Drug-related toxicity in breast cancer patients: a new path towards tailored treatment?—a narrative review
Review Article

Drug-related toxicity in breast cancer patients: a new path towards tailored treatment?—a narrative review

Eleonora Lai1, Mara Persano1, Marco Dubois1, Dario Spanu1, Clelia Donisi1, Marta Pozzari1, Giulia Deias1, Giorgio Saba1, Marco Migliari1, Nicole Liscia2, Mariele Dessì1, Mario Scartozzi1, Francesco Atzori1

1Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy; 2Medical Oncology Unit, Sapienza University of Rome-University Hospital and University of Cagliari, Cagliari, Italy

Contributions: (I) Conception and design: E Lai, F Atzori; (II) Administrative support: None; (III) Provision of study materials or patients: E Lai, M Persano, M Dubois, D Spanu, C Donisi, M Pozzari, G Deias, G Saba, M Migliari; (IV) Collection and assembly of data: E Lai, N Liscia, M Dessì, M Scartozzi, F Atzori; (V) Data analysis and interpretation: E Lai, N Liscia, M Dessì, M Scartozzi, F Atzori; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Francesco Atzori, MD. Medical Oncology Unit, University Hospital and University of Cagliari, AOU Cagliari, PO Policlinico Universitario “Duilio Casula”, SC Oncologia Medica, Blocco Q 4° Piano, SS 554 Km 4.500 Bivio per Sestu-09042 Monserrato, Cagliari, Italy. Email: francescoatzori74@yahoo.it.

Background and Objective: Side effects of drugs administered for breast cancer (BC) according to cancer biology and patients’ clinical features can limit patient’s compliance and consequently the benefit. Precision medicine is a growing research field to improve the management of BC patient’s care and evaluating toxicities profile could be an interesting feature in the choice a personalised treatment. This review aims to explore the implications of a tailored anti-cancer therapy knowing the safety profile and predisposing factors for potential specific drug-related toxicities. More specifically, this review aims to focus on personalised medicine and patients’ selection according to clinical, laboratory and genetic features involved in adverse events (AEs) development.

Methods: We performed an extensive literature research on PubMed and available Medical Oncology Congresses resources regarding tailored anti-cancer therapy for BC, toxicity profile and predisposing factors for potential specific drug-related toxicities, selecting publications in English in a timeframe from January 1, 1997 to December 31, 2021. Furthermore, we provide a focus on personalised medicine with potential implications on patients’ selection.

Key Contents and Findings: Literature review focused on the role of anti-cancer agents toxicity profile and AEs predisposing factors in the personalisation of BC patients treatment. For most anti-cancer agents, potential safety-related biomarkers and the implications of clinical features of BC patients for a tailored treatment were investigated.

Conclusions: A safety profile-tailored treatment combined with the clinical characteristics of BC patients and potential biomarkers predisposing to specific treatment-related toxicities might be particularly helpful in the therapeutic choice in the context of precision oncology. In this perspective, the knowledge and the application of these factors would be crucial for better choice and management of the best care for the right patient.

Keywords: Toxicity; precision oncology; breast cancer (BC); patient selection


Received: 10 September 2021; Accepted: 23 March 2022; Published: 30 June 2022.

doi: 10.21037/pcm-21-38


Introduction

Breast cancer (BC) is the most frequently diagnosed tumour and one of the leading causes of cancer death in women worldwide (1). Over the years, several agents have been introduced for the management of both early and advanced stage of BC, thus leading to a substantial improvement of survival outcomes. Indeed, the combination of conventional chemotherapy, endocrine therapy, target agents and immunotherapy are currently an integral part of clinical practice and have been validated in clinical trials (2). Treatment is tailored on the basis of tumour biologic profile and disease burden, patient’s clinical features, comorbidities and preferences. Unfortunately, no drug is void of adverse events (AEs) and not all patients respond to treatment; moreover, some of them develop significant toxicities without obtaining clinical benefit. For these reasons, it crucial to identify factors which might improve the selection of patients who are candidate to a specific treatment.

Recently, the concept of “precision oncology”, on the basis of which therapy is delivered to patients according to unique patient clinical and molecular features, has gained growing importance (3). The scientific rational is mainly represented by the identification of an oncogenic mutation in a patient’s cancer genome that drives cancer growth, followed by treatment with target-selective drugs inhibiting that specific mutation product (4). Genomic sequencing results can be useful to classify cancer, predict prognosis and target therapies. Next-generation sequencing allows rapid and cost-effective sequencing of large portions of the genome, becoming crucial in the field of cancer genomics (5).

The identification of clinically useful gene expression signatures might be used to personalise treatment not only with the aim to improve survival, but also to reduce the risk of toxicity (6). However, not all Cancer Centres are provided with genomic testing that can be offered to BC patients in clinical practice. This leads to the need to identify further clinical and easy-to-use factors which might help improving patients’ selection.

In this perspective, the toxicity profile is an interesting point to be investigated to personalise treatment. In this review, we aim to focus on personalised medicine and in particular to give an overview of the safety profile of the main anti-cancer agents for the treatment of BC patients with potential implications on patients’ selection, taking into consideration their clinical characteristics. We present the following article in accordance with the Narrative Review reporting checklist (available at https://pcm.amegroups.com/article/view/10.21037/pcm-21-38/rc).


Methods

We performed an extended literature research on PubMed and available main Medical Oncology Congresses resources on tailored anti-cancer therapy for BC, toxicity profile and predisposing factors for potential specific drug-related toxicities. We selected papers published in a timeframe from January 1, 1997 to December 31, 2021. Our review was limited to manuscripts in the English language (Table 1). We provide a focus on personalised medicine with potential implications on patients’ selection.

Table 1

The search strategy summary

Items Specification
Date of search (specified to date, month and year) 15 February 2022
Databases and other sources searched Extensive literature research on PubMed, Medical oncology congresses resources (ESMO, ASCO)
Search terms used (including MeSH and free text search terms and filters) Breast cancer, toxicity profile, drug related-toxicities and predisposing factors
Timeframe January 1, 1997 to December 31, 2021
Inclusion and exclusion criteria (study type, language restrictions, etc.) English language
Selection process (who conducted the selection, whether it was conducted independently, how consensus was obtained, etc.) The authors conducted independently the selection of articles. Consensus was not required

ESMO, European Society for Medical Oncology; ASCO, American Society of Clinical Oncology.


Anthracyclines

Anthracyclines represent a cornerstone of BC treatment (7). Cardiotoxicity, myelosuppression, nausea and vomiting are the main AEs, for which various studies assessed potential predisposing factors. An analysis by Chen et al. in 211 BC patients treated with epirubicin-cyclophosphamide-docetaxel chemotherapy reported a significant correlation between fibroblast growth factor receptor 2 (FGFR2) rs2420946 CC genotype and higher AEs occurrence vs. TT (P=0.038) and CT/TT genotypes (P=0.019); similar results were found for FGFR2 rs2981578 AG genotype vs. GG genotype (P<0.0001) (8).

Cardiotoxicity

Vaitiekus et al. identified a significant association between HFE gene H63D single nucleotide polymorphism (SNP) and subclinical cardiac damage in 81 BC patients treated with doxorubicin-based chemotherapy (P<0.005) (7). Eighteen SNPs of NFKBIL1, TNF-a, ATP6V1G2-DDX39B, MSH5, MICA, LTA, BAT1, and NOTCH4 were suggested as potentially related to doxorubicin-induced cardiotoxicity (9). A genome-wide association study in 3,431 patients of three phase III adjuvant BC trials found an association of rs28714259 SNP with congestive heart failure (CHF) induced by anthracyclines (10). Vulsteke et al. identified 6 cycles of 5-fluorouracil, epirubicin and cyclophosphamide vs. 3 cycles (OR 1.3, 95% CI: 1.1–1.4, P<0.001) and heterozygous status for ABCC1 rs246221 T-allele vs. homozygous (OR 1.6, 95% CI: 1.1–2.3, P=0.02) as significantly related to left ventricular ejection fraction (LVEF) reduction >10% in early BC (EBC) (11). Another study detected UGT2B7-161 T allele as a potential independent biomarker of low occurrence of cardiotoxicity during adjuvant epirubicin-cyclophosphamide-docetaxel chemotherapy (P=0.004) (12).

Haematological toxicity

In a study by Cui et al. CBR1 rs20572 (C>T), ABCG2 rs2231142 (G>T) SNPs involved in anthracyclines pharmacokinetics or the combination of two polymorphic alleles were significantly associated to reduced risk of leukopenia (OR 0.412, 95% CI: 0.187–0.905, P=0.025) and neutropenia (OR 0.354, 95% CI: 0.148–0.846, P=0.018) in 194 BC patients receiving adjuvant anthracyclines. Moreover, patients carrying polymorphic allele T of CBR1 rs20572, or polymorphic allele C of AKR1A1 rs2088102 combined with ABCG2 rs2231142 (G>T) plus SLC22A16 rs6907567 (A>G) mutations showed an extremely low risk of grade 3–4 anaemia (OR 0.058, 95% CI: 0.006–0.554, P=0.008; OR 0.065, 95% CI: 0.006–0.689, P=0.022; OR 0.037, 95% CI: 0.004–0.36, P=0.015, respectively). Thus, these SNPs might be useful to identify which patients who are less likely to develop haematological AEs (13).

Gastrointestinal toxicity: nausea and vomiting

A study conducted in 110 BC patients treated with epirubicin +/− cyclophosphamide exploring the role of 5-hydroxytryptamine receptor 3 (HTR3C) genes for chemotherapy-induced nausea and vomiting (CINV), the variant genotype of K163N (HTR3C) was associated with vomiting (P=0.009) (14). Tsuji et al. suggested that TACR1 1323TT SNP, involving the gene encoding the neurokinin 1 receptor, might be a genetic risk factor for the development of delayed CINV (OR, 2.57; P=0.014) (15).


Fluoropyrimidines

Over 30% of patients treated with fluoropyrimidines have severe treatment-related side effects, such as diarrhoea, hand-foot syndrome, myelosuppression and mucositis (16). Fluoropyrimidine-related toxicity is often due to the presence of genetic variants in the gene encoding the enzyme dihydropyrimidine dehydrogenase (DPYD), the leading enzyme involved in fluoropyrimidine degradation (17,18). A proportion of 3–5% of the European and North American have a DYPD deficient activity (~50% reduction), resulting in major risk of severe fluoropyrimidine-related AEs occurrence if treated with full dosage (19). Actually, four DPYD variants are considered most clinically relevant for their statistically significant association with severe toxicity: c.1905+1G>A, c.2846A>T, c.1679T>G, and c.1236G>A (20). For several years routine DPYD genotype screening prior to fluoropyrimidine administration has not been the standard of care for BC management. However, recent studies have shown significant clinical and financial benefits of routine DPYD genotype screening application also in this malignancy (21-25). Notably, this has increasing importance with the introduction of adjuvant capecitabine at similar doses as those for colorectal cancer in human epidermal growth factor receptor 2 (HER2)-negative, stage I–IIIB BC without complete pathologic response or with a complete response with positive lymph nodes after neoadjuvant chemotherapy and surgery (26).

For these reasons, EMA strongly recommends DYPD testing before starting treatment with infusional fluorouracil or with the related pro-drugs, capecitabine and tegafur (27). In case of DYPD variants, the dose of fluoropyrimidines should be adapted according to guidelines and recommendations.


Anti-HER2 agents

Anti-HER2-targeted drugs in the last 20 years dramatically changed the clinical outcome of HER2 positive BC patients (28).

Cardiotoxicity

Cardiotoxicity is one of most concerning AE associated with the anti-HER2 therapy, both in terms of symptomatic events, such CHF, and asymptomatic, such the decrease of LVEF. Cardiac AEs have been extensively studied in both EBC and metastatic BC (MBC) (29).

