00668-6. [DOI] [PubMed] [Google Scholar]
- 39.Sougiannis AT, VanderVeen BN, Davis JM, Fan D, Murphy EA. Understanding chemotherapy-induced intestinal mucositis and strategies to improve gut resilience. Am J Physiol Gastrointest Liver Physiol. 2021;320((5)):G712–G719. doi: 10.1152/ajpgi.00380.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Reyes-Habito CM, Roh EK. Cutaneous reactions to chemotherapeutic drugs and targeted therapies for cancer part I. conventional chemotherapeutic drugs. J Am Acad Dermatol. 2014;71((2)) doi: 10.1016/j.jaad.2014.04.014. [DOI] [PubMed] [Google Scholar]
- 41.Reyes-Habito CM, Roh EK. Cutaneous reactions to chemotherapeutic drugs and targeted therapy for cancer part II. Targeted therapy. J Am Acad Dermatol. 2014;71((2)) doi: 10.1016/j.jaad.2014.04.013. [DOI] [PubMed] [Google Scholar]
- 42.Carlesimo M, Cortesi E, D'arino A, Pigliacelli F, Caro G, Zancla S, et al. Isochemotopic response the first case on a scar. G Ital Dermatol Venereol. 2021;155((6)) doi: 10.23736/S0392-0488.18.06149-7. [DOI] [PubMed] [Google Scholar]
- 43.Martin KI, Glaser DA. Cosmeceuticals the new medicine of beauty. Mo Med. 2011;108((1)):60–63. [PMC free article] [PubMed] [Google Scholar]
- 44.Goldie K, Kerscher M, Fabi SG, Hirano C, Landau M, Lim TS, et al. Skin quality a holistic 360° view: consensus results. Clin Cosmet Investig Dermatol. 2021;14:643–654. doi: 10.2147/CCID.S309374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zannini L, Verderame F, Cucchiara G, Zinna B, Alba A, Ferrara M. ‘My wig has been my journey's companion’ perceived effects of an aesthetic care programme for Italian women suffering from chemotherapy-induced alopecia. Eur J Cancer Care. 2012;21((5)):650–660. doi: 10.1111/j.1365-2354.2012.01337.x. [DOI] [PubMed] [Google Scholar]
- 46.Berger A, Regueiro C, Hijal T, Pasquier D, De La Fuente C, Le Tinier F, et al. Interest of supportive and barrier protective skin care products in the daily prevention and treatment of cutaneous toxicity during radiotherapy for breast cancer. Breast Cancer. 2018;12:1178223417752772. doi: 10.1177/1178223417752772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Richmond JM, Harris JE. Immunology and skin in health and disease. Cold Spring Harb Perspect Med. 2014;12:a015339. doi: 10.1101/cshperspect.a015339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.McGarvey EL, Baum LD, Pinkerton RC, Rogers LM. Psychological sequelae and alopecia among women with cancer. Cancer Pract. 2001;9((6)):283–289. doi: 10.1046/j.1523-5394.2001.96007.x. [DOI] [PubMed] [Google Scholar]
- 49.Schnur JB, Ouellette SC, Dilorenzo TA, Green S, Montgomery GH. A qualitative analysis of acute skin toxicity among breast cancer radiotherapy patients. Psychooncology. 2011;20((3)):260–268. doi: 10.1002/pon.1734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Krzyszczyk P, Acevedo A, Davidoff EJ, Timmins LM, Marrero-Berrios I, Patel M, et al. The growing role of precision and personalized medicine for cancer treatment. Technology. 2018;6((3–4)):79–100. doi: 10.1142/S2339547818300020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lüftner D, Dell'Acqua V, Selle F, Khalil A, Leonardi MC, De La Torre Tomas A, et al. Evaluation of supportive and barrier-protective skin care products in the daily prevention and treatment of cutaneous toxicity during systemic chemotherapy. Onco Targets Ther. 2018;11:5865–5872. doi: 10.2147/OTT.S155438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Rosenberg SM, Tamimi RM, Gelber S, Ruddy KJ, Kereakoglow S, Borges VF, et al. Body image in recently diagnosed young women with early breast cancer. Psychooncology. 