5,679
Views
0
CrossRef citations to date
0
Altmetric
Review

Chemotherapy in patients with early breast cancer: clinical overview and management of long-term side effects

, , , , , & ORCID Icon show all
Pages 1341-1355 | Received 25 Aug 2022, Accepted 21 Nov 2022, Published online: 05 Dec 2022

References

  • Allemani C, Weir HK, Carreira H, et al. Global surveillance of cancer survival 1995-2009: analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries (Concord-2). Lancet. 2015;385(9972):977–1010.
  • Runowicz CD, Leach CR, Henry NL, et al. American Cancer Society/american Society of Clinical Oncology Breast Cancer survivorship care guideline. J Clin Oncol. 2016;34(6):611–635.
  • Mock V, Atkinson A, Barsevick A, et al. NCCN practice guidelines for cancer-related fatigue. Oncology (Williston Park). 2000;14(11A):151–161.
  • Schmidt ME, Chang-Claude J, Vrieling A, et al. Fatigue and quality of life in breast cancer survivors: temporal courses and long-term pattern. J Cancer Surviv. 2012;6(1):11–19.
  • Goedendorp MM, Andrykowski MA, Donovan KA, et al. Prolonged impact of chemotherapy on fatigue in breast cancer survivors: a longitudinal comparison with radiotherapy-treated breast cancer survivors and noncancer controls. Cancer. 2012;118(15):3833–3841.
  • Martín M, Ruiz A, Ruiz Borrego M, et al. Fluorouracil, doxorubicin, and cyclophosphamide (FAC) versus FAC followed by weekly paclitaxel as adjuvant therapy for high-risk, node-negative breast cancer: results from the GEICAM/2003-02 study. J Clin Oncol. 2013;31(20):2593–2599.
  • Fisher MI, Davies C, Lacy H, et al. Oncology section EDGE task force on cancer: measures of cancer-related fatigue—a systematic review. Rehab Oncol. 2018;36(2):93–105.
  • Fabi A, Bhargava R, Fatigoni S, et al. Cancer-related fatigue: ESMO clinical practice guidelines for diagnosis and treatment. Ann Oncol. 2020;31(6):713–723.
  • Vaz-Luis I, Di Meglio A, Havas J, et al. Long-term longitudinal patterns of patient-reported fatigue after breast cancer: a group-based trajectory analysis. J Clin Oncol. 2022;40(19):2148–2162.
  • Puglisi F, Deroma L, Russo S, et al. Effect of age on hemoglobin levels and quality of life following treatment with epoetin alfa in cancer patients. Crit Rev Oncol Hematol. 2009;69(2):175–182.
  • Russo S, Cinausero M, Gerratana L, et al. Factors affecting patient’s perception of anticancer treatments side-effects: an observational study. Expert Opin Drug Saf. 2014;13(2):139–150.
  • Sobrero A, Puglisi F, Guglielmi A. et alFatigue: a main component of anemia symptomatology. Semin Oncol. 2001;28(2, Suppl 8):15–18.
  • Antonuzzo A, Ripamonti CI, Roila F, et al. Effectiveness of a phone-based nurse monitoring assessment and intervention for chemotherapy-related toxicity: a randomized multicenter trial. Front Oncol. 2022;12:925366.
  • Brown JC, Huedo-Medina TB, Pescatello LS, et al. Efficacy of exercise interventions in modulating cancer-related fatigue among adult cancer survivors: a meta-analysis. Cancer Epidemiol Biomarkers Prev. 2011;20(1):123–133.
  • Tomlinson D, Diorio C, Beyene J, et al. Effect of exercise on cancer-related fatigue: a meta-analysis. Am J Phys Med Rehabil. 2014;93(8):675–686.
  • Mock V, Pickett M, Ropka ME, et al. Fatigue and quality of life outcomes of exercise during cancer treatment. Cancer Pract. 2001;9(3):119–127.
  • Ligibel JA, Bohlke K, May AM, et al. Exercise, diet, and weight management during cancer treatment: ASCO guideline. J Clin Oncol. 2022;40(22):2491–2507.
  • Gielissen MFM, Verhagen S, Witjes F, et al. Effects of cognitive behavior therapy in severely fatigued disease-free cancer patients compared with patients waiting for cognitive behavior therapy: a randomized controlled trial. J Clin Oncol. 2006;24(30):4882–4887.
  • Greenlee H, DuPont-Reyes MJ, Balneaves LG, et al. Clinical practice guidelines on the evidence-based use of integrative therapies during and after breast cancer treatment. CA Cancer J Clin. 2017;67(3):194–232.
  • Buffart LM, van Uffelen JGZ, Riphagen II, et al. Physical and psychosocial benefits of yoga in cancer patients and survivors, a systematic review and meta-analysis of randomized controlled trials. BMC Cancer. 2012;12(1):559.
  • Danhauer SC, Addington EL, Cohen L, et al. Yoga for symptom management in oncology: a review of the evidence base and future directions for research. Cancer. 2019;125(12):1979–1989.
  • Barton DL, Liu H, Dakhil SR, et al. Wisconsin ginseng (Panax quinquefolius) to improve cancer-related fatigue: a randomized, double-blind trial, N07C2. J Natl Cancer Inst. 2013;105(16):1230–1238.