In the pivotal phase III randomised clinical trial (RCT) that led to its approval as first line treatment in MBC, trastuzumab, an anti-HER2 monoclonal antibody (mAb), showed a higher incidence of cardiac dysfunction and CHF of New York Heart Association class III or IV (27% vs. 16%) when associated with anthracycline-based therapy, compared to anthracycline-based chemotherapy alone (8% vs. 3%); incidence of these AEs was lower in the trastuzumab plus paclitaxel arm (13% and 2%, respectively) (30). These data were re-dimensioned by a metanalysis that included this RCT and other six subsequent RCTs, counting a total of 1,497 HER2-positive women; a significant increased risk of CHF and decreased LVEF in patients receiving trastuzumab [risk ratio (RR) 3.49 and 2.65, respectively], with severe cardiac AE occurring in 4.7% of patients trastuzumab-treated patients (31) were reported. Another metanalysis comparing RCTs with cohort studies, found that cohort studies patients, who more closely reflected the real-life treated population, have a higher risk of severe cardiac AEs than RCTs ones (4.4% vs. 2.8%), although, overall, severe cardiotoxicity was observed in 4.28% of MBC patients. This study also confirmed that trastuzumab administrated with anthracycline-based regimens is associated with a higher proportion of severe cardiotoxicity than with taxane-based schedules alone (2.9% vs. 0.9%) (32).

In the adjuvant setting, a metanalysis of eight RCTs, involving a total of 11,991 women with HER2-positive EBC, showed a significant higher risk of CHF and LVEF decrease in patients treated with trastuzumab-containing regimens compared with control arm (RR =5.11, P<0.00001 and RR =1.83, P=0.0008, respectively). CHF and LVEF decrease occurred in 2.5% and 11.2%, respectively, in trastuzumab arm vs. 0.4% and 5.6% in control arm (33). In another metanalysis of 6 RCTs, the overall RR of NYHA III/IV CHF with trastuzumab was found to be 3.04-fold higher (P<0.00001) than in patients who did not receive trastuzumab (34). Long-term safety analysis of major RCTs found also that the cumulative incidence of cardiotoxicity and the overall risk of cardiac AEs was higher in trastuzumab-containing regimens (35-37). Results from a combined analysis of three clinical trials investigating safety and efficacy of trastuzumab plus anthracycline-based regimens in neoadjuvant setting did not differ from adjuvant and metastatic, confirming an increased risk of cardiotoxicity for combination therapy (38).

The risk of cardiac AEs seems to significantly increase with a longer exposure to trastuzumab (35) and with administration of higher doses (34). Age ≥60 years, basal LVEF between 50% and 54.9% and use of antihypertensive medications are also associated with a significant increased risk of cardiotoxicity, so this category of BC patients deserves a special attention (36). Cardiac AEs mostly occur during trastuzumab administration and many of them are reversible, with a complete or partial recovery observed in 86.1% of trastuzumab-treated patients with symptomatic heart failure events (35,39).

Pertuzumab (a humanised anti-HER2 mAb) administered together with trastuzumab does not increase the rate of cardiac dysfunction compared to trastuzumab plus standard chemotherapy in metastatic, adjuvant and neoadjuvant settings (40-44). Lapatinib, a dual tyrosine kinase inhibitor (TKI) of EGFR and HER2, was not associated with a greater risk of cardiac AEs in MBC, as showed in the pivotal phase III trial and confirmed by a metanalysis involving 3,689 patients treated with lapatinib from different clinical trials, in which the incidence of cardiac AE was 1.6% (45,46). Data from RCTs in EBC were also consistent (47,48). Trastuzumab-emtansine (T-DM1), an antibody drug conjugate composed of an anti-HER2 mAb connected to a cytotoxic antimicrotubule agent, showed a favourable cardiotoxicity profile (49,50), as well as trastuzumab-deruxtecan (antibody-drug conjugate composed of an anti-HER2 mAb, a cleavable tetrapeptide-based linker, and a cytotoxic topoisomerase I inhibitor) (51) and tucatinib, a highly selective TKI (52). There was also no evidence of cardiac toxicity with neratinib, a TKI targeting EGFR, HER2 and HER4 (53).

Baseline ECG and baseline LVEF measurement through echocardiogram and/or multigated acquisition scan or magnetic resonance imaging are strongly recommended immediately prior to initiation of HER2-targeted therapy to identify individuals at higher risk of future CV complications. Asymptomatic patients undergoing adjuvant trastuzumab treatment should repeat routine surveillance consisting of cardiac imaging every 3 months, for the early detection of cardiac toxicity during treatment, and every 6 months following discontinuation of treatment until 2 years from the last administration. Asymptomatic patients undergoing anti-HER2 treatment of MBC should also have general surveillance with cardiac imaging. If the anti-HER2-targeted therapy is withheld for symptomatic left ventricular cardiac dysfunction the LVEF measurement should be repeat after 4 weeks. Serial monitoring should be carried out preferably with the same imaging modality and at the same facility (54-58). Serum enzymes of cardiac damage have also been investigated as potential biomarkers of cardiotoxicity. Troponin I has been shown to predict LVEF reduction and cardiac AEs in trastuzumab-treated patients, especially those who have been exposed to anthracyclines. A higher risk for development of trastuzumab-induced cardiotoxicity was observed in patients with troponin I levels ≥0.08 ng/mL (HR =22.9, non-recovery HR =2.88) and with elevated high sensitivity troponin T levels>14 at the end of anthracycline therapy (59-64). The NeoALTTO sub-study BIG 1–06 showed that troponin T and proBNP were detected only in a few anthracycline-naïve patients receiving trastuzumab and/or lapatinib; so, they might not be effective early predictors of cardiotoxicity city in this patients’ setting (65). Since most research on troponin focuses on anthracycline-pretreated patients, further studies are needed to explore the role of this biomarker and its application in clinical practice. As for the potential influence of FC gamma receptor (FCGR) polymorphisms, most studies focused on anti-HER2 efficacy and provided contrasting findings on FCGR2A and FCGR3A role (66,67). Limited data are available on FCGR SNP and toxicity. In a study by Cresti et al. in 101 HER2 positive EBC patients receiving trastuzumab every 3 weeks after adjuvant chemotherapy, FCGR2A His131Arg SNP was significantly related to trastuzumab-related cardiotoxicity occurrence (68). Roca et al. found a significant association between cardiotoxicity after trastuzumab and HER2–I655V genotype (P=0.025), but not with FCGR2A-H131R and FCGR3A–V158F SNPs (69). Though of interest, these findings require more extensive research to be confirmed.

Pulmonary toxicity

Trastuzumab-deruxtecan was associated with interstitial lung disease (ILD) incidence of 13.6% and four deaths were attributed to treatment-related ILD (51). Patients should be monitored for signs and symptoms of ILD/pneumonitis and suspected ILD/pneumonitis should be evaluated by computed tomography (CT) scan. In case of asymptomatic ILD/pneumonitis (grade 1), the administration should be withheld until recovery to grade 0 and it may be resumed, while for symptomatic ILD/pneumonitis (grade ≥2) it is recommended to permanently discontinued trastuzumab-deruxtecan, promptly administer corticosteroids for at least 14 days or until complete resolution of clinical and chest CT findings (51,70,71). In the KATHERINE trial, pneumonitis occurred in 2.6% of patients in the T-DM1 group compared to 0.8% in the trastuzumab group (50). ILD incidence is higher when trastuzumab is combined with mTOR inhibitors: in the BOLERO-3 trial, ILD incidence was 9.2% among patients who received trastuzumab with vinorelbine and everolimus compared with 3.9% of those who received trastuzumab vinorelbine and placebo, although the proportion of patients with grade 3–4 ILD was similar in the two arms (72).

Gastrointestinal and skin toxicity

HER2-targeted TKIs have a higher incidence of gastrointestinal and skin toxicity. In the lapatinib pivotal RCT for MBC, diarrhoea and cutaneous rash were the most common treatment-related AEs (any grade and grade ≥3) in lapatinib plus capecitabine arm (45). In neoadjuvant and adjuvant settings, lapatinib-containing regimens were associated most frequently with grade 3 diarrhoea and cutaneous rash compared to trastuzumab-containing regimens (47,48). Tucatinib and neratinib showed a higher incidence of gastrointestinal and skin toxicity compared to trastuzumab-containing regimens in MBC (52,73), as well as neratinib in adjuvant setting compared to placebo (37). Trastuzumab plus pertuzumab combination seems to be associated with higher number of any grade and grade ≥3 diarrhoea compared to trastuzumab in MBC and EBC (42,43,74).

Hepatotoxicity

Hepatotoxicity, predominantly in the form of asymptomatic increases in the concentrations of serum transaminases or bilirubin, has been observed during treatment with tucatinib (52), neratinib (53) and lapatinib. Serious hepatobiliary disorders, such as nodular regenerative hyperplasia of the liver have been observed in patients treated with T-DM1 (49,50); in these cases, T-DM1 must be permanently discontinued (75).

Haematological toxicity

One of the most commonly reported grade 3–4 AEs with T-DM1 was thrombocytopenia (49-51). Routinely monitoring with haematological complete evaluation is recommended before each treatment cycle.


PIK3CA-AKT-mTOR pathway inhibitors

Everolimus

The phosphoinositide 3-kinase (PI3K) pathway plays a crucial role in the cells' growth, proliferation, migration, and death. PI3K mutations are frequently involved in BC development. Mammalian target of rapamycin (mTOR) is one of the PI3K-related kinase proteins. Everolimus is an oral mTOR inhibitor used in postmenopausal women with hormone receptor (HR) positive and HER2-negative BC, with recurrence during adjuvant non-steroidal aromatase inhibitor (AI) or disease progression in the pre-treated advanced setting (76).

Several studies evaluated the safety and feasibility of everolimus combined with other agents (72,77-86) (Table 2).

Table 2

Main clinical trials on everolimus

Study BOLERO-2 phase III BOLERO-1 phase III BOLERO-3 phase III GINECO phase II PrE0102 phase II NCT00915603 phase II MANTA phase II VIcTORia phase II EVEREXES phase III EVA retrospective Real world experience
Study details HR+ HER2− HR+/− HER2+ HR+/− HER2+ HR+ HER2− HR+ HER2− HR+ HER2− HR+ HER2− HR+ HER2− HR+ HER2− HR+ HER2− HR+ HER2−
Treatment EVE-EXE EVE-TRAST-PCT EVE-TRAST-VNR EVE-TMX EVE-FUL EVE-BEV-PCT EVE-FUL EVE-VNR EVE-EXE EVE-EXE EVE-EXE
Setting UNR-MBC UNR-MBC MBC UNR-MBC UNR-MBC UNR-MBC UNR-MBC UNR-MBC UNR-MBC UNR-MBC UNR-MBC
Line of therapy Post AI or 2nd line Post AI or 1st line 2nd or more Post AI or 1st line Post AI or 2nd line Post AI or 1st line Post AI or 2nd line Post AI or 2nd line Post AI or 2nd line 1st or more Post AI or 2nd line
n of patients 482 480 284 54 66 56 65 68 235 404 44
G3-G4 AEs
   Stomatitis (%) 8 13 13 11 11 15 12 7 10 11 11
   Anemia (%) 6 30 19 2 3 11 NA 6 7 4 3
   Fatigue (%) 4 35 13 6 6 15 NA 6 3 3 2
   Dyspnea (%) 4 24 2 NA 3 2 0 NA 1 NA NA
   Hyperglicemia (%) 4 12 2 NA 2 NA 3 NA 7 1 0
   Pneumonitis (%) 3 15 <2 2 6 NA NA NA 2 4 0
Treatment discontinuation (%) 19 50 10 17 NA NA NA 27 10 NA 18
Treatment related death (n) 7 17 2 NA 0 1 NA NA 1 0 NA

AEs, adverse events; AI, aromatase inhibitor; BEV, bevacizumab; EVE, everolimus; EXE, exemestane; FUL, fulvestrant; MBC, metastatic breast cancer; PCT, paclitaxel; TMX, tamoxifen; TRAST, trastuzumab; UNR, unresectable; VNR, vinorelbine; NA, not available.

In the BOLERO-2 trial, 485 patients were randomized to receive everolimus plus exemestane. Everolimus discontinuation due to AEs occurred in 19% of the cases. Overall, 23% of patients referred to severe AEs. The most frequent grade 3 and grade 4 AEs were: stomatitis (8%), anaemia (6%), dyspnoea (4%), hyperglycaemia (4%), fatigue (4%), and pneumonitis (3%); seven treatment related-deaths occurred (77).

