ArticlePDF Available

ESO–ESMO fifth international consensus guidelines for breast cancer in young women (BCY5)

Authors:

Abstract and Figures

The 5th International Consensus Symposium for Breast Cancer in Young Women (BCY5) took place virtually in October 2020, organized by the European School of Oncology (ESO) and the European Society of Medical Oncology (ESMO). Consensus recommendations for the management of breast cancer in young women were updated from BCY4 with incorporation of new evidence to inform the guidelines. Areas of research priorities, as well as specificities in different geographic and minority populations were identified. This manuscript summarizes the ESO-ESMO international consensus recommendations, which are also endorsed by the European Society of Breast Specialists (EUSOMA).
Content may be subject to copyright.
SPECIAL ARTICLE
ESOeESMO fth international consensus guidelines for breast cancer in
young women (BCY5)
S. Paluch-Shimon
1*y
, F. Cardoso
2y
, A. H. Partridge
3
, O. Abulkhair
4
, H. A. Azim
5
, G. Bianchi-Micheli
6
, M. J. Cardoso
2
,
G. Curigliano
7,8
, K. A. Gelmon
9
, O. Gentilini
10
, N. Harbeck
11
, B. Kaufman
12
, S. B. Kim
13
, Q. Liu
14
, J. Merschdorf
15
,
P. Poortmans
16
, G. Pruneri
17
, E. Senkus
18
, B. Sirohi
19
, T. Spanic
20
, V. Sulosaari
21
, F. Peccatori
7,22z
& O. Pagani
23z
1
Hadassah University Hospital & Faculty of Medicine, Hebrew University, Jerusalem, Israel;
2
Breast Unit, Champalimaud Clinical Centre/Champalimaud Foundation,
Lisbon, Portugal;
3
Dana-Farber Cancer Institute, Boston, USA;
4
King Abdulaziz Medical City for National Guard, Riyadh, Saudi Arabia;
5
Breast Cancer Center, Hospital
Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico;
6
Breast Unit of Southern Switzerland, Lugano, Switzerland;
7
European
Institute of Oncology IRCCS, Milan;
8
Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy;
9
British Columbia Cancer, Vancouver, Canada;
10
San Raffaele Hospital, Milan, Italy;
11
Breast Center, Department of OB&GYN and CCCMunich, LMU University Hospital, Munich, Germany;
12
Sheba Medical Center,
Ramat Gan, Israel;
13
Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea;
14
Sun Yat-sen Memorial Hospital, Sun Yat-sen University,
Guangzhou, China;
15
Young Survival Coalition, New York, USA;
16
Iridium Netwerk, Department of Radiation Oncology & University of Antwerp, Faculty of Medicine and
Health Sciences, Wilrijk-Antwerp, Belgium;
17
National Cancer Institute, IRCCS Foundation, Milan, Italy;
18
Medical University of Gdansk, Gdansk, Poland;
19
Max Institute
of Cancer Care, New Delhi and Gurgaon, India;
20
Europa Donna Slovenia, Ljubljana, Slovenia;
21
European Oncology Nursing Society (EONS) and Turku University of
Applied Sciences, Turku, Finland;
22
European Institute of Oncology IRCCS & European School of Oncology, Milan, Italy;
23
Interdisciplinary Cancer Service Hospital
Riviera-Chablais Rennaz, Vaud, Geneva University Hospitals, Lugano University, Swiss Group for Clinical Cancer Research (SAKK), Lugano, Switzerland
Available online XXX
We dedicate this manuscript in memory of a dear friend and colleague Bella Kaufman.
The fth International Consensus Symposium for Breast Cancer in Young Women (BCY5) took place virtually in October
2020, organized by the European School of Oncology (ESO) and the European Society of Medical Oncology (ESMO).
Consensus recommendations for the management of breast cancer in young women were updated from BCY4 with
incorporation of new evidence to inform the guidelines. Areas of research priorities as well as specicities in
different geographic and minority populations were identied. This manuscript summarizes the ESOeESMO
international consensus recommendations, which are also endorsed by the European Society of Breast Specialists
(EUSOMA).
Key words: young women, breast cancer, fertility
INTRODUCTION
Only 4% of new breast cancer cases in the United States in
2019 occurred in women <40 years of age, with an esti-
mated cumulative risk of 1 in 65 by the age of 40 years and
1 in 209 before 30 years of age.
1
In low- and middle-income
countries (LMICs), breast cancer before menopause repre-
sents a much greater burden both in incidence (55% of total
breast cancer cases compared to 25% in high-income
countries) and death rates (8.5 and 3.3 deaths/100 000,
respectively),
2-4
the highest mortality being reported in
Africa (with the exception of southern Africa), Melanesia,
the Caribbean and parts of south-central Asia (e.g.
Afghanistan, Pakistan and Turkmenistan). Reports of
increasing incidence of premenopausal breast cancer in
some high-income industrialized countries (France, Italy,
New Zealand, Norway)
2,5
may reect changes in age de-
mographics. In the United States, an increasing incidence of
de novo metastatic disease has recently been reported in
young black women.
6
Compared with their older counterparts, breast cancers
arising in young women are characterized by higher pro-
portion of tumors with aggressive phenotypes
7
and less
favorable outcomes irrespective of stage at diagnosis,
8,9
particularly in luminal-A like tumors.
10
Possible explana-
tions for the poorer outcome in luminal-like tumors include
different tumor or host biology, less chemotherapy-induced
amenorrhea (CIA), suboptimal endocrine treatment and
decreased adherence to adjuvant endocrine therapy (ET).
Young women are under-represented in contemporary
research evaluating risk-stratication models and molecular
tools resulting in young patients at risk of being
*Correspondence to: Dr Shani Paluch-Shimon, Sharett Institute of Oncology,
Hadassah University Hospital & Faculty of Medicine, Hebrew University, Ein
Kerem, Jerusalem, 12000, Israel. Tel: þ972-26777755
E-mail: shanipal@hadassah.org.il (S. Paluch-Shimon).
y
Co-rst authors.
z
Co-last authors.
0923-7534/© 2022 European Society for Medical Oncology. Published by
Elsevier Ltd. All rights reserved.
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 1
over-treated based solely on age considerations. Prospec-
tive trials dedicated to young patients are key to answering
many of the outstanding questions to ensure optimal
management. Two such examples are the POSH cohort
study, conducted at 127 hospitals in the UK,
11
and the
Helping Ourselves, Helping Others: The Young Womens
Breast Cancer Study (HOHO) conducted in the United States
and Europe. These studies and others have demonstrated a
greater proportion of luminal B and estrogen receptor-
negative (ER) tumors in young patients, increased risk of
early relapse and a more unfavorable longer-term outcome
for young women with ERþtumors when compared to
older women.
8,11
Consistent with previous guidelines,
12-16
the panel
dened young womenas women under the age of 40
years at breast cancer diagnosis and dened advanced
breast cancer in young womenas diagnosis of inoperable
locally advanced or metastatic disease before the age of 40
years.
Due to the coronavirus disease 2019 (COVID-19)
pandemic, the fth International Consensus Symposium for
Breast Cancer in Young Women (BCY5) took place virtually
on 10-11 October 2020 with over 1200 participants
including health care professionals and patient advocates.
The BCY5 guidelines are developed by the European School
of Oncology (ESO) and European Society of Medical
Oncology (ESMO) and are endorsed by the European Soci-
ety of Breast Cancer Specialists (EUSOMA). All recommen-
dations are for standard care, outside of clinical trials. All
diagnostic and treatment recommendations should be
considered in the context of national regulatory approval,
availability and reimbursement.
METHODOLOGY
Recommendations from BCY4 formed the basis for the
current recommendations; new and updated statements
from BCY4 were circulated amongst the panelists before
BCY5 and were then presented, discussed, adapted and
voted on during the consensus session. All panel members
were instructed to vote on all questions; members with
potential conicts of interest or who did not feel comfort-
able responding (e.g. due to lack of expertise on the topic)
were instructed to abstain for that specic question. Sub-
stantial controversies or disagreements are noted in the
discussion of the recommendations. These recommenda-
tions were later circulated to panel members by email for
comments, updating based on recent reports and correc-
tions as needed.
Previous recommendations not requiring update or only
minor changes were not re-voted and remain part of the
recommendations of BCY5.
Tables 1-6 describe the grading system used as per ESMO
guidelines methodology (adapted from Dykewicz et al.
17
;see
http://www.esmo.org/Guidelines/ESMO-Guidelines-Methodo
logy), general guidelines for the management of young
women with breast cancer including early and advanced
disease, supportive care and follow-up and management of
women with germline pathogenic variants (PVs). Statements
without grading were considered justied standard clinical
practice by the panel experts.
Supplementary Appendix S1, available at https://doi.org/
10.1016/j.annonc.2022.07.007, presents the denition of
menopause following CIA and detailed supportive and
follow-up care issues, unchanged or slightly modied since
BCY4.
BCY5 consensus panel members and their disclosure of
any relationships with the pharmaceutical industry that
could be perceived as a potential conict of interest are
reported.
General considerations when caring for young women
with breast cancer
Management of young women with breast cancer is multi-
faceted and requires specic and dedicated multidisci-
plinary care (including but not limited to medical and
radiation oncologist, gynecologist, pathologist, radiologist,
breast and plastic surgeon, nurse specialist, geneticist,
physiotherapist, fertility, sexual therapy and psychosocial
experts), best provided in dedicated breast centers, whose
quality control assurance ensures qualied experience and
care.
18
The panel recommended that personalized psycho-
social support and counseling on genetic predisposition,
fertility, sexual health and socioeconomic consequences be
incorporated into individual treatment planning as well as
lifelong follow-up care given the improved long-term sur-
vival with modern therapies and risks of late side-effects.
Specic guidelines for post-treatment survivorship care
19
are available for health care providers.
Table 1. Levels of evidence and grades of recommendation
Levels of evidence
I Evidence from at least one large randomized, controlled
trial of good methodological quality (low potential for bias)
or meta-analyses of well-conducted randomized trials
without heterogeneity
II Small randomized trials or large randomized trials with a
suspicion of bias (lower methodological quality) or meta-
analyses of such trials or of trials with demonstrated
heterogeneity
III Prospective cohort studies
IV Retrospective cohort studies or case-control studies
V Studies without control group, case reports, experts
opinions
Grades of recommendation
A Strong evidence for efcacy with a substantial clinical
benet, strongly recommended
B Strong or moderate evidence for efcacy but with a limited
clinical benet, generally recommended
C Insufcient evidence for efcacy or benet does not
outweigh the risk or the disadvantages (adverse events,
costs, etc.), optional
D Moderate evidence against efcacy or for adverse
outcome, generally not recommended
E Strong evidence against efcacy or for adverse outcome,
never recommended
Adapted from the Infectious Diseases Society of America-United States Public Health
Service Grading System with their permission.
Annals of Oncology S. Paluch-Shimon et al.
2https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
Table 2. General guidelines
Guidelines LoE/GoR Consensus
Overall, the stage-specic outcome of young breast cancer patients has improved over the years due to diagnostic and
treatment advances.
Nonetheless, even in countries with universal health care, these improvements are signicantly lower for women with
low socioeconomic status (SES) compared to those with high SES.
Every young breast cancer patient must have access to optimal cancer treatment and supportive care according to the
highest standards of patient-centered care, irrespective of her social status.
Expert opinion 100%
Many specic issues in the treatment of young women with breast cancer, both in the early and in the advanced
settings, still lack denitive proven standards. Therefore, well-designed, independent, prospective randomized trials
should be a global research priority.
Expert opinion
The care of all young patients with breast cancer (either early stage, EBC, or advanced disease, ABC) should be
discussed within a multidisciplinary team before any treatment decision making, and ideally provided in specialized
breast clinics.
Expert opinion
Navigators/navigation tools are of great assistance in optimizing patient care. Navigators should ideally be breast
nurses but lay-health professionals with strong communication skills and sufcient experience may also address
complex care issues and mixed cultural settings.
Expert opinion
In view of the many specic aspects related to young age, personalized psychosocial support, counseling on genetic
predisposition, fertility, sexual health and socioeconomic impact are highly recommended as part of the individual
treatment planning.
Expert opinion
In young women, innovative and structured communication and supportive tools (e.g. online programs, web-based
interventions) should be developed and scientically validated and disseminated in different languages.
This would help young patients to overcome barriers to accessing support, such as child and family care, work
timetables and distance issues.
Expert opinion
In view of the many specic aspects related to young age, personalized psychosocial support, counseling on genetic
predisposition, fertility, sexual health and socioeconomic impact are highly recommended as part of the individual
treatment planning.