  • Semiglazov VF, Stepula VV, Dudov A, et al. Quality of life is improved in breast cancer patients by standardised mistletoe extract PS76A2 during chemotherapy and follow-up: a randomised, placebo-controlled, double-blind, multicentre clinical trial. Anticancer Res. 2006;26(2B):1519–1529.
  • Zhang Y, Lin L, Li H, et al. Effects of acupuncture on cancer-related fatigue: a meta-analysis. supportive care in cancer. J Multinat Assoc Support Care Cancer. 2018;26(2):415–425.
  • Roth AJ, Nelson C, Rosenfeld B, et al. Methylphenidate for fatigue in ambulatory men with prostate cancer. Cancer. 2010;116(21):5102–5110.
  • Kerr CW, Drake J, Milch RA, et al. Effects of methylphenidate on fatigue and depression: a randomized, double-blind, placebo-controlled trial. J Pain Symptom Manage. 2012;43(1):68–77.
  • Richard PO, Fleshner NE, Bhatt JR, et al. Phase II,randomised, double-blind, placebo-controlled trial of methylphenidate for reduction of fatigue levels in patients with prostate cancer receiving LHRH-agonist therapy. BJU Int. 2015;116(5):744–752.
  • Lower EE, Fleishman S, Cooper A, et al. Efficacy of dexmethylphenidate for the treatment of fatigue after cancer chemotherapy: a randomized clinical trial. J Pain Symptom Manage. 2009;38(5):650–662.
  • Moraska AR, Sood A, Dakhil SR, et al. Phase III, randomized, double-blind, placebo-controlled study of long-acting methylphenidate for cancer-related fatigue: North Central Cancer Treatment Group NCCTG-N05C7 trial. J Clin Oncol. 2010;28(23):3673–3679.
  • Jean-Pierre P, Morrow GR, Roscoe JA, et al. A phase 3 randomized, placebo-controlled, double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631 patients receiving chemotherapy: a university of Rochester cancer center community clinical oncology program research base study. Cancer. 2010;116(14):3513–3520.
  • Morrow GR, Jickok JT, Roscoe JA, et al. Differential effects of paroxetine on fatigue and depression: a randomized, double-blind trial from the University of Rochester Cancer Center Community Clinical Oncology Program. J Clin Oncol. 2003;21(24):4635–4641.
  • Roscoe A, Morrow GR, Hickok JT, et al. Effect of paroxetine hydrochloride (Paxil) on fatigue and depression in breast cancer patients receiving chemotherapy. Breast Cancer Res Treat. 2005;89(3):243–39.
  • Bruera E, El Osta B, Valero V, et al. Donepezil for cancer fatigue: a double-blind, randomized, placebo-controlled trial. J Clin Oncol. 2007;25(23):3475–3481.
  • Dimsdale JE, Ball ED, Carrier E, et al. Effect of eszopiclone on sleep, fatigue, and pain in patients with mucositis associated with hematologic malignancies. Support Care Cancer. 2011;19(12):2015–2020.
  • Bruera E, Ernst S, Hagen N, et al. Effectiveness of megestrol acetate in patients with advanced cancer: a randomized, double-blind, crossover study. Cancer Prevent Cont. 1998;2(2):74–78.
  • Lund Rasmussen C, Olsen MK, Johnsen AT, et al. Effects of melatonin on physical fatigue and other symptoms in patients with advanced cancer receiving palliative care: a double-blind placebo-controlled crossover trial. Cancer. 2015;121(20):3727–3736.
  • Jim HSL, Hoogland AI, Han HS, et al. A randomized placebo-controlled trial of bupropion for cancer-related fatigue: study design and procedures. Contemp Clin Trials. 2020;91:105976.
  • Palesh OG, Roscoe JA, Mustian KM, et al. Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center-community Clinical Oncology Program. J Clin Oncol. 2010;28(2):292–298.
  • Ancoli-Israel S, Moore PJ, Jones V. The relationship between fatigue and sleep in cancer patients: a review. Eur J Cancer Care (Engl). 2001;10(4):245–255.
  • Palesh OG, Collie K, Batiuchok D, et al. A longitudinal study of depression, pain, and stress as predictors of sleep disturbance among women with metastatic breast cancer. Biol Psychol. 2007;75(1):37–44.
  • Leysen L, Lahousse A, Nijs J, et al. Prevalence and risk factors of sleep disturbances in breast cancer survivors: systematic review and meta-analyses. Support Care Cancer. 2019;27(12):4401–4433.
  • Savard J, Ivers H. The initiation of chemotherapy, but not radiation therapy, coincides with increased insomnia. Psychooncology. 2012;21:1–130.
  • Palesh O, Peppone L, Innominato F, et al. Prevalence, putative mechanisms, and current management of sleep problems during chemotherapy for cancer. Nat Sci Sleep. 2012;4:151–162.
  • Sanford SD, Wagner LI, Beaumont JL, et al. Longitudinal prospective assessment of sleep quality: before, during, and after adjuvant chemotherapy for breast cancer. Support Care Cancer. 2013;21(4):959–967.
  • Sephton SE, Sapolsky RM, Kraemer HC, et al. Diurnal cortisol rhythm as a predictor of breast cancer survival. J Natl Cancer Inst. 2000;92(12):994–1000.