Likewise, various trials and a real-life retrospective study enrolling HR+ HER2- MBC patients treated with everolimus plus exemestane observed similar toxicities (84-86).

In clinical practice, toxicities are generally successfully managed with treatment interruption until symptoms improve to grade ≤1 and/or its dose reduction (87).

Willemsen et al. assessed the association of peripheral blood immunological cell subsets with antitumour response and pulmonary toxicity in 20 BC patients receiving everolimus plus exemestane. BC patients developing pulmonary toxicity compared to other patients had relatively more NKT cells (CD3+ CD56+) at baseline (6.0% versus 1.3%, P=0.0068, 59 k ×109/L versus 12 k ×109/L, P=0.0081) and at the moment of toxicity occurrence (5.2% versus 1.2%, P=0.0106 and 47 k ×109/L versus 16 k ×109/L, P=0.0466). Baseline percentage NKT cells predicted pulmonary toxicity with 0.78 sensitivity and 1.0 specificity, even if further validation is required to confirm these data (88).

Pascual et al. performed an exploratory analysis to assess the role of SNPs on AEs occurrence and outcomes by a pharmacogenetic study on 90 postmenopausal HR positive, HER2 negative MBC patients receiving exemestane-everolimus progressing after a non-steroidal AI. They conducted a genotyping analysis in 12 SNPs implicated in everolimus pharmacokinetics and pharmacodynamics and investigated the association with everolimus plasma concentrations, significant AEs and consequent drug schedule modifications, progression free survival and overall survival. Patients harbouring CYP3A4 rs35599367 SNP (CYP3A4*22 allele) showed increased drug plasma levels compared to other patients (P=0.019). ABCB1 rs1045642 carriers were exposed to increased risk of mucosal inflammation (P=0.031), whereas PIK3R1 rs10515074 and RAPTOR rs9906827 patients had an increased risk of hyperglycaemia (P=0.016) and non-infectious lung inflammation (P=0.024). These results show that SNPs might influence everolimus outcomes in MBC (89).

Paying attention to patients with comorbidity as diabetes or a history of lung disease is crucial. Nonetheless, these patients could benefit from treatment as well as those without these comorbidities.

Alpelisib

Alpelisib is an oral selective inhibitor of PI3K alpha (90). Alpelisib has been investigated in the SOLAR-1 phase III trial in postmenopausal women or men with HR positive, HER2 negative advanced chemo-naïve BC, pre-treated with an AI. Among 572 patients enrolled in SOLAR-1, 284 (169 PIK3CA-mutant and 115 wild type) received alpelisib combined with fulvestrant. The most common AEs of any grade were: hyperglycaemia (63.7%; 36.6% G3–4), diarrhoea (57.7%; 6.7% G3–4), nausea (44.7%), decreased appetite (35.6%), and rash (35.6%; 9.9% G3–4) or maculopapular rash (14.1%; 8.8% G3–4). AEs lead to permanent discontinuation in 25% of the cases. No treatment related deaths occurred (91).

The randomized, double-blind phase III NEO-ORB trial enrolled postmenopausal women with HR-positive HER2 negative resectable BC, including patients eligible for neoadjuvant therapy, and evaluated alpelisib combined with letrozole. No prior local or systemic treatment was allowed. AEs observed in the alpelisib plus letrozole arm were: hyperglycaemia (any grade 54%; G ≥3 27%), diarrhoea (any grade 52%), rash (any grade 45%; G ≥3 12%), nausea (any grade 44%), fatigue (any grade 41%), stomatitis (any grade 33%), decreased appetite (any grade 31%), alopecia (any grade 22%), headache (any grade 20%), and maculo-papular rash (G ≥3 8%). No treatment-related deaths occurred (92).

Recently, Rodon and colleagues reported the results of a pooled analysis of X2101 and SOLAR-1, a risk-analysis of alpelisib-induced hyperglycaemia according to baseline features of 505 solid cancers (including BC) patients. Risk modelling identified 5 baseline factors, namely fasting plasma glucose, body mass index, HbA1c, monocyte counts, and age which were associated with a higher probability of G3/4 hyperglycaemia. High risk patients showed higher rates of alpelisib modifications and anti-hyperglycaemic agents. This model might be useful to identify among BC patients candidate to alpelisib those who are at higher risk for alpelisib-induced hyperglycaemia (93).

Although the toxicities of grade 3/4 observed lead to discontinuation of treatment, they regress with the temporary suspension of treatment in the majority of cases. Thus, both alpelisib and everolimus appear safe.


Poly(ADP-ribose) polymerase inhibitors (PARPi)

PARPi represent one of the main innovative approaches in target therapy in BRCA-mutant BC patients. Several PARPi have been studied, including olaparib, talazoparib, niraparib, veliparib and rucaparib. Their mechanism of action is not univocal, so their efficacy is closely related to different pathways. In particular, their interaction with the PARP enzyme family is crucial (94).

Haematological toxicity

In clinical practice, haematological toxicities are very common during PARPi administration and they usually present in the early phases of treatment (95).

Anaemia is the most frequent, followed by thrombocytopenia and neutropenia. In the three phase 3 maintenance trials, all-grade anaemia was reported in 44% of patients receiving olaparib, 50% of patients receiving niraparib and in 37% of patients receiving rucaparib. Grade 3 and 4 AEs were more frequent with niraparib (25%), followed by rucaparib (19%) and olaparib (19%). Thrombocytopenia of any grade is more common with niraparib (61% vs. 28% with rucaparib and 14% with olaparib). All-grade neutropenia occurred in 18–30% of subjects, with grade 3–4 AEs higher with niraparib (20%) (96-98).

For all patients starting a PARPi or those requiring dose changes, a complete blood count once a month is recommended to assess haematological AEs. The FDA niraparib label recommends testing once per week in the first month to check haematological toxicity and especially platelet concentrations (99-101). Actually, no validated predictive biomarkers for this AEs are available. A retrospective analysis of the ENGOT-OV16/NOVA trial of maintenance niraparib in patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in complete or partial response to platinum-based chemotherapy was performed to assess clinical parameters predicting dose reductions. Baseline platelet count <150,000/µL and baseline body weight <77 kg were identified as risk factors for increased incidence of grade 3 thrombocytopenia and dose reduction to 200 or 100 mg. PFS was not influenced by dose changes in these patients, suggesting that they may benefit from a starting dose of 200 mg/day. Although retrospective, these data might be a starting point for further research on this topic in BC patients (102).

Gastrointestinal toxicity

Gastrointestinal AEs are quite common, the most frequent being nausea, followed by constipation, vomiting, and diarrhoea (99-101). Their management is similar to that of chemotherapy-induced gastrointestinal toxicities, using prokinetics and antiemetic drugs like metoclopramide, dexamethasone (103,104). Aprepitant, neurokinin-1 receptor antagonist, should not be administered with olaparib as it is a potent CYP3A4 inhibitor and may influence olaparib plasma concentrations (105).

Renal toxicity

The administration of rucaparib in ARIEL3 led to an increase of creatinine (any grade) levels in 15% of patients vs. 2% in the placebo group in the first weeks of treatment. Rucaparib inhibits the renal transporter proteins MATE1 and MATE2-K, that are involved in creatinine secretion (96). In study SOL0218, 21/195 olaparib-treated patients had a grade 1–2 increase in creatinine (no grade 3–4) vs. 1% in the placebo arm while niraparib did not induce an increase in serum creatinine (98). This alteration may not reflect a true decline in glomerular filtration rate (GFR). If GFR is appropriate (i.e., GFR is typical or inconsistent with elevated creatinine), dose reductions or interruptions are not strictly necessary (95).

Fatigue

A proportion of 59–69% of patients assuming PARPi had fatigue of any grade (96-98). Experts recommend non-pharmacological approach, namely exercise, massage therapy, and cognitive behavioural therapy (95).

Clinical laboratory abnormalities

The most common laboratory abnormalities are hypercholesterolemia and increased serum levels of alanine aminotransferase and aspartate aminotransferase. These effects are generally transient (98). Particular caution should be taken in patients with pre-existing liver dysfunctions and of lipid profile. In the case of persistent hypercholesterolemia, a statin-based treatment is indicated (95).

Other toxicities

Less frequent AEs include neurological symptoms which may comprise headaches and insomnia. The underlying mechanism is not yet fully understood but some preclinical studies have identified a role of PARP1 in maintaining the transcription of circadian genes, with PARP1 inhibition leading to a disconnect in key circadian rhythm transcriptional components (106).

For mild symptoms symptomatic therapy may be sufficient, while for more severe symptoms a dose reduction may be required, on the basis of the FDA label of each PARPi (99-101).

Reported respiratory symptoms include dyspnoea, cough, nasopharyngitis and more rarely pneumonia (96-98). The mechanism causing these symptoms has not been understood. Preclinical data only showed that PARP activation is related with bronchial hyper-reactivity and airway remodelling (107). The management of suspected or confirmed pneumonitis should be performed according accepted guidelines for drug-induced pneumonitis (108).

Other rarer side effects include musculoskeletal toxicities (arthralgia, back pain), skin toxicities (photosensitivity reactions, pruritis, rash, peripheral edema) and cardiovascular toxicities (hypertension, tachycardia, palpitations) (96-98). For the last mentioned, patients on niraparib should undergo blood pressure and heart rate monitoring once a month for the first year and regularly afterwards, especially in case of cardiovascular comorbidities (101).

Secondary malignancies

Since the primary mechanism of PARP inhibition involves interference with DNA repair pathways, myelodysplastic syndrome and acute myeloid leukaemia, are serious AEs requiring treatment discontinuation. Incidence is rare (0.5–1.4%) and after long-term treatment. In all clinical trials, all patients developing these AEs had been previously treated with platinum-based chemotherapy or other DNA-damaging drugs, making it difficult to define PARPi as responsible (96-98).


Immunotherapy

The clinical activity of programmed cell death-1/programmed death ligand-1 (PD-1/PD-L1) antagonists was demonstrated in the treatment of triple negative BC (TNBC) (109).

In the IMpassion 130 trial, the association of the anti-PD-L1 atezolizumab and nab-paclitaxel, showed an acceptable safety profile and it was approved as first-line treatment for patients with unresectable locally advanced or metastatic TNBC whose tumours have a PD-L1 >1% expression. In the atezolizumab group, 49% of patients had grade 3–4 AEs. Peripheral neuropathy occurred in 6% of patients in the atezolizumab arm and it was the leading cause for treatment discontinuation due to toxicity (4%), but it was also deemed to be taxane-related, which is known to be cumulative. The AEs of special interest that differed substantially between atezolizumab group and placebo group were any-grade rash, hypothyroidism, hyperthyroidism, pneumonitis, and adrenal insufficiency. Treatment-related deaths occurred in <1% patients in the atezolizumab group (one due to autoimmune hepatitis and one due to septic shock related to nab-paclitaxel only) and <1% patient in the placebo group (hepatic failure) (110,111).

In the neoadjuvant setting, atezolizumab showed a safety profile consistent with MBC. In the IMpassion031 trial, hypothyroidism occurred in 7% of patients in the atezolizumab arm versus 1% control arm. The number of patients who discontinued atezolizumab or placebo due to AEs was 13% versus 11% (112).

Data from the KEYNOTE-522 trials evaluating pembrolizumab plus chemotherapy, reported an incidence of immune-related AEs in 38.9% of patients and included hypothyroidism (any grade: 14.9%; grade >3: 0.4%), hyperthyroidism (any grade: 5.1%; grade >3: 0.3%), severe skin reaction (any grade: 4.4%; grade >3: 3.8%) and adrenal insufficiency (any grade: 2.3%; grade >3: 1.3%). Even if manageable, immune checkpoint inhibitors-related AEs might lead to persistent alterations, including thyroid disorders and adrenal failure, for which hormone replacement treatment might become necessary for undefined time (113,114). In GeparNuevo trial, the addition of durvalumab to standard neoadjuvant chemotherapy did not lead to more frequent incidence of AEs, with the exception of thyroid dysfunction (any grade), which was more frequently reported on durvalumab (47%). Seven patients had hypothyroidism and 9 patients hyperthyroidism; one patient had a hypophysitis (115).