Patient support groups should be developed and promoted.
Open discussion and shared decision making should be promoted in a clear, culturally appropriate manner encouraging
patients to be proactive in their cancer care.
Expert opinion
Young age by itself should not be the reason to prescribe more aggressive therapy than in other age groups. Factors
inuencing choice of treatment should include but not be limited to the biological characteristics of the tumor [ER/PR,
HER2, proliferation markers (e.g. Ki-67), histological grade], tumor stage, genetic status (if available) and patient s
comorbidities and preferences.
Expert opinion
Systematic research into age-specic host-tumor characteristics is needed.
In particular, the identication of age-specic molecular, biological, radiomics-based and/or genomic aberrations with
prognostic and predictive signicance could open the door for tailored therapeutic interventions.
Expert opinion
National reimbursement policies/algorithms rewarding treatment protocols per number of treatments, dosages,
administration route/use of day hospital or planning complexity (in the case of radiation treatment planning) should be
discouraged.
For exampledradiation therapy should not be reimbursed per fraction nor should physicians receive reimbursement
for administering intravenous chemotherapy.
Expert opinion
Risk factors
Although data about whether obesity is a risk factor for breast cancer in premenopausal women are not conclusive, it
can be said with certainty that obesity is a risk factor for a multitude of serious diseases. For the time being, young
women should be encouraged to maintain BMI 25 kg/m
2
.
Expert opinion Y ¼96%
A¼4%
Pharmaco-prevention with tamoxifen has proven to be effective in reducing breast cancer occurrence in high-risk
young women but is underutilized.
Pharmaco-prevention indication, schedule and potential adverse events should be discussed with all young women at
high risk of developing breast cancer.
IA Y ¼96%
A¼4%
There is limited evidence to suggest specic lifestyle approaches (e.g. dietary habits, weight management and physical
activity) for ovarian and breast cancer risk reduction among women with a germline BRCA1/2 mutation compared to
the general population.
At this time, before obtaining solid data, recommendations about lifestyle behaviors should follow available guidelines
for the general population.
Expert opinion 100%
Male breast cancer
Male breast cancer should be managed in accordance with international recommendations/guidelines. Expert opinion
Clinical trials should allow for inclusion of male breast cancer patients in both early and advanced settings. Expert opinion
Transgender (TG) and nonbinary (NB) persons
Knowledge about the risks of breast/gynecological cancers in TG and NB persons receiving gender-afrming hormone
therapy is limited.
Even less information exists regarding TG/NB individuals with known genetic predisposition.
Cancer registries should include TG/NB identities, and further education and research should be implemented to ll
the gap in information and counseling in this population.
Expert opinion 100%
In green, NEW/MODIFIED BCY5 guidelines with consensus voting.
ER, estrogen receptor; GoR, grades of recommendation; HER2, human epidermal growth factor receptor 2; LoE, levels of evidence; PR, progesterone receptor.
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 3
Table 3. Assessment and treatment guidelines in early breast cancer
Guidelines LoE/GoR Consensus
Screening, diagnosis and imaging for staging and follow-up
There is no clear role for routine screening by any imaging for early breast cancer detection in healthy, average-risk
young women.
Additional consideration may be given to ultrasound and breast MRI in young women particularly in the setting of very
dense breast tissue or consideration of a genetic predisposition or other higher-risk individuals (i.e. radiation therapy
for childhood or young adulthood malignancy).
IA
Expert opinion
Diagnosis, imaging and staging in young women should follow standard algorithms consistent with older women.
Additional consideration may be given to ultrasound and breast MRI in young women with very dense breast tissue.
IIC
Digital breast tomosynthesis (DBT) is more sensitive than digital mammography in dense breasts, is associated with
marginal increase in radiation dose and should be the preferred diagnostic tool.
Expert opinion Y ¼80%
N¼5%
A¼15%
In patients with dense breasts, abbreviated magnetic resonance imaging (AB-MRI) may represent a valid alternative to
conventional MRI with the advantages of short examination/interpretation times and low costs.
Expert opinion Y ¼45%
N¼15%
A¼40%
For BRCA 1/2 mutation carriers and others at high risk based on family history or predisposing mutations in other
genes (e.g. p53, PALB2, CHEK2, ATM), and for those at increased risk because of a personal history of therapeutic
radiation, annual surveillance with MRI and mammography with or without ultrasound is recommended.
IIA
For BRCA 1/2 mutation and other cancer susceptibility genes carriers (e.g. RAD51C, p53, BRIP1) who have not
undergone salpingo-oophorectomy, gynecologic surveillance every 6 months is recommended.
Expert opinion
Annual whole body MRI and brain MRI should be offered to women harboring germline p53 pathogenic variants
(LieFraumeni syndrome) for staging and follow-up.
Other diagnostic tools (e.g. FDGePETeCT) are still under evaluation in LieFraumeni patients as well as in patients
harboring other germline pathogenic gene variants (e.g. ATM carriers) and should not be part of routine staging and
follow-up.
Expert opinion
Expert opinion
Y¼75%
A¼25%
Y¼75%
A¼25%
Risk-adapted early detection and surveillance tools should be researched in young women. Expert opinion
Genetic counseling and testing
Recent literature suggests that health disparities remain amongst young women from minority or disadvantaged
ethnic/racial backgrounds in utilizing and seeking genetic counseling services based on family history of malignancy,
before a breast cancer diagnosis.
Risk-reducing measures are also underutilized in minority BC patients harboring BRCA1/2 pathogenic gene variants.
Strategies to minimize racial/ethnic and social disparities in early access to genetic testing and risk management should
be implemented to optimize preventive interventions
Expert opinion Y ¼96%
A¼4%
Every young woman with breast cancer should be offered genetic counseling preferably before starting treatment.
Practice should follow national/international guidelines on a country-by-country basis. For those women who are not
ready to consider genetic issues at diagnosis, access to genetic counseling should be offered again during follow-up, to
address issues of surveillance and risk reduction of additional primary tumors for the patient, and risk issues for
relatives.
Expert opinion
Genetic testing should be performed only after adequate information is provided by an appropriately trained health
professional who explains the implications of the results, according to national/international regulations.
The patient must be made aware that the presence of a predisposing mutation may have an impact on her
management, follow-up and decision making, as well as family members.
A fast-track process should be available when the identication of a pathogenic gene variant could change the
therapeutic approach (e.g. indication for risk-reducing surgery, platinum derivatives, PARP inhibitors, etc.).
Little is known about the clinical/psychosocial factors affecting the communication of genetic test results by young BC
patients with parents and siblings, especially in non-Caucasian women.
Further research and strategies to improve communication among families are needed and should be developed.
Expert opinion 100%
Genes to be tested for depend on personal and family history.
Although BRCA1/2 are the most frequently mutated genes, other additional moderate- to high-penetrance genes may
be considered, if deemed appropriate by the geneticist/genetic counselor or if they will impact therapeutic
interventions.
Development of quality-controlled genetic counseling services is strongly encouraged.
Expert opinion 100%
When a hereditary cancer syndrome is suspected and a mutation in BRCA1/2 has not been identied, multi-gene panel
testing may be considered. Practice should be guided by high quality national/international guidelines.
As commercially available multi-gene panels include different panels of genes, the choice of the specic panel and
quality-controlled laboratory is crucial.
Expert opinion
Risk communication and clinical recommendations need to be adapted to the increased complexity and uncertainty of
multi-gene testing.
Health professionals should also be trained to address the related complex psychological scenarios.
Expert opinion 100%
The clinical utility (including risk assessment, screening and prevention recommendations) of moderate-risk genes
identied on multi-gene panel testing is not yet established and this needs to be clearly communicated to patients in
both the pre- and post-testing counseling consultations.
Expert opinion
Multi-gene panel testing (when available) should be proposed when either a hereditary cancer syndrome is suspected
and a pathogenic gene variant in BRCA1/2 has not been identied and/or if the personal/family history can be
explained by more than one gene. Practice should be guided by national/international guidelines.
Expert opinion
As commercially available multi-gene panels include different genes, the choice of the specic panel should be
performed in quality-controlled laboratories.
Expert opinion
For the time being somatic BRCA1/2 testing should not be used as an alternative or in addition to germline pathogenic
gene variant identication. The therapeutic implications of somatic BRCA1/2 mutations in breast tumors need to be
further explored within a research setting before they can be used in routine clinical practice.
Expert opinion
Amongst individuals who do not have an identied low-/moderate-/high-risk germline mutation, polygenic risk score
models, evaluating multiple factors for estimating breast cancer risk, are not yet ready for implementation in routine
clinical practice.
Expert opinion Y ¼87%
A¼13%
Continued
Annals of Oncology S. Paluch-Shimon et al.
4https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
Table 3. Continued
Early breast cancer locoregional treatment
Surgical treatment of young patients with EBCdwhile being tailored to the individual patientdshould in general not
differ from that of older patients.
Breast-conserving surgery should be performed as the rst option whenever suitable, as it provides the same overall
survival than mastectomy.
I
IA
Onco-plastic repair techniques should be discussed with all patients treated by BCS in order to maximize cosmetic
results and optimize self-image whenever an obvious post-operative asymmetry can be estimated by a dedicated
breast surgical team. Immediate breast reconstruction after mastectomy offers the same survival rates as mastectomy
without reconstruction and should be offered to all patients except those with inammatory breast cancer.
IC
Immediate breast reconstruction after mastectomy is less frequent in patients with low socioeconomic status (SES)
compared to those with high SES, even in countries with universal health care where the inuence of reimbursement
should be negligible.
Immediate breast reconstruction is rarely available in patients from low- and middle-income countries.
Every young BC patient should have access to immediate breast reconstruction and oncoplastic surgery if medically
appropriate.
Expert opinion 100%
There is no evidence of an increased false-negative rate or a worse outcome in young patients undergoing SNLB,
therefore the indications for SNLB are the same as in older patients.
IB
In young women with the diagnosis of either invasive disease or pre-invasive lesions, who are not high-risk mutation
carriers, there is no evidence for improved OS by performing risk-reducing bilateral mastectomy.
IB
For all surgical decisions and particularly for risk-reducing mastectomy, patients must be given proper, thorough and
unbiased information based on the available data, and adequate time to decide. Once an informed decision is made by
the patient it should be respected. Additional psychosocial support should be offered given the potentially high anxiety
and long-term sequela of patients making these difcult decisions.
Expert opinion
Decisions about locoregional treatment after neoadjuvant chemotherapy should be made independent of age. Expert opinion
Mutation status should be part of the individual decision-making algorithm. Sufcient time to discuss the different
options and adequate psychological support should be offered given the potential long-term sequela and implications.
Expert opinion
Indications for adjuvant RT are the same as for older patients.
After breast-conserving surgery, breast radiation + boost are recommended. Young patients should be informed about
the high local recurrence risk if RT is avoided after BCS and about the benet of RT on reduction of local recurrence and
improvement in OS. This must be balanced with information about the potential long-term toxicities.
Partial breast irradiation (PBI) or accelerated PBI has not been sufciently studied in young patients and should not be
performed in this age group.
IB
Indications for post-operative RT are independent of BRCA status.
Limited and inconclusive evidence is available in presence of pathogenic gene variants in other predisposing genes
(e.g. CHEK2, ATM): in these patients the riskebenet ratio needs to be individually discussed.
For patients with a germline TP53 mutation, post-operative RT is relatively contraindicated and mastectomy is
preferred.
In these patients, post-mastectomy RT should be discussed only in cases of signicant risk of locoregional recurrence.
This underscores the importance of early genetic testing at the time of diagnosis to aid optimal treatment planning.
Expert opinion Y ¼87%
A¼13%
Despite the fact that data on the efcacy and safety of hypofractionation are accumulating in premenopausal women,
hypofractionation is not widely adopted as standard radiation therapy for young patients in many countries.
Hypofractionated WBI schedules should replace standard fractionated WBI as gold standard for most patients
irrespective of their age.
IB Y ¼74%
A¼26%
Ultra-hypofractionated WBI or for the lymph node regions (such as in FAST-Forward) is not yet standard for young
patients.
Expert opinion Y ¼61%
N¼4%
A¼35%
Indications and extension of nodal irradiation are the same as in other age groups. IB
Indications for adjuvant RT are the same as for older patients.
Data are stronger for benets of post-mastectomy RT for young women.
IB
IB
Partial breast irradiation (PBI), or accelerated PBI, has not been sufciently studied in young patients and should not be
performed in this age group.
