Home » NuclearMedicine » SJNMRT-ID14.php Scientific Journal of Nuclear Medicine & Radiation Therapy


Review Article

Special Issue on Radiation Therapy for Breast Cancer - Radiation Side Effects

Mariacarla Valli1,2*, Elona Cekani1, Simona Cima1,2 and Antonella Richetti1,2

1Radiation Oncology, Oncology Institute of Southern Switzerland, Bellinzona-Lugano, Switzerland
2Senology Center of Italian Switzerland

*Address for Correspondence: Mariacarla Valli, Senior Consultant in Radiation Oncology, Oncology Institute of Southern Switzerland, via Ospedale, 6500 Bellinzona CH, Tel no: +4191-8119430/7613, E-mail: mariacarla.valli@eoc.ch

Submitted: 18 September 2017; Approved: 20 October 2017; Published: 23 October 2017

Citation this article: Valli M, Cekani E, Cima S, Richetti A. Special Issue on Radiation Therapy for Breast Cancer - Radiation Side Effects. Sci J Nucl Med Radiat Ther. 2017;1(1): 016-031.

Copyright: © 2017 Valli M, et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited

Keywords: Radiotherapy; Breast Cancer; Cutaneous Toxicity; Cardiac Toxicity; Pulmonary Toxicity; Thyroid Toxicity; Brachial Plexus Toxicity

Download Fulltext PDF

Post-operative radiotherapy after conservative surgery or mastectomy improves disease free and overall survival. Collateral effects and toxicity dramatically decreased in the last decades due to technological innovations (i.e. conformal techniques, respiratory gating, image-guided radiotherapy, tracking systems), less demolitive surgical approaches, new reconstructive techniques and better knowledge of radiotherapy interaction with chemo/hormono/immunotherapies.

A more careful consideration of patients co-morbidities and the collaboration of specialized personnel improves its safety too.

Even when loco-regional lymphnodes have to be irradiated, according to tumor stage and age of the patient, limited acute and late toxicity has been detected. The irradiation of regional lymphnodes draws our attention to thyroid and brachial plexus toxicities in addition to the cutaneous, cardiac and pulmonary ones.

Introduction

Breast cancer is the most common female malignancy in the western countries and is the second leading cause of malignant deaths. A large proportion of breast cancer patients receive post-operative radiation therapy after conservative surgery and mastectomy to decrease locoregional recurrence rates and improve overall survival [1,2].The irradiated volumes i.e. whole breast, tumoral bed, chest wall, regional lymph nodes (internal mammary chain, supra/infraclavicular and axillary nodes) could vary, according to international guidelines, taking into account patient and tumor characteristics.

The duration of radiotherapy strictly depends on the irradiated volume, dose/fraction and total dose and lasts between 1 day, in case of intraoperative RT, and nearly 7 weeks, in case of conventional whole breast RT plus booster dose on surgical bed.

Side effects and toxicities widely vary as a consequence of patients features and attitudes, irradiated volume, total dose, dose/fraction, radiotherapy technique and systemic treatments.

Several grading scales exist to aid in the reproducible quantification of acute radiation toxicities. The Radiation Therapy Oncology Group/European Organization for Research and

Treatment of Cancer (RTOG/EORTC) toxicity criteria and National Cancer Institute Common Toxicity Criteria for Adverse Events (NCI CTCAE) systems are the most commonly used [3,4]. This review focuses on the main radiation side effects in adjuvant breast cancer treatment and discusses the strategies for the prevention and management of radiation-related toxicity.

Skin Toxicity

The effects of radiation on skin have been recognized as soon as radiation was introduced as a treatment modality [5-7]. Consequently researches have begun examining techniques to reduce radiation induced skin reactions, aided by the radio-biology evidences on fractionation modalities for improving normal tissue repair [6]. The introduction of the megavoltage units improved the skin sparing if compared with orthovoltage ones and consequently reduced toxicity [8].

Despite the effort, Radiation Dermatitis (RD) still continue to be the most common side effect of radiotherapy and could condition worse cosmesis and subsequently QoL[5].

Radiation affects the two main components of skin, epidermis and dermis, each of which respond variably to radiation exposure. High energy X-Rays leads to DNA breaks within the epidermis and dermis causing the so called syndrome of radiation dermatitis [6]. Radiation induced DNA damage within the epidermis disrupts normal proliferation and differentiation of basal keratinocytes, leading to the physical barrier lost [9-11].

By the other hand, the effects on dermis consists mainly of microvascular injury, activating proinflammatory cytokines, tTNF alfa and transforming growth factor: they contribute in both acute and late side effects of radiation, starting with inflammatory edema, erythema, desquamation, ulceration, telangiectasia , late tissue fibrosis [12-19].

Mechanism of RD developes in a dose dependent manner with predictable timing [12,20,21]. Acute skin reactions (defined as occurring within 30-90 days from radiation exposure) has a dose threshold development, starting with erythema at 6-10 Gy up and worse toxicity at 40 Gy or higher dose. Nearly around day 10 of treatment the re-epithelisation begins and competes with ongoing radiation damage to maintain the homeostasis of the epidermal layer. The majority of symptoms usually relieves in 2-4 weeks after the end of treatment, hyperpigmentation lasts some more months. Late side effects can appear months or years after the end of therapy [20-24].

Risk Factors

Many risk factors have been identified and categorized in treatment and patient related ones.

Treatment related factors

The negative impact of adjuvant systemic chemotherapy on acute and late toxicity has been spoted out and underlined in studies enrolling patients mainly receiving taxanes and/or anthracyclines based regimen [25-27], so that the concomitant use of radiotherapy and anthracyclines/taxanes based chemotherapy was not recommended.

Surgical approach is crucial for the cosmetic outcome too. The amount of resected tissue is considered one of the most relevant features concerning breast cosmesis. The kind of surgery, more or less radical, the new oncoplastic techniques and the different reconstructive surgery (i.e.expander, prosthesis or flap) are strictly related to radiotherapy toxicity. Nevertheless no clear data still exist on the best sequence of reconstruction and radiotherapy, the safer break between these procedures and the more appropriate radiotherapy techniques in this group of patients [28,29].

The evolution in radiotherapy delivery techniques, from 2dimension (2D) to 3D treatment planning, resultes in significantly less acute and long term toxicity. The 3D technique, taking into account changes in breast contour, reduces radiation induced skin reaction. Data from retrospective studies suggest that large dose inhomogeneity, occurring with the 2D techniques, predisposes to more severe acute skin reactions [30-33].

Advanced 3D techniques, Volumetric Arc Therapy (VMAT) and Intensity Modulated RadioTherapy (IMRT) are the main methods that allow multiple radiation fields for decreasing breast dose inhomogeneity. Prospective trials of 3D and IMRT techniques have shown decreased rates of acute and late radiation skin toxicity [34-36].

Radiotherapy delivered to patient in prone position could improve the dose distribution, reduce radiation hotspots and lower the incidence of breast dermatitis including moist desquamation [37].

Alternative treatment schedules, such as hypofractionated breast irradiation, is increasingly used [38] and safe in terms of local control and acute/late toxicity profile without negative impact on cosmesis. Three randomized trials (START A, START B and Canadian), which compared standard vs Hypofractionation (HF) in post-operative radiotherapy, suggest that HF reduces the rate of dermatitis, pruritus and hyperpigmentation [39,41]. In a recently published study [42] the authors conclude that HF is associated with a significantly reduction of severe acute skin reaction compared to the standard fractionation one: Grade 2 skin reactions were observed in 19% of the patients treated with CF compared to 2 % treated with HF.

Even and moreover with such a fractionation, large dose inhomogeneity (V >107%) has to be carefully detected and corrected for avoiding severe acute skin reactions.

The role of boost on cosmesis is still controversial, but a correlation with higher late toxicity is widely demonstrated by the results of the EORTC trial 22881-10882 [43,44].

Accelerated partial breast Irradiation (APBI) has been introduced as an alternative treatment method for selected patients with early stage breast cancer. Results concerning cosmetic outcome differ widely, ranging from good/excellent cosmetic results in 90% of the patients to 21% unacceptable cosmesis [45]. This variability strongly depends on the RT technique and moreover randomized trials are still ongoing for confirming the efficacy of this treatment in terms of local control and toxicity.

Interestingly some authors have investigated time of radiotherapy in circadian cycle, suggesting that there are no difference on failure outcomes between early morning and late afternoon radiotherapy delivery, but afternoon radiotherapy was associated with higher-grade 2-skin toxicity [46].

Patient related factors

Patient related factors have been investigated too for establishing a correlation with radiotherapy toxicity. Age and postmenopausal status seem to be correlated with a worse cosmetic outcome [47]. Large breast size women have been reported to experience acute skin reactions five times more than small breast size ones [48,50]. Body mass index is independently associated with increased risk of acute skin toxicity too [51,52].

The worst skin reactions are seen in inframammary and axillary folds, due probably to a greater self bolusing effect in these regions [52]. Racial differences too have been demonstrated to be associated with radiation dermatitis, black skin population experiencing a higher toxicity [53]. No conclusions can be drawn from existing data on the effect of smoking on RT related toxicity [51,54]. Genetic syndromes, such as Ataxia-Teleangioectasia, Fanconi anemia, etc may be associated with higher acute radio sensitivity [55].

Strategies for Prevention

The high incidence of radiation-induced skin reactions has generated interest in methods for preventing and effectively treating such reactions. Although a general consensus among radiotherapy centres is lacking, it is generally agreed that the ideal method for preventing and minimizing skin reactions is moisturization of the irradiated area

Several products have been used for the prevention of radiation-induced toxicity. Topical steroids like momethasone or bethametasone were found to improve the outcome in diminishing dermatitis when compared with placebo [56,57]. Non steroidal agents, like sucralfate or aqueous crème, aloe vera and hyaluronic acid have failed to show any benefit when tested prospectively [58]. Some other nonsteroidal agents like silver sulfadiazine or calendula officinalis have shown to result in overall reduced dermatitis, even though these results were not replicated in more recent larger trials [59]. The barrier products, like Mepitel filmappears promising in reducing moist desquamation rate [60].

Management During Treatment

Lack of high quality evidence and disagreements regarding the optimal management of skin toxicity still remain. Skin washing with soap and water has showed significant reduction of acute erythema and desquamation 6-8 weeks after the treatment, the authors recommend mild pH –neutral or nonalcaline soaps [61]. An important study by Burch [62] challenged the previous assumption that products used as deodorants containing aluminum or zinc, would increase skin reaction, by demonstrating that there is no difference between metallic and nonmetallic deodorant products and no evidence in worsening skin outcomes with deodorant use [63,64]. Mepilex dressing too reduces skin erythema for dermatitis treatment [65]. Management of desquamation is also critical, especially the moist one. While the dry desquamation is not a cause of concern, the moist one should be monitored carefully and closely because it may worsen causing fever, infections, smelling, and pain. The use of saline soaks four times daily is encouraged [66-68], also moisture-retentive, barrier ointments after each saline soak.

Pulmonary Toxicity

Lung is one of the relevant Organs at Risk (OaR) in planning radiotherapy for breast cancer. Acute and late lung toxicities have been widely described as Radiation Pneumonitis (RP) and radiation fibrosis. The acute one known as an inflammatory reaction occurring four to twelve weeks after completion of RT, while the late one is usually observed beyond six months [69].

Definition and grading of RP differs depending on the scoring system used e.g. grade 2 can be dyspnea, notinterfering with Activities Of Daily Living (ADL) according to CTCAE scoring system, meanwhile it needs medication with corticosteroids according to SWOG scale.

RP reflects the response to the activation of a rapid cascade of genetic and molecular events evolving during a subclinical time, including loss of pneumocytes, increased capillary permeability, interstitial and alveolar edema and access of inflammatory cell into the intra-alveolar spaces [70]. These changes are observed on chest X rays or Computed Tomography (CT). This latter examination allows a better evidence of parenchymal changes and better demonstrates the changes restricted to the irradiated area. The most common findings are ground glass opacities and/or airspace consolidation [71,72]. Marks et al did not show any association between the presence or absence of radiotherapy induced pulmonary symptoms and the frequency of radiotherapy induced radiological changes [73]. Hernberg et al. reported clinical signs of suspected pneumonia in 29% of patients with radiological changes on CT, in nearly 68% these changes were more common three months after radiotherapy [74].

Many specific gene loci have been studied as responsibles of a greater susceptibility to radiation [75]. The association between single nucleotide polymorphisms in TGFβ1 gene and risk of RP was studied in patients with NSCLC. In multivariate analysis CT/CC genotypes of TGFβ1 at rs1982073:T869C were found to be associated with a lower risk of RP grades ≥ 2 (P=0.013) and grades ≥ 3 (P=0.007) respectively, compared to the TT genotype, after adjustment for Karnofsky performance status, smoking habit, pulmonary function and dosimetrical parameters [76].

Radiation induced lung injury occurs in almost 10-15 % of patients treated with adjuvant radiotherapy for breast cancer [77]. In retrospective and prospective studies the incidence of lung injury varies widely, ranging between 4.5-63 % [78-80], probably due to several biases [81]. A meta-analysis on the incidence of early lung toxicity in patients treated with a 3D conformal radiotherapy for breast cancer analyzed ten different studies and reported the overall incidence of clinical and radiological RP as 14% and 42 % respectively [82].

It is crucial to minimize radiotherapy related complications as most of the breast cancer patients are long survivors. If the incidence of symptomatic RP is reduced, not only QoL but also the compliance of breast cancer patients may be improved.

Risk factors

Several risk factors for radiation induced lung injury have been studied including age, BMI, irradiated lung volume, radiation dose, central lung distance, pre-radiotherapy functional level and concurrent chemotherapy [83-87].

Patient age seems to be the strongest predicting factor for radiotherapy lung toxicity [80]. Increased age has been correlated with both clinical and radiological lung changes [85]. Many studies have suggested that patients older than 55 or 59 years have to be treated cautionary with post-operative RT [81]. Furthermore BMI have been shown by different authors to be a good indicator of the risk of pneumonitis, suggesting a low BMI to be associated with higher incidence of symptomatic RP. Conversely some other authors did not show a significant association between RP and BMI [88-91].

Smoking habits represent another baseline risk factor. Association between smoking and lung radiotherapy injury is still debated, since published studies showed conflicting results [80]. A history of smoking increases the risk of RP as a result of pre-existing lung damage, but active smoking seems to protect the lung from radiotherapy-induced damage [92].