In The TONIC trial, after a 2-week induction with chemotherapy or irradiation in metastatic TNBC patients, nivolumab was not associated with any previously unreported toxicity. Induction treatment-related AEs of any grade occurred in 28% of patients (3% grade 3) and immune-related AEs of grade 3–5 occurred in 19% of patients (116).

Moat available data on potential predictive factors for toxicities that might help the selection of patients derive from studies in melanoma and non-small-cell lung cancer. In a retrospective analysis by Krishnan et al., patients who developed eosinophilia during treatment were more likely to have toxicity (P=0.042), thus suggesting further prospective investigation (117). Increased white blood cells count and decreased relative lymphocyte count have been reported to be independently associated with lung/gastrointestinal toxicities from nivolumab (118). Baseline anti-thyroglobulin antibodies and anti-thyroid peroxidase antibodies levels and their early increase during treatment with anti-PD-1 were associated with the development of thyroiditis and thyroid dysfunction (119-121). Additionally, cutaneous toxicity was observed more frequently among patients with pre-existing rheumatoid factor (120,122). Further larger studies with prospective design are needed to confirm these findings and to assess their potential application in clinical practice.


Endocrine treatment

Tamoxifen acts as Selective Estrogen Receptor Modulator (123). In breast tissue, it exerts an anti-estrogenic effect by competitive binding to estrogen receptors. In other tissues, tamoxifen has an estrogen agonistic effect, e.g., by stimulating endometrium proliferation with subsequent higher risk of endometrial malignancy. Other reported side effects are: dizziness, headache, depression, confusion, fatigue and muscle cramps (124). The increased thromboembolic risk might be related to the tamoxifen-induced altered circulating coagulation inhibitors, namely reduced antithrombin III, protein C and protein S levels (125,126). Scientific evidence shows that long-term use of tamoxifen is related to secondary endometrial cancer in women. Based on the available results, the risk of endometrial cancer increases from 2 to 4 times with longer therapy duration with tamoxifen (127). In the ATLAS study, which evaluated continuation of adjuvant tamoxifen therapy for a total of 10 years, the cumulative risk of endometrial cancer during years 5–14 was 3.1% (mortality 0.4%) in patients who continued treatment and 1.6% (0.2% mortality) for patients who stopped treatment at 5 years (128).

The CYP2D6 enzyme is essential to convert tamoxifen into endoxifen, the main active metabolite. CYP2D6 gene alterations might be responsible for abnormal enzyme activity, thus configuring the profile of ultrarapid metabolizer (increased activity), intermediate metabolizer (decreased activity), poor metabolizer (absent activity). These two last conditions may result in reduced endoxifen blood levelsand consequently in decreased tamoxifen efficacy (129).

Third-generation AI—anastrozole, letrozole and exemestane are an effective endocrine treatment for HR positive EBC and MBC patients (130,131).

Estrogens exert their physiologic action on several tissues including bone, immune system, central nervous system and cardiovascular system (132). They may induce a protective cardiovascular effect, as suggested by lower incidence of coronary heart disease by older age at the first cardiovascular event compared with men (133,134). The protective action of tamoxifen on cardiovascular system is related to estrogen-like activity (agonist on alfa receptor) leading to decreased low-density lipoprotein (LDL) cholesterol and homocysteine serum levels. In fact, a meta-analysis of 12 studies comparing tamoxifen with placebo, revealed a lower incidence of heart attack (HR =0.62, 95% CI: 0.41–0.93) with tamoxifen (135). In a combined analysis of two trials evaluating up-front AI versus up-front tamoxifen, AI were significantly associated with cardiovascular disease (OR =1.30, 95% CI: 1.06–1.61, P=0.01) (135-137). Consistent findings were reported in the study comparing switching from tamoxifen to AI versus up-front AI (OR =1.37, 95% CI: 1.05–1.79, P=0.02) (138). The biologic rationale for a potential negative effect of AI on cardiovascular system is mainly related to their action on lipid metabolism. Contrary to tamoxifen, AI raise the serum cholesterol levels and this may lead to higher cardiovascular risk, especially in case of pre-existing arterial hypertension, diabetes and obesity (139).

Extended adjuvant endocrine therapy with either tamoxifen or AI after 5 years of initial tamoxifen treatment has been shown to improve the disease-free survival in EBC (108,140-142). The EBCTCG meta-analysis has shown that administration of AI in the first 5 years of adjuvant therapy was superior to tamoxifen monotherapy (143). In a literature-based meta-analysis published in 2019, comprising eight trials, longer treatment with AI was related to higher RR of bone pain (RR =1.26, RD =0.04, P=0.003), bone fractures (RR =1.59, RD =0.02, P=0.002), osteoporosis (RR =1.53, RD =0.07, P=0.005), myalgia (RR =1.26, RD =0.04, P=0.02), and therapy discontinuation for AEs (RR =1.51, RD =0.06, P=0.0009) (144).

AI administration might be associated with hot flushes and musculoskeletal AEs affecting quality of life. rs10046 variant T/T of CYP19A1 seemed to be associated to lower occurrence of hot flashes/sweating with exemestane and ovarian function suppression in premenopausal patients enrolled in the TEXT trial, thus improving patients’ compliance to AI treatment (145). Borrie et al. found that BC patients with higher body mass index (P=0.001) and those receiving letrozole vs. anastrozole (P=0.018) were more likely to develop arthralgia and subsequently discontinue AI. Moreover, the Authors found that CYP19A1 rs4775936 and ESR1 rs9322336, rs2234693, rs9340799 SNPs were associated with occurrence of arthralgia (P=0.016, 0.018, 0.017, 0.047) and that CYP19A1 rs4775936 SNP was related to AI discontinuation for intolerable arthralgia (146). rs2073618 SNP in osteoprotegerin gene was found to be related with higher risk of muscoloskeletal symptoms and pain in 254 AI-treated (147). In a nested case-control correlative study by Niravath et al. in BC patients enrolled in the MA.27 trial, VDR Fok-I variant genotype was associated to lower incidence of arthralgia after 6 months of AI vs. wild type VDR (P<0.0001) (148).

Fulvestrant is the first pure anti-estrogen approved to treat MBC postmenopausal patients. Fulvestrant acts as both a competitive antagonist and a Selective Estrogen Receptor Degrader (149). The acute toxicity of fulvestrant is low. Some reported AEs include injection site reactions, nausea, pain, headaches, asthenia, and increased liver enzymes. A review analysed data from the main available studies to assess the efficacy and safety of fulvestrant for postmenopausal hormone-sensitive locally advanced or MBC patients versus other standard endocrine agents. There was no significant difference in vasomotor toxicity (RR =1.02, 95% CI: 0.89–1.18, 3,544 women, 8 studies), arthralgia (RR =0.96, 95% CI: 0.86–1.09, 3,244 women, 7 studies), and gynaecological toxicities (RR =1.22, 95% CI: 0.94–1.57, 2,848 women, 6 studies) (150).


CDK4/6 inhibitors

Currently, three cyclin-dependent kinase (CDK) 4 and 6 inhibitors (CDK4/6) inhibitors are approved for HR positive MBC patients in combination with AI and fulvestrant: palbociclib, ribociclib and abemaciclib (128,151).

The enzymes primarily involved in the metabolism of CDK4/6 inhibitors, which are in turn time-dependent CYP3A-inhibitors, are represented by CYP3A and SULT2A1 (152-154). Administration with strong CYP3A inhibitors (e.g., itraconazole) and with strong (e.g., phenytoin, clarithromycin) or moderate (e.g., modafinil, diltiazem) CYP3A inducers (152-154) is strongly discouraged. Thus, it is crucial to investigate on eventual concomitant medications in BC patients who are candidate to these agents, especially elderly patients with multiple comorbidities and polipharmacy. The safety profile is similar for all CDK4/6 inhibitors, except for some aspects (155-158). In general, the most frequent AE (all grades) in the group treated with the combination of CDK4/6 inhibitor plus endocrine therapy was neutropenia (65%) followed by diarrhoea (49%), infections (44%), nausea (40%), fatigue (39%), and leukopenia (35%) (159). Other safety issues reported in clinical trials include hepatobiliary toxicity (ribociclib, abemaciclib), prolongation of the QT interval on ECG (ribociclib), and venous thromboembolism (160,161). To date, no prospective factors predicting toxicity have been validated and can be used in clinical practice to identify which patients are more likely to develop AEs. However, some available data discussed afterwards might deserve further investigation.

Haematological toxicity

Neutropenia and leukopenia represent the most common grade 3/4 CDK4/6-related AEs. Anaemia or thrombocytopenia are less frequent (155-158,162,163). The rate of all-grade neutropenia with abemaciclib is 50% lower than palbociclib and ribociclib due to the greater CDK4 selectivity (164). CDK4/6 inhibitors cause cell-cycle stop by reducing hematopoietic stem cells division, which is regained after reducing or interrupting the dose; for this reason, neutropenia has a quick recover, as opposed to the same AE induced by chemotherapy (165). In the PALOMA-3 trial with palbociclib and fulvestrant, grade 3/4 neutropenia generally recovered in a week timeframe (166). granulocyte-colony stimulating factor is not required and febrile neutropenia reported in CDK4/6 inhibitors studies is significantly lower than chemotherapy (156-158,162,164,167,168). Timing for neutropenia occurrence is usually 15 days after the first dose for palbociclib and ribociclib and within the first two cycles (155,164,166,169) with abemaciclib. A complete blood count is recommended prior to treatment start, at the beginning of each further cycle and on day 14 of cycle 1 and 2 (170). In palbociclib-treated patients from PALOMA-2 (n=584) and PALOMA-3 (n=442), low baseline absolute neutrophil count was a strong independent risk factor for C1D15 grade 3/4 neutropenia. ABCB1_rs1128503 (C/C vs. T/T: OR =0.57, 95% CI: 0.311−1.047, P=0.070) and ERCC1_rs11615 (A/A vs. G/G: OR =1.75, 95% CI: 0.901−3.397, P=0.098) SNPs were identified as potential independent risk factors for C1D15 grade 3/4 neutropenia in non-Asian patients; therefore, pharmacogenetic testing might be informative on potentially increased risk of developing severe neutropenia (171). A study by Modi et al. pooling the data from MONARCH-1, 2 and 3 demonstrated the ability of a clinical prediction tool including ethnicity, Eastern Cooperative Oncology Group Performance Status and pre-treatment white blood cell count, in identifying subgroups with significantly different risks of grade ≥3 neutropenia after abemaciclib initiation. This tool might be useful to assess personalised risks and the risk-benefit ratio of abemaciclib (172).

Gastrointestinal toxicity

Abemaciclib has a higher rate of grade 3 diarrhoea compared to palbociclib and ribociclib. In the MONARCH-1 trial, 90% of the patients receiving abemaciclib monotherapy had diarrhoea, (generally within 1 week of treatment initiation), that required to dose reductions in 21% of the patients. The vast majority of episodes had a short duration (median: 7.5 days for grade 2 and 4.5 days for grade 3) (173). In the MONARCH-2 grade 1 and 2 diarrhoea was reported in 73% and grade 3 in 13.4% and, occurred, consistently with MONARCH-1, in the first treatment cycle, with a median duration of 6 days, without requiring treatment modifications in 70.1% of the patients (168). Advanced age has been identified as significantly correlated to an increased risk of grade ≥3 diarrhoea [HR for age >70: 1.72 (95% CI: 1.14–2.58); P=0.009] (172). Particular caution is required also for patients with inflammatory bowel disease (e.g., ulcerative colitis and Chron).

QTc prolongation

Treatment with ribociclib is strongly discouraged in patients at risk of developing QTc prolongation, since this drug may induce prolonged QT interval according to its concentration. In the MONALEESA-2 trial, 3.3% of patients treated with ribociclib plus letrozole experienced QTc prolongation to >480 ms, mostly in the first cycle and limited by proactive dose interruption or reduction (162). Caution should be taken when prescribing symptomatic therapies because of potential drug interactions. In clinical practice, it is recommended to check patients eligible for ribociclib on the basis of their cardiac status and their potentially QTc-prolonging concomitant medication. Electrocardiograms at baseline, day 14 in cycle 1 and day 1 in cycle 2, and careful monitoring should be performed to limit the incidence of this AE (174). Particular caution should be kept when ribociclib is administered with antiemetics (e.g., intravenous ondansetron, dolasetron, metoclopramide, diphenhydramine, haloperidol) because of the risk of QT interval prolongation (175,176).