2013;22((8)):1849–1855. doi: 10.1002/pon.3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Di Franco R, Sammarco E, Calvanese MG, De Natale F, Falivene S, Di Lecce A, et al. Preventing the acute skin side effects in patients treated with radiotherapy for breast cancer the use of corneometry in order to evaluate the protective effect of moisturizing creams. Radiat Oncol. 2013;8:57. doi: 10.1186/1748-717X-8-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.González Sanchis A, Brualla Gonzalez L, Sanchez Carazo JL, Gordo Partearroyo JC, Esteve Martinez A, Vicedo Gonzalez A. Evaluation of acute skin toxicity in breast radiotherapy with a new quantitative approach. Radiother Oncol. 2017;122((1)):54–59. doi: 10.1016/j.radonc.2016.09.019. [DOI] [PubMed] [Google Scholar]
- 55.Warren S. The immediate causes of death in cancer. Am J Med Sci. 1932;184((5)):610–615. [Google Scholar]
- 56.Cassileth B. Gerson regimen. Oncology. 2010;24((2)):201. [PubMed] [Google Scholar]
- 57.Copeland EM. Historical perspective on nutritional support of cancer patients. CA Cancer J Clin. 1998;48((2)):67–68. doi: 10.3322/canjclin.48.2.67. [DOI] [PubMed] [Google Scholar]
- 58.Dewys WD, Begg C, Lavin PT, Band PR, Bennett JM, Bertino JR, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern cooperative oncology group. Am J Med. 1980;69((4)):491–497. doi: 10.1016/s0149-2918(05)80001-3. [DOI] [PubMed] [Google Scholar]
- 59.Freedman RJ, Aziz N, Albanes D, Hartman T, Danforth D, Hill S, et al. Weight and body composition changes during and after adjuvant chemotherapy in women with breast cancer. J Clin Endocrinol Metab. 2004;89((5)):2248–2253. doi: 10.1210/jc.2003-031874. [DOI] [PubMed] [Google Scholar]
- 60.Sheean PM, Hoskins K, Stolley M. Body composition changes in females treated for breast cancer a review of the evidence. Breast Cancer Res Treat. 2012;135((3)):663–680. doi: 10.1007/s10549-012-2200-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kumar NB, Cantor A, Allen K, Cox CE. Android obesity at diagnosis and breast carcinoma survival evaluation of the effects of anthropometric variables at diagnosis, including body composition and body fat distribution and weight gain during life span, and survival from breast carcinoma. Cancer. 2000;88((12)):2751–2757. doi: 10.1002/1097-0142(20000615)88:12<2751::aid-cncr13>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
- 62.Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev. 2009;89((2)):381–410. doi: 10.1152/physrev.00016.2008. [DOI] [PubMed] [Google Scholar]
- 63.Roubenoff R. Excess baggage sarcopenia, obesity, and cancer outcomes. Lancet Oncol. 2008;9((7)):605–607. doi: 10.1016/S1470-2045(08)70160-8. [DOI] [PubMed] [Google Scholar]
- 64.Demark-Wahnefried W, Campbell KL, Hayes SC. Weight management and its role in breast cancer rehabilitation. Cancer. 2012;118((8 Suppl l)):2277–2287. doi: 10.1002/cncr.27466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Monroy Cisneros K, Astiazaran Garcia H, Esparza Romero J, Guevara Torres AG, Valencia Juillerat ME, Mendez Estrada RO, et al. Antineoplastic treatment impact on nutritional status in patients with breast cancer. Nutr Hosp. 2014;30((4)):876–882. doi: 10.3305/nh.2014.30.4.7646. [DOI] [PubMed] [Google Scholar]
- 66.Danaei G, Vander Hoorn S, Lopez AD, Murray CJL, Ezzati M. Comparative Risk Assessment collaborating group Cancers. Causes of cancer in the world comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005;366((9499)):1784–1793. doi: 10.1016/S0140-6736(05)67725-2. [DOI] [PubMed] [Google Scholar]
- 67.Mauricio SF, Ribeiro HS, Correia MI. Nutritional status parameters as risk factors for mortality in cancer patients. Nutr Cancer. 2016;68((6)):949–957. doi: 10.1080/01635581.2016.1188971. [DOI] [PubMed] [Google Scholar]