Pre-treatment lung inflammation seems to make pulmonary tissue more susceptible to radiation damage. This hypothesis has recently been demonstrated: volumes within the lung (excluding tumour) that show an avid uptake of 18F-Deoxyglucose (FDG) before radiotherapy are more susceptible to radiation damage. Therefore, the risk of radiation-induced lung toxicity may be decreased by applying sophisticated radiotherapy techniques to spare volumes in the lung with high FDG uptake [93]. Early lung damage in patients who later develop RP can be demonstrated by an increased FDG uptake during the first two weeks of irradiation [94].

Dosimetric parameters, total dose, dose per fraction and irradiated lung volume are the main predictive factors. Despite the improvement of radiotherapy techniques and the increasing use of conformal radiation therapy radiation-induced injuries are still registrated.

Recent technique developments, such as IMRT, VMAT, helical tomotherapy, Image-Guided Radiotherapy (IGRT), breathing adapted techniques, etc have helped to optimize the dose distribution into the PTV and to reduce doses to the OAR. Concerning unconventional RT schedules, major clinical trials of hypofractionated whole breast radiotherapy have reported no significant differences in lung injury rate [95].

Reported risk of pulmonary toxicity using APBI is low and is mainly correlated with the kind of radiotherapy technique; the 3D conformal one seems to involve a slightly higher volume of lung in comparison to the others [96].

Lung is known to be very sensitive to both dose per fraction and total dose so that well defined dose parameters are standardized and used in daily practice.

Ipsilateral Mean Lung Dose (MLD) and V20 are considered the most important ones. MLD values\20 Gy and V20\20 % for the ipsilateral lung are considered acceptable in case of whole breast RT. A significant relation between V13 and radiological changes on CT images was reported [97-99]. Prone position seems to be associated with a reduction in the amount of irradiated lung [100].

Ramella et al. [101] showed that adding ipsilateral lung volume received 20 Gy (IV20) and ipsilateral lung volume received 30 Gy (IV30) to the classical total lung constraints reduced pulmonary toxicity in concurrent chemoradiation treatment. They suggested that not all beam entrances should be on the ipsilateral lung. A conservative approach would be to use the constraint settings IV20 and IV30 as simple predictive factors of lung injury. According to a recent publication IV20 is the greates risk predictor factor [102].

Goldman et al. [103] showed that minimizing the percentage of the ipsilateral lung dose to V20 < 30% can significantly reduce moderate to severe radiological changes and symptomatic pneumonitis. Lind et al. [78,85] concluded that the incidence of short-term moderate pulmonary complications in adjuvant locoregional 3Dradiotherapy for breast cancer is clinically significant. Their results also suggested an association among pulmonary complications, older age and incidentally irradiated lung volume. Another recent report suggests that utilizing multiple variables including V20 to the ipsilateral lung, age and BMI could better estimate the risk of pneumonitis [102]. Lymphnode treatment increases the irradiated lung volume and the dose to the lung. Several studies have demonstrated an increased risk of RP in case of loco-regional (breast/chest wall+lymphnodes) radiotherapy compared to brest/chest wall radiotherapy alone [104-106]. Several authors and meta-analysis have found a strong association between supraclavicular field irradiation and incidence of RP [82]. The Internal Mammary Nodes (IMN) irradiation too is associated with an increased incidence of RP [78,85]. Another study found a strong correlation between using electrons beams for the chest wall and symptomatic RP, probably related to the inhomogeneity caused by the scattering effect of electrons [104]. Chest/Lung Distance (CLD), easy to measure at the time of simulation [107], has been shown to be predictive of lung toxiticy because of its correlation with the irradiated lung volume [108]. This paremeter gain impontance especially in case of tangential fields treatments.

The recent publication of the EORTC trial 22922-10925 results confirms that radiotherapy on lymphatic drainage improves the risk of lung fibrosis, even if in less than 5% of the population, at a median follow-up of 10 years. Only 3% of the 3866 eligible patients experienced pulmonary fibrosis (4.4% in the nodal-irradiation group vs. 1.7% in the control group, P < 0.001). No significant difference was observed between the two study groups with respect to performance status. The author registered an increased risk of pulmonary fibrosis at 10 years if compared with 3 year follow-up in both groups (from 0.9% to 1.7% in the control group and from 2.8% to 4.4% in the nodal-irradiation group [109].

The use of breath holding techniques even for right sided breast cancers, if lymphatic drainage radiotherapy is prescribed, lowers the risk of pneumonitis and secondary lung cancer (even in smoking patients) suggesting to apply breath-hold for locoregional irradiation of right-sided breast cancer patients [110].

Several studies concerning mortality from RT-related lung cancer showed that the risk is correlated with total dose to the lungs, thus recommending a mean dose of 7-18 Gy for the ipsilateral, and a dose of 0.1-3 Gy for the contralateral lung [114].

Systemic Treatment Related Factors

Many studies have shown that concomitant RT and endocrine therapy (tamoxifen) could represent a pulmonary fibrosis risk factor [111]. In patients receiving adjuvant hormonal manipulation, tamoxifen and RT is routinely associated, but should be administered with caution in case of potentially radiosensitive patients. Conversely concomitant RT and aromatase inhibitors seems to be safe and no toxicity was observed in the irradiated lung tissue [112], although estrogen deprivation has theoretically a negative effect on post-radiation tissue remodeling. Several chemotherapeutic agents are able to induce pulmonary toxicity, independently by RT. The concomitant administration of paclitaxel and RT significantly increases the risk of lung injury due to the radio-sensitizing effect of taxanes, and should be therefore avoided [113].

Prevention

The effect of giving a radioprotective agent concomitantly with radiation to prevent normal lung tissue toxicity is the object of research in several studies. Amifostine, an aminothiol with broad-spectrum cytoprotection, did not reduce the incidence of RP in a large randomised Phase III RTOG study. Neither there was evidence of a tumour-protecting effect [115]. Pentoxifilline taken during radiation therapy significantly decreased pulmonary toxicity in a small study [116].

Incidentally the ACE inhibitor use decreased the risk of RP in lung cancer patients receiving thoracic irradiation in a retrospective study and in animal models [117].

Treatment

There are no controlled, randomised trials on the treatment of RP. In case of grade I toxicity patients are mostly asymptomatic with only radiographic findings and seldom a dry cough. No treatment is recommended. In case of grade II toxicity corticosteroids are suggested if patients have more severe complaints. However, mild forms of grade II can be treated with inhalation corticosteroids and bronchodilators. More severe grade II and grade III toxicities are treated with Prednisolone 30-40 mg daily for two weeks followed by a slow dose reduction for six to twelve weeks [118]. A relapse may occur after discontinuation of corticosteroids. A substantial reduction in symptoms is normally seen, as well as an improvement of the radiological abnormalities. For patients with grade IV RP and severe respiratory insufficiency, continuous oxygen or even assisted ventilation is required. Some patients with RP are resistant to corticosteroids. This is due to more than 1.5-times increase of the lung epithelium-specific protein Krebs von den Lungen-6 (KL-6), which is produced by and secreted by type II pneumocytes. It is suggested that these patients can be treated with azathioprine or cyclosporine A [119]. There is no proven effective treatment for radiation-induced lung fibrosis. It occurs months to years after the end of RT, in response to the initial tissue injury, and leads to permanent impairment of oxygen transfer [120].

Cardiovascular Toxicity

It is well known that postoperative radiotherapy may have adverse effects in long-term survivors and an increased mortality was observed in long-term survivors since the first meta-analysis by Cuzick and colleagues [121]. The Scandinavian studies and cancer registries identified a high rate of cardiac deaths in patients treated with post-operative RT for left sided breast cancer and underlined its role in the increased long-term mortality of this population. Interestingly a clear dose-response relationship was defined according to Scandinavian trials analysis. This dose-effect relationship has been confirmed by the meta-analysis of the Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) in 2005 [122]. Since then, these consolidated findings have been consistently taken into account for modern radiotherapy treatment planning [123].

The exact causes of cardiotoxicity after radiotherapy in breast cancer patients is not clear [124].

The potential role of radiotherapy in causing cardiotoxicity derives from the evidence that patients, treated with mantle field irradiation for Hodgkin lymphoma, experienced pericarditis, as well as congestive heart failure, ischemic coronary artery disease, arytmia or miocardial infarction [125].

Experimental evidence suggests indirect harmful effects of microvascular and macrovascular damage on the myocytes. It has been postulated that radiotherapy leads to an acute inflammation within the heart blood capillaries and to continuous inflammatory processes, resulting in endothelial cell proliferation and formation of fibrin thrombi with obstruction of the myocardial capillary lumen. A consequently reduction of collateral flow and vascular reserve causes ischemia and macrovascular injury. This phenomena could accelerate an age related atheroschlerosis leading to CAD (Coronary Artery Disease) [126]. Therefore, it is quite comprehensible that epidemiologic studies detected baseline cardiac risk factors as independent risk factor for cardiovascular disease after radiotherapy. Those risk factors are age, hypertension, diabetes mellitus, high total cholesterol levels, family history of early myocardial infarction and smoking [127]. Concomitant administration of cardio-toxic systemic treatments should be strongly considered when prescribing RT. Cardiotoxicity of anthracycline-based chemotherapy regimens is well-known and studied; therefore their concomitant use with radiotherapy is not recommended. Safety of adding trastuzumab to standard adjuvant systemic therapy for Human Epidermal Growth Receptor 2 Factor (HER2) positive early breast cancer was largely demonstrated [128]. Among patients who received radioherapy, there were no significant differences across treatment arms in the incidence of acute skin reaction, pneumonitis, dyspnea, cough, dysphagia, or neutropenia. In particular, the 3-year cumulative incidence of cardiac events was 2.7 % with or without radiotherapy. Safety of trastuzumab, given concomitantly to radiotherapy in the adjuvant setting, was further investigated in large observational serie and no significant impact on cardiac toxicity profile was detected [129,130].

Asian studies have demonstrated, on the contrary, a higher risk of acute LVEF dysfunction [131] suggesting that trastuzumab and left sided radiotherapy have at least a combined effect on cardiac toxicity. The authors suggest that a LVEF > 50% before the initiation of trastuzumab and RT is required. The main limitation of this study is that LVEF is not sensitive enought to detect early and minor cardiac disfunction, so that the actual toxicity might be underestimated [131].

The cardiac toxicity of radiotherapy in breast cancer is certainly less than the one of several drug therapies. However, up to now, it still remains unclear if late effects after combination of both therapies can become manifest in old age with decreasing compensation ability of the heart. Finally, this is not a specific radiotherapy related toxicity but a multifactorial one [132]. Several studies deal with the effect of dose on specific cardiac structures in patients undergoing radiotherapy. Lind et al. [133] evaluated post-radiation regional myocardial heart perfusion changes with Single-Photon Emission Tomography (SPECT) in 69 patients. They correlated the Left Anterior Descending artery (LAD) distribution SPECT changes with the percentage of irradiated Left Ventricle (LV) volume and risk factors for coronary artery disease. The meta-analysis of Darby et al. [134] confirms well-known data on cardiac risks in old studies and has identified a dose-response relationship. Unfortunately it gives no conclusive answers to the most important question of how best to protect the heart against radiation toxicity. CAD rate increased linearly with the mean dose to the heart, with no apparent threshold. Critical doses and volume relationships are not well defined, mainly due to wide variation in structures delineation [135]. Current recommended mean heart dose is around 2-7 Gy for left-sided, and 1.5 Gy for right-sided patients [136]. Despite a significant reduction in the mean heart dose has been observed in the last decades with the development of more sophisticated raditherapy techniques, an increased risk of cardiac toxicity has been registered [137].

Due to this reason national and international guidelines highlight the sparing of the Organs at Risk (OaR), with peculiar attention to the heart. The Danish Breast Cancer Cooperative Group suggests to administer the prescribed therapeutic dose to the tumor bed sparing LAD, heart and lung [138]. The authors suggest, in case of standard treatments, to respect the following dose constrains: for LAD maximum 20 Gy, for heart V20\10 %, and V40\5 %, using standard fractionation.

Hypofractionation for whole breast RT is approved, recommended, increasingly used and often considered a standard treatment schedule [139]. No evidence of a higher incidence of adverse cardiac events is reported, and some authors suggest that hypofractionated RT might be even safer, in terms of heart sparing, than conventional regimens [140]. An innovative fractionation schedule (hypofractionation with integrated boost) was tested, and doses to organs at risk were routinely determined and documented during treatment planning. Interestingly the mean heart dose was only 1.48 + 0.9 Gy (n=151) [141].

The IMN irradiation is associated with an increased dose delivered to heart and coronary artery. The EORTC 22922-10925 study results showed that, after 10 years of follow up, 1% of patients had cardiac fibrosis (1.2% if lymphnodes RT vs. 0.6%, P = 0.06), and 6% had cardiac disease (6.5% if lymphnodes RT vs. 5.6%, p = 0.25) [109].

On the contrary, no increased rate of cardiac disease was observed in the nodal-irradiation group of the Canadian trial at a median follow up of 10 years [142,143].

The Danish Population-Based Cohort Study evaluated 3089 patients on the effect of internal mammary node irradiation in early node-positive breast cancer. The authors registered equal numbers of patients who died of ischemic heart disease in both groups with a median follow-up of 8 years [144].

Prevention

Innovative techniques have been tested for avoiding inappropriate high doses to heart and LAD. Conventional 3DCRT uses tangential beams toavoid high dose region in the Ipsilateral Lung (IL) and heart. However, it results in poor conformity, homogeneity and hotspots outside the target volume. On the other hand Intensity Modulated Radiotherapy (IMRT) and Volumetric Arc radiotherapy (VMAT) improve the dose conformity inside the target at the cost of an increased low dose spread to Contralateral Lung (CL) and Contralateral Breast (CB) which could increase the risk of secondary cancer [145,146]. Additional advantages of these techniques is their ability to provide differential dose distributions, allowing the Simultaneous Integrated Boost (SIB) delivery [147,148] Of note IMRT plans employ higher Monitor Units (MU) and hence increases the delivery time by 3-4 folds compared to VMAT and conventional 3DCRT. Increase in treatment time, MU, leakage, scatter radiation in IMRT has implications on tumor cell repair and repopulation [149,150].