Expert opinion
Post-mastectomy radiation therapy (PMRT) to implant-based breast reconstruction may be associated with a higher
risk of capsular contracture.
When PMRT is foreseen the timing and technique of the procedure should be discussed pre-operatively on an
individual basis by all the specialists involved.
Expert opinion Y ¼85%
N¼5%
A¼10%
Adjuvant systemic treatment
Endocrine therapy
All young women should be counseled, before the onset of systemic therapy (either CT or ET), about the risks,
associated symptoms and outcomes of treatment-related amenorrhea and premature menopause, referred for special
fertility counseling/consultation and informed of available and approved ameliorative therapies.
Expert opinion
Neoadjuvant ET should not be used in young women outside clinical trials. Expert opinion
All patients with HR-positive disease should receive adjuvant ET.
Tamoxifen alone for 5 years is indicated for low-risk patients.
Switching to an AI, after 5 years of tamoxifen, should be considered for women who have become denitively
postmenopausal.
Tamoxifen for 10 years should be considered in high-risk patients, if tolerated.
The addition of a GnRH agonist (or ovarian ablation) to tamoxifen or an aromatase inhibitor is indicated in patients at
higher risk of relapse.
IA
IA
IA
IA
IA
AIs without ovarian function suppression are contraindicated in premenopausal women.
The combination of an AI and a GnRH agonist (or oophorectomy) confers a signicant absolute benet in terms of
freedom from distant recurrence and should be the preferred option in higher-risk patients.
IA Y ¼90%
N¼5%
A¼5%
Continued
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 5
Table 3. Continued
Adjuvant systemic treatment
Endocrine therapy
Young women with stage I or II breast cancer who cannot take tamoxifen (due to contraindications or severe side-
effects) may receive a GnRH agonist alone, oophorectomy or an aromatase inhibitor + GnRH agonist.
IA
Recommendations for adjuvant GnRH agonist use are based on data from trials with monthly administration. Thus,
current guidelines support monthly use to optimize ovarian function suppression, particularly in very young women
(<35 years of age) and in those receiving an AI.
3-6 monthly use may be considered on a case-by-case basis with very close monitoring of ovarian function, when
monthly use is not feasible or accepted by the patient.
Expert opinion Y¼90%
N¼5%
A¼5%
Estradiol levels should be checked if there are concerns ovarian function is not suppressed, especially if a breakthrough
bleeding occurs and/or the patient is on an AI; if done, the analysis should preferably be performed in the same
laboratory, and when possible in a central reference laboratory. In cases of inadequate suppression alternative
strategies should be discussed (oophorectomy or continuation of tamoxifen alone).
Expert opinion
The method of ovarian suppression (surgical versus medical) requires balancing patients wish for potentially
preserving fertility, compliance with frequent injections over a long period of time and cost/availability.
Expert opinion
Radiation to ovaries as a method of ovarian function suppression should be discouraged. Expert opinion Y ¼40%
N¼30%
A¼30%
The addition of a GnRH agonist to tamoxifen can be considered in women at higher risk of relapse resuming ovarian
function within 2 years after chemotherapy completion.
IIB
Adjuvant CDK4/6 inhibitors
Early data suggest that adjuvant abemaciclib combined with ET may be benecial in patients with high-risk ER+ disease
(3.5% absolute risk reduction in invasive disease-free survival), however follow-up time is short.
43% of patients were premenopausal with similar magnitude of benet observed irrespective of menopausal status.
High-risk features were dened in the clinical trial as 4 LN or 1-3 involved LN with other high-risk features (tumor size
5 cm, grade 3, Ki67 20%).
Abemaciclib could be considered for use in the described high-risk groups, when approved.
IB Y ¼61%
N¼13%
A¼26%
Chemotherapy
Many factors, including patient and tumor characteristics and genomic signatures, should be considered when deciding
whether to administer adjuvant chemotherapy in young women with HR+ early BC. Most young women enrolled in
these studies were not treated with modern, risk-adapted ET. Available data in premenopausal women support the role
for gene expression tests in predicting additional benet of chemotherapy over ET alone but further data are needed in
order to guide clinical practice and to determine whether the observed benets are from the cytotoxic therapy or the
chemotherapy-induced OFS.
IB Y ¼91%
A¼9%
Commercially available prognostic genomic assays in HR+ early breast cancer have not been developed to predict
which endocrine therapy is more appropriate according to genomic risk. Therefore, they should not be used at this
time for selecting type or duration of endocrine therapy.
Expert opinion
Available data suggest that a discussion of omitting adjuvant chemotherapy in very young women (35 years of age at
diagnosis) with low-risk ER+ disease is appropriate in highly selected cases with favorable clinical and pathological
features including low gene expression proles where available.
Expert opinion
The indications for and the choice of adjuvant systemic treatment for invasive breast cancer should be driven, as for
women in other age categories, by extent of disease and the biological characteristics of the tumor (including, but not
limited to, ER/PR and HER2 receptors, proliferation and grade) and patients comorbidities.
IA
For the time being, the type of systemic treatment of EBC is independent of BRCA or any other constitutional genetic
status.
Expert opinion
The optimal (neo)adjuvant CT regimen specically for young women in terms of efcacy and long-term toxicity is
currently unknown. As for all stage I-III breast cancer patients, the preferred regimens are standard anthracycline,
alkylating and taxane-based regimens.
The indication for dose-dense chemotherapy is independent of age.
IA
Young age by itself should not be an indication to prescribe a more intense combination of cytotoxic agents.
Standard duration of chemotherapy (minimum of 4 and maximum of 8 cycles) should be prescribed.
Sequential regimens have at least equal or superior efcacy over combinations and are better tolerated.
The indication for dose-dense chemotherapy is independent of age.
IA
In patients with TNBC or BRCA-associated tumors the incorporation of platinum agents increases pCR rates and may be
considered when neoadjuvant chemotherapy is indicated.
Data on the impact of incremental increases in pCR on long-term outcome are not conclusive.
The use of platinum derivatives has potential additional impact on fertility and increased toxicity that may compromise
standard duration and dosing of systemic treatment, and this needs to be clearly communicated to patients.
IB
For patients with TNBC not achieving a pCR after standard neoadjuvant regimens, the routine addition of adjuvant
chemotherapy with 6-8 cycles of capecitabine may be considered
IA
There are no data on the use of platinum derivatives in the adjuvant setting and therefore these cannot be
recommended.
IA
In patients with TNBC with indications for pre-operative chemotherapy, the addition of pembrolizumab (with the
chemotherapy and for completion of 1 year following surgery) can be considered in selected young patients with high-
risk disease, where approved, after careful consideration and discussion with the patient about the possibility of long-
term side-effects.
IA Y ¼90%
N¼10%
Continued
Annals of Oncology S. Paluch-Shimon et al.
6https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
The National Comprehensive Cancer Network (NCCN) in
Oncology and the Breast Health Global Initiative developed
resource-stratied and harmonized guidelines/recommen-
dations
20,21
and strategies to address the poor outcomes in
LMICs for young women with breast cancer.
22
Prospective
cohort studies are collecting data on young patientscon-
cerns, in particular about fertility preservation,
23,24
selection
of ovarian function preservation strategies
23
and psychoso-
cial and quality-of-life (QoL) issues after diagnosis.
25
Young women with a lower socioeconomic status (SES) have
worse outcomes compared to those with a higher SES,
26
even
in countries with universal health care
27,28
thought to be due
to a higher frequency of comorbidities and limited access to
health services. In some studies, differences persist after
adjusting for all these factors. In particular, a large Norwegian
report in premenopausal women (7501 patients aged 30-48
years at diagnosis) showed that 5-year relative survival
improved steadily for patients with high SES (by 9% and 36%,
for regional and distant disease, respectively), but remained
unchanged for patients with low SES.
27
The BCY5 panel
strongly and unanimously stated that every young breast
cancer patient must have access to optimal cancer treatment
and supportive care according to the highest standards of
patient-centered care, irrespective of her social status.
Panel members re-emphasized that national reimburse-
ment policies/algorithms rewarding treatment protocols
per number of treatments, dosages, administration route/
use of day hospital or planning complexity (in the case of
radiation treatment) should be discouraged. For example,
radiation therapy (RT) should not be reimbursed per frac-
tion, nor should physicians receive higher reimbursement
for administering intravenous agents compared to oral
chemotherapy or ET. To this end, the panel endorses the
12th European Breast Cancer Conference Manifesto.
29
Asignicant proportion of young patients experience work-
and insurance-related challenges in the rst 2 years after
diagnosis,
30-33
but little is known about the impact of late ef-
fects of cancer treatment (e.g. fatigue and arm morbidity) on
Table 3. Continued
Adjuvant PARP inhibitors
Early data suggests that 1 year of adjuvant olaparib following the completion of (neo)adjuvant chemotherapy provides
signicant benet in disease-free survival amongst women harboring a germline BRCA1/2 mutation who have high-risk
early breast cancer. High-risk features were dened in the clinical trial as stage 2-3, HER2 negative-TNBC: pT2Nx or
pTxN1-3 or residual disease after NAST; HR+:pTxN2-3 or signicant residual disease after NAST. It may also reduce the
risk of further primary malignancies. Olaparib, once approved, should be offered to germline BRCA1/2 mutation
carriers that meet the inclusion criteria for the OLYMPIA trial and modications which may be approved by the
regulatory bodies.
IA Y ¼91%
A¼9%
Anti-HER2 therapy
One year treatment with adjuvant trastuzumab, together with chemotherapy, is indicated for women with HER2-
positive, node-positive or high-risk node-negative breast cancer (tumor size >0.5 cm), who have a left ventricular
ejection fraction within normal limits and without signicant cardiovascular risk factors, irrespective of age.
IA
In highly selected patients with small, node-negative, HER2+ breast cancer, the administration of 12 weeks of weekly
paclitaxel and trastuzumab without anthracyclines can be considered
The incorporation of neoadjuvant/adjuvant pertuzumab should be in keeping with current standards, as for older
patients, in women with high-risk HER2+ breast cancer.
IIB
IA
In case of pathological residual disease (non-pCR) after pre-operative chemotherapy plus anti-HER2 therapy the
patient should be offered to complete 1 year of adjuvant anti-HER2 therapy with TDM-1.
IA Y ¼85%
N¼5%
A¼10%
In HER2+ patients at high risk of relapse (e.g. N+) 1 year adjuvant pertuzumab + trastuzumab can be discussed, as in
other age groups.
Limited data are available on the efcacy of such treatment in women who received pertuzumab + trastuzumab as part
of their pre-operative systemic therapy.
IC Y ¼90%
N¼5%
A¼5%
In HER2+/HR+ patients at high risk of relapse (e.g. signicant nodal involvement), 1 year of treatment with neratinib
after trastuzumab can be discussed, as in other age groups.
There is no data about the efcacy of neratinib after 1 year of adjuvant trastuzumab AND pertuzumab or after adjuvant
TDM1.
IC Y ¼80%
A¼20%
General considerations in the adjuvant setting
In view of the long potential life expectancy, particular attention should be paid to possible long-term toxicities of
adjuvant treatments (e.g. secondary cancers, cardiovascular toxicity, irreversible ovarian failure, weight gain, cognitive
function, bone health).
Clinics dedicated to the assessment and management of early and late treatment side-effects and adherence to
treatment and follow-up guidelines should be developed.
Expert opinion
The management of inammatory breast cancer in young women should be the same as in the older breast cancer
population.
Expert opinion
Data are accumulating about disease-free survival improvement with adjuvant bisphosphonates among
premenopausal women under OFS.
Zoledronic acid q6 months may be discussed in young women receiving OFS. Optimal duration of treatment is
uncertain, and risks and benets should be considered on a case-by-case basis.
IA Y ¼57%
N¼17%
A¼26%
In green, NEW/MODIFIED BCY5 guidelines with consensus voting.
A, abstain; AIs, aromatase inhibitors; BC, breast cancer; BCS, breast-conserving surgery; CT, computed tomography; EBC, early breast cancer; ER, estrogen receptor; FDG, [
18
F]2-
uoro-2-deoxy-D-glucose; GnRH, gonadotropin-releasing hormone; GoR, grades of recommendation; HER2, human epidermal growth factor receptor 2; HR, hormone receptor;
LoE, levels of evidence; MRI, magnetic resonance imaging; N, no; OFS, ovarian function suppression; OS, overall survival; PARP, poly (ADP-ribose) polymerase; pCR, pathological
complete response; PET, positron emission tomography; RT, radiation therapy; SNLB, sentinel lymph node biopsy; TDM-1, trastuzumab-emtansine; TNBC, triple-negative breast
cancer; WBI, whole breast irradiation; Y, yes.