  • Kondratova AA, Kondratov RV. The circadian clock and pathology of the ageing brain. Nat Rev Neurosci. 2012;13(5):325–335.
  • Garaulet M, Madrid J. A. Chronobiological aspects of nutrition, metabolic syndrome and obesity. Adv Drug Deliv Rev. 2010;62(9–10):967–978.
  • Savard J, Morin CM. Insomnia in the context of cancer: a review of a neglected problem. J Clin Oncol. 2001;19(3):895–908.
  • Berger AM, Wielgus K, Hertzog M, et al. Patterns of circadian activity rhythms and their relationships with fatigue and anxiety/depression in women treated with breast cancer adjuvant chemotherapy. Support Care Cancer. 2010;18(1):105–114.
  • Rich T, Innominato PF, Boerner J, et al. Elevated serum cytokines correlated with altered behavior, serum cortisol rhythm, and dampened 24-hour rest-activity patterns in patients with metastatic colorectal cancer. Clin cAncer Res Off J Am Assoc Cancer Res. 2005;11(5):1757–1764.
  • Opp MR. Cytokines and sleep. Sleep Med Rev. 2005;9(5):355–364.
  • Liu L, Mills PJ, Rissling M, et al. Fatigue and sleep quality are associated with changes in inflammatory markers in breast cancer patients undergoing chemotherapy. Brain Behav Immun. 2012;26(5):706–713.
  • Panossian LA, Veasey SC. Daytime sleepiness in obesity: mechanisms beyond obstructive sleep apnea—a review. Sleep. 2012;35(5):601–615.
  • Lucassen EA, Rother KI, Cizza G. Interacting epidemics? Sleep curtailment, insulin resistance, and obesity. Ann N Y Acad Sci. 2012;1264(1):110–134.
  • Berger AM, Gerber LH, Mayer DK. Cancer-related fatigue: implications for breast cancer survivors. Cancer. 2012;118(8 Suppl):2261–2269.
  • Gerber LH, Stout N, McGarvey C, et al. Factors predicting clinically significant fatigue in women following treatment for primary breast cancer. Support Care Cancer. 2011;19(10):1581–1591.
  • Koffel E, Khawaja IS, Germain A. Sleep disturbances in posttraumatic stress disorder: updated review and implications for treatment. Psychiatr Ann. 2016;46(3):173–176.
  • King AC, Oman RF, Brassington GS, et al. Moderate-intensity exercise and self-rated quality of sleep in older adults. a randomized controlled trial. JAMA. 1997;277(1):32–37.
  • Tworoger SS, Yasui U, Vitiello MV, et al. Effects of a year long moderate-intensity exercise and a stretching intervention on sleep quality in postmenopausal women. Sleep. 2003;26(7):830–836.
  • Humpel N, Iverson DC. Sleep quality, fatigue and physical activity following a cancer diagnosis. Eur J Cancer Care (Engl). 2010;19(6):761–768.
  • Young-McCaughan S, Mays MZ, Arzola SM, et al. Research and commentary: change in exercise tolerance, activity and sleep patterns, and quality of life in patients with cancer participating in a structured exercise program. Oncol Nurs Forum. 2003;30(3):441–454.
  • Mustian KM, Palesh O, Sprod L, et al. Effect of YOCAS yoga on sleep, fatigue, and quality of life: a URCC CCOP randomized, controlled clinical trial among 410 cancer survivors. J Clin Oncol. 2010;28(15_suppl):9013.
  • Mustian KM, Sprod LK, Janelsins M, et al. Multicenter, randomized controlled trial of yoga for sleep quality among cancer survivors. J Clin Oncol. 2013;31(26):3233–3241.
  • Morgenthaler T, Kramer M, Alessi C, et al. Practice parameters for the psychological and behavioral treatment of insomnia: an update. an American Academy of Sleep Medicine Report. Sleep. 2006;29(11):1415–1419.
  • Savard J, Simard S, Ivers H, et al. Randomized study on the efficacy of cognitive-behavioral therapy for insomnia secondary to breast cancer, part I: sleep and psychological effects. J Clin Oncol. 2005;23(25):6083–6096.
  • Espie CA, Fleming L, Cassidy J, et al. Randomized controlled clinical effectiveness trial of cognitive behavior therapy compared with treatment as usual for persistent insomnia in patients with cancer. J Clin Oncol. 2008;26(28):4651–4658.
  • Berger AM, Kuhn BR, Farr LA, et al. Behavioral therapy intervention trial to improve sleep quality and cancer-related fatigue. Psychooncology. 2009;18(6):634–646.
  • Berger AM, Kuhn BR, Farr LA, et al. One-year outcomes of a behavioral therapy intervention trial on sleep quality and cancer-related fatigue. J Clin Oncol. 2009;27(35):6033–6040.
  • Barsevick A, Beck SL, Dudley WN, et al. Efficacy of an intervention for fatigue and sleep disturbance during cancer chemotherapy. J Pain Symptom Manage. 2010;40(2):200–216.
  • Chung K, Lee C, Yeung W, et al. Sleep hygiene education as a treatment of insomnia: a systematic review and meta-analysis. Fam Pract. 2018;35(4):365–375.