Another issue of post-operative radiotherapy is the breast motion due to breathing. At present, the Deep Inspiration Breath Hold Technique (DIBH) is increasingly used for reducing the target motion and primarily for reducing the hearth dose [151-153]. In deep inspiration, the heart sinks down and the distance from the chest wall increases. First dosimetric studies on single patients showed a significant reduction in heart and lung dose using this modality. It has to be clarified whether the unique dosimetric advantage is clinically relevant or not, as of today there are no prospective studies or data demonstrating a possible clinical benefit of DIBH irradiation on the late cardiac toxicity rate for left breast irradiation. Noteworthy preliminary retrospective data on CT based “calcium score” of the coronary arteries provided some evidence that radiotherapy of left sided breast cancer using breath hold may be associated with less calcification [154]. Due to the long interval between radiotherapy and the appearance of heart toxicity, we need many years to assess the clinical impact of DIBH on cardiac toxicity. Even though the best clinical practice is to reduce the heart and LAD dose to as low as reasonably achievable. Data from the major retrospective series indicate that an increasing mean cardiac dose was clearly associated with a proportional increased rate of major coronary events [155,156]. Sardaro et al. reported a 4% increase in the late heart disease risk per 1 Gy in mean heart dose [156]. Darby et al. have estimated that a 1 Gy increase in mean heart dose will result in a 7.4% higher rate of major coronary events, defined as myocardial infarction and death from ischemic heart disease [155]. Obviously, mean heart dose sufficiently reflectes coronary artery exposure in patients treated with DIBH in a recent analysis [157,158].

A recent study [159] showed a significant reduction in mean heart dose of 20% with IMRT and 23% with VMAT. (In this study also was seen an additional decrease in heart and LADCA dose by IMRT in both FB and DIBH irradiation, compared with VMAT) Therefore, it has been ongoing and continuing institutional policy to use the DIBH technique whenever feasible as the standard irradiation method for all left-sided breast cancer patients in order to keep the heart and LAD dose as low as possible.

Irradiation in prone position is another promising treatment option. Several publications are in favor of replacing the supine standard position by the prone one for whole-breast irradiation, especially in patients with large breasts [160-162]. Recently, Mulliez et al. [160] reported a phase III randomized trial in this context, in which skin desquamation, dermatitis and edema were significantly reduced. Moreover the dose to the ipsilateral lung and the LAD were significantly reduced. Comparing the prone and supine treatment positions in 400 patients, Formenti et al. [162] evaluated the in field volumes of the heart receiving the full dose as a surrogate for normal-tissue exposure. They described a considerable anatomical variability of the volume range, but were also able to show a significantly lower mean dose to the heart in the prone position.

Another way to reduce heart toxicity should be the use of Accelerated Partial Breast Irradiation (APBI), which exclusively targets only the lumpectomy site plus some margin. Current techniques for APBI are Intraoperative Radiotherapy (IORT), multi-interstitial brachytherapy, intracavitary brachytherapy and external beam radiotherapy. The available data on APBI suggests acceptable local control and survival in selected patients with low-risk breast cancer [163].

Thyroid toxicity

The true prevalence of radiotherapy-induced Hypothyroidism (HT) in patients who receive radiotherapy is not known because Thyroid Function Tests (TFT) are not routinely assessed prior and after the treatment. Additionally, the radiation dose to the thyroid gland is not routinely evaluated because this organ is neglected as OaR, especially if only the breast is irradiated [164].

HT is one of the late toxicities of curative RT to the neck region, and the reported incidences of HT ranges from 20% to 52% in patients treated with radiotherapy for head and neck cancers and Hodgking disease [165-168].

Conversely only few studies have performed clinical DVH analysis for thyroid dysfunction after radiotherapy in breast cancer patients and, according to their results, the incidence of HT varies from 6% to 21% [169-173].

Thyroid dysfunction develops in nearly 15% of patients, it is detectable usually 3 months after the end of radiotherapy and progressively increases at nearly 6 years from the end of radiotherapy, the authors registered the maximum amount of HT in nearly 66% of patients [174-177]. Therefore they considered this toxicity as a late side effect of radiotherapy. The cumulative incidences [178] of HT in patients irradiated in the supraclavicular region at 3, 4, and 5 years after treatment were 10%, 22%, and 40%, respectively, whereas the cumulative incidences of HT in patients not irradiated in the supraclavicular region at 2, 3, and 5 years after RT were 3%, 8%, and 27%, respectively.

HT can be easily diagnosed with laboratory tests, including serum Free Thyroxin (FT4), Free Triiodothyronine (FT3) and Thyrotropin (TSH). Clinically evident HT is generally characterized as a reduction in serum FT4 concentration with a high serum TSH concentration and the existence of clinical symptoms like fatigue/weakness, weight gain, cognitive dysfunction, edema, cold intolerance, myalgia/paresthesia, depression, decreased hearing, constipation, dry skin, arthralgia, menorrhagia, and hoarseness. In the subclinical HT, on the contrary, the patients have no sympthoms but increased TSH concentration while FT4 level could be low or within normal limits.

Radiotherapy induced hypothyroidism is caused by the damage of small thyroid vessels and artheriosclerosis of larger vessels, additionally parenchymal injury of thyroid cells and secondary capsular fibrosis contribute toimprove toxicity [179].

The irradiation dose is a significant factor for the prediction of thyroid dysfunction [180-184] but, as previously stated, few researchers consider routinely thyroid as OaR and evaluate the Dose Volume Hystogram (DVH) of this organ. Kuten et al underlined a correlation between the dose to the tyrhoid and HT [185]. Yoden et al. [186] too analysed DVHs to evaluate the correlation between the percentage volume of the thyroid gland receiving doses and the thyroid function. These researchers have suggested that the percentage volume of the thyroid gland receiving doses between 10 and 60 Gy (V10 to V60) would represent a predictor of HT, and the percentage volume of the thyroid gland receiving doses V10 to V30 had a significant impact on the peak level of TSH. Similarly, Cella et al. [182] showed that the V30 value was the only predictor for HT. Additionally, Akgun et al. [181] reported that V30 was a statistically significant predictor for the development of HT. Kim, et al. [183] revealed that the thyroid V45 value can predict the development of HT and a V45 of 50% can be used as a threshold in RT planning. In contrast, Diaz et al. [187] reported no clear correlation between Dmean, Dmin, Dmax, V10 to V70 and HT. These data are confirmed by Alterio et al: in their experience too Dmean, Dmin, Dmax, V10, V30, V50, and 5 other point doses were not associated with HT [168]. In the study by Johansen et al. [173] 15 patients with small thyroid volumes experienced a higher risk to develop HT after RT, as less thyroid tissue receiving doses less than 30 Gy is available for adequate thyroxin making; anyway in their study no statistically significant intergroup variations were found between V20 and V50. According to the study published recently by Kanyilmaz G.et al. [178] HT group had smaller thyroid gland volume than those with normally functioning glands (13.25 cm3 vs 19.63 cm3, respectively, p = 0.08), but the results were not significant. There were no relationships observed between Dmin or V10 to V50 and HT, but Dmean and Dmax were statistically significant dosimetric predictors for HT. According to multivariate regression analysis, Dmean was the only prognostic factor that predicted HT. The authors concluded that Dmean > 21 Gy was a threshold value for the development of HT.

Some others risk factors were identified as predictors for thyroid disfunction. Radiotherapy volumes are statistically significant predictors in the study of Kanyilmaz et al. [178] 84 % of patients with HT were in the supraclavicular region irradiated group. Smith at al. [171] found no difference in the incidence of HT between breast cancer patients with more than 4 positive lymphnodes and therefore irradiated on the supraclavicular fossa if compared with patients treated on the breast/chest wall only.

Age is associated with the development of postradiation HT. Radiosensitivity of the thyroid gland is believed to decrease with increasing age [171,187] even if in the cohort of Kanyilmaz G, et al. [178] age did not have a statistically significant effect on HT.

Chemotherapy impact on HT is not clearly proven. According to Jereczek-Fossa, et al.’s review [179], the impact of chemotherapy and hormone therapies on the risk of HT was controversial. In Kanyilmaz et al. hemotherapy administration was not a statistically significant predictor of HT [178]. Hormonotherapy too has no impact on the risk of HT. This study confirmed that the extent of surgery and the nodal status, related with supraclavicular fossa irradiation, were also a statistically significan predictor for HT.

Prevention and treatment: The practice of nodal radiotherapy for early breast cancer is increasing in many centres. Sentinel node biopsy is now standard practice and there is a move towards considering radiotherapy instead of further surgery for some lymph node positive patients. This policies derive particularly from the results of the EORTC 10891-22023/ AMAROS trial which showed very low recurrence rates in the axilla with radiotherapy and surgery [188].

The EORTC trial 22922/10925 investigated 4004 patients for detecting the role of lymphnode irradiationin low-inermediate risk breast cancer patients [142]. They report that disease-free, distant disease-free survival and breast-cancer mortality were improved and a marginal effect on overall survival was detected in the group of patients irradiated on the regional lymphnode volumes. The Canadian MA20 study reported on 1832 women who were node positive or high-risk node negative following conservative breast surgery and either an axillary dissection or sentinel-lymph-node biopsy [143]. They report that the addition of regional nodal irradiation did not improve overall survival but reduced the rate of breast-cancer recurrence.

Both these trials have lead conseguently to an increased rate of irradiation of regional lymphnodes in breast cancer patiens. The technology improvement of radiation therapy surely allows a better dosimetric coverage of the lymphnodes PTV with the possibility to erogate a lower dose to the OaR.

This dose coverage was greatly improved for plans produced with modulated techniques, saving in this way as much as possible the thyroid gland.

Clinical HT is an irreversible condition, requiring lifelong treatment with thyroxin supplementation. Replacement treatment reverses all symptoms and signs of HT [180]. However, if replacement treatment is not carried out, both clinical and subclinical HT are related to worse perception of quality of life.

According to the American Association of Clinical Endocrinologists and the American Thyroid Association Guideline for HT [189] patients who had clinical or subclinical HT with elevated serum TSH concentration above 10 μU/mL were planned to require hormon replacement therapy.

Brachial plexus toxicity

Neurological symptoms and signs of brachial plexus dysfunction may occur as a complication of radiotherapy for the treatment of breast carcinoma, but may also result from metastatic spread of tumour, unrelated acute brachial neuritis, traumato the plexus during surgery or radiation-induced plexus tumours. The most common causes are Metastatic Brachial Plexopathy (MBP) and Radiation-Induced Brachial Plexopathy (RBP).

The wider indications to adjuvant radiotherapy on regional lymph nodes and specifically on suvraclavicular fossa highlight the risk of improved neurological toxicity caused by the unavoidable irradiation of the brachial plexus [142,143]. The recent publication of the results of the EORTC 22922/19025-MA20 and Danish trials indicates that locoregional radiotherapy has an impact on DFS, DMFS andOS, even not significative with a median FU of ten years. This benefit is detected even in patients with 1-3 positive axillary lymphnodes [190-193].

The timing to onset of brachial plexopathy and the first symptom of plexus disorder variy widely. The average interval rangesbetween 7.5 months tand 6 years while symptoms may develop decades after treatmet or with a relatively short latency as indicated by other authors [194-198]. Kori et al reported that symptoms from brachial plexopathy arose within 4 years from treatment [194], while Fathers et al. reported that the median time to onset of symptoms was 1.5 years [196]. The median follow-up time since RT completion was over 5.5 years in the report of Leong et al. [199] enough to identify arm morbidity, including plexopathy-associated symptoms and functional deficits, if present.

Conversely we will have to consider that several previous studies, in which larger doses per fraction was prescribed, demonstrated a latency period up to 20 years [195,197,198].

The mechanism of plexophaty is believed to be a combination of failure of cellular proliferation and localized ischemia. The net result is fibrosis of the neural and perineural soft tissues secondary to microvascular insufficiency. This, in turn, leads to ischemic damage to the axons and Schwann cells [200].

Sensory symptoms, such as numbness, paresthesia and dysesthesia, swelling and weakness of the arm are the predominant clinical evidence of such a damage. These neurologic symptoms can be progressive and may lead to a weak and edematous arm.

Most radiation plexopathies are painless, but when present, pain symptoms are usually limited to the shoulder and proximal arm. Such pain usually is rated as mild to moderate in intensity. Significant pain complaints are more commonly associated with recurrent tumor than with radiation plexopathy [200].

The frequency of radiation-induced brachial plexopathy has declined over the past 60 years and depends significantly on both the radiation dose and the proximity of the radiation volume to the underlying plexus. In the 1950s, the incidence was as high as 66% after 60-Gy total dose to the axilla and supraclavicular area using 5 Gy/fraction. The current incidence is 1-2% in patients receiving a typical dose of less than 55 Gy [201].

Breast carcinoma accounts for 40-75% of reported cases of brachial plexophaty, followed by lung carcinoma and lymphoma [194-196].

Paresthesia is a fairly common sympton among women treated with post-operative radiotherapy for breast cancer in Sweden, with a prevalence of approximately 17% [202].

Lundstedt et al reported 25% of paresthesia after RT of supraclavicular lymphnodes, 13% in patients without RT [203].

Risk Factors

The radiation dose, treatment technique, concomitant use of chemotherapy, surgical lymph node dissection and underlying comorbidities such as diabetes, hypertension, obesity, and vascular disease have been demonstrated significant association with the development of radiation injury to the brachial plexus [204,205].

Given that breast cancer often is treated with radiation therapy, women experience a greater incidence and prevalence of radiation-induced brachial plexopathy than men [206].

Advanced age may be a risk factor for the development of brachial plexopathy after radiation treatment [207].

It is well known that high doses to the brachial plexus increase the risk of neuropathy, as shown by Powell, et al [208], Olsen, et al [209] and Johansson, et al [195] in their comparisons of different fractionation schedules.

The effects of nodal RT total dose and dose per fraction on the risk of brachial plexopathy have been evaluated in studies conducted before the era of CT planning. In a retrospective study of 449 patients treated from 1982-1984, Powell et al reported that symptoms of brachial plexopathy increased from 1% with 54 Gy in 30 fractions (1.8 Gy/fx) to 6% with 45 Gy in 15 fx (3 Gy/fx). (p = 0.09) [208].