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 7
long-term work ability.
34
Given the length of working years
ahead for these women, the BCY5 panel rmly stated that
awareness and referral to appropriate resources should be
available and further research in this setting is needed.
Panel members re-emphasized that many specic issues
in the treatment of young women with breast cancer, in all
settings of the disease, still lack evidence-based standards
and that systematic research into age-specic tumor char-
acteristics is needed which could open the door for more
tailored therapeutic interventions.
Risk factors
Lifestyle-associated risk factors should be discussed with
young women as part of public health initiatives.
35
Data are
inconclusive on whether or not obesity is a risk factor for
breast cancer in premenopausal women and increasing
body mass index (BMI) seems inversely correlated with
breast cancer risk in young women.
36
Increases in the waist-
to-hip ratio, which measures central adiposity, are associ-
ated with increased risk of premenopausal breast cancer. As
obesity is associated with increased risk of many serious
health issues, the BCY5 panel stated that health care
professionals should inform young women about breast
health and other modiable breast cancer risk factors in
general (e.g. maintaining a BMI 25 kg/m
2
, as well as
limiting alcohol consumption, smoking and physical
inactivity).
35
Diagnostic imaging for screening, staging and follow-up
1. Screening: There is no indication for routine screening
by any imaging technique in healthy, average-risk
young women. Surveillance in high-risk women,
based on family history or pathogenic gene variants in
predisposing genes, and for those at increased risk
because of a personal history of therapeutic radiation
in childhood or young adulthood,
37
should follow
available guidelines.
2. Staging and follow-up: The panel reinforced the recom-
mendation that imaging and staging in young women
including axillary assessment should in principle follow
standard algorithms as for older women.
Breast ultrasound remains the rst diagnostic approach for
clinical abnormalities in this age group and in pregnant/
Table 4. Assessment and treatment general guidelines in advanced breast cancer
Guidelines LoE/GoR Consensus
In ABC, age alone is not a reason to prescribe more aggressive therapy and International Consensus Guidelines for
management of advanced breast cancer must be applied (ABC guidelines, NCCN guidelines, evidence-based national
guidelines).
Therapeutic recommendations should not differ from those for older women with the same disease characteristics and
extent.
Expert opinion
IC
The BCY5 panel endorses the ESOeESMO ABC5 guidelines for the management of ABC in premenopausal women. IA 100%
Clinical and pathologic characteristics predicting for CNS recurrence often overlap with factors that indicate increased
risk for general metastatic dissemination (i.e. young age, ER- and PR-negativity, HER2 overexpression, high proliferation
and genomic instability). Although young age has been associated with an increased risk of CNS metastases,
surveillance and therapeutic recommendations should not differ from those for older women with the same disease
characteristics and extent.
IC
Preliminary data suggest BC patients harboring BRCA mutations present a higher incidence of CNS involvement and a
worse outcome compared to non-carriers, irrespective of BC subtype.
No data on the efcacy of PARPi on CNS involvement are available as none of the phase III PARP trials included patients
with active CNS metastases.
In subgroup analyses, talazoparib benet persisted in patients who had stable/treated brain metastases on study entry.
Inclusion of patients with active CNS metastases should be encouraged in all studies investigating new therapies
(unless medically contraindicated)
Expert opinion Y ¼87%
N¼4%
A¼9%
Many trials in HR+ ABC have not included premenopausal women.
Despite this, we recommend that young women with ER+ ABC have adequate ovarian suppression or ablation and then
be treated in the same way as postmenopausal women with endocrine agents targeted therapies.
Future trials exploring new endocrine/endocrine-biological strategies should be designed to allow for enrollment of
both pre- and postmenopausal women and men.
IA
Platinum agents have been demonstrated to be superior to taxanes in BRCA-associated advanced breast cancer. IB
The BCY5 panel endorses all the ABC5 ESMOeESO statements on PARPi for patients with a germline BRCA mutation.
Platinum and PARP inhibitors have not been compared in the advance setting and preferential use of either or optimal
sequencing of these treatments is unknown
IA Y ¼83%
N¼0
A¼17%
Phase II data suggest a benet for PARPi in patients with ABC harboring somatic BRCA1/2 mutations or a germline
PALB2 mutation.
Somatic BRCA1/2 mutations and germline PALB2 mutations are not common; however, tailoring of treatment based on
these alterations may be considered with caution on a case-by-case basis.
This data underscores the importance of molecular tumor boards and of pooling of data in international registries.
IIB
Expert opinion
Y¼87%
N¼0
A¼13%
Very little is known about psychosocial challenges and dying concerns in young parents with ABC. Most of the data
refer to Caucasian, upper-, middle-class women within nuclear families.
In general, patients express concerns for their children and their co-parent, and personal concerns which impact their
QoL contribute to the emotional and psychological distress, and increase family dysfunction.
Further research in this setting is needed on patients from diverse backgrounds, non-nuclear families, on the co-
parent, parents and caregivers.
Expert opinion 100%
In green, NEW/MODIFIED BCY5 guidelines with consensus voting.
A, abstain; BC, breast cancer; BCY5, fth International Consensus Symposium for Breast Cancer in Young Women; CNS, central nervous system; ESMO, European Society of
Medical Oncology; ESO, European School of Oncology; GoR, grades of recommendation; HER2, human epidermal growth factor receptor 2; HR, hormone receptor; LoE, levels of
evidence; N, no; NCCN, National Comprehensive Cancer Network; PARPi, poly (ADP-ribose) polymerase inhibitors; QoL, quality of life; Y, yes.
Annals of Oncology S. Paluch-Shimon et al.
8https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
lactating women.
38
Given the data on diagnostic superiority
of digital breast tomosynthesis (DBT) over digital mammog-
raphy in young women and in those with dense breasts,
39
with only marginal increase in radiation dose, a majority of
the BCY5 panel stated that DBT is the preferred diagnostic
tool.
Pre-operative magnetic resonance imaging (MRI) is asso-
ciated with increased rates of ipsilateral mastectomy and
contralateral prophylactic mastectomy in newly diagnosed
breast cancer patients, irrespective of age
40
: its indications
should strictly follow available recommendations.
41
MRI is
generally superior to other clinical and imaging assessments
after pre-operative chemotherapy.
42
The optimal timing for
carrying out mammography and MRI is the rst half of the
menstrual cycle (days 7-14).
43
Recent data show that
abbreviated MRI (AB-MRI), i.e. the acquisition of only two
sequences, before and immediately after the administration
of gadolinium, has equivalent performances to standard MRI
protocols in diagnostic clinical settings.
44
Although validation
with prospective multicenter trials is pending, almost half of
the BCY5 panelists agreed that AB-MRI may represent a valid
alternative to conventional MRI for persons with dense
breasts with the advantages of short examination/interpre-
tation times and low costs.
The panel re-conrmed that in average-risk patients,
imaging surveillance after primary breast cancer treatment
should follow the same guidelines as in older women.
Genetic counseling and testing
The panel conrmed that genetic counseling should be
offered to every young woman, irrespective of tumor subtype
[e.g. triple-negative (TN) disease] or family history of breast
cancer as studies have reported that if testing is carried out
based on traditional testing guidelines (largely guided by
personal and family history of malignancy), close to 50% of
persons with a germline mutation would not be identied.
45
A fast-track process, which enables testing before
commencement of therapy, should be available when the
identication of a pathogenic gene variant could change the
therapeutic approach [e.g. indication for risk-reducing sur-
gery, platinum derivatives, poly (ADP-ribose) polymerase
(PARP) inhibitors (PARPi)]. Although BRCA1/2 genes are the
most frequently mutated genes, testing for other additional
moderate- to high-penetrance genes using a multi-gene
panel may be considered, if they will impact therapeutic in-
terventions. In a recent study, genes most commonly asso-
ciated with breast cancer in woman aged 45 years were
BRCA, BRCA2, ATM, CHEK2 and PALB2
46
due to prevalence
and impact. The clinical utility (including risk assessment,
screening and prevention recommendations) of moderate-
risk genes identied on multi-gene panel testing and poly-
genic risk score models are not yet established or ready for
clinical practice.
Multidisciplinary management of mutation carriers and
high-risk individuals should ideally be provided in dedicated
high-risk clinics, when available. Clinical trials focusing on
risk reduction and optimal screening strategies for this
group of women are strongly needed. Unaffected carriers
should be encouraged to participate in clinical trials evalu-
ating risk-reducing strategies such as the BRCA-P study
evaluating denosumab in women harboring a PV in BRCA1
who have not undergone risk-reducing mastectomy
(NCT04711109). With the recent shift to oncologist-led
mainstreamtesting where the surgeon or oncologist refers
for testing, and counseling and interpretation are provided
following test results,
47,48
education and training of the
involved health care professionals is needed to provide
optimal risk communication and clinical recommendations.
For women who are not ready to consider genetic testing
at the time of diagnosis, access to genetic counseling should
Table 5. Additional considerations in women with hereditary-associated breast cancer
Guidelines LoE/GoR Consensus
For survivors harboring a BRCA 1/2 or (other) strongly predisposing mutation, bilateral risk-reducing mastectomy may
be considered, although there is no denite evidence that it leads to a survival benet. Therapeutic decisions should
reect a balance between the risk of recurrence of the diagnosed breast cancer and the potential benet of preventing
an additional primary tumor.
IIB
For the time being, the radiotherapy treatment of EBC is independent of BRCA or any other constitutional genetic
status, with the exception of germline TP53 and ATM mutations, for which a very high risk of secondary cancers has
been described after radiation therapy.
Radiation therapy should be carefully discussed on an individual basis for these patients.
IB
IIC
In the absence of evidence-based recommendations for risk-reducing surgery in patients harboring pathogenic variants
in lowemoderate-penetrance genes, decisions must be taken individually, mainly based on family history.
For breast cancer survivors and asymptomatic carriers harboring a BRCA 1/2 mutation, risk-reducing salpingo-
oophorectomy (RRSO) should be discussed from the age of 35 years provided that the woman has completed family
planning. For BRCA1 mutation carriers RRSO is recommended between age 35 and 40 years and for BRCA2 mutation
carriers around age 40 years, always respecting patients preferences and considering the family history.
Indications and timing of risk-reducing salpingo-oophorectomy for other highly penetrant mutations should follow
available international/national guidelines.
Expert opinion
Salpingectomy with delayed oophorectomy remains investigational and should preferably be carried out only within a
clinical trial.
Expert opinion Y ¼85%
N¼10%
A¼5%
Currently, the evidence on the reduction of BC risk and mortality by oophorectomy is conicting and likely limited to
BRCA1 carriers.
Expert opinion Y ¼53%
N¼17%
A¼30%
In green, NEW/MODIFIED BCY5 guidelines with consensus voting.
A, abstain; BC, breast cancer; EBC, early breast cancer; GoR, grades of recommendation; LoE, levels of evidence; N, no; y, yes.
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 9
Table 6. Supportive and follow-up care guidelines
Guidelines LoE/GoR Consensus
Young women with breast cancer face specic physical, psychosocial and sexual issues that should be addressed by a
multidisciplinary group of providers including breast medical, surgical and radiation oncologists, breast care nurses,
social workers, psycho-oncologists, gynecologists and fertility experts, among others.
Expert opinion
Young women with breast cancer are at higher risk for psychosocial distress. Patientsdistress and psychosocial needs
should be regularly assessed.
Psychosocial care should be available and integrated in routine cancer treatments and follow-up.
Partners and family members should be involved early on and couple-based psychosocial interventions should be
promptly proposed if needed.
IIB
The specic psychosocial distress pertaining to body image, sexuality and sexual dysfunction resulting from premature
menopause, treatment-related amenorrhea, weight gain, hair loss and breast surgery should be routinely addressed by
the health care team in order to ensure provision of appropriate information and support.
This needs to be carried out in a culturally appropriate manner.
Expert opinion 100%
All young women should be counseled regarding the risk of getting pregnant while on chemotherapy, endocrine or
anti-HER2 therapy, despite developing amenorrhea, and of the need for adequate non-hormonal contraception if they
are sexually active and could become pregnant.
Exogenous hormonal contraception is generally contraindicated in young cancer survivors, irrespective of disease
subtype, and alternative strategies should be considered.
IB
Expert opinion
All young women (irrespective of stage of disease) should be informed about approved fertility preservation options
and referred for specialist counseling/consultation if interested in fertility preservation before commencement of any
therapy.
Women should be informed that currently available methods of fertility preservation generally allow for timely start of
chemotherapy schedules.