- 68.Baena Ruiz R, Salinas Hernández P. Diet and cancer risk factors and epidemiological evidence. Maturitas. 2014;77((3)):202–208. doi: 10.1016/j.maturitas.2013.11.010. [DOI] [PubMed] [Google Scholar]
- 69.Hébuterne X, Lemarié E, Michallet M, De Montreuil CB, Schneider SM, Goldwasser F. Prevalence of malnutrition and current use of nutrition support in patients with cancer. J Parenter Enter Nutr. 2014;38((2)):196–204. doi: 10.1177/0148607113502674. [DOI] [PubMed] [Google Scholar]
- 70.Carmichael AR. Obesity as a risk factor for development and poor prognosis of breast cancer. BJOG Int J Obstet Gynaecol. 2006;113((10)):1160–1166. doi: 10.1111/j.1471-0528.2006.01021.x. [DOI] [PubMed] [Google Scholar]
- 71.Demark-Wahnefried W, Kenyon AJ, Eberle P, Skye A, Kraus WE. Preventing sarcopenic obesity among breast cancer patients who receive adjuvant chemotherapy results of a feasibility study. Clin Exerc Physiol. 2002;4((1)):44–49. [PMC free article] [PubMed] [Google Scholar]
- 72.Ravasco P, Monteiro-Grillo I, Vidal PM, Camilo ME. Nutritional deterioration in cancer the role of disease and diet. Clin Oncol. 2003;15((8)):443–450. doi: 10.1016/s0936-6555(03)00155-9. [DOI] [PubMed] [Google Scholar]
- 73.LeVasseur N, Cheng W, Mazzarello S, Clemons M, Vandermeer L, Jones L, et al. Optimising weight-loss interventions in cancer patients a systematic review and network meta-analysis. PLoS One. 2021;16((2)):e0245794. doi: 10.1371/journal.pone.0245794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Limon-Miro AT, Valencia ME, Lopez-Teros V, Guzman-Leon AE, Mendivil-Alvarado H, Astiazaran-Garcia H. Bioelectric impedance vector analysis (Biva) in breast cancer patients a tool for research and clinical practice. Medicina (Kaunas) 2019;55((10)):663. doi: 10.3390/medicina55100663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ferreira IB, Marinho EC, Custodio IDD, Gontijo CA, Paiva CE, Crispim CA. Consumo alimentar e estado nutricional de mulheres em quimioterapia. Cienc Saude Coletiva. 2016;21((7)):2209–2218. doi: 10.1590/1413-81232015217.05412015. [DOI] [PubMed] [Google Scholar]
- 76.Custódio IDD, Marinho EDC, Gontijo CA, Pereira TSS, Paiva CE, Maia YCd. P. Impact of chemotherapy on diet and nutritional status of women with breast cancer a prospective study. PLoS One. 2016;11((6)):e0157113. doi: 10.1371/journal.pone.0157113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Dierssen-Sotos T, Gómez-Acebo I, Gutiérrez-Ruiz N, Aragonés N, Amiano P, et al. Dietary constituents relationship with breast cancer prognostic (MCC-Spain follow-up) Int J Environ Res Public Health. 2021;18((1)):84. doi: 10.3390/ijerph18010084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Reeves MM, Terranova CO, Erickson JM, Job JR, Brookes DSK, McCarthy N, et al. Living well after breast cancer randomized controlled trial protocol evaluating a telephone-delivered weight loss intervention versus usual care in women following treatment for breast cancer. BMC Cancer. 2016;16((1)):830. doi: 10.1186/s12885-016-2858-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG, Donato KA, et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults a report of the American College of cardiology/American Heart Association task force on practice guidelines and the obesity society. Circulation. 2014;129((25 Suppl 2)) doi: 10.1161/01.cir.0000437739.71477.ee. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS, Schwartz AL, et al. Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin. 2012;62((4)):243–274. doi: 10.3322/caac.21142. [DOI] [PubMed] [Google Scholar]
- 81.Food, nutrition, Physical activity and the prevention of cancer a global perspective Choice Rev Online. 2008.