Galecki et al. suggested that the rate of radiation induced plexophaty increased sharply with doses beyond 55 Gy (delivered at 2 Gy/fx) [206].

As shown in a recent publication by Leong et al. [199], limiting hypofractionation to fraction sizes 2.25 to 2.5 Gy/day with a compensatory reduction in total dose may reduce overall treatment time, without increasing the risk of shoulder or brachial plexus fibrosis. In the UK Standardisation of Breast Radiotherapy (START) A and B trials, nodal radiotherapy was prescribed in 14% and 7% of subjects respectively [210]. Hypofractionation in the START trials was not associated with increased risks of arm symptoms, brachial plexopathy or lymphedema [210]. The START A trial reported one case of brachial plexopathy (0.1%) if a schedule of 41.6 Gy in 13 fractions (3.2Gy/fx) was used. The START B trial reported no case of plexopathy among 82 women treated with 40 Gy in 15 fractions (2.67 Gy/fx). These findings were confirmed by a retrospective series of 257 patients treated with HFRT 42 Gy in 15 fx (2.8Gy/fx) which reported no cases with brachial plexopathy with a median follow-up of 6 year [211].

Another randomized trial [212] reinforce these data. In the British Columbia post-mastectomy RT (PMRT) randomized trial [213], locoregional radiotherapy was delivered over 16 fractions in which the mid-axillary dose was 35 Gy, a dose lower than the HF nodal RT prescriptions evaluated in the current study. The short PMRT regimen resulted in a significant reductions in the risk of loco-regional recurrence and improvements in 20-year breast cancer survival. Although limited by cross-trial comparison, the locoregional control and survival outcomes observed in the British Columbia trial were consistent with outcomes in the Danish PMRT trial in which a conventiona fractionation was used. In a recent study published by Nelson Leong, et al. [199], hypofractionation was not associated with increased patient reported arm symptoms or functional deficits compared to conventional one.

In the report by Lundstedt, et al. [203] paresthesia in relation to dose/volume was evaluated. Paresthesia was reported in 25% of patients treated with raditherapy to the supraclavicular lymphnodes, with a V40Gy ≥ 13.5cm3, compared with 13% if no radiotherapy was prerscribed, RR1.83 (95% CI 1.13-2.95).

A irradiated volume effect was suggested by Emami, et al [214] when they irradiated the brachial plexus by thirds: radiotherapy on one third induced less toxicity probably because one third was a little more radio-resistant than all 3 at the same time. Volume effects after irradiation of the brachial plexus has also been suggested by Amini et al [215] and Eblan et al [216].

Rehabilitation Program

Therapeutic modalities should focus on reducing pain, strengthening, preservation of range of motion and limiting lymphedema. The interventions and modalities stricly depend on the kind of impairments (i.e. weakness, pain, lymphedema, range of motion) and are based on occupational therapy and sensorial/motorial re-education techniques [217]. Anticonvulsants and antidepressants are used to manage severe muscle spasms and provide pain relief in neuralgia.

Prognosis

One third of patients experience severe progression of their radiation-induced plexopathy, whereas the remainder a gradual one. A mild form of reversible radiation plexopathy may present rarely.

Patient Education

Patients should be awared on the expected progressive course of radiation plexopathy. A home exercise program should be considered to preserve strength and range of motion of shoulder and arm. Susceptibility to trauma and infection due to altered sensation, edema and fibrotic tissue should be discussed.

Rare Toxicity and Secondary Tumors

The risk of secondary malignancies has been analysed by the Danish Breast Cancer Collaborative Group: 18% of the pre-menopausal breast cancer patients treated with surgery alone developed a second non breast cancer: ovarian, endometrial and biliar tract cancer were the most frequent ones [218].

The annual risk ratio of radio-induced secondary cancer is 1.2, increases with follow up and is strictly correlated with the age at radiation treatment [219].

Two SEER studies evidenced no increased risk of secondary cancer in breast cancer patients treated with radiotherapy in the volumes receiving if < 1Gy, while the risk increased if dose overcame 1 Gy in organs such as pleura, esophagus, lung, bone, soft tissue and contralateral breast. Only 5% of controlateral tumors and 6% of the other cancers could be correlated to the previous radiation treatment in a population of 182000 women [220,221].

The risk of radio-induced sarcoma is nearly 0.2% at 10 years, tipically are induced by high doses and so arise usually inside the irradiated volumes. In case of angiosarcoma the interval should be lower, between1 and 2.5 years from the end of radiotherapy [222].

The risk of controlateral breast cancer depends on several factors (hormonal, genetics) while radiotherapy seems to have a role in improving the risk only in young women if irradiated on the internal breast quadrants [223].

A slightly improvement of lung cancer has been detected if high volume of lung has been irradiatiated especially in smokers patients and if high lung volumes have been irradiated [224,225].

Conclusions

Radiation side effects have been extensively analyzed and categorized usig international scales (EORTC/RTOG, NCI CTCAE). The prevention and treatment of the most frequent radiotherapy toxicities are nowadays better standardized, due to technical improvement and dedicated personnel (phisicians, nurses, radiographers). Conversely we lack clear data on the interaction between radiotherapy and the other specific treatments (i.e. chemo/hormono/immunotherapy, surgery, reconstructive surgery) and their best sequences for reducing toxicity. Moreover, due to recent publications, we will have to consider the irradiation of regional lymphnodes even in intermediate risk patients. This new attitude enforce more attention to neglected toxicities, such as the thyroid and brachial plexus ones, especially in these long term survival patients.

The patients characteristics have to be taken into account too for prescribing a tailored radiotherapy with the lowest collateral effects and toxicities.

Technological improvements allow us to obtain more dedicated treatments, image guided radiotherapy assures a proper positioning of the patients during the treatment and the respiratory gating allows a reduced dose to the hearth and the anterior coronary artery.

We aim that a wider diffusion of these new tools will further decrease acute and late radiotherapy toxicity.