Conclusions

Several advances have been introduced in the recent years for the management of BC in the perspective of personalised treatment, on the basis of tumour biology, genetics and patients’ clinical features (2). Despite intensive research, no validated prospective factors able to identify the best treatment for each category of BC patients to guide the therapeutic choice are available yet.

In the era of precision medicine and tailored therapy, genomic testing and the identification of potential biomarkers are a growing field of research in BC patients. In that regard, the exploration of drugs’ toxicity profile is increasingly appealing, due to the potential application in everyday clinical practice (5,6). Indeed, a safety profile-tailored treatment combined with the clinical characteristics of BC patients might be particularly helpful in the therapeutic choice (Table 3). Therefore, the awareness of the patients’ comorbidities and potential biomarkers predisposing to specific treatment-related toxicities could be crucial for better choice and management of the best care for the right patient. Currently, the upcoming application of these findings in clinical practice is urgently needed to reach this objective, but further research is needed. We believe that these factors, if confirmed in the near future and in further studies, might be helpful in the individualisation of treatment; on one hand, they would allow a better selection of patients; on the other hand, they will permit to tailor the patients’ monitoring for toxicities in the perspective of an individualised I and improvement of clinical outcome.

Table 3

Main drug-related adverse events and application in patients’ selection

Treatment Drug Main AEs Risk factors Management References
Anthracyclines Doxorubicin and epirubicin Cardiotoxicity; myelosuppression; nausea and vomiting Longer treatment duration, FGFR2 rs2420946 CC genotype, FGFR2 rs2981578 AG genotype, HFE H63D SNP, NFKBIL1, TNF-a, ATP6V1G2-DDX39B, MSH5, MICA, LTA, BAT1, and NOTCH4 SNPs, rs28714259 SNP, heterozygous status for ABCC1 rs246221 T-allele, UGT2B7-161 T allele for cardiotoxicity Baseline and routine monitoring with ECG and Echocardiogram and/or MUGA for LVEF evaluation; baseline and routine monitoring of complete blood count; prophylaxis and monitoring of nausea and vomiting (7-15)
Fluoropyrimidines Capecitabine and 5-fluorouracil Diarrhoea; HFS; myelosuppression; mucositis DYPD variants c.1905+1G>A, c.2846A>T, c.1679T>G, and c.1236G>A DYPD testing before treatment start; identification of variants associated with severe toxicity: dose adaptation according to guidelines (10-27)
Anti-HER2 Trastuzumab Cardiotoxicity Longer exposure; high doses; age ≥60 years; baseline LVEF 50–54.9%; anti-hypertensive drugs Baseline ECG and Echocardiogram and/or MUGA or MRI for LVEF evaluation immediately prior to initiation; asymptomatic patients: routine surveillance with cardiac imaging every 3 months during treatment and every 6 months following discontinuation of treatment until 2 years from the last administration Overview (30,31,33,42,43,45,47-55), cardiotoxicity specific references (29,32,34-41,44,46,56-58)
Neratinib, lapatinib and tucatinib Diarrhoea; cutaneous rash Monitoring for gastrointestinal symptoms (45,47,48,53)
T-DM1 Thrombocytopenia Baseline and routine monitoring of complete blood count (49-51,74)
Trastuzumab deruxtecan ILD-pneumonitis Chest CT scan at baseline and strict monitoring for signs and symptoms of ILD during treatment (51,70,71)
PIK3CA-AKT-mTOR inhibitors Everolimus Stomatitis; anaemia; dyspnoea; hyperglycaemia; fatigue; pneumonitis High number of NKT cells: pulmonary toxicity; ABCB1 rs1045642: risk of mucositis; PIK3R1 rs10515074 and RAPTOR rs9906827: hyperglycaemia and non-infectious pneumonitis Caution in case of comorbidities as diabetes or history of lung disease; monitoring for signs and symptoms of toxicity (72,77,89)
Alpelisib Hyperglycaemia; diarrhoea; nausea; decreased appetite; maculopapular rash Fasting plasma glucose, body mass index, HbA1c, monocyte counts and age: hyperglycaemia Caution in patients with diabetes mellitus; monitoring for signs and symptoms of toxicity (91-93)
PARP-inhibitors Olaparib, niraparib, rucaparib, talazoparib, veliparib Anaemia; thrombocytopenia; neutropenia; nausea; constipation; vomiting; diarrhoea; fatigue; hypercholesterolemia; AST/ALT increased serum levels Baseline platelet count <150,000/mL and baseline body weight <77 kg: grade 3 thrombocytopenia with niraparib Complete blood count to monitor haematological toxicity; caution in patients with pre-existing liver disfunction and dyslipidemia (94-101)
Immunotherapy Atezolizumab nivolumab, pembrolizumab Rash; hypothyroidism; hyperthyroidism; pneumonitis; adrenal insufficiency; diarrhoea; skin toxicity Eosinophilia development during treatment; increased WBC count and decreased relative lymphocyte count: lung/gastrointestinal toxicities with nivolumab; baseline and early increase of anti-Tg Ab and anti-TPO Ab: thyroiditis and thyroid dysfunction with anti-PD-1; pre-existing rheumatoid factor: cutaneous toxicity Clinical monitoring for toxicities; laboratory assessments including thyroid function; caution in patients with pre-existing autoimmune disorders (109-122)
Endocrine therapy Tamoxifen Dizziness; headache; depression; confusion; fatigue; muscle cramps; thromboembolic events; endometrial cancer Longer duration for endometrial cancer Regular gynecological visits and monitoring for signs and symptoms of toxicity (124-128)
AI Bone pain; bone fracture; osteoporosis; myalgia; hypercholesterolemia; cardiovascular disease Longer treatment duration; higher body mass index; letrozole vs. anastrozole; CYP19A1 rs4775936 and ESR1 rs9322336, rs2234693, rs9340799 SNPs, osteoprotegerin gene SNP Caution in patients with severe osteoporosis; vitamin D and calcium supplementation; caution in patients with arterial hypertension, diabetes, obesity, dyslipidemia (128,130,131,135-142,146,147)
CDK4/6 inhibitors Palbociclib Neutropenia; diarrhoea; infections; nausea; fatigue Low baseline absolute neutrophil count: cycle 1 day 15 grade 3/4 neutropenia; ABCB1_rs1128503 and ERCC1_rs11615 SNPs: cycle 1 day 15 grade 3/4 neutropenia in non-Asian patients Complete blood count prior to the start of treatment, at the beginning of each new treatment cycle and on day 14 of cycle 1 and 2 (156-158,165-171)
Ribociclib Neutropenia; Tc prolongation; diarrhoea; infections; nausea; fatigue; hepatobiliary toxicity Concomitant administration of intravenous ondansetron, dolasetron, metoclopramide, diphenhydramine, haloperidol and other known QTc potentially prolonging drugs: QTc prolongation Complete blood count prior to the start of treatment, at the beginning of each new treatment cycle and on day 14 of cycle 1 and 2; ECG at baseline, day 14 in cycle 1 and day 1 in cycle 2 and careful monitoring; caution in patients with polipharmacy for potential interactions with QTc potentially prolonging agents (159,162,174-176)
Abemaciclib Diarrhoea; neutropenia; infections; nausea; fatigue; hepatobiliary toxicity Advanced age, inflammatory bowel disease: grade ≥3 diarrhoea; ethnicity, ECOG PS and pre-treatment WBC count Particular caution in patients with inflammatory bowel disease (e.g., ulcerative colitis and Chron); complete blood count prior to the start of treatment, at the beginning of each new treatment cycle and on day 14 of cycle 1 and 2 (159,168,172,173)

AEs, adverse events; ALT, alanine aminotransferase; anti-Tg Ab, anti-thyroglobulin antibodies; anti-TPO Ab, anti-thyroid peroxidase antibodies, AST, aspartate aminotransferase; CT, computed tomography; DYPD, dihydropyrimidine dehydrogenase; ECG, electrocardiogram; ECOG PS, Eastern Cooperative Oncology Group Performance Status; HER2, human epidermal growth factor receptor 2; HFS, hand-foot syndrome; ILD, interstitial lung disease; LVEF, left ventricular ejection fraction, MRI, magnetic resonance imaging; MUGA, multigated acquisition; PD-1, programmed cell death-1; SNPs, single nucleotide polymorphisms; WBC, white blood cells.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Luca Moscetti) for the series “New Insights in Precision Oncology in Breast Cancer” published in Precision Cancer Medicine. The article has undergone external peer review.

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://pcm.amegroups.com/article/view/10.21037/pcm-21-38/rc