- 82.Jiralerspong S, Goodwin PJ. Obesity and breast cancer prognosis evidence, challenges, and opportunities. J Clin Oncol. 2016;34((35)):4203–4216. doi: 10.1200/JCO.2016.68.4480. [DOI] [PubMed] [Google Scholar]
- 83.Sanft T, Harrigan M, Cartmel B, Ferrucci LM, Li FY, McGowan C, et al. Effect of healthy diet and exercise on chemotherapy completion rate in women with breast cancer the Lifestyle, exercise and nutrition early after diagnosis (LEANer) study: study protocol for a randomized clinical trial. Contemp Clin Trials. 2021;109:106508. doi: 10.1016/j.cct.2021.106508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Toriola AT, Liu J, Ganz PA, Colditz GA, Yang L, Izadi S, et al. Effect of weight loss on bone health in overweight/obese postmenopausal breast cancer survivors. Breast Cancer Res Treat. 2015;152((3)):637–643. doi: 10.1007/s10549-015-3496-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.National Institute of Health NHLBI obesity education initiative expert panel on the identification, evaluation, and treatment of overweight and obesity in adults. NHLBI Obes. Educ. Initiat. 2000.
- 86.Levin RM. Developing a culture of nutrition at a community cancer center [Google Scholar]
- 87.Limon-Miro AT, Lopez-Teros V, Astiazaran-Garcia H. Dietary guidelines for breast cancer patients a critical review. Adv Nutr. 2017;8((4)):613–623. doi: 10.3945/an.116.014423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.American Cancer Society . Atlanta (GA), US: Am. Cancer Soc; 2018. Nutrition for the person with cancer. Available from: http://www.cancer.org/docroot/MBC/MBC_6.asp?sitearea=MBC. [Google Scholar]
- 89.Velentzis LS, Keshtgar MR, Woodside JV, Leathem AJ, Titcomb A, Perkins KA, et al. Significant changes in dietary intake and supplement use after breast cancer diagnosis in a UK multicentre study. Breast Cancer Res Treat. 2011;128((2)):473–482. doi: 10.1007/s10549-010-1238-8. [DOI] [PubMed] [Google Scholar]
- 90.Adams LA, Shepard N, Caruso RA, Norling MJ, Belansky H, Cunningham RS. Putting evidence into practice® evidence-based interventions to prevent and manage anorexia. Clin J Oncol Nurs. 2009;13((1)):95–102. doi: 10.1188/09.CJON.95-102. [DOI] [PubMed] [Google Scholar]
- 91.Arends J, Bachmann P, Baracos V, Barthelemy N, Bertz H, Bozzetti F, et al. ESPEN guidelines on nutrition in cancer patients. Clin Nutr. 2017;36((1)):11–48. doi: 10.1016/j.clnu.2016.07.015. [DOI] [PubMed] [Google Scholar]
- 92.Griñan-Lison C, Blaya-Canovas JL, Lopez-Tejada A, Avalos-Moreno M, Navarro-Ocon A, Cara FE, et al. Antioxidants for the treatment of breast cancer are we there yet? Antioxidants. 2021;10((2)):205. doi: 10.3390/antiox10020205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.González-González A, Mediavilla MD, Sánchez-Barceló EJ. Melatonin a molecule for reducing breast cancer risk. Molecules. 2018;23((2)):336. doi: 10.3390/molecules23020336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Veiga ECd. A, Simoes R, Valenti VE, Cipolla-Neto J, Abreu LC, Barros EPM, et al. Repercussions of melatonin on the risk of breast cancer a systematic review and meta-analysis. Rev Assoc Med Bras. 2019;65((5)):699–705. doi: 10.1590/1806-9282.65.5.699. [DOI] [PubMed] [Google Scholar]
- 95.Nooshinfar E, Safaroghli-Azar A, Bashash D, Akbari ME. Melatonin an inhibitory agent in breast cancer. Breast Cancer. 2017;24((1)):42–51. doi: 10.1007/s12282-016-0690-7. [DOI] [PubMed] [Google Scholar]
- 96.Xiang S, Dauchy RT, Hoffman AE, Pointer D, Frasch T, Blask DE. Epigenetic inhibition of the tumor suppressor ARHI by light at night-induced circadian melatonin disruption mediates STAT3-driven paclitaxel resistance in breast cancer. J Pineal Res. 2019;67((2)):e12586. doi: 10.1111/jpi.12586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wang W, Zhang L, Chen T, Guo W, Bao X, Wang D, et al. Anticancer effects of resveratrol-loaded solid lipid nanoparticles on human breast cancer cells. Molecules. 2017;22((11)):1814. doi: 10.3390/molecules22111814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Poschner S, Maier-Salamon A, Thalhammer T, Jäger W. Resveratrol and other dietary polyphenols are inhibitors of estrogen metabolism in human breast cancer cells. J Steroid Biochem Mol Biol. 2019;190:11–18. doi: 10.1016/j.jsbmb.2019.03.001. [DOI] [PubMed] [Google Scholar]