  1. Early Breast Cancer Trialists’ Collaborative Group, Darby S, McGale P, Correa C, Taylor C, Arriagada R, Clarke M, et al. Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: meta-analysis of individual patient data for 10,801 women in 17 randomised trials. Lancet. 2011; 378: 1707-1716. https://goo.gl/NzM7yA
  2. Early Breast Cancer Trialists’ Collaborative Group, McGale P, Taylor C, Correa C, Cutter D, Duane F, et al. Effect of radiotherapy after mastectomy and axillary surgery on 10-year recurrence and 20-year breast cancer mortality: meta-analysis of individual patient data for 8135 women in 22 randomised trials. Lancet. 2014; 383: 2127-2135. https://goo.gl/tafjh7
  3. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995; 31: 1341-1346. https://goo.gl/Xtq8jS
  4. National Cancer Institute. Common Terminology Criteria for Adverse Events v4.0. NIH; 2009. NIH publication # 09-7473. Available from: Accessed July 10, 2016.
  5. Waljee JF, Hu ES, Ubel PA, Smith DM, Newman LA, Alderman AK. Effect of esthetic outcome after breast-conserving surgery on psychosocial functioning and quality of life. J Clin Oncol. 2008; 20: 3331-3337. https://goo.gl/yj7WGQ
  6.  Zeman EM. The biological basis of radiation oncology. In: Gunderson LL, Tepper JE, Bogart JA, editors. Clinical Radiation Oncology. 4th ed. Philadelphia. PA; 2016: 2-40. https://goo.gl/s7Eg94
  7. Quimby E, Maccomb W. Further studies on the rate of recovery of humanskin from the effects of roentgen or gamma rays. Radiology. 1937; 29: 305–312. https://goo.gl/fLcBD6
  8. Bourland JD. Radiation oncology physics. In: Gunderson LL, TepperJE, Bogart JA, editors. Clinical Radiation Oncology. 4th ed. Philadelphia. PA; 2016: 93-147.
  9. Liu K, Kasper M, Trott KR. Changes in keratinocyte differentiation during accelerated repopulation of the irradiated mouse epidermis. Int J Radiat Biol. 1996; 69: 763-769. https://goo.gl/F9cddo
  10. Schmuth M, Sztankay A, Weinlich G, Linder DM, Wimmer MA, Fritsch PO, et al. Permeability barrier function of skin exposed to ionizing radiation. Arch Dermatol. 2001; 137: 1019-1023. https://goo.gl/yX2pVL
  11. Trott KR, Shirazi A, Heasman F. Modulation of accelerated repopulationin mouse skin during daily irradiation. Radiother Oncol. 1999; 50: 261-266. https://goo.gl/4e1UXS
  12. Koenig TR, Wolff D, Mettler FA, Wagner LK. Skin injuries fromfluoroscopically guided procedures: part 1, characteristics of radiation injury. AJR Am J Roentgenol. 2001; 177: 3-11. https://goo.gl/ZJx56x
  13. Archambeau JO, Ines A, Fajardo LF. Response of swine skin microvasculature to acute single exposures of X rays: quantification of endothelial changes. Radiat Res. 1984; 98: 37-51. https://goo.gl/zH9Rpa
  14. Hopewell J, Calvo W, Jaenke R, Reinhold H, Robbins M, Whitehouse E. Microvasculature and Radiation Damage. In: Hinkelbein W, Bruggmoser G, Frommhold H, Wannenmacher M, editors. Acute andLong-Term Side-Effects of Radiotherapy Biological Basis and ClinicalRelevance. Berlin. Springer. 1993; 1–16. https://goo.gl/eddjMs
  15. Muller K, Meineke V. Radiation-induced alterations in cytokine productionby skin cells. Exp Hematol. 2007; 35: 96-104. https://goo.gl/a8QHYL
  16. Petit-Frere C, Capulas E, Lyon DA, Norbury CJ, Lowe JE, Clingen PH, et al. Apoptosis and cytokine releaseinduced by ionizing or ultraviolet B radiation in primary and immortalized human keratinocytes. Carcinogenesis. 2000; 21: 1087-1095. https://goo.gl/qAiJMx
  17. Beetz A, Messer G, Oppel T, van Beuningen D, Peter RU, Kind P. Inductionof interleukin 6 by ionizing radiation in a human epithelial cell line: control by corticosteroids. Int J Radiat Biol. 1997; 72: 33-43. https://goo.gl/dttse4
  18. Hallahan DE, Spriggs DR, Beckett MA, Kufe DW, Weichselbaum RR. Increased tumor necrosis factor alpha mRNA after cellular exposure to ionizing radiation. Proc Natl Acad Sci U S A. 1989; 86: 10104-10107. https://goo.gl/ogxNGM
  19. Martin M, Lefaix J, Delanian S. TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys. 2000; 47: 277-290. https://goo.gl/SEGqA4
  20.  Pohlers D, Brenmoehl J, Loffler I, Müller CK, Leipner C, Schultze-Mosgau S, et al. TGF-beta and fibrosis in different organs – molecular pathway imprints. Biochim Biophys Acta. 2009; 1792: 746-756. https://goo.gl/nJFvZe
  21. Mendelsohn FA, Divino CM, Reis ED, Kerstein MD. Wound care after radiation therapy. Adv Skin Wound Care. 2002; 15: 216-224. https://goo.gl/pa9XMm
  22. Brown KR, Rzucidlo E. Acute and chronic radiation injury. J Vasc Surg. 2011; 53: 15-21. https://goo.gl/W8Q4TJ
  23. Harper JL, Franklin LE, Jenrette JM, Aguero EG. Skin toxicity during breast irradiation: pathophysiology and management. South Med J. 2004; 97: 989-993. https://goo.gl/vSWL9G
  24. Ryan JL. Ionizing radiation: the good, the bad, and the ugly. J InvestDermatol. 2012; 132: 985-993. https://goo.gl/uFA8Yn
  25. McQuestion M. Evidence-based skin care management in radiation therapy: clinical update. Semin Oncol Nurs. 2011; 27: 1-17. https://goo.gl/B13cw7
  26. Ismaili N, Mellas N, Masbah O, Elmajjaoui S, Arifi S, Bekkouch I, et al. Concurrent chemoradiotherapy in adjuvant treatment of breast cancer. Radiat Oncol. 2009; 4: 12. https://goo.gl/DBEsge
  27. Livi L, Meattini I, Scotti V, C Saieva, G Simontacchi, L Marrazzo, et al. Concomitant adjuvant chemoradiation therapy with anthracycline-based regimens in breast cancer: a single centre experience. Radiol Med. 2011; 116: 1050-1058. https://goo.gl/jErXBn
  28. Hanna YM, Baglan KL, Stromberg JS, Vicini FA, A Decker D. Acute and subacute toxicity associated with concurrent adjuvant radiation therapy and paclitaxel in primary breast cancer therapy. Breast J. 2002; 8: 149-153. https://goo.gl/2udVTF
  29. Chawla AK, Kachnic LA, Taghian AG, Niemierko A, Zapton DT, Powell SN. Radiotherapy andbreast reconstruction: complications and cosmesis with TRAM versus tissue expander/implant. Int J Radiat Oncol Biol Phys. 2002; 54: 520-526. https://goo.gl/RR8Uf9
  30. Albornoz CR, Matros E, McCarthy CM, Klassen A, Cano SJ, Alderman AK, et al. Implant breast reconstruction and radiation: a multicenter analysis of long-term health-related quality of life and satisfaction. Ann Surg Oncol. 2014; 21: 2159-2164. https://goo.gl/9xH2Bq
  31. Rutter C, Qin L, Higgins S, Moran M, Evans S. Dosimetric and clinicalpredictors of the development of moist desquamation in breast cancer irradiation. J Radiat Oncol. 2014; 3: 147-152. https://goo.gl/JQ4mmq
  32. Macmillan MS, Wells M, MacBride S, Raab GM, Munro A, MacDougall H. Randomized comparison of dry dressings versus hydrogelin management of radiation-induced moist desquamation. Int J Radiat Oncol Biol Phys. 2007; 68: 864-872. https://goo.gl/RCax2e
  33. Pollard JM, Gatti RA. Clinical radiation sensitivity with DNA repair disorders: an overview. Int J Radiat Oncol Biol Phys. 2009; 74: 1323–1331. https://goo.gl/nXX1KE
  34. Chen MF, Chen WC, Lai CH, Hung CH, Liu KC, Cheng YH. Predictive factors of radiation-induced skin toxicity in breast cancer patients. BMC Cancer. 2010; 10: 508. https://goo.gl/nRdzec
  35. Harsolia A, Kestin L, Grills I, Wallace M, Jolly S, Jones C, et al. Intensity-modulated radio therapy results in significant decrease in clinical toxicities compared with conventional wedge-based breast radiotherapy. Int J Radiat OncolBiol Phys. 2007; 68: 1375-1380. https://goo.gl/DMZrAd
  36.  Pignol JP, Olivotto I, Rakovitch E, Gardner S, Sixel K, Beckham W, et al. A multicenter randomizedtrial of breast intensity-modulated radiation therapy to reduce acuteradiation dermatitis. J Clin Oncol. 2008; 26: 2085–2092. https://goo.gl/tMuVsM
  37.  Freedman GM, Li T, Nicolaou N, Chen Y, Ma CC, Anderson PR. Breastintensity-modulated radiation therapy reduces time spent with acute dermatitis for women of all breast sizes during radiation. Int J Radiat Oncol Biol Phys. 2009; 74: 689–694. https://goo.gl/iCQ7e4
  38. Mulliez T, Speleers B, Madani I, De Gersem W, Veldeman L, De Neve W. Whole breast radiotherapy in prone and supine position: is there aplace for multi-beam IMRT? Radiat Oncol. 2013; 8: 151. https://goo.gl/d5gdeU
  39. Wang EH, Mougalian SS, Soulos PR, Rutter CE, Evans SB, Haffty BG et al. Adoption of hypofractionated whole-breast irradiation for early-stage breast cancer: a National Cancer Data Base analysis. Int J Radiat Oncol Biol Phys. 2014; 90: 993-1000. https://goo.gl/Jkec51
  40. Whelan TJ, Pignol JP, Levine MN, Julian JA, MacKenzie R, Parpia S, et al. Long-term results of hypofractionated radiation therapy for breast cancer. N Engl J Med. 2010; 362: 513-520. https://goo.gl/hQBmu3
  41.  Bentzen SM, Agrawal RK, Aird EG, Barrett JM, Barrett-Lee PJ, Bliss JM, et al. The UK Standardisation of Breast Radiotherapy (START) Trial A of radiotherapy hypofractionation for treatment of early breast cancer: a randomised trial. Lancet Oncol. 2008; 9: 331-341. https://goo.gl/TspkjK
  42. Bentzen SM, Agrawal RK, Aird EG, Barrett JM, Barrett-Lee PJ, Bentzen SM, et al. The UK Standardisation of Breast Radiotherapy (START) trial B of radiotherapy hypofractionation for treatment of early breast cancer: a randomised trial. Lancet. 2008; 371: 1098-1107. https://goo.gl/1P4VDy
  43. Volker Rudat, Alaa Nour, Salam Abou Ghaida, Aziz Alaradi. Impact of hypofractionation and tangential beam IMRT on the acute skin reaction in adjuvant breast cancer radiotherapy 2016 Radiat Oncol. 2016; 11: 100. Published online. 2016; 11: 100. https://goo.gl/GfBXDu
  44. Poortmans PM, Collette L, Bartelink H, Struikmans H, Van den Bogaert WF, Fourquet A, et al. The addition of a boost dose on the primary tumour bed after lumpectomy in breast conserving treatment for breast cancer. A summary of the results of EORTC 22881-10882 "boost versus no boost" trial. Cancer Radiother. 2008; 12: 565-570. https://goo.gl/zbHmUt
  45. Collette S, Collette L, Budiharto T, Horiot JC, Poortmans PM, Struikmans H, et al. Predictors of the risk offibrosis at 10 years after breast conserving therapy for earlybreast cancer: a study based on the EORTC Trial 22881-10882 ‘boost versus no boost’. Eur J Cancer. 2008; 44: 2587-2699. https://goo.gl/WpMmre
  46. Jagsi R, Ben-David MA, Moran JM, Marsh RB, Griffith KA, Hayman JA, et al. Unacceptable cosmesisin a protocol investigating intensity-modulated radiotherapy withactive breathing control for accelerated partial-breast irradiation. Int J Radiat Oncol Biol Phys. 2010; 76: 71–78. https://goo.gl/88XaT8
  47. Jae Myoung Noh, Doo Ho Choi, Hyojung Park, Seung Jae Huh, Won Park, Seung Won Seol, et al. Comparison of acute skin reaction following morning versus late afternoon radiotherapy in patients with breast cancer who have undergone curative surgical resection. J Radiat Res. 2014; 55: 553-558. https://goo.gl/eBZoxU
  48. Bartelink H, Maingon P, Poortmans P, Weltens C, Fourquet A, Jager J, et al. Whole-breast irradiationwith or without boost for patients treated with breastconservingsurgery for early breast cancer: 20-year follow-up of a randomised phase 3 trial. Lancet Oncol. 2015; 16: 47-56. https://goo.gl/KFZNeU
  49. Fernando IN, Ford HT, Powles TJ, Ashley S, Glees JP, Torr M, et al. Factors affecting acute skin toxicity in patients having breast irradiation after conservative surgery: a prospective study of treatment practice at the Royal Marsden Hospital. Clin Oncol. 1996; 8: 226-233. https://goo.gl/JGwTmF
  50.  Fisher J, Scott C, Stevens R, Marconi B, Champion L, Freedman GM, et al. Randomized phase III study comparing Best Supportive Care to Biafine as a prophylactic agent for radiation-induced skin toxicity for women undergoing breast irradiation: Radiation Therapy Oncology Group (RTOG) 97-13. Int J Radiat Oncol Biol Phys. 2000; 48: 1307-1310. https://goo.gl/9ifVQr
  51. Back M, Guerrieri M, Wratten C, Steigler A. Impact of radiation therapy on acute toxicity in breast conservation therapy for early breast cancer. Clin Oncol. 2004; 16: 12-16. https://goo.gl/PdJaUt
  52. Rutter C, Qin L, Higgins S, Moran M, Evans S. Dosimetric and clinical predictors of the development of moist desquamation in breast cancer irradiation. J Radiat Oncol. 2014; 3: 147–152. https://goo.gl/7C66yb
  53. Twardella D, Popanda O, Helmbold I, Ebbeler R, Benner A, von Fournier D, et al. Personal characteristics, therapy modalities and individual DNA repair capacity as predictive factors of acute skin toxicity in an unselected cohort of breast cancer patients receiving radiotherapy. Radiother Oncol. 2003; 69: 145-153. https://goo.gl/gUG72i
  54. Wright JL, Takita C, Reis IM, Zhao W, Lee E, Hu JJ. Racial variations in radiation-induced skin toxicity severity: data from a prospective cohort receiving postmastectomy radiation. Int J Radiat Oncol Biol Phys. 2014; 90: 335-343. https://goo.gl/eCzmuH
  55. Macmillan MS, Wells M, MacBride S, Raab GM, Munro A, MacDougall H. Randomized comparison of dry dressings versus hydrogel in management of radiation-induced moist desquamation. Int J Radiat Oncol Biol Phys. 2007; 68: 864-872. https://goo.gl/RovXPi
  56. Pollard JM, Gatti RA. Clinical radiation sensitivity with DNA repair disorders: an overview. Int J Radiat Oncol Biol Phys. 2009; 74: 1323-1331. https://goo.gl/7mScAJ
  57. Omidvari S, Saboori H, Mohammadianpanah M, Mosalaei A, Ahmadloo N, Mosleh-Shirazi MA, et al. Topical betamethasone for prevention of radiation dermatitis. Indian J Dermatol Venereol Leprol. 2007; 73: 209. https://goo.gl/s6BYxn
  58. Ulff E, Maroti M, Serup J, Falkmer U. A potent steroid cream is superior to emollients in reducing acute radiation dermatitis in breast cancer patients treated with adjuvant radiotherapy. A randomised study of betamethasone versus two moisturizing creams. Radiother Oncol. 2013; 108: 287-292. https://goo.gl/tjRvE4