The BCY5 panel endorses the ABC guidelines statement that the impact of the anticancer therapies on fertility should
be discussed with all women with ABC of childbearing age and their partners, before the start of treatment.
The discussion must also include appropriate information about the prognosis of the disease and the potential
consequences of pregnancy (e.g. stopping ongoing treatment).
Expert opinion Y ¼96%
A¼4%
Physiciansknowledge and familiarity with the available international guidelines on fertility preservation, pregnancy
after BC and management of BC during pregnancy should be increased in order to facilitate and improve evidence-
based individualized management of young patients with BC.
Expert opinion Y ¼96%
A¼4%
The use of GnRH analog concomitant with (neo)adjuvant CT should be offered to reduce the risk of premature ovarian
failure, possibly preserve ovarian function and reduce damage to fertility.
Concomitant GnRHa use during chemotherapy does not replace established fertility preservation methods, which
should still be offered to all young patients.
The effectiveness of GnRHa does not seem to depend on the time of administration in relation to commencement of
chemotherapy.
IB Y ¼70%
N¼20%
A¼10%
Biomarkers such as AMH levels may predict ovarian function after chemotherapy though data are limited. Expert opinion Y ¼85%
A¼15%
All young women should be counseled about the risks and associated symptoms and outcomes and management of
treatment-related amenorrhea and premature menopause before the onset of systemic therapy (either CT or ET) and
informed of available ameliorative therapies.
Expert opinion
Premature menopause and/or treatment-related amenorrhea increase the risk of bone thinning and patients should be
counseled, monitored and treated accordingly.
IA
Pregnancy after breast cancer should not be discouraged even in patients with HR-positive disease or those harboring
a germline BRCA mutations. While pregnancy itself does not appear to increase the risk of recurrence the discussion
about pregnancy should take into account the patients prognosis based on initial stage and biology.
Pending results of prospective clinical trials, patients with HR+ disease who are unwilling to wait till the end of
adjuvant ET should complete at least 18-24 months of ET before attempting pregnancy. Treatment should be resumed
and completed according to initial planning after delivery and breast-feeding.
IIA
Expert opinion
Y¼95%
A¼5%
Y¼95%
A¼5%
There are limited data on breast-feeding after BC. Lactation can be carried out from the unaffected breast and data
suggest that lactation from the previously affected breast may sometimes be feasible, depending on the type of
surgery and radiation. Women should be counseled at the time of receipt of radiation therapy that they most likely will
not be able to breast-feed from the irradiated breast.
Young BC survivors who pursue a pregnancy and are interested in breast-feeding should have access to lactation
counseling by trained health professionals.
Expert opinion Y ¼96%
A¼4%
Breast cancer during pregnancy
Treatment of patients with breast cancer during pregnancy should be decided on an individual basis according to
international guidelines within an expert multidisciplinary team, expanded to include obstetricians and perinatologists,
and according to patientspreferences.
Whenever possible, women with a diagnosis of pregnancy during breast cancer should be managed by a
multidisciplinary teams with expertise in this eld.
Expert opinion
Y¼91%
N¼4%
A¼5%
Pregnancy termination has not been shown to improve patient prognosis and should not be promoted for such reason. IVA 100%
In principle, standard chemotherapy can be offered to most patients, based on the gestational age, tumor stage and
biology.
IIB 100%
Exposure to ET, bone-modifying agents and anti-HER2 therapies should be avoided during pregnancy. If treatment
cannot be delayed until delivery (e.g. in case of ABC), treatment choices should be carefully discussed with the patient
to ensure maternal benet and reduce fetal risk.
IIB Y ¼91%
A¼9%
Patients diagnosed in the few years after pregnancy have worse prognosis. Further research is warranted to better
understand their biology and dene treatment strategies accordingly.
Expert opinion Y ¼85%
A¼15%
Continued
Annals of Oncology S. Paluch-Shimon et al.
10 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
be offered on an ongoing basis, and women with previous
limited testing should be considered for a more compre-
hensive contemporary panel in survivorship. Strategies to
minimize recently documented racial/ethnic and social dis-
parities in early access to genetic testing and risk manage-
ment should be implemented to optimize risk-reducing
interventions.
49
Research is needed to assess the psycho-
social factors affecting the communication of genetic test
results by young breast cancer patients with parents and
siblings, especially in non-Caucasian women.
Screening for other malignancies and risk-reducing surgery
recommendations for women harboring a pathogenic
gene variant
In patients harboring a pathogenic germline variant in TP53
(LieFraumeni syndrome, LFS), the use of ionizing radiation
should be discouraged to avoid increasing the riskof secondary
radio-induced malignancies.
50
Annual brain and whole body
MRI (WB-MRI) is recommended as contrast-free WB-MRI has
shown to be effective for cancer detection in asymptomatic
carriers and during follow-up in breast cancer patients.
51,52
[
18
F]
2-uoro-2-deoxy-D-glucoseepositron emission tomographye
computed tomography (FDGePETeCT) scan has also been
proven to be effective in cancer screening of patients with LFS,
but the availability of safer modalities (e.g. contrast-free WB-
MRI) has limited its incorporation in surveillance protocols.
53
For those with a BRCA1/2 mutation and other breast cancer-
associated susceptibility genes, timing of risk-reducing salpingo-
oophorectomy (RRSO) and gynecologic surveillance should
follow international guidelines. Ongoing trials will possibly help
clarify the role for salpingectomy with delayed oophorectomy
in women wishing to delay RRSO and early onset of menopause
(ClinicalTrials.gov Identier: NCT02321228, NCT01907789,
NCT01608074). For now salpingectomy with delayed oopho-
rectomy remains investigational.
Transgender and nonbinary persons
Knowledge about the risks of breast/gynecological cancers in
transgender (TG) and nonbinary (NB) persons receiving
gender-afrming hormone therapy is limited.
54
Even less in-
formation exists regarding TG/NB individuals with known ge-
netic predisposition. Cancer registries should include TG/NB
identities, and further education and research is needed to ll
the gap in information and counseling in this population.
EARLY BREAST CANCER
Locoregional treatment
Surgery. Although young age remains an independent risk
factor for increased locoregional recurrence, irrespective of
the type of surgery carried out, especially in patients with
human epidermal growth factor receptor 2-positive
(HER2þ) and TN disease, a decreasing trend in locore-
gional recurrence is reported across continents. The panel
remains concerned about the expanding international trend
for bilateral mastectomies, particularly in younger women
55
despite no improvement in long-term survival.
56
Commu-
nication strategies to optimize decision making for surgical
treatment of breast cancer should be implemented to
encourage appropriate breast-conserving surgery (BCS).
Decision aid tools focused on the needs of young, average-
risk breast cancer patients are being developed and evalu-
ated and should become a research priority.
57
Oncoplastic surgical techniques should be discussed with
all patients scheduled for BCS if post-operative asymmetry
is anticipated and should always be carried out by a dedi-
cated breast surgical team. When mastectomy is indicated,
skin- and nipple-sparing techniques with immediate breast
reconstruction (IBR) can provide oncological control while
also addressing cosmetic needs.
58
IBR (irrespective of
technique) following mastectomy offers the same survival
outcome rates as mastectomy without reconstruction
59
and
should be offered to all patients except those with inam-
matory breast cancer for whom delayed reconstruction is
recommended (given the need for extensive radiation and
the recommendation to wait until the highest relapse risk
passes) and those with locally advanced disease at pre-
sentation with poor response to primary systemic therapy.
The increasing use of pre-operative therapy for stage 2-3
Table 6. Continued
Other issues
Young patients should be strongly encouraged to adopt the following healthy lifestyle changes:
maintain BMI 25
perform regular aerobic exercise
not to smoke
to limit daily alcohol intake
Expert opinion
Young BC survivors experience signicant job- and insurance-related issues, and potential nancial toxicity following
diagnosis.
Awareness and referral to appropriate resources should be available for all young patients.
Further research in this setting is needed in order to plan targeted interventions and avoid unnecessary difculties.
Expert opinion
Expert opinion
Y¼96%
A¼4%
Y¼96%
A¼4%
Many young BC patients use integrative medicine (complementary and alternative therapies). Health professionals
should proactively promote open communication about integrative medicine with their young BC patients.
To ensure patient safety and quality of treatments, when possible, supervised and established integrative medicine
services should be provided within the oncology service at the treating institution.
Expert opinion
Expert opinion
Y¼91%
N¼9%
Y¼78%
A¼17%
N¼5%
In green, NEW/MODIFIED BCY5 guidelines with consensus voting.
A, abstain; ABC, advanced breast cancer; AMH, anti-Müllerian hormone; BC, breast cancer; BCY5, fth International Consensus Symposium for Breast Cancer in Young Women;
BMI, body mass index; CT, computed tomography; ET, endocrine therapy; GnRHa, gonadotropin-releasing hormone agonist; GoR, grades of recommendation; HER2, human
epidermal growth factor receptor 2; HR, hormone receptor; LoE, levels of evidence; N, no; Y, yes.
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 11
disease particularly in the more aggressive subtypes has
reduced past concerns that wound healing complications
from IBR will delay any necessary systemic therapy. Higher
SES is associated with higher IBR rates among both young
and older women, including in countries with universal
health care.
60,61
IBR is rarely available for patients from
LMICs.
62
The BCY5 panel unanimously stated that every
young breast cancer patient should have access to IBR and
oncoplastic surgery if medically appropriate.
The panel conrmed that the indications for sentinel lymph
node biopsy and surgical management of patients with
sentinel lymph node involvement should be the same as in
older patients and that, despite the fact that optimal locore-
gional treatment after pre-operative chemotherapy remains
controversial, decisions should be made irrespective of age.
Germline mutation status should be part of the individual
decision-making algorithm when young women are making
breast surgery choices. In the absence of evidence-based
recommendations for risk-reducing surgery in patients
with PVs in lowemoderate-penetrance genes, decisions
must be tailored to the individual, guided by family history
and patient preference.
Radiation therapy. Indications for post-operative RT are the
same as for older patients
63
; however, data are more robust
for benets of post-mastectomy radiation therapy (PMRT)
amongst young women. PMRT after implant-based breast
reconstruction is associated with higher risk of reconstruc-
tive failure (>15%) and capsular contracture (>30%). Rates
of capsular contracture are lower with PMRT with tissue
expanders, but reconstructive failure is more common.
64
These complications may translate into poor aesthetic
outcomes, decreased satisfaction and lower QoL. Given the
lack of denitive evidence for optimal reconstructive algo-
rithms and techniques (implant-based or autologous tissue
reconstructions), shared decision making with the patient
and optimal integration of plastic surgery and radiation
oncology are crucial.
65
When PMRT is foreseen, the panel
recommended that timing and technique of the recon-
structive procedure should be discussed pre-operatively on
an individual basis by all the specialists involved. PMRT is
not a stand-alone reason to postpone reconstruction.
Indications and extent of nodal irradiation are the same as
in other age groups.
63
Modern techniques to minimize long-
term side-effects are necessitated. The use of respiratory
control is the most common approach to reduce the dose to
heart and lungs, offering a more favorable irradiation ge-
ometry by inating the lungs and increasing the distance
between the heart and the chest wall.
66
A boost to the site
of the radical local excision in case of BCS remains standard
pending the results of modern randomized trials (e.g. the
Young Boost Phase III trialdNCT00212121).
The BCY5 panel afrmed that moderately hypofractio-
nated whole breast irradiation (WBI) schedules should
replace standard fractionated WBI as gold standard for most
patients. Ultra-hypofractionated schedules were either not
tested in premenopausal women, such as in the FAST trial
(once-weekly ve fractions),
67
or not mature yet for long-
term adverse effects as in the FAST-Forward Trial (ve frac-
tions in 1 week with 15% of premenopausal patients
enrolled).
68
The BCY5 panel therefore recommended against
ultra-hypofractionated WBI or for irradiation of the lymph
node regions (such as in FAST-Forward) for young patients.
As partial breast irradiation (PBI), or accelerated PBI, has
not been sufciently studied in young patients,
63
the panel
recommended against its use outside of clinical trials.
Indications of post-operative RT are independent of BRCA
status. Prophylactic radiation to the unaffected contralat-
eral breast in BRCA carriers who decline contralateral
mastectomy should not be carried out outside of clinical
trials.