  • Neikrug AB, Rissling M, Trofimenko V, et al. Bright light therapy protects women from circadian rhythm desynchronization during chemotherapy for breast cancer. Behav Sleep Med. 2012;10(3):202–216.
  • Costantini C, Ale-Ali A, Helsten T. Sleep aid prescribing practices during neoadjuvant or adjuvant chemotherapy for breast cancer. J Palliat Med. 2011;14(5):563–566.
  • Chen WY, Giobbie-Hurder A, Gantman K, et al. A randomized, placebo-controlled trial of melatonin on breast cancer survivors: impact on sleep, mood, and hot flashes. Breast Cancer Res Treat. 2014;145(2):381–388.
  • Cavaletti G, Marmiroli P, and Marmiroli P. Chemotherapy-induced peripheral neurotoxicity. Nat Rev Neurol. 2010;6(12):657–666.
  • Jordan B, Margulies A, Cardoso F, et al. Systemic anticancer therapy-induced peripheral and central neurotoxicity: ESMO-EONS-EANO clinical practice guidelines for diagnosis, prevention, treatment and follow-up. Ann Oncol. 2020;31(10):1306–1319.
  • Bao T, Basal C, Seluzicki C, et al. Long-term chemotherapy-induced peripheral neuropathy among breast cancer survivors: prevalence, risk factors, and fall risk. Breast Cancer Res Treat. 2016;159(2):327–333.
  • Rivera DR, Ganz PA, Weyrich MS, et al. Chemotherapy-associated peripheral neuropathy in patients with early-stage breast cancer: a systematic review. J Natl Cancer Inst. 2018;110(2).
  • Argyriou AA, Koltzenburg M, Polychronopoulos P, et al. Peripheral nerve damage associated with administration of taxanes in patients with cancer. Crit Rev Oncol Hematol. 2008;66(3):218–228.
  • Rivera E, Cianfrocca M. Overview of neuropathy associated with taxanes for the treatment of metastatic breast cancer. Cancer Chemother Pharmacol. 2015;75(4):659–670.
  • Biganzoli L, Cinieri S, Berardi R, et al. EFFECT: a randomized phase II study of efficacy and impact on function of two doses of nab-paclitaxel as first-line treatment in older women with advanced breast cancer. BCR. 2020;22(1):83.
  • Tofthagen C, Visovsky CM, Hopgood R. Chemotherapy-induced peripheral neuropathy: an algorithm to guide nursing management. Clin J Oncol Nurs. 2013;17(2):138–144.
  • Crevenna R, Ashbury FD. Physical interventions for patients suffering from chemotherapy-induced polyneuropathy. Support Care Cancer. 2018;26(4):1017–1018.
  • Beijers AJM, Bonhof CS, Mols F, et al. Multicenter randomized controlled trial to evaluate the efficacy and tolerability of frozen gloves for the prevention of chemotherapy-induced peripheral neuropathy. Ann Oncol. 2020;31(1):131–136.
  • Kleckner IR, Kamen C, Cole C, et al. Effects of exercise on inflammation in patients receiving chemotherapy: a nationwide NCORP randomized clinical trial. Support Cancer Ther. 2019;27(12):4615–4625.
  • Kleckner IR, Kamen C, Gewandter JS, et al. Effects of exercise during chemotherapy on chemotherapy-induced peripheral neuropathy: a multicenter, randomized controlled trial. Support Care Cancer. 2018;26(4):1019–1028.
  • Lu W, Giobbie-Hurder A, Freedman RA, et al. Acupuncture for chemotherapy-induced peripheral neuropathy in breast cancer survivors: a randomized controlled pilot trial. Oncologist. 2020;25(4):310–318.
  • Kolb NA, Smith G, Singleton JR, et al. The association of chemotherapy-induced peripheral neuropathy symptoms and the risk of falling. JAMA Neurol. 2016;73(7):860–866.
  • Tamburin S, Park SB, Schenone A, et al. Rehabilitation, exercise, and related non-pharmacological interventions for chemotherapy-induced peripheral neurotoxicity: systematic review and evidence-based recommendations. Crit Rev Oncol Hematol. 2022;171:103575.
  • Li Y, Lustberg MB, Hu S. Emerging pharmacological and non-pharmacological therapeutics for prevention and treatment of chemotherapy-induced peripheral neuropathy. Cancers (Basel). 2021;13(4):766.
  • Smith EML, Pang H, Cirrincione C, et al. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. JAMA. 2013;309(13):1359.
  • Durand JP, Deplanque G, Montheil V, et al. Efficacy of venlafaxine for the prevention and relief of oxaliplatin-induced acute neurotoxicity: results of EFFOX, a randomized, double-blind, placebo-controlled phase iii trial. Ann Oncol. 2012;23(1):200–205.
  • Kautio A, Haanpää M, Saarto T, et al. Amitriptyline in the treatment of chemotherapy-induced neuropathic symptoms. J Pain Symptom Manage. 2008;35(1):31–39.
  • Desforges AD, Hebert CM, Spence AL, et al. Treatment and diagnosis of chemotherapy-induced peripheral neuropathy: an update. Biomed Pharmacother. 2022;147:112671.
  • Szklener K, Szklener S, Michalski A, et al. Dietary supplements in chemotherapy-induced peripheral neuropathy: a new hope? Nutrients. 2022;14(3):625.