  59. Wells M, Macmillan M, Raab G, MacBride S, Bell N, MacKinnon K, et al. Does aqueous or sucralfate cream affect the severity of erythematous radiation skin reactions? A randomised controlled trial. Radiother Oncol. 2004; 73: 153-162.
  60. Pommier P, Gomez F, Sunyach MP, D’Hombres A, Carrie C, Montbarbon X. Phase III randomized trial of Calendula officinalis compared with trolamine for the prevention of acute dermatitis during irradiation for breast cancer. J Clin Oncol. 2004; 22: 1447–1453. https://goo.gl/QeEGv7
  61. Herst PM, Bennett NC, Sutherland AE, Peszynski RI, Paterson DB, Jasperse ML. Prophylactic use of Mepitel Film prevents radiation induced moist desquamation in an intra-patient randomised controlled clinical trial of 78 breast cancer patients. Radiother Oncol. 2014; 110: 137–143. https://goo.gl/iUqU7q
  62. D’Haese S, Van Roy M, Bate T, Bijdekerke P, Vinh-Hung V. Management of skin reactions during radiotherapy in Flanders (Belgium): a study of nursing practice before and after the introduction of a skincare protocol. Eur J Oncol Nurs. 2010; 14: 367–372. https://goo.gl/u5Hz8W
  63. Burch SE, Parker SA, Vann AM, Arazie JC. Measurement of 6-MVX-ray surface dose when topical agents are applied prior to external beam irradiation. Int J Radiat Oncol Biol Phys. 1997; 38: 447–451. https://goo.gl/5D2xkj
  64. Watson LC, Gies D, Thompson E, Thomas B. Randomized control trial: evaluating aluminum-based antiperspirant use, axilla skin toxicity, and reported quality of life in women receiving external beam radiotherapy for treatment of Stage 0, I, and II breast cancer. Int J Radiat Oncol Biol Phys. 2012; 83: 29-34. https://goo.gl/7ikaks
  65. Gee A, Moffitt D, Churn M, Errington R. A randomised controlled trial to test a non-metallic deodorant used during a course of radiotherapy. J Radiother Pract. 2000; 1: 205–212. https://goo.gl/drgSY7
  66.  Herst PM, Bennett NC, Sutherland AE, Peszynski RI, Paterson DB, Jasperse ML. Prophylactic use of Mepitel Film prevents radiation induced moist desquamation in an intra-patient randomised controlled clinical trial of 78 breast cancer patients. Radiother Oncol. 2014; 110:137-143. https://goo.gl/eBE2VF
  67. BC Cancer Agency. Symptom Management Guidelines: Radiation Dermatitis. Available from: Accessed November 8, 2016. https://goo.gl/zzKvf9
  68. Chen MF, Chen WC, Lai CH, Hung CH, Liu KC, Cheng YH. Predective factors of radiation-induced skin toxicity in breast cancer patients. BMC Cancer. 2010; 10: 508. https://goo.gl/cjBA4G
  69.  Dorn PL, Corbin KS, Al-Hallaq H, Hasan Y, Chmura SJ. Feasibility and acute toxicity of hypofractionated radiation in large-breasted patients. Int J Radiat Oncol Biol Phys. 2012; 83: 79-83. https://goo.gl/6nsMF7
  70. Tsoutsou PG, Koukourakis MI. Radiation pneumonitis and fibrosis: mechanisms underlying its pathogenesis and implications for future research. Int J Radiat Oncol Biol Phys. 2006; 66: 1281-1293. https://goo.gl/WtXkoh
  71. Hassaballa HA, Cohen ES, Khan AJ, Ali A, Bonomi P, Rubin DB. Positron emission tomography demonstrates radiation-induced changes to nonirradiated lungs in lung cancer patients treated with radiation and chemotherapy. Chest. 2005; 128: 1448-1452. https://goo.gl/qZKMHr
  72. Choi YW, Munden RF, Erasmus JJ, Park KJ, Chung WK, Jeon SC, et al. Effects of radiation therapy on the lung: radiologic appearances and differential diagnosis. Radiographics: a review publication of the Radiological Society of North America, Inc. 2004; 24: 985–997. https://goo.gl/tY9Frc
  73. Ikezoe J, Takashima S, Morimoto S, Kadowaki K, Takeuchi N, Yamamoto T, et al. CT appearance of acute radiation-induced injury in the lung. AJR American journal of roentgenology. 1988; 150: 765–770. https://goo.gl/eZNpp3  
  74. Marks LB, Fan M, Clough R, Munley M, Bentel G, Coleman RE, et al. Radiation-induced pulmonary injury: symptomatic versus subclinical endpoints. Int J Radiat Biol. 2000; 76: 469-475. https://goo.gl/KFHzy9
  75. Hernberg M, Virkkunen P, Maasilta P, Keyriläinen J, Blomqvist C, Bergh J, et al. Pulmonary toxicity after radiotherapy in primary breast cancer patients: results from a randomized chemotherapy study. Int J Radiat Oncol Biol Phys. 2002; 52: 128-136. https://goo.gl/VrSauB   
  76. Haston CK, Zhou X, Gumbiner-Russo L, Irani R, Dejournett R, Gu X, et al. Universal andradiation-specific loci influence murine susceptibility to radiation- induced pulmonary fibrosis. Cancer Res. 2002; 62: 3782-3788. https://goo.gl/ixNRmg
  77. Yuan X, Liao Z, Liu Z, Wang LE, Tucker SL, Mao L, et al. Single Nucleotide Polymorphism at rs1982073: T869C of the TGFβ1 Gene Is Associated With the Risk of Radiation Pneumonitis in patients with non-small-cell lung cancer treated with definitive radiotherapy. https://goo.gl/9T3Zg4
  78. Livi L, Meattini I, Marrazzo L, Simontacchi G, Pallotta S, Saieva C, et al. Accelerated partial breast irradiation using intensity-modulated radiotherapy versus whole breast irradiation: 5-year survival analysis of a phase 3 randomised controlled trial. Eur J Cancer. 2015; 51: 451-463. https://goo.gl/zv79iy
  79.  Lind PA, Wennberg B, Gagliardi G, Fornander T. Pulmonary complicationsfollowing different radiotherapy techniques for breastcancer, and the association to irradiated lung volume and dose. Breast Cancer Res Treat. 2001; 68: 199–210. https://goo.gl/WoQZTu
  80. Holli K, Pitka¨nen M, Ja¨rvenpa¨a¨ R, Rajala J, Lahtela S, Hyödynmaa S, et al. Early skin and lung reactions in breast cancer patients after radiotherapy: prospectivestudy. Radiother Oncol. 2002; 64: 163-169. https://goo.gl/4Lv9ms
  81. Gagliardi G, Bjohle J, Lax I, Ottolenghi A, Eriksson F, Liedberg A, et al. Radiation pneumonitis afterbreast cancer irradiation: analysis of the complication probabilityusing the relative seriality model. Int J Radiat Oncol Biol Phys. 2000; 46: 373-381. https://goo.gl/VaDT79
  82. Kaha´n Z, Csenki M, Varga Z, Szil E, Cserháti A, Balogh A, et al. The risk of early and late lung sequelae after conformal radiotherapy in breast cancer patients.Int J Radiat Oncol Phys. 2007; 68: 673-681. https://goo.gl/ZijNrJ
  83. Gokula K, Earnest A, Wong LC. Meta-analysis of incidence of early lung toxicity in 3-dimensional conformal irradiation of breast carcinomas. Radiat Oncol. 2013; 8: 268. https://goo.gl/j4bDKE
  84. Lingos TI, Recht A, Vicini F, Abner A, Silver B, Harris JR. Radiation pneumonitis [3]in breast cancer patients treated with conservative surgery and radiation therapy. Int J Radiat Oncol Biol Phys. 1991; 21: 355-360. https://goo.gl/QVebi3
  85. Tokatli F, Kaya M, Kocak Z, Ture M, Mert S, Unlu E, et al. Sequential pulmonary [6]effects of radiotherapy detected by functional and radiological end points in women with breast cancer. Clin Oncol (R Coll Radiol). 2005; 17: 39-46. https://goo.gl/ni6Xwz
  86.  Lind PA, Marks LB, Hardenbergh PH, Clough R, Fan M, Hollis D, et al. Technical factors associated with radiation pneumonitis after local +_ regional radiation therapy for breast cancer. Int J Radiat Oncol Biol Phys. 2002; 52: 137-143. https://goo.gl/5Fop7w
  87. Bornstein BA, Cheng CW, Rhodes LM, Rashid H, Stomper PC, Siddon RL, et al. Can simulation measurements be used to predict the irradiated lung volume in the tangential fields in patients treated for breast cancer? Int J Radiat Oncol Biol Phys. 1990; 18: 181-187. https://goo.gl/pzwTGt
  88.  Kong FM, Klein EE, Bradley JD, Mansur DB, Taylor ME, Perez CA, et al. The impact of central lung distance, maximal heart distance, and radiation technique on the volumetric dose of the lung and heart for intact breast radiation. Int J Radiat Oncol Biol Phys. 2002; 54: 963-971. https://goo.gl/cqxz5D
  89. Dehing-Oberije C, De Ruysscher D, Van Baardwijk A, Yu S, Rao B, Lambin P. The Importanceof patient characteristics for the prediction of radiation-induced lung toxicity. Radiother Oncol. 2009; 91: 421-426. https://goo.gl/yADP7X
  90. Hernando ML, Marks LB, Bentel GC, Zhou S-M, Hollis D, Das SK, et al. Radiation-induced pulmonary toxicity: a dose-volume histogram analysis in 201 patients with lung cancer. Int J Radiat Oncol Biol Phys. 2001; 51: 650-659. https://goo.gl/rR84iA
  91. Abe M, Miwa H, Ema R, Miki Y, Tomita K, Nakamura H, et al. Risk Factors Of Symptomatic Radiation Pneumonitis In Post-Operative Breast Cancer Patients. Am J Resp Crit Care. 2012; 185: 5727. https://goo.gl/UnWyqN
  92. Allen AM, Prosnitz RG, Ten Haken RK, Normolle DP, Yu X, Zhou S-m, et al. Body mass index predicts the incidence of radiation pneumonitis in breast cancer patients. The Cancer Journal. 2005; 11: 390-398. https://goo.gl/A36ZHK
  93. Mehta V. Radiation pneumonitis and pulmonary fibrosis in Non-Small-Celllung cancer: pulmonary function, prediction and prevention. Int J Radiat Oncol Biol Phys. 2005; 63: 5-24. https://goo.gl/2GA4uE
  94.  Petit S, Van Elmpt W, Oberije C, Vegt E, Dingemans AM, Lambin P, et al. [18F] fluorodeoxygluose uptake patternsin lung before radiotherapy identify areas susceptible to radiation-induced lungtoxicity in non-small- cell lung cancer patients. Int J Radiat Oncol Biol Phys 2011; 81: 698-705. https://goo.gl/6puoSC
  95. De Ruysscher D, Houben A, Aerts HJ, Dehing C, Wanders R, Ollers M, et al. Increased 18F-deoxyglucoseuptake in the lung during the first weeks of radiotherapy is correlated with subsequent Radiation-Induced Lung Toxicity (RILT): A prospective pilot study. Radiother Oncol. 2009; 91: 415-420. https://goo.gl/As8hix
  96. Yarnold J, Bentzen SM, Coles C, Haviland J. Hypofractionated wholebreastradiotherapy for women with early breast cancer: mythsand realities. Int J Radiat Oncol Biol Phys. 2011; 79: 1-9. https://goo.gl/vjUP72
  97. Mast ME, van Kempen-Harteveld L, Heijenbrok MW, Kalidien Y, Rozema H, Jansen WP, et al. Leftsidedbreast cancer radiotherapy with and without breath-hold: does IMRT reduce the cardiac dose even further. Radiother Oncol. 2013; 108: 248-253. https://goo.gl/m7WDgk
  98. Kubo A, Osaki K, Kawanaka T, Furutani S, Ikushima H, Nishitani H. Risk factors for radiation pneumonitis caused by whole breast irradiation following breast conserving surgery. J Med Invest. 2009; 56: 99-110. https://goo.gl/siYike
  99. Blom Goldman U, Wennberg B, Svane G, Bylund H, Lind P. Radiat Oncol.Reduction of radiation pneumonitis by V20-constraints in breast cancer. Radiat Oncol. 2010; 5: 99. https://goo.gl/LkKyS7
  100. Rancati T, Wennberg B, Lind P, Svane G, Gagliardi G. Early clinical and radiologicalpulmonary complications following breast cancer radiationtherapy: nTCP fit with four different models. Radiother Oncol. 2007; 82: 308-316. https://goo.gl/pEs43r
  101. Formenti SC, DeWyngaert JK, Jozsef G, Goldberg JD. Prone vs. supinepositioning for breast cancer radiotherapy. JAMA. 2012; 308: 861-863. https://goo.gl/8JXAbh
  102. Ramella S, Trodella L, Mineo TC, Pompeo E, Stimato G, Gaudino D, et al. Adding Ipsilateral V20 and V30 to Conventional Dosimetric Constraints Predicts Radiation Pneumonitis in Stage IIIA–B NSCLC Treated With Combined-Modality Therapy. Int J Radiat Oncol Biol Phys. 2010; 76: 110-115. https://goo.gl/ZtJsca
  103. Lee TF, Chao PJ, Chang L, Ting HM, Huang YJ. Developing multivariable normal tissue complication probability model to predict the incidence of symptomatic radiation pneumonitis among breast cancer patients. PLoS ONE. 2015; 10: 0131736. https://goo.gl/RFkgkf
  104. Blom Goldman U1, Wennberg B, Svane G, Bylund H, Lind P. Reduction of radiation pneumonitis by V 20-constraints in breast cancer. Radiat Oncol. 2010; 5: 1-6. https://goo.gl/VSXNDk
  105. Wennberg B, Gagliardi G, Sundbom L, Svane G, Lind P. Early response of lung in breast cancer irradiation: radiologic density changes measured by CT and symptomatic radiation pneumonitis. Int J Radiat Oncol Biol Phys. 2002; 52: 1196-1206. https://goo.gl/J1eez3
  106. Lind PA, Marks LB, Hardenbergh PH, Clough R, Fan M, Hollis D, et al. Technical factors associated with radiation pneumonitis after local +_ regional radiation therapy for breast cancer. Int J Radiat Oncol Biol Phys. 2002; 52: 137-143. https://goo.gl/DWcWYY
  107. Ooi GC, Kwong DL, Ho JC, Lock DT, Chan FL, Lam WK, et al. Pulmonary  sequelae of treatment for breast cancer: A prospective study. Int J Radiat Oncol Biol Phys. 2001; 50: 411-419. https://goo.gl/CN3sx4
  108. Bornstein BA, Cheng CW, Rhodes LM, Rashid H, Stomper PC, Siddon RL, et al. Can simulation measurements be used to predict the irradiated lung volume in the tangential fields in patients treated for breast cancer? Int J Radiat Oncol Biol Phys. 1990; 18: 181-187. https://goo.gl/pRAQiA
  109. Lingos TI, Recht A, Vicini F, Abner A, Silver B, Harris JR. Radiation pneumonitis in breast cancer patients treated with conservative surgery and radiation therapy. Int J Radiat Oncol Biol Phys. 1991; 21: 355-360. https://goo.gl/d1Tihd
  110. Philip M Poortmans, Sandra Collette,  Carine Kirkove, Erik Van Limbergen, Volker Budach, Henk Struikmans, et al. Internal Mammary and Medial Supraclavicular Irradiation in Breast Cancer. EORTC Radiation Oncology and Breast Cancer Groups* New England Journal of Medicine. 2015; 373: 317-327. https://goo.gl/912BrP
  111. Essers M, Poortmans PM, Verschueren K, Hol S, Cobben DC. Should breathing adapted radiotherapy also be applied for right-sided breast irradiation? Acta Oncol. 2016; 55: 460-465. https://goo.gl/X57DHM
  112. Koc M, Polat P, Suma S. Effects of tamoxifen on pulmonary fibrosis after cobalt-60 radiotherapy in breast cancer patients. Radiother Oncol. 2002; 64: 171-175. https://goo.gl/BC4HP6
  113. Azria D, Larbouret C, Cunat S, Ozsahin M, Gourgou S, Martineau P, et al. Letrozole sensitizes breast cancer cells to ionizing radiation. Breast Cancer Res. 2005; 7: 156-163. https://goo.gl/AtWYXN