69
Although a recent study demonstrated fewer ipsi-
lateral breast recurrences amongst BRCA1/2 carriers who
had undergone PMRT compared to those without PMRT,
70
further data are needed. There is limited evidence about
the safety of radiation for those harboring a moderate-
penetrance pathogenic gene variant (e.g. CHEK2, ATM)
71
and the riskebenet ratio needs to be individually dis-
cussed, but there is no clear contraindication to RT. PMRT
may be discussed in cases of signicant risk of locoregional
recurrence in patients with a germline TP53 mutation, for
whom radiotherapy is otherwise contraindicated to the high
risk of secondary malignancies. These recommendations
underscore the importance of early genetic testing at the
time of diagnosis to aid optimal treatment planning.
Adjuvant systemic treatment
Adjuvant systemic treatment decisions for invasive breast
cancer should be based on the extent of disease and bio-
logical characteristics of the tumor (including, but not
limited to, tumor size, nodal status, hormone receptor and
HER2 overexpression/amplication, Ki67, proliferation and
grade), patients comorbidities and preferences similar to as
in older women.
Gene expression signatures
Although gene expression signatures, such as Oncotype Dx,
MammaPrint, Prosigna, Endopredict and Breast Cancer In-
dex, provide additional information on an individuals
recurrence risk and some signatures have demonstrated
clinical utility for adjuvant chemotherapy decision mak-
ing,
72-74
women younger than 40 are grossly under-
represented in both retrospective and prospective studies.
Additionally, most of the premenopausal patients in these
studies did not receive contemporary risk-adapted ET.
In TAILORx, patients with a low-risk recurrence score (RS),
dened as RS 0-10 (30% of whom were premenopausal but
only 4% aged <40 years), had a 5-year distant recurrence-
free survival of 99%
72
with ET alone. For those with an
intermediate-risk RS (RS 11-25), the 9-year distant
recurrence-free survival was 94.5% for the ET-only group
and 95% for those who received chemotherapy and ET.
73
Exploratory, unplanned subgroup analyses of women aged
50 years suggested a benet from chemotherapy amongst
those with an intermediate RS within the range of 16-25
73
and further analyses suggested that clinical risk level
Annals of Oncology S. Paluch-Shimon et al.
12 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
combined with RS identied women aged 50 years to be
more likely to benet from the addition of chemotherapy to
ET alone.
75
Only 13% of premenopausal women received
ovarian function suppression (OFS).
The RxPONDER trial randomized patients with lowein-
termediate-risk RS (0-25) with 1-3 lymph nodes to either ET
alone or chemotherapy plus ET: 33% of participants were
premenopausal, 2.9% were <40 years of age and 21.5%
were aged 40-49 years. While no benet was demonstrated
in the postmenopausal population with the addition of
chemotherapy, in the premenopausal women an absolute
benet of 5.2% favoring chemotherapy was demonstrated
both for low and intermediate RS, with an invasive disease-
free survival (iDFS) of 94.2% versus 89% [hazard ratio (HR)
0.54, 95% condence interval (CI) 0.38-0.76].
76
In the ET-
only arm, 16% received OFS compared to only 3% in the
chemotherapy arm.
In the WGS ADAPT ERþ/HER2study, all patients with
0-3 involved lymph nodes and a low RS of 0-11 received ET
alone [mostly tamoxifen in pre- and aromatase inhibitor
(AI) in postmenopausal patients]. Those with intermediate
RS (12-25) were given 3 weeks of ET before surgery. Pa-
tients with a Ki67 10% in the surgical specimen were
considered endocrine responders and received ET alone (of
whom 23.3% were aged 50 years), while those with a
Ki67 >10% were classied as endocrine non-responsive
and received chemotherapy in addition to ET (of whom
64.7% were aged 50 years). Amongst endocrine re-
sponders with an RS of 12-25 and limited nodal burden (up
to two nodes), despite no data in women <40 years of age,
there was no difference in outcome between the low and
intermediate RS groups, with a 5-year distant DFS (DDFS) of
96.8% and 97.4%, respectively, in patients aged 50 years.
Outcome was also similar to that of patients aged >50
years with 0-3 nodes and RS 0-25 who received ET alone.
77
Derived from the ADAPT data, the ENREP algorithm
(https://enrep.info) can help to estimate endocrine
responsiveness based on clinical and immunohistochemical
factors.
The MINDACT trial assessed all patients for recurrence
risk by both clinicalepathological factors (clinical risk) and
MammaPrint (genomic risk) and assigned those with clinical
low/genomic low to ET alone. Patients with high clinical/
high genomic risk were assigned to CT followed by ET. Pa-
tients with discordant risk proles underwent randomiza-
tion to determine use of chemotherapy.
74
Only 6.2% of the
study population was aged <40 years.
74
An exploratory
analysis demonstrated a 5% benet in distant metastases-
free survival favoring the addition of chemotherapy in
women 50 years of age who were clinically high risk,
genomic low risk
78
but it is unclear if these results indicate
true benet from chemotherapy or a chemoendocrine ef-
fect given among the 50-year-old patients who did not
receive chemotherapy, 55% received tamoxifen alone for 5
years, and only 20% received OFS.
79
Favorable outcomes were seen in the SOFT and TEXT
studies and the Austrian Breast and Colorectal Cancer
Study Group (ABCSG) 12 trial, among premenopausal
patients with low-risk disease who did not receive
chemotherapy.
80,81
In SOFTeTEXTdamongst women aged
<35 years with HER2disease only 57 women (out of
442) did not receive chemotherapyd94% were node
negative, 84% had T1 and 23% grade 1 tumors. In this
group, 14% had an early invasive breast cancer event
(including three distant recurrences and one death) at 6-
year and 5.6-year follow-up in TEXT and SOFT, respec-
tively.
82
Thus, omitting adjuvant chemotherapy in young
and very young women (35 years old at diagnosis) may
be appropriate in selected cases with favorable clinical,
genomic and pathological features.
Pharmaco-prevention
Five years of tamoxifen 20 mg daily reduces breast cancer
risk by 35%-40% and is recommended as pharmaco-
prevention for women with an elevated breast cancer
risk.
83
Despite the riskebenet ratio being in favor of
tamoxifen in all women <50 years of age, irrespective of
the degree of risk, the uptake of tamoxifen for breast cancer
prevention is low (10%-12%).
84
Higher-risk premenopausal
women appear more likely to accept tamoxifen,
84
with the
main reasons for non-initiation being concerns about side-
effects, tamoxifen being perceived as a cancer drug, min-
imal benet and fertility concerns.
84,85
Low-dose tamoxifen
(5 mg/day for 3 years) may be an alternative to full-dose
tamoxifen in women with breast intraepithelial neoplasia
given its efcacy and limited toxicity.
86,87
The BCY5 panel
recommends discussing pharmaco-prevention with young
women at high risk of developing breast cancer.
Pre-operative endocrine therapy
There are no new data regarding pre-operative ET in young
women since BCY4. The BCY5 panel agreed with the pub-
lished American Society of Clinical Oncology (ASCO) guide-
lines
88
that pre-operative ET should not be routinely
recommended for young women outside of clinical trials.
Nevertheless, a short pre-operative ET (2-4 weeks) with
subsequent Ki67 determination in the surgical specimen for
assessing endocrine responsiveness may help adjuvant
treatment decision making as demonstrated by the ADAPT
trial also for young women.
89
Trials evaluating the efcacy
of cyclin-dependent kinase (CDK) 4/6 inhibitors plus ET in
the pre-operative setting are ongoing.
Adjuvant endocrine therapy
Based on the updated results of the SOFT and TEXT
studies
80
and in line with existing guidelines,
16,90,91
BCY5
conrmed that tamoxifen alone remains the standard of
care in premenopausal women at low risk of relapse, as
dened by clinical and immunohistochemical parameters,
who did not receive adjuvant chemotherapy. More than
97% of these women are free of distant recurrence and
alive at 8 years, with no additional benet by escalating ET
to OFS plus tamoxifen or exemestane.
In women at higher risk of relapse, adding OFS to ET is
associated with a signicant improvement in outcomes
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 13
including distant recurrence compared to tamoxifen
alone.
80
In high-risk patients, the combination of OFS and
an AI should be the preferred option and OFS and tamox-
ifen for those with toxicity to the AI. An overall survival (OS)
benet was evident in the SOFT study amongst patients
who received adjuvant chemotherapy followed by OFS plus
oral ET; however, this was not yet demonstrated in the TEXT
study. Given that the risk of disease recurrence continues
for >20 years,
92
long-term follow-up will be key to ascertain
if the positive effects on DFS will translate into improve-
ments in OS and also to dene potential longer-term safety
issues (e.g. second primaries), particularly relevant in young
patients.
An online tool (https://rconnect.dfci.harvard.edu/Compo
siteRiskSTEPP/) has been developed for clinicians to use in
daily practice to estimate individual risk-based benetof
escalating ET for women with HER2disease. This is a
composite measure of the distant recurrence risk according
to traditional prognostic features (i.e. patient age, tumor
size, grade, lymph node status, ER, progesterone receptor
and Ki67 expression) derived from the SOFTeTEXT popu-
lation.
93
Although the relative efcacy of escalating ET is
independent of age, women aged <35 years have the
largest magnitude of absolute improvement in outcomes
with OFS.
80,82
OFS timing (concurrent versus sequential) in
women receiving adjuvant chemotherapy does not impact
breast cancer outcomes. Notably, in women <40 years old
who are less likely to develop CIA,
94,95
gonadotropin-
releasing hormone agonist (GnRHa) concomitant with
chemotherapy has the added benet of ovarian function
protection.
96
GnRHa in combination with tamoxifen or an AI should be
prescribed for 5 years based on the SOFT/TEXT data. A
shorter duration was associated with excellent mid- and
long-term outcomes in women with lower or intermediate
risk who did not necessarily receive chemotherapy (ABCSG-
12 in which only 5% received chemotherapy and the AST-
TRA trials).
81,97
Adding OFS to tamoxifen signicantly im-
proves the 5-year DFS, compared to tamoxifen alone,
including in women with late (within 2 years) resumption of
ovarian function after chemotherapy,
97
suggesting that
ovarian function should be monitored long term. Switching
from 2-3 years of tamoxifen to Ais plus goserelin for a total
treatment duration of 5 years versus continuing tamoxifen
alone was associated with more adverse events but no
improvement in disease outcomes in a small phase II trial
with short follow-up.
98
Extending tamoxifen beyond 5 years should be considered
in high-risk patients
99,100
as the riskof recurrence continues for
>20 years.
92
The impact ofextended OFS and tamoxifen or an
AI beyond 5 years is unknown.
OFS always needs to be given in young women with
otherwise intact ovarian function who receive an AI. AIs
alone are contraindicated in premenopausal women:
caution must be taken when considering an AI in premen-
opausal women who became amenorrheic during the
course of treatment due to the potential for recovery of
ovarian function.
101,102
The criteria for dening menopausal
status following CIA are dened in the BCY2 paper
14
and
reported in Supplementary Appendix S1, available at
https://doi.org/10.1016/j.annonc.2022.07.007.
BCY5 conrmed that hormone levels should be checked
under GnRH therapy if there are concerns that ovarian
function is not suppressed, especially if breakthrough
bleeding occurs and/or the patient is on an AI. A gas
chromatography/mass spectroscopy method, if available, is
preferred to monitor therapy.
103,104
The updated results of
the SOFT-EST sub-study, at over 4 years of treatment, were
consistent with the rst-year results showing that OFS does
not achieve optimal estrogen suppression in up to 17% of
patients.
105
Based on the limited available data
106-108
and concerns
about suboptimal OFS with depot formulations, monthly
formulations of GnRHa are preferred,
90
especially in women
<35 years of age and in those receiving an AI. However,
when monthly use is not feasible or accepted by the pa-
tient, 3-6 monthly administration may be considered on a
case-by-case basis with close monitoring of ovarian
function.
107
The method of ovarian suppression (surgical versus
medical) requires balancing patients wish for potentially
preserving fertility and compliance with frequent injections
over a long period of time and cost/availability. The BCY5
panel was divided when discussing radiation to ovaries as a
method of OFS: a narrow majority voted that it should be
discouraged if alternatives exist. New RT techniques better
delineate the position of the ovaries, thus improving
effectiveness and minimizing the adverse events.
109
Younger age is associated with lower adherence and
persistence to adjuvant ET,
82,110
which has been associated
with reduced OS.
111
Determinants of treatment persis-
tence
112
may vary according to race and ethnicity
113,114
:
potentially modiable factors should be identied with
targeted interventions.
115,116
Three recently reported neo/adjuvant trials investigating
the impact of adding the CDK4/6 inhibitors abemaciclib
117
and palbociclib
118,119
to ET have provided mixed results.