Peer Review File: Available at https://pcm.amegroups.com/article/view/10.21037/pcm-21-38/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://pcm.amegroups.com/article/view/10.21037/pcm-21-38/coif). The series “New Insights in Precision Oncology in Breast Cancer” was commissioned by the editorial office without any funding or sponsorship. MS reports that he has the following disclosures: consultant, advisory board and speakers’ bureau fees from Amgen, Sanofi, MSD, Eisai, Merck, Bayer. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Ferlay J, Colombet M, Soerjomataram I, et al. Cancer statistics for the year 2020: An overview. Int J Cancer 2021; [Epub ahead of print]. [Crossref] [PubMed]
  2. Caswell-Jin JL, Plevritis SK, Tian L, et al. Change in Survival in Metastatic Breast Cancer with Treatment Advances: Meta-Analysis and Systematic Review. JNCI Cancer Spectr 2018;2:pky062. [Crossref] [PubMed]
  3. Chae YK, Pan AP, Davis AA, et al. Path toward Precision Oncology: Review of Targeted Therapy Studies and Tools to Aid in Defining “Actionability” of a Molecular Lesion and Patient Management Support. Mol Cancer Ther 2017;16:2645-55. [Crossref] [PubMed]
  4. Brock A, Huang S. Precision Oncology: Between Vaguely Right and Precisely Wrong. Cancer Res 2017;77:6473-9. [Crossref] [PubMed]
  5. Brown NA, Elenitoba-Johnson KSJ. Enabling Precision Oncology Through Precision Diagnostics. Annu Rev Pathol 2020;15:97-121. [Crossref] [PubMed]
  6. Fernández XM. Untangling Data in Precision Oncology - A Model for Chronic Diseases? Yearb Med Inform 2020;29:184-7. [Crossref] [PubMed]
  7. Vaitiekus D, Muckiene G, Vaitiekiene A, et al. HFE Gene Variants’ Impact on Anthracycline-Based Chemotherapy-Induced Subclinical Cardiotoxicity. Cardiovasc Toxicol 2021;21:59-66. [Crossref] [PubMed]
  8. Chen L, Qi H, Zhang L, et al. Effects of FGFR gene polymorphisms on response and toxicity of cyclophosphamide-epirubicin-docetaxel based chemotherapy in breast cancer patients. BMC Cancer 2018;18:1038. [Crossref] [PubMed]
  9. Todorova VK, Makhoul I, Dhakal I, et al. Polymorphic Variations Associated With Doxorubicin-Induced Cardiotoxicity in Breast Cancer Patients. Oncol Res 2017;25:1223-9. [Crossref] [PubMed]
  10. Schneider BP, Shen F, Gardner L, et al. Genome-Wide Association Study for Anthracycline-Induced Congestive Heart Failure. Clin Cancer Res 2017;23:43-51. [Crossref] [PubMed]
  11. Vulsteke C, Pfeil AM, Maggen C, et al. Clinical and genetic risk factors for epirubicin-induced cardiac toxicity in early breast cancer patients. Breast Cancer Res Treat 2015;152:67-76. [Crossref] [PubMed]
  12. Li H, Hu B, Guo Z, et al. Correlation of UGT2B7 Polymorphism with Cardiotoxicity in Breast Cancer Patients Undergoing Epirubicin/Cyclophosphamide-Docetaxel Adjuvant Chemotherapy. Yonsei Med J 2019;60:30-7. [Crossref] [PubMed]
  13. Cui L, Huang J, Zhan Y, et al. Association between the genetic polymorphisms of the pharmacokinetics of anthracycline drug and myelosuppression in a patient with breast cancer with anthracycline-based chemotherapy. Life Sci 2021;276:119392. [Crossref] [PubMed]
  14. Fasching PA, Kollmannsberger B, Strissel PL, et al. Polymorphisms in the novel serotonin receptor subunit gene HTR3C show different risks for acute chemotherapy-induced vomiting after anthracycline chemotherapy. J Cancer Res Clin Oncol 2008;134:1079-86. [Crossref] [PubMed]
  15. Tsuji D, Matsumoto M, Kawasaki Y, et al. Analysis of pharmacogenomic factors for chemotherapy-induced nausea and vomiting in patients with breast cancer receiving doxorubicin and cyclophosphamide chemotherapy. Cancer Chemother Pharmacol 2021;87:73-83. [Crossref] [PubMed]
  16. Froehlich TK, Amstutz U, Aebi S, et al. Clinical importance of risk variants in the dihydropyrimidine dehydrogenase gene for the prediction of early-onset fluoropyrimidine toxicity. Int J Cancer 2015;136:730-9. [PubMed]
  17. Diasio RB, Harris BE. Clinical pharmacology of 5-fluorouracil. Clin Pharmacokinet 1989;16:215-37. [Crossref] [PubMed]
  18. Johnson MR, Diasio RB. Importance of dihydropyrimidine dehydrogenase (DPD) deficiency in patients exhibiting toxicity following treatment with 5-fluorouracil. Adv Enzyme Regul 2001;41:151-7. [Crossref] [PubMed]
  19. Meulendijks D, Henricks LM, Sonke GS, et al. Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity: a systematic review and meta-analysis of individual patient data. Lancet Oncol 2015;16:1639-50. [Crossref] [PubMed]
  20. Henricks LM, Lunenburg CATC, de Man FM, et al. DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol 2018;19:1459-67. [Crossref] [PubMed]
  21. Stavraka C, Pouptsis A, Okonta L, et al. Clinical implementation of pre-treatment DPYD genotyping in capecitabine-treated metastatic breast cancer patients. Breast Cancer Res Treat 2019;175:511-7. [Crossref] [PubMed]
  22. Deenen MJ, Meulendijks D, Cats A, et al. Upfront Genotyping of DPYD*2A to Individualize Fluoropyrimidine Therapy: A Safety and Cost Analysis. J Clin Oncol 2016;34:227-34. [Crossref] [PubMed]
  23. Henricks LM, Opdam FL, Beijnen JH, et al. DPYD genotype-guided dose individualization to improve patient safety of fluoropyrimidine therapy: call for a drug label update. Ann Oncol 2017;28:2915-22. [Crossref] [PubMed]
  24. Lunenburg CATC, Henricks LM, Guchelaar HJ, et al. Prospective DPYD genotyping to reduce the risk of fluoropyrimidine-induced severe toxicity: Ready for prime time. Eur J Cancer 2016;54:40-8. [Crossref] [PubMed]
  25. Henricks LM, Lunenburg CATC, de Man FM, et al. A cost analysis of upfront DPYD genotype-guided dose individualisation in fluoropyrimidine-based anticancer therapy. Eur J Cancer 2019;107:60-7. [Crossref] [PubMed]
  26. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med 2017;376:2147-59. [Crossref] [PubMed]
  27. EMA recommendations on DPD testing prior to treatment with fluorouracil, capecitabine, tegafur and flucytosine. Available online: https://www.ema.europa.eu/en/documents/press-release/ema-recommendations-dpd-testing-prior-treatment-fluorouracil-capecitabine-tegafur-flucytosine_en.pdf (accessed Apr 30, 2020).
  28. Loibl S, Gianni L. HER2-positive breast cancer. Lancet 2017;389:2415-29. [Crossref] [PubMed]
  29. Pondé NF, Lambertini M, de Azambuja E. Twenty years of anti-HER2 therapy-associated cardiotoxicity. ESMO Open 2016;1:e000073. [Crossref] [PubMed]
  30. 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;344:783-92. [Crossref] [PubMed]
  31. Balduzzi S, Mantarro S, Guarneri V, et al. Trastuzumab-containing regimens for metastatic breast cancer. Cochrane Database Syst Rev 2014;CD006242. [PubMed]
  32. Mantarro S, Rossi M, Bonifazi M, et al. Risk of severe cardiotoxicity following treatment with trastuzumab: a meta-analysis of randomized and cohort studies of 29,000 women with breast cancer. Intern Emerg Med 2016;11:123-40. [Crossref] [PubMed]
  33. Moja L, Tagliabue L, Balduzzi S, et al. Trastuzumab containing regimens for early breast cancer. Cochrane Database Syst Rev 2012;CD006243. [PubMed]
  34. Long HD, Lin YE, Zhang JJ, et al. Risk of Congestive Heart Failure in Early Breast Cancer Patients Undergoing Adjuvant Treatment With Trastuzumab: A Meta-Analysis. Oncologist 2016;21:547-54. [Crossref] [PubMed]
  35. de Azambuja E, Procter MJ, van Veldhuisen DJ, et al. Trastuzumab-associated cardiac events at 8 years of median follow-up in the Herceptin Adjuvant trial (BIG 1-01). J Clin Oncol 2014;32:2159-65. [Crossref] [PubMed]
  36. Advani PP, Ballman KV, Dockter TJ, et al. Long-Term Cardiac Safety Analysis of NCCTG N9831 (Alliance) Adjuvant Trastuzumab Trial. J Clin Oncol 2016;34:581-7. [Crossref] [PubMed]
  37. Romond EH, Jeong JH, Rastogi P, et al. Seven-year follow-up assessment of cardiac function in NSABP B-31, a randomized trial comparing doxorubicin and cyclophosphamide followed by paclitaxel (ACP) with ACP plus trastuzumab as adjuvant therapy for patients with node-positive, human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2012;30:3792-9. [Crossref] [PubMed]
  38. Bozovic-Spasojevic I, Azim HA Jr, Paesmans M, et al. Neoadjuvant anthracycline and trastuzumab for breast cancer: is concurrent treatment safe? Lancet Oncol 2011;12:209-11. [Crossref] [PubMed]
  39. Russell SD, Blackwell KL, Lawrence J, et al. Independent adjudication of symptomatic heart failure with the use of doxorubicin and cyclophosphamide followed by trastuzumab adjuvant therapy: a combined review of cardiac data from the National Surgical Adjuvant breast and Bowel Project B-31 and the North Central Cancer Treatment Group N9831 clinical trials. J Clin Oncol 2010;28:3416-21. [Crossref] [PubMed]
  40. Swain SM, Ewer MS, Cortés J, et al. Cardiac tolerability of pertuzumab plus trastuzumab plus docetaxel in patients with HER2-positive metastatic breast cancer in CLEOPATRA: a randomized, double-blind, placebo-controlled phase III study. Oncologist 2013;18:257-64. [Crossref] [PubMed]
  41. Valachis A, Nearchou A, Polyzos NP, et al. Cardiac toxicity in breast cancer patients treated with dual HER2 blockade. Int J Cancer 2013;133:2245-52. [Crossref] [PubMed]
  42. 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;377:122-31. [Crossref] [PubMed]
  43. Gianni L, Pienkowski T, Im YH, 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;13:25-32. [Crossref] [PubMed]
  44. Schneeweiss A, Chia S, Hickish T, et al. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol 2013;24:2278-84. [Crossref] [PubMed]
  45. Cameron D, Casey M, Oliva C, et al. Lapatinib plus capecitabine in women with HER-2-positive advanced breast cancer: final survival analysis of a phase III randomized trial. Oncologist 2010;15:924-34. [Crossref] [PubMed]
  46. Perez EA, Koehler M, Byrne J, et al. Cardiac safety of lapatinib: pooled analysis of 3689 patients enrolled in clinical trials. Mayo Clin Proc 2008;83:679-86. [Crossref] [PubMed]
  47. Piccart-Gebhart M, Holmes E, Baselga J, et al. Adjuvant Lapatinib and Trastuzumab for Early Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Results From the Randomized Phase III Adjuvant Lapatinib and/or Trastuzumab Treatment Optimization Trial. J Clin Oncol 2016;34:1034-42. [Crossref] [PubMed]
  48. de Azambuja E, Holmes AP, Piccart-Gebhart M, et al. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): survival outcomes of a randomised, open-label, multicentre, phase 3 trial and their association with pathological complete response. Lancet Oncol 2014;15:1137-46. [Crossref] [PubMed]
  49. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med 2012;367:1783-91. [Crossref] [PubMed]
  50. von Minckwitz G, Huang CS, Mano MS, et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N Engl J Med 2019;380:617-28. [Crossref] [PubMed]
  51. Modi S, Saura C, Yamashita T, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N Engl J Med 2020;382:610-21. [Crossref] [PubMed]
  52. Murthy RK, Loi S, Okines A, et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N Engl J Med 2020;382:597-609. [Crossref] [PubMed]
  53. Chan A, Moy B, Mansi J, et al. Final Efficacy Results of Neratinib in HER2-positive Hormone Receptor-positive Early-stage Breast Cancer From the Phase III ExteNET Trial. Clin Breast Cancer 2021;21:80-91.e7. [Crossref] [PubMed]
  54. Available online: https://www.ema.europa.eu/en/documents/product-information/lynparza-epar-product-information_en.pdf
  55. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/761106s000lbl.pdf
  56. Curigliano G, Lenihan D, Fradley M, et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol 2020;31:171-90. [Crossref] [PubMed]
  57. Armenian SH, Lacchetti C, Barac A, et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2017;35:893-911. [Crossref] [PubMed]
  58. Eschenhagen T, Force T, Ewer MS, et al. Cardiovascular side effects of cancer therapies: a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2011;13:1-10. [Crossref] [PubMed]