  114. Varga Z, Cserhati A, Kelemen G, Boda K, Thurzó L, Kahán Z. Role of systemic therapy in the development of lung sequelae after conformal radiotherapy in breast cancer patients. Int J Radiat Oncol Biol Phys. 2011; 80: 1109-1116. https://goo.gl/qmP7wN
  115. Henson KE, McGale P, Taylor C, Darby SC. Radiation-related mortality from heart disease and lung cancer more than 20 years after radiotherapy for breast cancer. Br J Cancer. 2013; 108: 179-182. https://goo.gl/66Drpn
  116. Werner-Wasik M, Scott C, Movsas B, C Langer, L Sarna, N Nicolaou, et al. Amifostine as mucosal protectant inpatients with locally advanced non–small cell lung cancer (NSCLC) receiving intensive chemotherapy and thoracic radiotherapy (RT): Results of the RadiationTherapy Oncology Group (RTOG) 98 – 01study. Int J Radiat Oncol BiolPhys. 2003; 57: 216. https://goo.gl/uJcFZY
  117. Ozturk B, Egehan I, Atavci S, Kitapci M. Pentoxifylline in prevention of radiation induced lung toxicity in patients with breast and lung cancer: a double-blind randomized trial. Int J Radiat Oncol Biol Phys. 2004; 58: 213-219. https://goo.gl/diya2J
  118. Kharofa J, Cohen E, Tomic R, Xiang Q, Gore E. Decreased Risk of Radiation PneumonitisWith Incidental Concurrent Use of Angiotensin-Converting Enzyme Inhibitorsand Thoracic Radiation Therapy. Int J Radiat Oncol Biol Phys. 2012; 84: 238-243. https://goo.gl/255JbY
  119. Sekine I, Sumib M, Itob Y, Nokihara H, Yamamoto N, Kunitoh H, et al. Retrospective analysis of steroid therapy forradiation-induced lung injury in lung cancer patients. Radiother Oncol. 2006; 80: 93-97. https://goo.gl/zkYWck
  120. Muraoka T, Bandoh S, Fujita J, Horiike A, Ishii T, Tojo Y, et al. Corticosteroid refractory radiationpneumonitis that remarkably responded to cyclosporin A. Intern Med. 2002; 41: 730-733. https://goo.gl/MuorcG
  121. Pass H, Carbone D, Johnson D, John D. Minna, Giorgio V, Andrew T Turrisi. A Text Book: Principles and Practiceof Lung Cancer: The Official Reference Text of the International Association forthe Study of Lung Cancer (IASLC) 2010. https://goo.gl/yyLzsN
  122. Cuzick J, Stewart H, Peto R, Baum M, Fisher B, HostH, et al. Overview of randomized trials of postoperative adjuvantradiotherapy in breast cancer. Cancer Treat Rep. 1987; 71: 15-29. https://goo.gl/yz6pn6
  123. Clarke M, Collins R, Darby S, Davies C, Elphinstone P, Evans E, et al. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG): Effects of radiotherapy and of differencesin the extent of surgery for early breast cancer on localrecurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005; 366: 2087-2106. https://goo.gl/SncT7B
  124. Sautter-Bihl ML, Sedlmayer F, Budach W, Dunst J, Feyer P, Fietkau R, et al; Breast Cancer ExpertPanel of the German Society of Radiation Oncology (DEGRO): DEGRO practical guidelines: radio therapy of breast cancer III – radiotherapy of the lymphatic pathways. Strahlenther Onkol. 2014; 190: 342-351. https://goo.gl/ojuKY3
  125. Schultz-Hector S, Trott KR. Radiation-induced cardiovascular diseases: is the epidemiologic evidence compatiblewith the radiobiologic data? Int J Radiat Oncol Biol Phys. 2007; 67: 10-18. https://goo.gl/QMmfx5
  126. Carver JR, Shapiro CL, Ng A, Jacobs L, Schwartz C, Virgo KS, et al. American Society of Clinical Oncology clinical evidence review on the ongoing care of adult cancer survivors: cardiac and pulmonary late effects. J Clin Oncol. 2007; 25: 3991-4008. https://goo.gl/ccB5ai
  127. Darby S, Cutter D, Boerma M, Constine LS, Fajardo LF, Kodama K, et al. Radiation-related heart disease: current knowledge and future prospects. Int J Radiat Oncol Biol Phys. 2010; 76: 656-665. https://goo.gl/atA2vC
  128. Sardaro A, Petruzzelli MF, D’Errico MP, Grimaldi L, Pili G, Portaluri M: Radiation-induced cardiac damage in early left breast cancer patients: risk factors, biological mechanisms, radiobiology, and dosimetric constraints. Radiother Oncol. 2012; 103: 133-142. https://goo.gl/9ToHyg
  129. Halyard MY, Pisansky TM, Dueck AC, Suman V, Pierce L, Solin L, et al. Radiotherapy and adjuvant trastuzumab in operable breast cancer: tolerability and adverse event data from the NCCTG Phase III Trial N9831. J Clin Oncol. 2009; 27: 2638-2644. https://goo.gl/SQ83HE
  130. Belkacemi Y, Gligorov J, Ozsahin M, Marsiglia H, De Lafontan B, Laharie-Mineur H, et al. Concurrent trastuzumab with adjuvant radiotherapy in HER2-positive breast cancer patients: acute toxicity analyses from the French multicentric study. Ann Oncol. 2008; 19: 1110-1116. https://goo.gl/pzvR6w
  131. Meattini I, Cecchini S, Muntoni C, Scotti V, De Luca Cardillo C, Mangoni M, et al. Cutaneous and cardiac toxicity of concurrent trastuzumab and adjuvant breast radiotherapy: a single institution series. Med Oncol. 2014; 31: 891. https://goo.gl/gGcZXa
  132. Lu Cao, Gang Cai, Cai Chang, Zhao-Zhi Yang, Yan Feng, Xiao-Li Yu, et al. Early cardiac toxicity following adjuvant radiotherapy of left-sided breast cancer with or without concurrent trastuzumab. Oncotarget. 2016; 7: 1042-1054. https://goo.gl/uWYp7W
  133. Schlitt A, Jordan K, Vordermark D, Schwamborn JR, Langer T, Thomssen C. Cardiotoxicity and oncological treatments. Dtsch Arztebl Int. 2014; 111: 161-168. https://goo.gl/Vp7kpx
  134. Lind PA, Pagnanelli R, Marks LB, Borges-Neto S, HuC, Zhou SM, et al . Myocardial perfusion changes in patients irradiated for left-sided breast cancer and correlation with coronary artery distribution. Int J Radiat Oncol Biol Phys. 2003; 55: 914-920. https://goo.gl/Q5aVZS
  135. Darby SC, Ewertz M, McGale P, Bennet AM, Blom- Goldman U, Bronnum D, et al. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med. 2013; 368: 987-998. https://goo.gl/S9V6Wz
  136. Feng M, Moran JM, Koelling T, Chughtai A, Chan JL, Freedman L, et al. Development and validation of a heart atlas to study cardiac exposure to radiation following treatment for breast cancer. Int J Radiat Oncol Biol Phys. 2011; 79: 10-18. https://goo.gl/Ft7HrR
  137. Taylor CW, McGale P, Povall JM, Thomas E, Kumar S, Dodwell D, et al. Estimating cardiac exposure from breast cancer radiotherapy in clinical practice. Int J Radiat Oncol Biol Phys. 2009; 73: 1061-1068. https://goo.gl/bgY14p
  138. Azizova TV, Muirhead CR, Moseeva MB, Grigoryeva ES, Vlasenko EV, Hunter N, et al. Ischemic heart disease in nuclear workers first employed at the Mayak PA in 1948-1972. Health Phys. 2012; 103: 3-14. https://goo.gl/rPjFX7
  139. Nielsen MH, Berg M, Pedersen AN, Andersen K, Glavicic V, Jakobsen EH, et al. Delineation of target volumes and organs at risk in adjuvant radiotherapy of early breast cancer: national guidelines and contouring atlas by the Danish Breast Cancer Cooperative Group. Acta Oncol. 2013; 52: 703-710. https://goo.gl/oJHU3d
  140. Haviland JS, Owen JR, Dewar JA, Agrawal RK, Barrett J, Barrett-Lee PJ, et al. The UK Standardisation of Breast Radiotherapy (START) trials of radiotherapy hypo fractionation for treatment of early breast cancer: 10-year follow-up results of two randomised controlled trials. Lancet Oncol. 2013; 14: 1086-1094. https://goo.gl/XAGjuP
  141. Appelt AL, Vogelius IR, Bentzen SM. Modern hypo fractionation schedules for tangential whole breast irradiation decrease the fraction size-corrected dose to the heart. Clin Oncol (R Coll Radiol). 2013; 25: 147-152. https://goo.gl/EJb8vo
  142. Dellas K, Vonthein R, Zimmer J, Dinges S, Boicev AD, Andreas P, et al. Hypo fractionation with simultaneous integrated boost for early breast cancer: results of the German multicenter phase II trial (ARO-2010–01). Strahlenther Onkol. 2014; 190: 646-653. https://goo.gl/q7vpnf
  143. Poortmans PM, Collette S, Kirkove C, Van Limbergen E, Budach V, Struikmans H, et al. Internal Mammary and Medial Supraclavicular Irradiation in Breast Cancer. N Engl J Med. 2015; 373: 317-327. https://goo.gl/PGXrLM
  144. Whelan TJ1, Olivotto IA, Parulekar WR, Ackerman I, Chua BH, Nabid A, et al. Regional Nodal Irradiation in Early-Stage Breast Cancer. N Engl J Med. 2015; 373: 307-316. https://goo.gl/3btrDD
  145. Thorsen LB, Offersen BV, Dano H, Berg M, Jensen I, Pedersen AN, et al. DBCG-IMN: A Population-Based Cohort Study on the Effect of Internal Mammary Node Irradiation in Early Node-Positive Breast Cancer. J Clin Oncol. 2016; 34: 314-320. https://goo.gl/QzSx7d
  146. Johansen S, Cozzi L, Olsen D R. A planning comparison of dose patterns in organ a trisk and predicted risk for radiation induced malignancy in the contralateral breast following radiation therapy of primary breast using conventional, IMRT and Volumetric modulated arc treatment techniques. Acta Oncol. 48: 495-503; https://goo.gl/HnNTFA
  147. Vishruta A Dumane, Margie A Hunt, Sheryl Green, Yeh-Chi LoRichard L Bakst. Dosimetric comparison of volumetric modulated arctherapy, static field intensity modulated radiationtherapy, and 3D conformal planning for the treatment of a right-sided reconstructed chest wall and regional nodal case. J Radiat. 2014; 12. https://goo.gl/PAxRDb
  148. Teoh M, Clark CH, Wood K, Whitaker S, Nisbet A. Volumetric modulatedarctherapy: a review of current literature and clinical use in practice. Br J Radiol. 2011; 84: 967-996. https://goo.gl/oWVnaq
  149. Aly M, Glatting G, Jahnke L, Wenz F, Abo Madyan Y. Comparison ofbreastsimultaneous integratedboost(SIB)radiotherapy techniques. Radiat Oncol. 2015; 10: 139. https://goo.gl/jJvmMA
  150. Wang JZ, Li XA, D’Souza WD, Stewart RD. Impact of prolonged fraction delivery times on tumor control: anote of caution for intensity-modulated radiation therapy (IMRT). Int J Radiat Oncol Biol Phys. 2003; 57: 543-552. https://goo.gl/572JQ5
  151. Withers HR, Taylor JM, Maciejewski B. The hazard of accelerated tumor clonogen repopulation during radiotherapy. Acta Oncol. 27: 131-146; 1988. https://goo.gl/Zjn7C1
  152. Pedersen AN, Korreman S, Nyström H, Specht L. Breathing adapted radiotherapy of breast cancer: reduction of cardiac and pulmonary doses using voluntary inspirationbreath-hold. Radiother Oncol. 2004; 72: 53–60. https://goo.gl/NVyfzx
  153. Hjelstuen MH, Mjaaland I, Vikstrom J, Dybvik KI. Radiation during deep inspiration allows loco-regional treatment of left breast and axillary-, supraclavicular and internal mammary lymph nodes without compromising target coverage or dose restrictions to organs at risk. Acta Oncol. 2012; 51: 333-344. https://goo.gl/hZ5rVD
  154. Nissen HD, Appelt AL. Improved heart, lung and target dose with deep inspiration breath hold in a large clinical series of breast cancer patients. Radiother Oncol. 2013; 106: 28–32. https://goo.gl/fhB62T
  155. MastME, HeijenbrokMW, van Kempen Harteveld ML, Petoukhova AL, Scholten AN,Wolterbeek R, et al. Less increase of CT-based calcium scores of the coronary arteries: Effect three years after breast-conserving radiotherapy using breath-hold. Strahlenther Onkol. 2016; 192: 696-704. https://goo.gl/pMGpy3
  156. Darby SC, Ewertz M, McGale P, Bennet AM, Blom-Goldman U, Bronnum D, et al. Risk of ischemic heart disease in women after radiotherapy for breastcancer. N Engl J Med. 2013; 368: 987-998. https://goo.gl/yhqdR7
  157. Nemoto K, Ogushi M, Nakajima M, Kozuka T, Nose T, Yamashita T. Cardiac sparing radiotherapy for left breast cancer with deep breath-holding. Jpn J Radiol. 2009; 27: 259-263. https://goo.gl/E9t5Ld
  158. Bahrainy M, Kretschmer M, Jost V, Kasch A, Würschmidt F, Dahle J, et al. Treatment of breast cancer with simultaneous integrated boost in hybrid plan technique: Influence of flattening filter-free beams. Strahlenther Onkol. 2016; 192: 333-341. https://goo.gl/BeFQDm
  159. Becker Schiebe M, Stockhammer M, Hoffmann W, Wetzel F, Franz H. Does mean heart dose sufficiently reflect coronary artery exposure in left-sided breast cancer radiotherapy? Influence of respiratory gating. Strahlenther Onkol. 2016; 192: 624-631. https://goo.gl/Tj7Xxz
  160. Sakka M, Kunzelmann L, Martin Metzger, Gerhard GG. Cardiac dose-sparing effects of deep-inspiration breath-hold in leftbreast irradiation Is IMRT more beneficial than VMAT? Strahlenther Onkol. 2017; 23. https://goo.gl/Y36ho7
  161. Mulliez T, Veldeman L, van Greveling A, Speleers B, Sadeghi S, Berwouts D, et al. Hypofractionated whole breast irradiation for patients with large breasts: a randomized trial comparing prone and supine positions. Radiother Oncol. 2013; 108: 203-208. https://goo.gl/MKEAPo
  162. Lymberis SC, deWyngaert JK, Parhar P, Chhabra AM, Fenton-Kerimian M, Chang J, et al. Prospective assessment of optimal individual position (prone versus supine) for breast radiotherapy: volumetric and do simetric correlations in 100 patients. Int J Radiat Oncol Biol Phys. 2012; 84: 902-909. https://goo.gl/SzJ4Sv
  163. Formenti SC, DeWyngaert JK, Jozsef G, Goldberg JD. Prone vs supine positioning for breast cancer radiotherapy. JAMA. 2012; 308: 861–863. https://goo.gl/6SWYs3
  164. Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, Luini A, et al. Twenty-year follow-up of a randomized study comparing breastconserving surgery with radical mastectomy for early breast cancer. N Engl J Med. 2002; 347: 1227–1232. https://goo.gl/fVFDw9
  165. Tunio MA1, Al Asiri M, Bayoumi Y, Stanciu LG, Al Johani N, Al Saeed EF. Is thyroid gland an organ at risk in breast cancer patients treated with locoregional radiotherapy? Results of a pilot study. J Cancer Res Ther. 2015; 11: 684-689. https://goo.gl/7AFVBy
  166. Kuten A, Lubochitski R, Fishman G, Dale J, Stein ME. Postradiotherapy hypothyroidism: radiation dose response and chemotherapeutic radiosensitization at less than 40 Gy. J Surg Oncol. 1996; 61: 281-283. https://goo.gl/MdcrpQ
  167. Koc M, Capoglu I. Thyroid dysfunction in patients treated with radiotherapy for neck. Am J Clin Oncol. 2009; 32: 150-153. https://goo.gl/WCX8qf
  168. Jereczek Fossa BA, Alterio D, Jassem J, Gibelli B, Tradati N, Orecchia R. Radiotherapy-induced thyroid disorders. Cancer Treat Rev. 2004; 30: 369-384. https://goo.gl/UvrPNd