Almost 50% of enrolled patients were premenopausal in
each trial. In the PALLAS adjuvant trial, 3-year iDFS did not
differ between the two arms (88.2% with palbociclib and
88.5% with ET). Similarly, in PENELOPE-B, at a median
follow-up of 42.8 months, palbociclib did not improve the 3-
year iDFS in women with residual invasive disease after pre-
operative chemotherapy (81.2% with palbociclib, 77.7%
with placebo). However, the monarchE study which added
abemaciclib to standard ET demonstrated an absolute 3.5%
improvement in the 2-year iDFS (92.2% versus 88.7%), and
5.4% at 3 years.
117,120
There are several possible reasons for
different ndings including the patient populations, rates of
discontinuation and the specic CDK4/6 inhibitor. Longer
follow-up and the results of ongoing trials (NATALEE triald
NCT03701334; ADAPTcycle trialdNCT4055493; ADAPTlate
trialdNCT4565054) may provide more data but the BCY5
panel stated that abemaciclib could be considered in high-
risk patients similar to those enrolled in the monarchE
study (4 involved nodes, or 1-3 involved nodes with other
Annals of Oncology S. Paluch-Shimon et al.
14 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
high-risk features, i.e. tumor size 5 cm, grade 3, Ki67
20%).
GnRH agonists and ovarian function preservation
In line with the most recent guidelines focused on fertility
after cancer,
121,122
BCY5 conrmed that the use of GnRHa
concomitant with (neo)-adjuvant chemotherapy should
be offered to all patients to reduce the risk of premature
ovarian insufciency and possibly preserve ovarian func-
tion. It is important to note that there are no clinical data
whether use of a GnRHa for the purpose of ovarian
function suppression is necessary or recommended dur-
ing use of post-neoadjuvant capecitabine, olaparib and
immunotherapy (noteworthy, in the CREATE-X study
evaluating post-neoadjuvant capecitabine, over half of
the patients were premenopausal; however, no data are
available about GnRHa use). The recently reported results
from the OPTION trial
123
suggest that patients experience
a short-term decrease in QoL from the addition of
goserelin to chemotherapy to preserve ovarian function,
a price that women may decide to pay if adequately
informed.
Because fertility outcome data after temporary OFS
during chemotherapy are still insufcient,
96
the BCY5 panel
conrmed that GnRHa use during chemotherapy does not
replace established fertility preservation methods, which
should be offered to all young patients interested in sub-
sequent pregnancies. The efcacy of GnRHa in preserving
the ovarian reserve seems unrelated to the timing of
administration before chemotherapy,
124,125
but scheduling
after oocyte cryopreservation should be coordinated be-
tween the fertility and oncology teams to avoid a potential
ovarian hyperstimulation syndrome.
126
Anti-Müllerian hor-
mone may predict ovarian function recovery after GnRHa
during chemotherapy.
124,127-129
Neo/adjuvant chemotherapy
Proportional risk reductions with taxane- and/or
anthracycline-based adjuvant regimens are not signicantly
affected by age
130
: since no studies have specically
investigated different chemotherapy regimens/scheduling
in young women previous statements remain valid. At the
time of BCY5, the majority of the panel did not support
routine use of non-anthracycline-based regimens. While
anthracyclines have long been the backbone of neo/adju-
vant chemotherapy, they carry long-term risks of cardiac
failure and leukemia/myelodysplastic syndromes. In light of
these risks, numerous studies have evaluated non-
anthracycline-based regimens and there is now a growing
body of evidence supporting non-anthracycline-based regi-
mens for endocrine-responsive tumors with a low tumor
burden that require chemotherapy and in HER2 over-
expressing tumors.
131,132
The recent shift in practice and
the emergent data will be formally discussed and voted
upon in a consensus statement at BCY6.
The question of whether to incorporate platinum agents
in the pre-operative setting for TN- or BRCA-associated
tumors remains unresolved.
16,133
A recent study comparing
Acx4 to 4 cycles of cisplatin in the neoadjuvant setting for
BRCA-associated triple-negative breast cancer (TNBC)
demonstrated that Doxorubicin and cyclophosphamide was
superior to the 4 cisplatin cycles for achieving pathological
complete response (pCR).
134
The BRIGHTNESS neoadjuvant
study reported that pCR and event-free survival (EFS) were
inferior for paclitaxel monotherapy compared to either
carboplatin/paclitaxel or carboplatin/paclitaxel/veliparib, all
followed by 4 cycles of cyclophosphamide and doxorubicin
in TNBC.
135
While platinum agents may be considered in
selected patients, risk of additional gonado-toxicity should
be considered. There are still no data on the use of platinum
agents in the adjuvant setting. For patients with TN disease
without a pCR after standard pre-operative regimens,
addition of 6-8 cycles of capecitabine may be considered, as
in other age groups.
136
Recent data demonstrated that
substituting a platinum for capecitabine in this setting did
not improve outcomes.
137
The phase III KEYNOTE 522 study evaluated the incor-
poration of immunotherapy in the pre-operative setting.
Pembrolizumab with chemotherapy was compared to pla-
cebo with chemotherapy followed by a year of pem-
brolizumab or placebo, respectively. In the most recent
update, a benet is seen for pCR (64.8% versus 51.2%, P¼
0.00055) and for EFS [91.3% versus 85.3%, HR 0.63 (95% CI
0.43-0.93)] favoring the pembrolizumab arm.
138
An impor-
tant consideration is immune-related endocrinopathies,
some of which are irreversible, and the impact on female
fertility and ovarian function recovery is for the time being
unknown. The IMPASSION-031 randomized patients to pre-
operative chemotherapy with or without atezolizumab fol-
lowed by a year of atezolizumab or placebo. pCR was
superior for the atezolizumab arm, 58% versus 41%
(P¼0.0044),
139
but outcome data are awaited. Gepar-
Nuevo, a phase II study with 174 patients, evaluated
incorporation of durvalumab in the pre-operative setting
alone, and demonstrated superior pCR, iDFS, DDFS and OS
favoring the durvalumab-containing arm.
140
The incorpo-
ration of pembrolizumab in this setting may be considered
in young women.
Adjuvant anti-HER2 therapy
The benet of adjuvant trastuzumab appears independent
of age
141
and anti-HER2 therapies should be the same as for
other age groups. No additional data from randomized
studies were published after BCY4; therefore, all recom-
mendations remain unchanged.
Adjuvant bisphosphonates
As new data on DFS improvement with adjuvant
bisphosphonates among premenopausal women emerged
after BCY4,
142
the panel stated that zoledronic acid q6
months may be discussed in young women receiving OFS
but given the limited data on long-term outcomes,
143
risks
and benets should be balanced on a case-by-case basis.
Optimal treatment duration and dosing interval are
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 15
uncertain and alternative schedules are under investiga-
tion.
144
As a result of the long half-life of bisphosphonates,
caution is needed in women interested in future concep-
tion, given the increased rates of neonatal complications
and spontaneous abortions in a recent case-control study
on pregnant women.
145
There are no data for benet from
adjuvant denosumab in young women.
Side-effects of adjuvant therapy
In view of the long life expectancy of young women, the
panel reinforced the need to monitor potential long-term
toxicities (i.e. cardiovascular, bone morbidity, cognitive
impairment, secondary malignancies).
INFLAMMATORY BREAST CANCER
Inammatory breast cancer should be managed the same as
for the older breast cancer population.
Advanced breast cancer
The BCY5 panel endorses the ESOeESMO ABC5 guidelines
for the management of advanced breast cancer (ABC)
146
and reiterated that young age alone should not be a
reason to prescribe more aggressive therapy including
combination chemotherapy over sequential use of
monotherapy. BCY5 panelists endorse the ABC5 statements
that (i) young women with ERþABC should have adequate
OFS/ovarian function ablation and be treated as post-
menopausal women, with endocrine agents, with or
without targeted therapies and (ii) that future trials
exploring new endocrine-based strategies should allow
enrolment of pre- and postmenopausal women, and men,
recognizing nonetheless that this eld has evolved in recent
years.
Young women with ABC have unique medical and psy-
chosocial concerns that need to be considered and
addressed.
147
While pregnancy in the setting of ABC is not
considered safe or desirable from a medical perspective,
concerns for fertility and family planning need to be
cautiously discussed and explored even in the setting of
advanced disease. In patients with long-term responses and
prolonged progression-free survival (e.g. in HER2þdisease),
the safety of interrupting anti-HER2 therapies for patients
who may be interested in a pregnancy is unknown.
Locoregional relapse
Young age is a risk factor for local relapse. Therefore, careful
attention to margin status is warranted in young women.
148
Following locoregional relapse, BCY5 conrmed that
chemotherapy should be considered in women with ER
tumors, as demonstrated in the CALOR study.
149
For ERþ
disease, ET should be given and for HER2þdisease,
trastuzumab-based therapy is recommended albeit based
only on expert opinion level of evidence.
Unique populations
BRCA mutation carriers. There remains no denitive
conclusion on the optimal chemotherapy regimen for BRCA-
associated breast cancer in the neo/adjuvant setting and
the panel recommended that standard prognostic features
should be used to decide treatment in early disease.
The role of PARPi in the early breast cancer setting has
been established by the OlympiA study which randomized
patients with BRCA1/2-associated breast cancer to a year of
adjuvant olaparib or placebo. The study included patients
with high-risk features dened as stages 2 and 3, TNBC with
residual disease after pre-operative chemotherapy or node
positive or pT2 irrespective of nodal status or ERþdisease
with four or more involved lymph nodes or signicant re-
sidual disease after pre-operative chemotherapy.The median
age in this study was 42 (range 36-50) years, most patients
being premenopausal. At a median follow-up of 2.5 years, the
3-year iDFS was improved by 8.8% in the olaparib group.
150
Noteworthy is the observation of fewer new primary malig-
nancies in the olaparib treatment arm, which may be
particularly important for younger patients. The BCY5 panel
stated that olaparib should be offered to germline BRCA1/2
mutation carriers who meet the OLYMPIA inclusion criteria or
the regulatory approval criteria.
The BCY5 panel endorses all the ABC5 statements on
systemic treatment for patients with a germline BRCA mu-
tation, germline PALB2 mutation and somatic BRCA1/2
mutations, recognizing the importance of molecular tumor
boards and of data pooling in international registries.
Preliminary data suggest that breast cancer patients
harboring BRCA mutations present a higher incidence of
central nervous system (CNS) involvement. Although sub-
group analyses in both OlympiAD and EMBRACA showed a
benet in persons with stable/treated brain metastases,
none of the phase III PARPi trials included patients with
active CNS metastases.
151
BCY5 panelists encourage the
inclusion of patients with active CNS metastases in studies
investigating new therapies.
MALE BREAST CANCER
Male breast cancer accounts for w1% of all breast cancers;
the lifetime risk is w1 : 1 000, and represents a small mi-
nority in males <40 years.
152,153
Breast cancer in young
men is rare; however, it would likely amplify the psycho-
logical distress already experienced by men with a breast
cancer diagnosis. Treatment should follow international
guidelines,
154
men should be included in clinical trials,
particularly in trials exploring ET as the expected differential
efcacy in males versus females is greatest in this setting,
germline testing carried out to aid in treatment and pre-
vention measures and distinctive histopathologic and
genomic features investigated.
155,156
Supportive and follow-up care
Many breast cancer patients use integrative medicine
(complementary and alternative therapiesdCAM)
157
but
very few data are available in young women. In a recent
Annals of Oncology S. Paluch-Shimon et al.
16 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
German study, 62.5% of 827 young breast cancer patients
had used CAM. Its use was signicantly higher in women
with higher educational level and employment status, and
in those feeling they had not received sufcient informa-
tion.
158
The BCY5 panel stated that health professionals
should proactively promote open communication about
integrative medicine with their young breast cancer pa-
tients. As effectiveness and safety of many therapies remain
under-explored, the BCY5 panel also approved that, if
possible, supervised and established integrative medicine
services should be provided within the oncology service.
BCY5 conrmed that follow-up care and supportive
treatment/prevention of specic symptoms and side-effects
in young women should follow the same guidelines as in
older women. New shared-care models involving cancer
specialists, general practitioners and breast nurses are being
evaluated to improve cost-effectiveness.
The panel also reiterated that standardized patient-
reported outcome measurements may enable timely
collection of treatment side-effects, enabling the develop-
ment of targeted interventions.
Dedicated survivorship clinics that assess and manage
early and late treatment toxicities and treatment adherence
are valuable in this population.
Psychosocial issues. Young breast cancer patients are at
greater risk of psychosocial morbidity, not only during the
active treatment period but also long term
159
and when
facing ABC.