  • Zandbergen N, de Rooij BH, Vos MC, et al. Changes in health-related quality of life among gynecologic cancer survivors during the two years after initial treatment: a longitudinal analysis. Acta Oncol. 2019;58(5):790–800.
  • Ahles TA, Root JC, Ryan EL. Cancer- and cancer treatment-associated cognitive change: an update on the state of the science. J Clin Oncol. 2012;30(30):3675–3686.
  • Chen VC, Lin T, Yeh D, et al. Predicting chemo-brain in breast cancer survivors using multiple MRI features and machine-learning. Magn Reson Med. 2019;81(5):3304–3313.
  • Minisini A, Atalay G, Bottomley A, et al. What is the effect of systemic anticancer treatment on cognitive function? Lancet Oncol. 2004;5(5):273–282.
  • Minisini AM, De Faccio S, Ermacora P, et al. Cognitive functions and elderly cancer patients receiving anticancer treatment: a prospective study. Crit Rev Oncol Hematol. 2008;67(1):71–79.
  • Nguyen LD, Ehrlich BE. Cellular mechanisms and treatments for chemobrain: insight from aging and neurodegenerative diseases. EMBO Mol Med. 2020;12(6):e12075.
  • Breuer B, Anderson R, and Anderson R. The relationship of tamoxifen with dementia, depression, and dependence in activities of daily living in elderly nursing home residents. Women Health. 2000;31(1):71–85.
  • Paganini-Hill A, Clark LJ. Preliminary assessment of cognitive function in breast cancer patients treated with tamoxifen. Breast Cancer Res Treat. 2000;64(2):165–176.
  • Phillips K, Regan MM, Ribi K, et al. Adjuvant ovarian function suppression and cognitive function in women with breast cancer. Br J Cancer. 2016;114(9):956–964.
  • van Dam FS, Schagen SB, Muller MJ, et al. Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy. J Natl Cancer Inst. 1998;90(3):210–218.
  • Marín AP, Sánchez AR, Arranz EE, et al. Adjuvant chemotherapy for breast cancer and cognitive impairment. South Med J. 2009;102(9):929–934.
  • Saykin AJ, Ahles TA, McDonald BC. Mechanisms of chemotherapy-induced cognitive disorders: neuropsychological, pathophysiological, and neuroimaging perspectives. Semin Neurol. 2003;8(4):201–216.
  • Chiesa A, Calati R, Serretti A. Does mindfulness training improve cognitive abilities? a systematic review of neuropsychological findings. Clin Psychol Rev. 2011;31(3):449–464.
  • Duval A, Davis CG, Khoo EL, et al. Mindfulness-based stress reduction and cognitive function among breast cancer survivors: a randomized controlled trial. Cancer. 2022;128(13):2520–2528.
  • Hartman SJ, Nelson SH, Myers E, et al. Randomized controlled trial of increasing physical activity on objectively measured and self-reported cognitive functioning among breast cancer survivors: the memory & motion study. Cancer. 2018;124(1):192–202.
  • Von Ah D, Carpenter JS, Saykin A, et al. Advanced cognitive training for breast cancer survivors: a randomized controlled trial. Breast Cancer Res Treat. 2012;135(3):799–809.
  • Alvarez J, Meyer FL, Granoff DL, et al. The effect of EEG biofeedback on reducing postcancer cognitive impairment. Integr Cancer Ther. 2013;12(6):475–487.
  • Kohli S, Fisher SG, Tra Y, et al. The effect of modafinil on cognitive function in breast cancer survivors. Cancer. 2009;115(12):2605–2616.
  • Shaw EG, Rosdhal R, D’Agostino RB, et al. Phase II study of donepezil in irradiated brain tumor patients: effect on cognitive function, mood, and quality of life. J Clin Oncol. 2006;24(9):1415–1420.
  • Dinas KD. Impact of breast cancer treatment on fertility. Adv Exp Med Biol. 2020;1252:175–179.
  • Goodwin PJ, Ennis M, Pritchard KI, et al. Risk of menopause during the first year after breast cancer diagnosis. J Clin Oncol. 1999;17(8):2365–2370.
  • Goldfarb SB, Turan V, Bedoschi G, et al. Impact of adjuvant chemotherapy or tamoxifen-alone on the ovarian reserve of young women with breast cancer. Breast Cancer Res Treat. 2021;185(1):165–173.
  • Arecco L, Ruelle T, Martelli V, et al. How to protect ovarian function before and during chemotherapy? J Clin Med. 2021;10(18):4192.
  • Moore HCF, Unger JM, Phillips K, et al. Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. N Engl J Med. 2015;372(10):923–932.
  • Franzoi MA, Agostinetto E, Perachino M, et al. Evidence-based approaches for the management of side-effects of adjuvant endocrine therapy in patients with breast cancer. Lancet Oncol. 2021;22(7):e303–13.
  • Bober SL, Fine E, Recklitis CJ. Sexual health and rehabilitation after ovarian suppression treatment (SHARE-OS): a clinical intervention for young breast cancer survivors. J Cancer Survivorship. 2020;14(1):26–30.
  • Enemchukwu EA. CO2 laser treatment is effective for symptoms of vaginal atrophy: no. J Urol. 2017;198(6):1228–1229.