  169. Alterio D, Jereczek Fossa BA, Franchi B, D'Onofrio A, Piazzi V, Rondi E, et al. Thyroid disorders in patients treated with radiotherapy for head-and-neck cancer: a retrospective analysis of seventy-three patients. Int J Radiat Oncol Biol Phys. 2007; 67: 144-150. https://goo.gl/fCG81X
  170. Bruning P, Bonfrer J, De Jong Bakker M, Nooyen W, Burgers M. Primary hypothyroidism in breast cancer patients with irradiated supraclavicular lymph nodes. Br J Cancer. 1985; 51: 659-663. https://goo.gl/io2ojn
  171. Joensuu H, Viikari J. Thyroid function after postoperative radiation therapy in patients with breast cancer. Acta Radiol Oncol. 1986; 25: 167-170. https://goo.gl/2szVut
  172. Smith GL, Smith BD, Giordano SH, Shih YC, Woodward WA, Strom EA, et al. Risk of hypothyroidism in older breast cancer patients treated with radiation. Cancer. 2008; 112: 1371-1379. https://goo.gl/GWLL86   
  173. Reinertsen KV, Cvancarova M, Wist E, Bjoro T, Dahl AA, Danielsen T, et al. Thyroid function in women after multimodal treatment for breast cancer stage II/III: comparison with controls from a population sample. Int J Radiat Oncol Biol Phys. 2009; 75: 764-770. https://goo.gl/o15TA8
  174. Johansen S, Reinertsen KV, Knutstad K, Olsen DR, Fossa SD. Dose distribution in the thyroid gland following radiation therapy of breast cancer-a retrospective study. Radiat Oncol. 2011; 6: 68. https://goo.gl/6JXp5e
  175. Glatstein E, McHardy Young S, Brast N, Eltringham JR, Kriss JP. Alterations in serum thyrotropin (TSH) and thyroid function following radiotherapy in patients with malignant lymphoma. J Clin Endocrinol Metab. 1971; 32: 833-841. https://goo.gl/KJR4My
  176. B A Laway, Kuchay M Shafi, Sabiya Majid, M Maqbool Lone, F Afroz,  Showkat Khan, et al. Incidence of primary hypothyroidism in patients exposed to therapeutic external beam radiation, where radiation portals include a part or whole of the thyroid gland. Indian J of Endocrinol Metab. 2012; 16: 329-331. https://goo.gl/2cTpJc
  177. Schimpff SC, Diggs CH, Wiswell JG, Salvatore PC, Wiernik PH. Radiation-related thyroid dysfunction. Implications for the treatment of Hodgkin’s disease. Ann Intern Med. 1980; 92: 91-98. https://goo.gl/97k6Pw
  178. Tamura K, Shimaoka K, Friedman M. Thyroid abnormalities associated with treatment of malignant lymphoma. Cancer. 1981; 47: 2704-2711. https://goo.gl/97k6Pw
  179. Kanyilmaz G, Aktan M, Koc M, Demir H, Demir LS. Radiation-induced hypothyroidism in patients with breast cancer: a retrospective analysis of 243 cases. Med Dosim. 2017; 42: 190-196. https://goo.gl/QzU3qS
  180. Jereczek-Fossa BA, Alterio D, Jassem J, Gibelli B, Tradati N, Orecchia R. Radiotherapy-induced thyroid disorders. Cancer Treat Rev. 2004; 30: 369–384. https://goo.gl/BeU8J6
  181. Johansen S, Reinertsen KV, Knutstad K, Olsen DR, Fossa SD. Dose distribution in the thyroid gland following radiation therapy of breast cancer – A retrospective study. Radiat Oncol. 2011; 6: 68. https://goo.gl/v2tRVY
  182. Yoden E, Soejima T, Maruta T, Demizu Y, Nishimura H, Ejima Y, et al. Hypothyroidism after radiotherapy to the neck. Nippon Acta Radiol. 2004; 64: 146-150. https://goo.gl/oLpwNq
  183. Akgun Z, Atasoy BM, Ozen Z, Yavuz D, Gulluoglu B, Sengoz M, et al. V30 as a predictor for radiation-induced hypothyroidism: a dosimetric analysis in patients who received radiotherapy to the neck. Radiat Oncol. 9: 104; 2014. https://goo.gl/HFV8oy
  184. Cella L, Conson M, Caterino M, De Rosa N, Liuzzi R, Picardi M, et al. Thyroid V30 predicts radiation-induced hypothyroidism in patients treated with sequential chemo-radiotherapy for Hodgkin’s lymphoma. Int J Radiat Oncol Biol Phys. 2012; 82: 1802-1808. https://goo.gl/GS4ZsW
  185. Kim MY, Yu T, Wu HG. Dose-volumetric parameters for predicting hypothyroidism after radiotherapy for head and neck cancer. Jpn J Clin Oncol. 2014; 4: 331-337. https://goo.gl/wG9eAQ
  186. 184   Fujiwara M, Kamikonya N, Odawara S, Suzuki H, Niwa Y, Takada Y et al. The threshold of hypothyroidism after radiation therapy for head and neck cancer: a retrospective analysis of 116 cases. J Radiat Res. 2015; 56: 577-582. https://goo.gl/e8bWsi
  187. Kuten A, Lubochitski R, Fishman G, Dale J, Stein ME. Post radiotherapy hypothyroidism: radiation dose response and chemotherapeutic radiosensitization at less than 40 Gy. J Surg Oncol. 1996; 61: 281–283. https://goo.gl/G83P75
  188. Yoden E, Soejima T, Maruta T, Demizu Y, Nishimura H, Ejima Y, et al. Hypothyroidism after radiotherapy to the neck. Nippon Acta Radiol. 2004; 64: 146-150. https://goo.gl/oLpwNq
  189. Diaz R, Jaboin JJ, Morales-Paliza M, Koehler E, Phillips JG, Stinson S, et al. Hypothyroidism as a consequence of intensity-modulated radiotherapy with concurrent taxane-based chemotherapy for locally advanced head and neck cancer. Int J Radiat Oncol Biol Phys. 2010; 77: 468-476.
  190. Donker M, Slaets L, van Tienhoven G, Rutgers EJ. Axillary lymph node dissection versus axillary radiotherapy in patients with a positive sentinel node: the AMAROS trial. Ned Tijdschr Geneeskd. 2015; 159: 9302.
  191. Garber JR, Cobin RH, Gharib H, Hennessey JV, Klein I, Mechanick JI, et al. Clinical practice guidelines for hypothyroidism in adults: cosponsored by the American Association of Clinical Endocrinologists and the American Thyroid Association. Endocr Pract. 2012; 18: 988-1028. https://goo.gl/TgZ3pn
  192. Wai ES, Lesperance M, Speers CH, Truong PT, Jones S, Tyldesley S, et al. Increased use of regional radiotherapy is associated with improved outcome in a population based cohort of women with breast cancer with 1-3 positive nodes. Radiother Oncol.  2010; 97: 301-306. https://goo.gl/igTuU1
  193. T J Whelan, I Olivotto, I Ackerman, J W Chapman, B Chua, A Nabid, et al. NCIC-CTG MA. 20: An intergroup trial of regional nodal irradiation in early breast cancer. https://goo.gl/YQ93Bo
  194. R Cote, A E Giuliano, D Hawes, K V Ballman, P W Whitworth, P W Blumencranz, et al. ACOSOG Z0010: A multicenter prognostic study of sentinel node (SN) and bone marrow (BM) micrometastases in women with clinical T1/T2 N0 M0 breast cancer. J Clin Oncol. 2010; 28: 504. https://goo.gl/zm5KR5
  195. Giuliano AE, Hunt KK, Ballman KV, Beitsch PD, Whitworth PW, Blumencranz PW, et al. Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: a randomized clinical trial. JAMA. 2011; 305: 569-575. https://goo.gl/fE5fPB
  196. Kori SH, Foley KM, Posner JB. Brachial plexus lesions in patients with cancer: 100 cases. Neurology. 1981; 31: 45-50. https://goo.gl/nYAQZe
  197. Johansson S, Svensson H, Denekamp J. Dose response and latency for radiation-induced fibrosis, edema, and neuropathy in breast cancer patients. Int J Radiat Oncol. 2002; 52: 1207-1219. https://goo.gl/QNNU64
  198. Fathers E, Thrush D, Huson SM, Norman A. Radiation-induced brachial plexopathy in women treated for carcinoma of the breast. Clin Rehabil. 2002; 16: 160-165. https://goo.gl/nPU5w5
  199. Bates TD. The 10-year results of a prospective trial of post-operative radiotherapy delivered in 3 fractions per week versus 2 fractions per week in breast carcinoma. Br J Radiol. 1988; 61: 625-630. https://goo.gl/o1ETvX
  200. Rodger A, Jack WJL, Kerr G. A change in post mastectomy radiotherapy fractionation: an audit of tumour control, acute and late morbidity. The Breast. 1996; 5: 244-250. https://goo.gl/GoeKwd
  201. NelsonLeong, pauline T Truong, Keith tankel, winkle kwan, LornaWeir, Ivo A.Olivotto. Hypofractionated nodal radiation therapy for breast cancer was not associated with increased patient-reported arm or brachial plexopathy symptoms. https://goo.gl/xZGqf1   
  202. Dumitru D, Zwarts MJ. Brachial plexopathies and proximal mononeuropathies. Dumitru D, Amato AA, Zwarts MJ. Electrodiagnostic Medicine. 2nd. Philadephia: Hanley & Belfus. 2002; 777-836. https://goo.gl/7dNx9i
  203. Delanian S, Lefaix JL, Pradat PF. Radiation-induced neuropathy in cancer survivors. Radiother Oncol. 2012; 105: 273-282. https://goo.gl/ReuNiG
  204. Atroshi I, Gummesson C, Johnsson R, Ornstein E, Ranstam J, Rosen I. Prevalence of carpal tunnel syndrome in a general population. JAMA. 1999; 282: 153-158. https://goo.gl/4KK2p7
  205. Lundstedt D, Gustafsson M, Steineck G, Sundberg A, Wilderang U, Holmberg E. Radiation Therapy to the Plexus Brachialisin Breast Cancer Patients: Analysis of Paresthesia in Relation to Dose and Volume. https://goo.gl/Xr9nZT
  206. Lundstedt D, Gustafsson M, Steineck G, Sundberg A, Wilderäng U, Holmberg E, et al. Brachial plexopathy after chemo radiotherapy for head and neck squamous cell carcinoma. Strahlenther Onkol. 2010; 186: 517-520. https://goo.gl/tMFozP
  207. Dropcho EJ. Neurotoxicity of radiation therapy. Neurol Clin. 2010; 28: 217-234. https://goo.gl/8tEsPP  
  208. Gałecki J, Hicer-Grzenkowicz J, Grudzien-Kowalska M, Michalska T, Zalucki W. Radiation-induced brachial plexopathy and hypofractionated regimens in adjuvant irradiation of patients with breast cancer-a review. Acta Oncol. 2006; 45: 280-284. https://goo.gl/QhReZx
  209. Delanian S, Lefaix JL. The radiation-induced fibroatrophic process: therapeutic perspective via the antioxidant pathway. Radiother Oncol. 2004; 73: 119-131. https://goo.gl/HjJhu7
  210. Powell S, Cooke J, Parsons C. Radiation-induced brachial plexus injury: Follow-up of two different fractionation schedules. Radiother Oncol. 1990; 18: 213-220. https://goo.gl/WNdit1
  211. Olsen NK, Pfeiffer P, Johannsen L, Schroder H, Rose C. Radiation-induced brachial plexopathy: Neurological follow-up in 161 recurrencefreebreast cancer patients. Int J Radiat Oncol Biol Phys. 1993; 26: 43-49. https://goo.gl/3jBo6t
  212. Haviland JS, Owen JR, Dewar JA, Agrawal RK, Barrett J, Barrett-Lee PJ, et al. The UK Standardisation of Breast Radiotherapy (START) trials of radiotherapy hypofractionation for treatment of early breast cancer: 10-yearfollow-up results of two randomised controlled trials. Lancet Oncol. 2013; 14: 1086-1094. https://goo.gl/WWTDSS
  213. Chan EK, Woods R, Virani S, Speers C, Wai ES, Nichol A, et al. Long-term mortality from cardiac causes after adjuvanthypofractionated vs. conventional radiotherapy for localized left-sided breast cancer. Radiother Oncol. 2015; 114: 73-78. https://goo.gl/AfujYa
  214. Overgaard M, Jensen MB, Overgaard J, Hansen PS, Rose C, Andersson M, et al. Postoperative radiotherapy in high-riskpostmenopausal breast-cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet. 1999; 353: 1641-1648. https://goo.gl/TiwyiD
  215. Ragaz J, Olivotto IA, Spinelli JJ, Phillips N, Jackson SM, Wilson KS, et al. Locoregional Radiation Therapy in Patients With High-Risk Breast Cancer Receiving Adjuvant Chemotherapy: 20-YearResults of the British Columbia Randomized Trial. J Natl Cancer Inst. 2005; 97: 116-126. https://goo.gl/ZTqWVX
  216. Emami B, Lyman J, Brown A, Coia L, Goitein M, Munzenrider JE, et al. Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys. 1991; 21: 109-122. https://goo.gl/bABUhc
  217. Amini A, Yang J, Williamson R, McBurney ML, Erasmus J Jr, Allen PK, et al. Dose constraints to prevent radiation-induced brachial plexopathy in patients treated for lung cancer. Int J Radiat Oncol Biol Phys. 2012; 82: 391-398. https://goo.gl/rhgLh7
  218. Eblan MJ, Corradetti MN, Lukens JN, Xanthopoulos E, Mitra N, Christodouleas JP, et al. Brachial plexopathy inapical non-small cell lung cancer treated with definitive radiation: Dosimetric analysis and clinical implications. Int J Radiat Oncol Biol Phys. 2013; 85: 175-181. https://goo.gl/oUMxYk
  219. Jaeckle KA. Plexopathies in cancer patients. Levin, Victor A. 2nd edition. Cancer in the Nervous System. New York, NY: Churchill Livingstone; 1996. 347-360. https://goo.gl/3UfuLb
  220. Langballe R, Olsen JH, Andersson M, Mellemkjær L. Risk of second primary non breast cancer in pre- and postmenopausal women with breast cancer not trated with chemotherapy, radiotherapy or endocrine therapy. Eur J Cancer. 2011; 47: 946-952. https://goo.gl/vbJXfi
  221. Clarke M, Collins R, Darby S, Davies C, Elphinstone P, Evans V, et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005; 366: 2087-2106. https://goo.gl/B2MiuA   
  222. Berrington de Gonzalez A, Curtis RE, Gilbert E, Berg CD, Smith SA, Stovall M, et al. Second solid cancers after radiotherapy for breast cancer in SEER registries. Br J Cancer. 2010; 102: 220-226. https://goo.gl/ebfvUe
  223. Amy Berrington de Gonzalez, Rochelle E Curtis, Stephen F Kry, Ethel Gilbert, Stephanie Lamart, Christine D Berg, et al. The proportion of second cancers attributable to radiotherapy treatments in adults: a prospective cohort study in the US SEER cancer registries. Lancet Oncol. 2011; 12: 353-360. https://goo.gl/ZG3oaV  
  224. Tubiana M. can we reduce the incidence of second primary malignancies occurring after radiotherapy? A critical review. Radiother Oncol. 2009; 91: 4-15. https://goo.gl/83BJqu
  225. Hooning MJ, Aleman BM, Hauptmann M, Baaijens MH, Klijn JG, Noyon R, et al. Roles of radiotherapy and chemotherapy in the development of contralateral breast cancer. J Clin Oncol. 2008; 26: 5561-5568. https://goo.gl/eJbgqP
  226. Deutsch M, Land SR, Begovic M, Wieand HS, Wolmark N, Fisher B. The incidence of lung carcinoma after surgery for breast carcinoma with and without postoperative radiotherapy. Results of National Surgical Adjuvant Breast and Bowel Project (NSABP) Clinical Trial B-04 and B-06. Cancer. 2003; 98: 1362-1368. https://goo.gl/HnpTbT
  227. Kaufman EL1, Jacobson JS, Hershman DL, Desai M, Neugut AI. Effect of breast cancer radiotherapy and cigarette smoking on risk of second primary lung cancer. J Clin Oncol. 2008; 26: 392-398. https://goo.gl/tfHZU8