  59. Dempsey N, Rosenthal A, Dabas N, et al. Trastuzumab-induced cardiotoxicity: a review of clinical risk factors, pharmacologic prevention, and cardiotoxicity of other HER2-directed therapies. Breast Cancer Res Treat 2021;188:21-36. [Crossref] [PubMed]
  60. Upshaw JN. The Role of Biomarkers to Evaluate Cardiotoxicity. Curr Treat Options Oncol 2020;21:79. [Crossref] [PubMed]
  61. Cardinale D, Colombo A, Torrisi R, et al. Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol 2010;28:3910-6. [Crossref] [PubMed]
  62. Cardinale D, Sandri MT, Martinoni A, et al. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J Am Coll Cardiol 2000;36:517-22. [Crossref] [PubMed]
  63. Cardinale D, Sandri MT, Colombo A, et al. Prognostic value of troponin I in cardiac risk ynparza ation of cancer patients undergoing high-dose chemotherapy. Circulation 2004;109:2749-54. [Crossref] [PubMed]
  64. Demissei BG, Hubbard RA, Zhang L, et al. Changes in Cardiovascular Biomarkers With Breast Cancer Therapy and Associations With Cardiac Dysfunction. J Am Heart Assoc 2020;9:e014708. [Crossref] [PubMed]
  65. Ponde N, Bradbury I, Lambertini M, et al. Cardiac biomarkers for early detection and prediction of trastuzumab and/or lapatinib-induced cardiotoxicity in patients with HER2-positive early-stage breast cancer: a NeoALTTO sub-study (BIG 1-06). Breast Cancer Res Treat 2018;168:631-8. [Crossref] [PubMed]
  66. Gavin PG, Song N, Kim SR, et al. Association of Polymorphisms in FCGR2A and FCGR3A With Degree of Trastuzumab Benefit in the Adjuvant Treatment of ERBB2/HER2–Positive Breast Cancer Analysis of the NSABP B-31 Trial. JAMA Oncol 2017;3:335-41. [Crossref] [PubMed]
  67. Mellor JD, Brown MP, Irving HR, et al. A critical review of the role of Fc gamma receptor polymorphisms in the response to monoclonal antibodies in cancer. J Hematol Oncol 2013;6:1. [Crossref] [PubMed]
  68. Cresti N, Jamieson D, Verrill MW, et al. Fcγ-receptor Iia polymorphism and cardiotoxicity in patients with breast cancer treated with adjuvant trastuzumab. J Clin Oncol 2011;29:2011. [Crossref]
  69. Roca L, Diéras V, Roché H, et al. Correlation of HER2, FCGR2A, and FCGR3A gene polymorphisms with trastuzumab related cardiac toxicity and efficacy in a subgroup of patients from UNICANCERPACS04 trial. Breast Cancer Res Treat 2013;139:789-800. [Crossref] [PubMed]
  70. Available online: https://www.ema.europa.eu/en/documents/product-information/enhertu-epar-product-information_en.pdf
  71. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761139s011lbl.pdf
  72. André F, O'Regan R, Ozguroglu M, et al. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol 2014;15:580-91. [Crossref] [PubMed]
  73. Awada A, Colomer R, Inoue K, et al. Neratinib Plus Paclitaxel vs. Trastuzumab Plus Paclitaxel in Previously Untreated Metastatic ERBB2-Positive Breast Cancer: The NEfERT-T Randomized Clinical Trial. JAMA Oncol 2016;2:1557-64. [Crossref] [PubMed]
  74. Swain SM, Miles D, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA): end-of-study results from a double-blind, randomised, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:519-30. [Crossref] [PubMed]
  75. Available online: https://www.ema.europa.eu/en/documents/product-information/kadcyla-epar-product-information_en.pdf
  76. Raphael J, Lefebvre C, Allan A, et al. Everolimus in Advanced Breast Cancer: A Systematic Review and Meta-analysis. Target Oncol 2020;15:723-32. [Crossref] [PubMed]
  77. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med 2012;366:520-9. [Crossref] [PubMed]
  78. Hurvitz SA, Andre F, Jiang Z, et al. Combination of everolimus with trastuzumab plus paclitaxel as first-line treatment for patients with HER2 positive advanced breast cancer (BOLERO-1): a phase 3, randomised, double-blind, multicentre trial. Lancet Oncol 2015;16:816-29. [Crossref] [PubMed]
  79. Bachelot T, Bourgier C, Cropet C, et al. Randomized phase II trial of everolimus in combination with tamoxifen in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer with prior exposure to aromatase inhibitors: a GINECO study. J Clin Oncol 2012;30:2718-24. [Crossref] [PubMed]
  80. Kornblum N, Zhao F, Manola J, et al. Randomized Phase II Trial of Fulvestrant Plus Everolimus or Placebo in Postmenopausal Women With Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer Resistant to Aromatase Inhibitor Therapy: Results of PrE0102. J Clin Oncol 2018;36:1556-63. [Crossref] [PubMed]
  81. Yardley DA, Bosserman LD, O’Shaughnessy JA, et al. Paclitaxel, bevacizumab, and everolimus/placebo as first-line treatment for patients with metastatic HER2-negative breast cancer: a randomized placebo controlled phase II trial of the Sarah Cannon Research Institute. Breast Cancer Res Treat 2015;154:89-97. [Crossref] [PubMed]
  82. Schmid P, Zaiss M, Harper-Wynne C, et al. Fulvestrant plus vistusertib vs. fulvestrant plus everolimus vs. fulvestrant alone for women with hormone receptor-positive metastatic breast cancer: the manta phase 2 randomized clinical trial. JAMA Oncol 2019;5:1556-64. [Crossref] [PubMed]
  83. Decker T, Marschner N, Muendlein A, et al. VicTORia: a randomised phase II study to compare vinorelbine in combination with the mTOR inhibitor everolimus versus vinorelbine monotherapy for second-line chemotherapy in advanced HER2-negative breast cancer. Breast Cancer Res Treat 2019;176:637-47. [Crossref] [PubMed]
  84. Im YH, Karabulut B, Lee KS, et al. Safety and efficacy of everolimus (EVE) plus exemestane (EXE) in postmenopausal women with locally advanced or metastatic breast cancer: final results from EVEREXES. Breast Cancer Res Treat 2021;188:77-89. [Crossref] [PubMed]
  85. Cazzaniga ME, Airoldi M, Arcangeli V, et al. Efficacy and safety of Everolimus and Exemestane in hormone-receptor positive (HR+) human-epidermal-growth-factor negative (HER2-) advanced breast cancer patients: New insights beyond clinical trials. The EVA study. Breast 2017;35:115-21. [Crossref] [PubMed]
  86. Li Y, Xie Y, Gong C, et al. Comparative Treatment Patterns and Outcomes of Fulvestrant versus Everolimus Plus Exemestane for Postmenopausal Metastatic Breast Cancer Resistant to Aromatase Inhibitors in Real-World Experience. Ther Clin Risk Manag 2020;16:607-15. [Crossref] [PubMed]
  87. Cazzaniga ME, Danesi R, Girmenia C, et al. Management of toxicities associated with targeted therapies for HR-positive metastatic breast cancer: a multidisciplinary approach is the key to success. Breast Cancer Res Treat 2019;176:483-94. [Crossref] [PubMed]
  88. Willemsen AECAB, He X, van Cranenbroek B, et al. Baseline effector cells predict response and NKT cells predict pulmonary toxicity in advanced breast cancer patients treated with everolimus and exemestane. Int Immunopharmacol 2021;93:107404. [Crossref] [PubMed]
  89. Pascual T, Apellániz-Ruiz M, Pernaut C, et al. Polymorphisms associated with everolimus pharmacokinetics, toxicity and survival in metastatic breast cancer. PLoS One 2017;12:e0180192. [Crossref] [PubMed]
  90. Armaghani AJ, Han HS. Alpelisib in the Treatment of Breast Cancer: A Short Review on the Emerging Clinical Data. Breast Cancer (Dove Med Press) 2020;12:251-8. [Crossref] [PubMed]
  91. André F, Ciruelos E, Rubovszky G, et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N Engl J Med 2019;380:1929-40. [Crossref] [PubMed]
  92. Mayer IA, Prat A, Egle D, et al. A Phase II Randomized Study of Neoadjuvant Letrozole Plus Alpelisib for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer (NEO-ORB). Clin Cancer Res 2019;25:2975-87. [Crossref] [PubMed]
  93. Rodon J, Demanse D, Rugo HS, et al. A risk analysis of alpelisib (ALP)-induced hyperglycemia (HG) using baseline factors in patients (pts) with advanced solid tumours and breast cancer (BC): A pooled analysis of X2101 and SOLAR-1. Ann Oncol 2021;32:S64. [Crossref]
  94. Patel M, Nowsheen S, Maraboyina S, et al. The role of poly(ADP-ribose) polymerase inhibitors in the treatment of cancer and methods to overcome resistance: a review. Cell Biosci 2020;10:35. [Crossref] [PubMed]
  95. LaFargue CJ, Dal Molin GZ, Sood AK, et al. Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol 2019;20:e15-28. [Crossref] [PubMed]
  96. Coleman RL, Oza AM, Lorusso D, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390:1949-61. [Crossref] [PubMed]
  97. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N Engl J Med 2016;375:2154-64. [Crossref] [PubMed]
  98. Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017;18:1274-84. [Crossref] [PubMed]
  99. Clovis Oncology. Full prescribing information for rubraca (rucaparib) 2021. Available online: https://clovisoncology.com/pdfs/RubracaUSPI.pdf (accessed Jan 8, 2018).
  100. AstraZeneca. Full prescribing information for Lynparza (ynparza). Available online: https://medicalinformation.astrazeneca-us.com/home/prescribing-information/lynparza-tablets-pi.html (accessed Jan 8, 2018).
  101. Tesaro. Full prescribing information for Zejula (niraparib). Available online: https://gskpro.com/content/dam/global/hcpportal/en_US/Prescribing_Information/Zejula_Capsules/pdf/ZEJULA-CAPSULES-PI-PIL.PDF
  102. Berek JS, Matulonis UA, Peen U, et al. Safety and dose modification for patients receiving niraparib. Ann Oncol 2018;29:1784-92. [Crossref] [PubMed]
  103. Gunderson CC, Matulonis U, Moore KN. Management of the toxicities of common targeted therapeutics for gynecologic cancers. Gynecol Oncol 2018;148:591-600. [Crossref] [PubMed]
  104. Moore KN, Monk BJ. Patient Counseling and Management of Symptoms During Olaparib Therapy for Recurrent Ovarian Cancer. Oncologist 2016;21:954-63. [Crossref] [PubMed]
  105. Moore K, Zhang ZY, Agarwal S, et al. The effect of food on the pharmacokinetics of niraparib, a poly(ADP-ribose) polymerase (PARP) inhibitor, in patients with recurrent ovarian cancer. Cancer Chemother Pharmacol 2018;81:497-503. [Crossref] [PubMed]
  106. Zhao H, Sifakis EG, Sumida N, et al. PARP1- and CTCF-Mediated Interactions between Active and Repressed Chromatin at the Lamina Promote Oscillating Transcription. Mol Cell 2015;59:984-97. [Crossref] [PubMed]
  107. Lucarini L, Pini A, Gerace E, et al. Poly(ADP-ribose) polymerase inhibition with HYDAMTIQ reduces allergen-induced asthma-like reaction, bronchial hyper-reactivity and airway remodelling. J Cell Mol Med 2014;18:468-79. [Crossref] [PubMed]
  108. Schwaiblmair M, Behr W, Haeckel T, et al. Drug induced interstitial lung disease. Open Respir Med J 2012;6:63-74. [Crossref] [PubMed]
  109. Emens LA. Breast Cancer Immunotherapy: Facts and Hopes. Clin Cancer Res 2018;24:511-20. [Crossref] [PubMed]
  110. FDA approves atezolizumab for PD-L1 positive unresectable locally advanced or metastatic triple-negative breast cancer. FDA U.S. Food and Drug. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-atezolizumab-pd-l1-positive-unresectable-locally-advanced-or-metastatic-triple-negative
  111. Schmid P, Rugo HS, Adams S, et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (Impassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2020;21:44-59. [Crossref] [PubMed]
  112. Mittendorf EA, Zhang H, Barrios CH, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet 2020;396:1090-100. [Crossref] [PubMed]
  113. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N Engl J Med 2020;382:810-21. [Crossref] [PubMed]
  114. Barroso-Sousa R, Tolaney SM. Pembrolizumab in the preoperative setting of triple-negative breast cancer: safety and efficacy. Expert Rev Anticancer Ther 2020;20:923-30. [Crossref] [PubMed]
  115. Loibl S, Untch M, Burchardi N, et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol 2019;30:1279-88. [Crossref] [PubMed]
  116. Voorwerk L, Slagter M, Horlings HM, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med 2019;25:920-8. [Crossref] [PubMed]
  117. Krishnan T, Tomita Y, Roberts-Thomson R. A retrospective analysis of eosinophilia as a predictive marker of response and toxicity to cancer immunotherapy. Future Sci OA 2020;6:FSO608. [Crossref] [PubMed]