  • Arêas F, Valadares Ana LR, Conde DM, et al. The effect of vaginal erbium laser treatment on sexual function and vaginal health in women with a history of breast cancer and symptoms of the genitourinary syndrome of menopause: a prospective study. Menopause. 2019;26(9):1052–1058.
  • Becorpi A, Campisciano G, Zanotta N, et al. Fractional CO2 laser for genitourinary syndrome of menopause in breast cancer survivors: clinical, immunological, and microbiological aspects. Lasers Med Sci. 2018;33(5):1047–1054.
  • Carson JW, Carson KM, Porter LS, et al. Yoga of awareness program for menopausal symptoms in breast cancer survivors: results from a randomized trial. Support Care Cancer. 2009;17(10):1301–1309.
  • Walker EM, Rodriguez AI, Kohn B, et al. Acupuncture versus venlafaxine for the management of vasomotor symptoms in patients with hormone receptor–positive breast cancer: a randomized controlled trial. J Clin Oncol. 2010;28(4):634–640.
  • Moegele M, Buchholz S, Seitz S, et al. Vaginal estrogen therapy in postmenopausal breast cancer patients treated with aromatase inhibitors. Arch Gynecol Obstet. 2012;285(5):1397–1402.
  • Sanchez-Rovira P, Hirschberg AL, Gil-Gil M, et al. A phase II prospective, randomized, double-blind placebo-controlled and multicenter clinical trial to assess the safety of 0.005% estriol vaginal gel in hormone receptor-positive postmenopausal women with early stage breast cancer in treatment with aromatase inhibitor in the adjuvant setting. Oncologist. 2020;25(12):e1846–1854.
  • Holmberg L, Anderson H. HABITS (Hormonal Replacement Therapy after Breast Cancer—Is it safe?), A randomised comparison: trial stopped. Lancet. 2004;363(9407):453–455.
  • Fahlén M, Fornander T, Johansson H, et al. Hormone replacement therapy after breast cancer: 10 year follow up of the Stockholm randomised trial. Eur J Cancer. 2013;49(1):52–59.
  • Sismondi P, Kimmig R, Kubista E, et al. Effects of tibolone on climacteric symptoms and quality of life in breast cancer patients—data from LIBERATE trial. Maturitas. 2011;70(4):365–372.
  • Wurz GT, Soe LH, DeGregorio MW. Ospemifene, vulvovaginal atrophy, and breast cancer. Maturitas. 2013;74(3):220–225.
  • Loprinzi CL, Sloan J, Stearns V, et al. Newer antidepressants and gabapentin for hot flashes: an individual patient pooled analysis. J Clin Oncol. 2009;27(17):2831–2837.
  • Goldberg RM, Loprinzi CL, O’Fallon JR, et al. Transdermal clonidine for ameliorating tamoxifen-induced hot flashes. J Clin Oncol. 1994;12(1):155–158.
  • Loprinzi CL, Kugler JW, Sloan JA, et al. Venlafaxine in management of hot flashes in survivors of breast cancer: a randomised controlled trial. Lancet. 2000;356(9247):2059–2063.
  • Boekhout AH, Vincent AD, Dalesio OB, et al. Management of hot flashes in patients who have breast cancer with venlafaxine and clonidine: a randomized, double-blind, placebo-controlled trial. J Clin Oncol. 2011;29(29):3862–3868.
  • Leon-Ferre RA, Novotny PJ, Wolfe EG, et al. Oxybutynin vs placebo for hot flashes in women with or without breast cancer: a randomized, double-blind clinical trial (ACCRU SC-1603). JNCI Cancer Spectrum. 2020;4(1):kz088.
  • Rachner TD, Coleman R, Hadji P, et al. Bone health during endocrine therapy for cancer. Lancet Diabetes Endocrinol. 2018;6(11):901–910.
  • Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials. Lancet. 2015;386(10001):1353–1361.
  • Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2019;30(8):1194–1220.
  • Gralow JR, Biermann JS, Farooki A, et al. NCCN task force report: bone health in cancer care. J National Compr Cancer Network. 2013;11(suppl 3):S-1-S–50.
  • Zamorano JL, Lancellotti P, Muñoz DR, et al. 2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC committee for practice guidelines: the task force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur Heart J. 2016;37(36):2768–2801.
  • Moliterni A, Bonadonna G, Valagussa P, et al. Cyclophosphamide, methotrexate, and fluorouracil with and without doxorubicin in the adjuvant treatment of resectable breast cancer with one to three positive axillary nodes. J Clin Oncol. 1991;9(7):1124–1130.
  • Bonadonna G, Zambetti M, Valagussa P. Sequential or alternating doxorubicin and CMF regimens in breast cancer with more than three positive nodes: ten-year results. JAMA. 1995;273(7):542–547.
  • Bonadonna G, Zambetti M, Bumma C, et al. Multimodal treatment with primary single-agent epirubicin in operable breast cancer: 5-year experience of the Michelangelo Cooperative Group. Ann Oncol. 2002;13(7):1049–1058.
  • Earl HM, Hiller L, Dunn JA, et al. for the NEAT Investigators and the SCTBG. Adjuvant epirubicin followed by cyclophosphamide, methotrexate and fluorouracil (CMF) vs CMF in early breast cancer: results with over 7 years median follow-up from the randomised phase III NEAT/BR9601 trials. Br J Cancer. 2012;107(8):1257–1267.