- 99.Sinha D, Sarkar N, Biswas J, Bishayee A. Resveratrol for breast cancer prevention and therapy preclinical evidence and molecular mechanisms. Semin Cancer Biol. 2016;40-41:209–232. doi: 10.1016/j.semcancer.2015.11.001. [DOI] [PubMed] [Google Scholar]
- 100.Kim YN, Choe SR, Cho KH, Cho DY, Kang J, Park CG. Resveratrol suppresses breast cancer cell invasion by inactivating a RhoA/YAP signaling axis. Exp Mol Med. 2017;49((2)):e296. doi: 10.1038/emm.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Wang X, Hang Y, Liu J, Hou Y, Wang N, Wang M. Anticancer effect of curcumin inhibits cell growth through miR-21/PTEN/Akt pathway in breast cancer cell. Oncol Lett. 2017;13((6)):4825–4831. doi: 10.3892/ol.2017.6053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hu S, Xu Y, Meng L, Huang L, Sun H. Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells. Exp Ther Med. 2018;16((2)):1266–1272. doi: 10.3892/etm.2018.6345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wang Y, Yu J, Cui R, Lin J, Ding X. Curcumin in treating breast cancer a review. J Lab Autom. 2016;21((6)):723–731. doi: 10.1177/2211068216655524. [DOI] [PubMed] [Google Scholar]
- 104.Banik U, Parasuraman S, Adhikary AK, Othman NH. Curcumin the spicy modulator of breast carcinogenesis. J Exp Clin Cancer Res. 2017;36((1)):98. doi: 10.1186/s13046-017-0566-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Harris HR, Orsini N, Wolk A. Vitamin C and survival among women with breast cancer a Meta-analysis. Eur J Cancer. 2014;50((7)):1223–1231. doi: 10.1016/j.ejca.2014.02.013. [DOI] [PubMed] [Google Scholar]
- 106.Sant DW, Mustafi S, Gustafson CB, Chen J, Slingerland JM, Wang G. Vitamin C promotes apoptosis in breast cancer cells by increasing TRAIL expression. Sci Rep. 2018;8((1)):5306. doi: 10.1038/s41598-018-23714-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.van Gorkom GNY, Lookermans EL, Van Elssen CHMJ, Bos GMJ. The effect of vitamin C (Ascorbic acid) in the treatment of patients with cancer a systematic review. Nutrients. 2019;11((5)):977. doi: 10.3390/nu11050977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Suhail N, Bilal N, Khan HY, Hasan S, Sharma S, Khan F, et al. Effect of vitamins C and e on antioxidant status of breast-cancer patients undergoing chemotherapy. J Clin Pharm Ther. 2012;37((1)):22–26. doi: 10.1111/j.1365-2710.2010.01237.x. [DOI] [PubMed] [Google Scholar]
- 109.de La Puente-Yagüe M, Cuadrado-Cenzual MA, Ciudad-Cabañas MJ, Hernández-Cabria M, Collado-Yurrita L. Vitamin D and its role in breast cancer. Kaohsiung J Med Sci. 2018;34((8)):423–427. doi: 10.1016/j.kjms.2018.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Carlberg C, Muñoz A. An update on vitamin D signaling and cancer. Semin Cancer Biol. 2022;79:217–230. doi: 10.1016/j.semcancer.2020.05.018. [DOI] [PubMed] [Google Scholar]
- 111.Giammanco M, Di Majo D, La Guardia M, Aiello S, Crescimannno M, Flandina C, et al. Vitamin D in cancer chemoprevention. Pharm Biol. 2015;53((10)):1399–1434. doi: 10.3109/13880209.2014.988274. [DOI] [PubMed] [Google Scholar]
- 112.Hossain S, Beydoun MA, Beydoun HA, Chen X, Zonderman AB, Wood RJ. Vitamin D and breast cancer a systematic review and meta-analysis of observational studies. Clin Nutr Espen. 2019;30:170–184. doi: 10.1016/j.clnesp.2018.12.085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Haque Ripon MS, Asadul Habib M, Hossain M, Ahmed N, Kibria T, Munira S, et al. Role of vitamin E in prevention of breast cancer an epidemiological review. Asian J Adv Res Rep. 2020;11((3)):37–47. [Google Scholar]