  118. Fujisawa Y, Yoshino K, Otsuka A, et al. Fluctuations in routine blood count might signal severe immunerelated adverse events in melanoma patients treated with nivolumab. J Dermatol Sci 2017;88:225-31. [Crossref] [PubMed]
  119. Kobayashi T, Iwama S, Yasuda Y, et al. Patients With Antithyroid Antibodies Are Prone To Develop Destructive Thyroiditis by Nivolumab: A Prospective Study. J Endocr Soc 2018;2:241-51. [Crossref] [PubMed]
  120. Toi Y, Sugawara S, Sugisaka J, et al. Profiling Preexisting Antibodies in Patients Treated With Anti-PD-1 Therapy for Advanced Non-Small Cell Lung Cancer. JAMA Oncol 2019;5:376-83. [Crossref] [PubMed]
  121. Kurimoto C, Inaba H, Ariyasu H, et al. Predictive and sensitive biomarkers for thyroid dysfunctions during treatment with immune-checkpoint inhibitors. Cancer Sci 2020;111:1468-77. [Crossref] [PubMed]
  122. Xu Y, Fu Y, Zhu B, et al. Predictive Biomarkers of Immune Checkpoint Inhibitors-Related Toxicities. Front Immunol 2020;11:2023. [Crossref] [PubMed]
  123. Yang G, Nowsheen S, Aziz K, et al. Toxicity and adverse effects of Tamoxifen and other anti-estrogen drugs. Pharmacol Ther 2013;139:392-404. [Crossref] [PubMed]
  124. Osborne CK. Tamoxifen in the treatment of breast cancer. N Engl J Med 1998;339:1609-18. [Crossref] [PubMed]
  125. Mandalà M, Ferretti G, Cremonesi M, et al. Venous thromboembolism and cancer: new issues for an old topic. Crit Rev Oncol Hematol 2003;48:65-80. [Crossref] [PubMed]
  126. Deitcher SR, Gomes MPV. The risk of venous thromboembolic disease associated with adjuvant hormone therapy for breast carcinoma. Cancer 2004;101:439-49. [Crossref] [PubMed]
  127. Hirsimäki P, Aaltonen A, Mäntylä E. Toxicity of antiestrogens. Breast J 2002;8:92-6. [Crossref] [PubMed]
  128. Davies C, Pan H, Godwin J, et al. Long-term effects of continuining adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013;381:805-16. [Crossref] [PubMed]
  129. Dean L. Tamoxifen Therapy and CYP2D6 Genotype. 2014 Oct 7 [updated 2019 May 1]. In: Pratt VM, Scott SA, Pirmohamed M, et al. editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US), 2012.
  130. Francis PA, Regan MM, Fleming GF, et al. Adjuvant ovarian suppression in premenopausal breast cancer. N Engl J Med 2015;372:436-46. [Crossref] [PubMed]
  131. Pagani O, Regan MM, Walley BA, et al. Adjuvant exemestane with ovarian suppression in premenopausal breast cancer. N Engl J Med 2014;371:107-18. [Crossref] [PubMed]
  132. Lee HR, Kim TH, Choi KC. Functions and physiological roles of two types of estrogen receptors, ERα and ERβ, identified by estrogen receptor knockout mouse. Lab Anim Res 2012;28:71-6. [Crossref] [PubMed]
  133. Barrett-Connor E. Sex differences in coronary heart disease. Why are women so superior? The 1995 Ancel Keys Lecture. Circulation 1997;95:252-64. [Crossref] [PubMed]
  134. Kalin MF, Zumoff B. Sex hormones and coronary disease: a review of the clinical studies. Steroids 1990;55:330-52. [Crossref] [PubMed]
  135. Braithwaite RS, Chlebowski RT, Lau J, et al. Meta-analysis of vascular and neoplastic events associated with tamoxifen. J Gen Intern Med 2003;18:937-47. [Crossref] [PubMed]
  136. Amir E, Seruga B, Niraula S, et al. Toxicity of adjuvant endocrine therapy in postmenopausal breast cancer patients: a systematic review and meta-analysis. J Natl Cancer Inst 2011;103:1299-309. [Crossref] [PubMed]
  137. Coates AS, Keshaviah A, Thürlimann B, et al. Five years of letrozole compared with tamoxifen as initial adjuvant therapy for postmenopausal women with endocrine-responsive early breast cancer: update of study BIG 1-98. J Clin Oncol 2007;25:486-92. [Crossref] [PubMed]
  138. van de Velde CJ, Rea D, Seynaeve C, et al. Adjuvant tamoxifen and exemestane in early breast cancer (TEAM): a randomised phase 3 trial. Lancet 2011;377:321-31. [Crossref] [PubMed]
  139. Wilson PW, D'Agostino RB, Sullivan L, et al. Overweight and obesity as determinants of cardiovascular risk: the Framingham experience. Arch Intern Med 2002;162:1867-72. [Crossref] [PubMed]
  140. Goss PE, Ingle JN, Martino S, et al. A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for early-stage breast cancer. N Engl J Med 2003;349:1793-802. [Crossref] [PubMed]
  141. Mamounas EP, Jeong JH, Wickerham DL, et al. Benefit from exemestane as extended adjuvant therapy after 5 years of adjuvant tamoxifen: intention-to-treat analysis of the National Breast Cancer Research and Treatment Surgical Adjuvant Breast And Bowel Project B-33 trial. J Clin Oncol 2008;26:1965-71. [Crossref] [PubMed]
  142. Gray RG, Rea D, Handley K, et al. aTTom: long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013;31:18. [Crossref]
  143. The Early Breast Cancer Trialists’ Collaborative Group. Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomized trials. Lancet 2015;386:1341-52. [Crossref] [PubMed]
  144. Qian X, Li Z, Ruan G, et al. Efficacy and toxicity of extended aromatase inhibitors after adjuvant aromatase inhibitors-containing therapy for hormone-receptor-positive breast cancer: a literature-based meta-analysis of randomized trials. Breast Cancer Res Treat 2020;179:275-85. [Crossref] [PubMed]
  145. Johansson H, Gray KP, Pagani O, et al. Impact of CYP19A1 and ESR1 variants on early-onset side effects during combined endocrine therapy in the TEXT trial. Breast Cancer Res 2016;18:110. [Crossref] [PubMed]
  146. Borrie AE, Rose FA, Choi YH, et al. Genetic and clinical predictors of arthralgia during letrozole or anastrozole therapy in breast cancer patients. Breast Cancer Res Treat 2020;183:365-72. [Crossref] [PubMed]
  147. Lintermans A, Van Asten K, Jongen L, et al. Genetic variant in the osteoprotegerin gene is associated with aromatase inhibitor-related musculoskeletal toxicity in breast cancer patients. Eur J Cancer 2016;56:31-6. [Crossref] [PubMed]
  148. Niravath P, Chen B, Chapman JW, et al. Vitamin D Levels, Vitamin D Receptor Polymorphisms, and Inflammatory Cytokines in Aromatase Inhibitor-Induced Arthralgias: An Analysis of CCTG MA.27. Clin Breast Cancer 2018;18:78-87. [Crossref] [PubMed]
  149. Fan M, Rickert EL, Chen L, et al. Characterization of molecular and structural determinants of selective estrogen receptor downregulators. Breast Cancer Res Treat 2007;103:37-44. [Crossref] [PubMed]
  150. Lee CI, Goodwin A, Wilcken N. Fulvestrant for hormone-sensitive metastatic breast cancer. Cochrane Database Syst Rev 2017;1:CD011093. [PubMed]
  151. Spring LM, Wander SA, Andre F, et al. Cyclin-dependent kinase 4 and 6 inhibitors for hormone receptor-positive breast cancer: past, present, and future. Lancet 2020;395:817-27. [Crossref] [PubMed]
  152. KISQUALI. Prescribing information: ribociclib. East Hanover, NJ: Novartis Pharmaceuticals Corporation, Available online: https://www.pharma.us.novartis.com/sites/www.pharma.us.novartis.com/files/kisqali.pdf (March 2017).
  153. VERZENIO. Prescribing information: abemaciclib. Indianapolis, IN: Eli Lilly and Company. Available online: http://pi.lilly.com/us/verzenio-uspi.pdf (September 2017).
  154. Dhillon S. Palbociclib: first global approval. Drugs 2015;75:543-51. [Crossref] [PubMed]
  155. Infante JR, Cassier PA, Gerecitano JF, et al. A Phase I Study of the Cyclin-Dependent Kinase 4/6 Inhibitor Ribociclib (LEE011) in Patients with Advanced Solid Tumors and Lymphomas. Clin Cancer Res 2016;22:5696-705. [Crossref] [PubMed]
  156. Finn RS, Crown JP, Lang I, et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study. Lancet Oncol 2015;16:25-35. [Crossref] [PubMed]
  157. Finn RS, Martin M, Rugo HS, et al. Palbociclib and Letrozole in Advanced Breast Cancer. N Engl J Med 2016;375:1925-36. [Crossref] [PubMed]
  158. Cristofanilli M, Turner NC, Bondarenko I, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol 2016;17:425-39. [Crossref] [PubMed]
  159. Ding W, Li Z, Wang C, et al. The CDK4/6 inhibitor in HR-positive advanced breast cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2018;97:e10746. [Crossref] [PubMed]
  160. Abemaciclib summary of product characteristics. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208855s000lbl.pdf (Accessed 31 Aug 2018).
  161. Ribociclib summary of product characteristics. Available online: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/004213/WC500233997.pdf (Accessed 31 Aug2018).
  162. Hortobagyi GN, Stemmer SM, Burris HA, et al. Ribociclib as First-Line Therapy for HR-Positive, Advanced Breast Cancer. N Engl J Med 2016;375:1738-48. [Crossref] [PubMed]
  163. DeMichele A, Clark AS, Tan KS, et al. CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment. Clin Cancer Res 2015;21:995-1001. [Crossref] [PubMed]
  164. Goetz MP, Toi M, Campone M, et al. MONARCH 3: Abemaciclib As Initial Therapy for Advanced Breast Cancer. J Clin Oncol 2017;35:3638-46. [Crossref] [PubMed]
  165. Hu W, Sung T, Jessen BA, et al. Mechanistic Investigation of Bone Marrow Suppression Associated with Palbociclib and its Differentiation from Cytotoxic Chemotherapies. Clin Cancer Res 2016;22:2000-8. [Crossref] [PubMed]
  166. Verma S, Bartlett CH, Schnell P, et al. Palbociclib in Combination With Fulvestrant in Women With Hormone Receptor-Positive/HER2-Negative Advanced Metastatic Breast Cancer: Detailed Safety Analysis From a Multicenter, Randomized, Placebo-Controlled, Phase III Study (PALOMA-3). Oncologist 2016;21:1165-75. [Crossref] [PubMed]
  167. Tripathy D, Sohn J, Im SA, et al. First-line ribociclib vs. placebo with goserelin and tamoxifen or a non-steroidal aromatase inhibitor in premenopausal women with hormone receptor-positive, HER2-negative advanced breast cancer: Results from the randomized phase III MONALEESA-7 trial. Cancer Res 2018;4:GS2-05.
  168. Sledge GW Jr, Toi M, Neven P, et al. MONARCH 2: Abemaciclib in Combination With Fulvestrant in Women With HR+/HER2- Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J Clin Oncol 2017;35:2875-84. [Crossref] [PubMed]
  169. IBRANCE. Prescribing information: palbociclib. New York, NY: Pfizer Inc, Available online: http://labeling.pfizer.com/ShowLabeling.aspX?id=2191 (March 2017).
  170. Rugo HS, Diéras V, Gelmon KA, et al. Impact of palbociclib plus letrozole on patient-reported health-related quality of life: results from the PALOMA-2 trial. Ann Oncol 2018;29:888-94. [Crossref] [PubMed]
  171. Iwata H, Umeyama Y, Liu Y, et al. Evaluation of the Association of Polymorphisms With Palbociclib-Induced Neutropenia: Pharmacogenetic Analysis of PALOMA-2/-3. Oncologist 2021;26:e1143-55. [Crossref] [PubMed]
  172. Modi ND, Abuhelwa AY, Badaoui S, et al. Prediction of severe neutropenia and diarrhoea in breast cancer patients treated with abemaciclib. Breast 2021;58:57-62. [Crossref] [PubMed]
  173. Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, A Phase II Study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a Single Agent, in Patients with Refractory HR+/HER2- Metastatic Breast Cancer. Clin Cancer Res 2017;23:5218-24. [Crossref] [PubMed]
  174. Thill M, Schmidt M. Management of adverse events during cyclin-dependent kinase 4/6 (CDK4/6) inhibitor-based treatment in breast cancer. Ther Adv Med Oncol 2018;10:1758835918793326. [Crossref] [PubMed]
  175. Barni S, Petrelli F, Cabiddu M. Cardiotoxicity of antiemetic drugs in oncology: An overview of the current state of the art. Crit Rev Oncol Hematol 2016;102:125-34. [Crossref] [PubMed]
  176. Olasińska-Wiśniewska A, Olasiński J, Grajek S. Cardiovascular safety of antihistamines. Postepy Dermatol Alergol 2014;31:182-6. [Crossref] [PubMed]
doi: 10.21037/pcm-21-38
Cite this article as: Lai E, Persano M, Dubois M, Spanu D, Donisi C, Pozzari M, Deias G, Saba G, Migliari M, Liscia N, Dessì M, Scartozzi M, Atzori F. Drug-related toxicity in breast cancer patients: a new path towards tailored treatment?—a narrative review. Precis Cancer Med 2022;5:15.

Download Citation