  • Von Hoff D. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med. 1979;91(5):710.
  • Bonadonna G, Gianni L, Santoro A, et al. Drugs ten years later: epirubicin. Ann Oncol. 1993;4(5):359–369.
  • Wojtukiewicz MZ, Omyla J, Kozlowski L, et al. Cardiotoxicity of anthracycline. Postepy Hig Med Dosw (Online). 2000;54(4):467–485.
  • Plana JC, Galderisi M, Barac A, et al. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Euro Heart J Cardiovascular Imaging. 2014;15(10):1063–1093.
  • Cardinale D, Colombo A, Bacchiani G, et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation. 2015;131(22):1981–1988.
  • Cardinale D, Colombo A, Lamantia G, et al. Anthracycline-induced cardiomyopathy. J Am Coll Cardiol. 2010;55(3):213–220.
  • Cardinale D, Colombo A, Sandri MT, et al. Prevention of high-dose chemotherapy–induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation. 2006;114(23):2474–2481.
  • Raccomandazioni pratiche. CARDIO-ONCOLOGIA 2019. N.d. n.d.
  • van Dalen EC, van der Pal HJH, Kremer LCM. Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Edited by Cochrane Gynaecological, Neuro-oncology and Orphan Cancer Group. Cochrane Database Syst Rev. 2016.
  • Rafiyath SM, Rasul M, Lee B, et al. Comparison of safety and toxicity of liposomal doxorubicin vs. conventional anthracyclines: a meta-analysis. Exp Hematol Oncol. 2012;1(1):10.
  • Yamaguchi N, Fujii T, Aoi S, et al. Comparison of cardiac events associated with liposomal doxorubicin, epirubicin and doxorubicin in breast cancer: a Bayesian network meta-analysis. Eur J Cancer. 2015;51(16):2314–2320.
  • O’Brien MER, Wigler N, Inbar M, et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl(CAELYXTM/Doxil®) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol. 2004;15(3):440–449.
  • Lyon AR, López-Fernández T, Couch LS, et al. ESC scientific document group, 2022 ESC guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS): developed by the task force on cardio-oncology of the European Society of Cardiology (ESC). Euro Heart J Cardiovascular Imaging. 2022;23(10):e333–e465.
  • van Dalen EC, Caron HN, Dickinson HO, et al. Cardioprotective interventions for cancer patients receiving anthracyclines. edited by Cochrane gynaecological, neuro-oncology and orphan cancer group. Cochrane Database Syst Rev. 2011;2016(9).
  • Smith LA, Cornelius VR, Plummer CJ, et al. Cardiotoxicity of anthracycline agents for the treatment of cancer: systematic review and meta-analysis of randomised controlled trials. BMC Cancer. 2010;10(1):337.
  • Asselin BL, Devidas M, Chen L, et al. Cardioprotection and safety of dexrazoxane in patients treated for newly diagnosed T-cell acute lymphoblastic leukemia or advanced-stage lymphoblastic non-Hodgkin lymphoma: a report of the Children’s Oncology Group randomized trial Pediatric Oncology Group 9404. J Clin Oncol. 2016;34(8):854–862.
  • Reichardt P, Tabone M, Mora J, et al. Risk–benefit of dexrazoxane for preventing anthracycline-related cardiotoxicity: re-evaluating the European labeling. Future Oncol. 2018;14(25):2663–2676.
  • Caspani F, Tralongo AC, Campiotti L, et al. Prevention of anthracycline-induced cardiotoxicity: a systematic review and meta-analysis. Intern Emerg Med. 2021;16(2):477–486.
  • Vaduganathan M, Hirji S, Qamar A, et al. Efficacy of neurohormonal therapies in preventing cardiotoxicity in patients with cancer undergoing chemotherapy. J Am Coll Cardiol CardioOnc. 2019;1(1):54–65.
  • Li X, Li Y, Zhang T, et al. Role of cardioprotective agents on chemotherapy-induced heart failure: a systematic review and network meta-analysis of randomized controlled trials. Pharmacol Res. 2020;151:104577.
  • Cardinale D, Colombo A, Sandri MT, et al. Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin converting enzyme inhibition. Circulation. 2006;114(23):2474–2481.
  • Gulati G, Heck SL, Ree AH, et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): a 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur Heart J. 2016;37(21):1671–1680.
  • Vaduganathan M, Jirji SA, Qamar A, et al. Efficacy of neurohormonal therapies in preventing cardiotoxicity in patients with cancer undergoing chemotherapy. JACC: CardioOncology. 2019;1(1):54–65.
  • Kaya MG, Ozkan M, Gunebakmaz O, et al. Protective effects of nebivolol against anthracycline-induced cardiomyopathy: a randomized control study. Int J Cardiol. 2013;167(5):2306–2310.
  • Shah P, Garris R, Abboud R, et al. Meta-analysis comparing usefulness of beta blockers to preserve left ventricular function during anthracycline therapy. Am J Cardiol. 2019;124(5):789–794.
  • Ma Y, Bai F, Qin F, et al. Beta-blockers for the primary prevention of anthracycline-induced cardiotoxicity: a meta-analysis of randomized controlled trials. BMC Pharmacol Toxicol. 2019;20(1):18.