- 114.Yang CS, Luo P, Zeng Z, Wang H, Malafa M, Suh N. Vitamin E and cancer prevention studies with different forms of tocopherols and tocotrienols. Mol Carcinog. 2020;59((4)):365–389. doi: 10.1002/mc.23160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kaidar-Person O, Marks LB, Jones EL. Pentoxifylline and vitamin E for treatment or prevention of radiation-induced fibrosis in patients with breast cancer. Breast J. 2018;24((5)):816–819. doi: 10.1111/tbj.13044. [DOI] [PubMed] [Google Scholar]
- 116.Khalefa HG, Shawki MA, Aboelhassan R, El Wakeel LM. Evaluation of the effect of N-acetylcysteine on the prevention and amelioration of paclitaxel-induced peripheral neuropathy in breast cancer patients a randomized controlled study. Breast Cancer Res Treat. 2020;183((1)):117–125. doi: 10.1007/s10549-020-05762-8. [DOI] [PubMed] [Google Scholar]
- 117.Monti D, Sotgia F, Whitaker-Menezes D, Tuluc M, Birbe R, et al. Pilot study demonstrating metabolic and anti-proliferative effects of in vivo anti-oxidant supplementation with N-acetylcysteine in breast cancer. Semin Oncol. 2017;44((3)):226–232. doi: 10.1053/j.seminoncol.2017.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Šalamon Š, Kramar B, Marolt TP, Poljšak B, Milisav I. Medical and dietary uses of n-acetylcysteine. Antioxidants. 2019;8((5)):111. doi: 10.3390/antiox8050111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Boots AW, Haenen GRMM, Bast A. Health effects of quercetin from antioxidant to nutraceutical. Eur J Pharmacol. 2008;585((2–3)):325–337. doi: 10.1016/j.ejphar.2008.03.008. [DOI] [PubMed] [Google Scholar]
- 120.Patisaul HB, Jefferson W. The pros and cons of phytoestrogens. Front Neuroendocrinol. 2010;31((4)):400–419. doi: 10.1016/j.yfrne.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Messina M, McCaskill-Stevens W, Lampe JW. Addressing the soy and breast cancer relationship review, commentary, and workshop proceedings. J Natl Cancer Inst. 2006;98((18)):1275–1284. doi: 10.1093/jnci/djj356. [DOI] [PubMed] [Google Scholar]
- 122.Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 2020;369((6510)):1481–1489. doi: 10.1126/science.abc3421. [DOI] [PubMed] [Google Scholar]
- 123.Misotti AM, Gnagnarella P. Vitamin supplement consumption and breast cancer risk a review. Ecancermedicalscience. 2013;7:365. doi: 10.3332/ecancer.2013.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Mokbel K, Mokbel K. Chemoprevention of breast cancer with vitamins and micronutrients a concise review. In Vivo. 2019;33((4)):938–997. doi: 10.21873/invivo.11568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ávila-Gálvez MÁ, Giménez-Bastida JA, Espín JC, González-Sarrías A. Dietary phenolics against breast cancer. A critical evidence-based review and future perspectives. Int J Mol Sci. 2020;21((16)):5718. doi: 10.3390/ijms21165718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Augimeri G, Montalto FI, Giordano C, Barone I, Lanzino M, Catalano S, et al. Nutraceuticals in the mediterranean diet potential avenues for breast cancer treatment. Nutrients. 2021;13((8)):2557. doi: 10.3390/nu13082557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Choi EK, Kim IR, Chang O, Kang D, Nam SJ, Lee JE, et al. Impact of chemotherapy-induced alopecia distress on body image psychosocial well-being and depression in breast cancer patients. Psychooncology. 2014;23((10)):1103–1110. doi: 10.1002/pon.3531. [DOI] [PubMed] [Google Scholar]
- 128.Münstedt K, Manthey N, Sachsse S, Vahrson H. Changes in self-concept and body image during alopecia induced cancer chemotherapy. Support Care Cancer. 1997;5((2)):139–143. doi: 10.1007/BF01262572. [DOI] [PubMed] [Google Scholar]
- 129.Rosman S. Cancer and stigma experience of patients with chemotherapy-induced alopecia. Patient Educ Couns. 2004;52((3)):333–339. doi: 10.1016/S0738-3991(03)00040-5. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplementary data