  • Huang S, Zhao Q, Yang ZG, et al. Protective role of beta-blockers in chemotherapy-induced cardiotoxicity-a systematic review and meta-analysis of carvedilol. Heart Fail Rev. 2019;24(3):325–333.
  • Polk A, Vaage-Nilsen M, Vistisen K, et al. Cardiotoxicity in cancer patients treated with 5-fluorouracil or capecitabine: a systematic review of incidence, manifestations and predisposing factors. Cancer Treat Rev. 2013;39(8):974–984.
  • Rowinsky EK, McGuire WP, Guarnieri T, et al. Cardiac disturbances during the administration of taxol. J Clin Oncol. 1991;9(9):1704–1712.
  • Martel S, Maurer C, Lambertini M, et al. Breast cancer treatment-induced cardiotoxicity. Expert Opin Drug Saf. 2017;16(9):1021–1038.
  • Pondé NF, Lambertini M, de Azambuja E. Twenty years of anti-HER2 therapy-associated cardiotoxicity. ESMO Open. 2016;1(4):e000073.
  • Moja L., Brambilla C., Compagnoni A., et al. Trastuzumab Containing Regimens for Early Breast Cancer. In Cochrane Database of Systematic Reviews, edited by The Cochrane Collaboration. 2006
  • Seidman A, Hudis C, Pierri MK, et al. Cardiac dysfunction in the trastuzumab clinical trials experience. J Clin Oncol. 2002;20(5):1215–1221.
  • Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344(11):783–792.
  • Advani PP, Ballman KV, Dockter TJ, et al. Long-term cardiac safety analysis of NCCTG N9831 (Alliance) adjuvant trastuzumab trial. J Clin Oncol. 2016;34(6):581–587.
  • Ewer MS, Ewer SM. Cardiotoxicity of anticancer treatments. Nat Rev Cardiol. 2015;12(9):547–558.
  • Jawa Z, Perez RM, Garlie L, et al. Risk factors of trastuzumab-induced cardiotoxicity in breast cancer: a meta-analysis. Medicine (Baltimore). 2016;95(44):e5195.
  • Cameron D, Piccart-Gebhart MJ, Gelber RD, et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin adjuvant (HERA) trial. Lancet. 2017;389(10075):1195–1205.
  • Goldhirsch A, Gelber RD, Piccart-Gebhart MJ, et al. 2 years versus 1 year of adjuvant trastuzumab for HER2-positive breast cancer (HERA): an open-label, randomised controlled trial. Lancet. 2013;382(9897):1021–1028.
  • Barish R, Gates E, Barac A. Trastuzumab-Induced Cardiomyopathy. Cardiol Clin. 2019 18;37(4):407–418.
  • Cardinale D, Colombo A, Torrisi R, et al. Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol. 2010;28(25):3910–3916.
  • von Minckwitz G, Procter M, de Azambuja E, et al. Adjuvant pertuzumab and trastuzumab in early HER2-positive breast cancer. N Engl J Med. 2017;377(2):122–131.
  • Gianni L, Pienkowski T, Im Y, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(1):25–32.
  • von Minckwitz G, Huang C, Mano MS, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019;380(7):617–628.
  • Baselga J, Bradbury I, Eidtmann H, et al. Lapatinib with Trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. 2012;379(9816):633–640.
  • Wood ME, Vogel V, Ng A, et al. Second malignant neoplasms: assessment and strategies for risk reduction. J Clin Oncol. 2012;30(30):3734–3745.
  • Smith RE, Bryant J, DeCillis A, et al. Acute myeloid leukemia and myelodysplastic syndrome after doxorubicin-cyclophosphamide adjuvant therapy for operable breast cancer: the national surgical adjuvant breast and bowel project experience. J Clin Oncol. 2003;21(7):1195–1204.
  • Wolff AC, Blackford AL, Visvanathan K, et al. Risk of marrow neoplasms after adjuvant breast cancer therapy: the national comprehensive cancer network experience. J Clin Oncol. 2015;33(4):340–348.
  • Leone G, Pagano L, Ben-Yehuda D, et al. Therapy-related leukemia and myelodysplasia: susceptibility and incidence. Haematologica. 2007;92(10):1389–1398.
  • Azarova AM, Lyu YL, Lin C, et al. Roles of DNA topoisomerase II isozymes in chemotherapy and secondary malignancies. Proc Natl Acad Sci U S A. 2007;104(26):11014–11019.
  • Travis LB, Andersson M, Gospodarowicz M, et al. Treatment-associated leukemia following testicular cancer. J Natl Cancer Inst. 2000;92(14):1165–1171.
  • Travis LB, Holowaty EJ, Bergfeldt K, et al. Risk of leukemia after platinum-based chemotherapy for ovarian cancer. N Engl J Med. 1999;340(5):351–357.
  • Schaapveld M, Visser O, Louwman MJ, et al. Risk of new primary nonbreast cancers after breast cancer treatment: a Dutch population-based study. J Clin Oncol. 2008;26(8):1239–1246.
  • Turcotte LM, Liu Q, Yasui Y, et al. Chemotherapy and risk of subsequent malignant neoplasms in the childhood cancer survivor study cohort. J Clin Oncol. 2019;37(34):3310–3319.