160
Psychosocial care should be available and
integrated in routine cancer treatments and follow-up. The
BCY5 panel recommended that the specic psychosocial
distress pertaining to body image, sexuality and sexual
dysfunction resulting from premature menopause,
treatment-related amenorrhea, weight gain, hair loss and
breast surgery should be routinely addressed by the health
care team in a culturally appropriate manner to ensure
provision of appropriate information and support.
Partners and family members should be involved early on
and couple-based and/or familial psychosocial interventions
should be promptly proposed during the different phases of
the disease. Offspring having a mother diagnosed with breast
cancer may experience high levels of psychological distress.
161
Partners of young breast cancer survivors (3-8 years post-
treatment) report poorer overall, physical, social, psychologi-
cal and spiritual QoL compared to partners of healthy
women.
162-164
Social issues that need to be addressed include
return to work, family planning and nancial loss. The scarce
data about end-of-life concerns of young patients with ABC
derive from Caucasian, upper-/middle-class women within
nuclear families and include worries about children and co-
parents which affect their QoL and family functioning.
165
The
BCY5 panelists recognized that further research in this setting
is needed on patients from diverse backgrounds, non-nuclear
families, on the co-parent, parents and caregivers.
Despite not being the topic of BCY5, we recognize the
impact of COVID-19 pandemic on patientspsychological
health
166
due to disruption of oncology service organization
and perceived increased loneliness.
Considerations and recommendations by the BCY5 panel
for fertility preservation, contraception and premature
menopause, sexual functioning, pregnancy after breast
cancer, bone health, cognitive impairment, lifestyle changes
and breast cancer during pregnancy are updated in
Supplementary Appendix S1, available at https://doi.org/
10.1016/j.annonc.2022.07.007.
Patient advocacy statements
BCY5 included the second patient advocacy workshop, and
for the third time, included a patient advocacy-dedicated
session for young women with breast cancer and the
consensus session included two young breast cancer survi-
vors as panelists. Since BCY3, a closed Facebook group for
young breast cancer survivors has been active and now has
over 53 active global members. The importance of the
worldwide breast cancer community to work together was
apparent throughout the meeting.
At BCY5, a presentation was made announcing Project
528, the rst-ever global needs assessment of young adults
diagnosed with breast cancer. This will be a global survey
that is created in true partnership with Young Survival
Coalition and Europa Donna Slovenia to learn the global
needs of young adults. Project 528 has four clear goals: (i)
identify the unmet needs of young adults diagnosed with
breast cancer and their caregivers; (ii) understand the
geographic difference in patient experiences; (iii) discover
existing services available to young adults diagnosed with
breast cancer globally; and (iv) understand the QoL burden
of young adults diagnosed with breast cancer.
Everyone interested in participating in this important
survey are encouraged to sign up and learn more at https://
project528.youngsurvival.org.
CONCLUSIONS
Since BCY4, progress has been made. More clinical trials in
the metastatic setting are incorporating young women with
breast cancer by allowing for OFS as an acceptable surrogate
for physiological menopause. The impact of germline muta-
tions in BRCA1/2 has now substantial therapeutic implica-
tions in both early breast cancer and ABC. One of the many
challenges in the treatment of premenopausal ERþearly
breast cancer is how to incorporate and interpret gene
expression signatures into treatment algorithms to deter-
mine which patients need chemotherapy in addition to
optimal ET. Urgent research is needed to address adherence
to available treatments, health disparities and needs of mi-
norities, and a global effort is required to help bridge the gaps
in LMICs. A multidisciplinary approach remains the backbone
of care to ensure optimal outcomes for young women with
breast cancer given their many concerns and care needs.
FUNDING
No external funding has been received for the preparation
of these guidelines. Production costs have been covered by
ESMO from central funds.
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 17
DISCLOSURE
SPS: Advisory Board consultancy, speakers bureau: Astra-
Zeneca, Pzer, Novartis, Lilly, Roche, MSD, Exact Sciences.
Research Funding: Pzer. HAA: Recipient of grants/research
supports: Amgen. Recipient of honoraria or consultation
fees: Novartis, Roche. Employment: Pierre Fabre. FC:
Recipient of honoraria or consultation fees: Amgen, Astel-
las/Medivation, AstraZeneca, Celgene, Daiichi-Sankyo, Eisai,
GE Oncology, Genentech, GlaxoSmithKline, Macrogenics,
Medscape, Merck-Sharp, Merus BV, Mylan, Mundipharma,
Novartis, Pzer, Pierre-Fabre, prIME Oncology, Roche,
Samsung Bioepis. GC: Recipient of grants/research sup-
ports: Merck, Astra Zeneca. Recipient of honoraria or
consultation fees: Daichii Sankyo. Participation in a spon-
sored speakersbureau: Novartis, Pzer, Lilly. KAG: Recipient
of grants/research supports: Pzer, Astra Zeneca. Recipient
of honoraria or consultation fees: Pzer, Lilly, Astrazeneca,
Novartis, Mylan, Benentech, Merck, Roche, Nanostring. NH:
Honoraria for lectures and/or consulting from Amgen, Astra
Zeneca, Daiichi-Sankyo, Exact Sciences, Lilly, MSD, Novartis,
Pierre Fabre, Pzer, Roche, Sandoz, Seagen (all outside the
submitted work). SBK: Research funding from Novartis,
Sano-Aventis and DongKook Pharm Co; consultant in
advisory boardsdNovartis, AstraZeneca, Lilly, Dae Hwa
Pharmaceutical Co. Ltd, ISU Abxis, Daiichi-Sankyo and Bei-
gene; and stocksdGenopeaks, NeogeneTC (all outside the
submitted work). FP: Recipient of honoraria or consultation
fees: Roche Diagnostics, Ipsen, Merck (all outside the sub-
mitted work). ES: Recipient of grants/research supports:
Amgen, AstraZeneca, Boehringer, Eli Lilly, Merck, Novartis,
Pzer, Roche, Samsung. Recipient of honoraria or consul-
tation fees: Amgen, AstraZeneca, Clinigen, Egis, Eli Lilly,
Genomic Health, Novartis, Pzer, Pierre Fabre, Roche, San-
doz, TLC Biopharmaceuticals. Travel support: Amgen,
AstraZeneca, Egis, Novartis, Pzer, Roche. All other authors
have declared no conicts of interest.
REFERENCES
1. DeSantis CE, Ma J, Gaudet MM, et al. Breast cancer statistics, 2019.
CA Cancer J Clin. 2019;69:438-451.
2. Heer E, Harper A, Escandor N, et al. Global burden and trends in
premenopausal and postmenopausal breast cancer: a population-
based study. Lancet Glob Health. 2020;8:e1027-e1037.
3. Joko-Fru WY, Jedy-Agba E, Korir A, et al. The evolving epidemic of
breast cancer in sub-Saharan Africa: results from the African Cancer
Registry Network. Int J Cancer. 2020;147:2131-2141.
4. McDonald JA, Rao R, Gibbons M, et al. Symposium report: breast
cancer in India-trends, environmental exposures and clinical impli-
cations. Cancer Causes Control. 2021;32:567-575.
5. Heer E, Ruan Y, Mealey N, et al. The incidence of breast cancer in
Canada 1971-2015: trends in screening-eligible and young-onset age
groups. Can J Public Health. 2020;111:787-793.
6. DeSantis CE, Ma J, Jemal A. Trends in stage at diagnosis for young
breast cancer patients in the United States. Breast Cancer Res Treat.
2019;173:743-747.
7. Azim HA Jr, Partridge AH. Biology of breast cancer in young women.
Breast Cancer Res. 2014;16:427.
8. Partridge AH, Hughes ME, Warner ET, et al. Subtype-dependent
relationship between young age at diagnosis and breast cancer sur-
vival. J Clin Oncol. 2016;34:3308-3314.
9. Fu J, Zhong C, Wu L, et al. Young patients with hormone receptor-
positive breast cancer have a higher long-term risk of breast cancer
specic death. J Breast Cancer. 2019;22:96-108.
10. Zhong W, Tan L, Jiang WG, et al. Effect of younger age on survival
outcomes in T1N0M0 breast cancer: a propensity score matching
analysis. J Surg Oncol. 2019;119:1039-1046.
11. Copson E, Eccles B, Maishman T, et al. Prospective observational
study of breast cancer treatment outcomes for UK women aged 18-
40 years at diagnosis: the POSH study. J Natl Cancer Inst. 2013;105:
978-988.
12. Cardoso F, Loibl S, Pagani O, et al. The European Society of Breast
Cancer Specialists recommendations for the management of young
women with breast cancer. Eur J Cancer. 2012;48:3355-3377.
13. Partridge AH, Pagani O, Abulkhair O, et al. First international
consensus guidelines for breast cancer in young women (BCY1).
Breast. 2014;23:209-220.
14. Paluch-Shimon S, Pagani O, Partridge AH, et al. Second international
consensus guidelines for breast cancer in young women (BCY2).
Breast. 2016;26:87-99.
15. Paluch-Shimon S, Pagani O, Partridge AH, et al. ESO-ESMO 3rd in-
ternational consensus guidelines for breast cancer in young women
(BCY3). Breast. 2017;35:203-217.
16. Paluch-Shimon S, Cardoso F, Partridge AH, et al. ESO-ESMO 4th In-
ternational Consensus Guidelines for Breast Cancer in Young Women
(BCY4). Ann Oncol. 2020;31:674-696.
17. Dykewicz CA, National Center for Infectious Diseases, Centers for
Disease Control and Prevention; Infectious Diseases Society of
America. American Society for Blood and Marrow Transplantation.
Guidelines for preventing opportunistic infections among hemato-
poietic stem cell transplant recipients: focus on community respira-
tory virus infections. Biol Blood Marrow Transplant. 2001;7(suppl):
19S-22S.
18. Biganzoli L, Cardoso F, Beishon M, et al. The requirements of a
specialist breast centre. Breast. 2020;51:65-84.
19. Runowicz CD, Leach CR, Henry NL, et al. American Cancer Society/
American Society of Clinical Oncology Breast Cancer Survivorship
Care Guideline. J Clin Oncol. 2016;34:611-635.
20. Koh WJ, Anderson BO, Carlson RW. NCCN resource-stratied and
harmonized guidelines: a paradigm for optimizing global cancer care.
Cancer. 2020;126(suppl 10):2416-2423.
21. Duggan C, Dvaladze A, Rositch AF, et al. The Breast Health Global
Initiative 2018 Global Summit on Improving Breast Healthcare
Through Resource-Stratied Phased Implementation: methods and
overview. Cancer. 2020;126(suppl 10):2339-2352.
22. Birnbaum JK, Duggan C, Anderson BO, et al. Early detection and
treatment strategies for breast cancer in low-income and upper
middle-income countries: a modelling study. Lancet Glob Health.
2018;6:e885-e893.
23. Lambertini M, Fontana V, Massarotti C, et al. Prospective study to
optimize care and improve knowledge on ovarian function and/or
fertility preservation in young breast cancer patients: results of the
pilot phase of the PREgnancy and FERtility (PREFER) study. Breast.
2018;41:51-56.
24. Ruggeri M, Pagan E, Bagnardi V, et al. Fertility concerns, preservation
strategies and quality of life in young women with breast cancer:
baseline results from an ongoing prospective cohort study in selected
European centers. Breast. 2019;47:85-92.
25. Rosenberg SM, Tamimi RM, Gelber S, et al. Treatment-related
amenorrhea and sexual functioning in young breast cancer survivors.
Cancer. 2014;120:2264-2271.
26. Coughlin SS. Social determinants of breast cancer risk, stage, and
survival. Breast Cancer Res Treat. 2019;177:537-548.
27. Trewin CB, Johansson ALV, Hjerkind KV, et al. Stage-specic survival
has improved for young breast cancer patients since 2000: but not
equally. Breast Cancer Res Treat. 2020;182:477-489.
28. Afshar N, English DR, Blakely T, et al. Differences in cancer survival by
area-level socio-economic disadvantage: a population-based study
using cancer registry data. PLoS One. 2020;15:e0228551.
Annals of Oncology S. Paluch-Shimon et al.
18 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
29. Cardoso F, MacNeill F, Penault-Llorca F, et al. Why is appropriate
healthcare inaccessible for many European breast cancer patients?
The EBCC 12 manifesto. Breast. 2021;55:128-135.
30. Dumas A, Vaz Luis I, Bovagnet T, et al. Impact of breast cancer
treatment on employment: results of a multicenter prospective
cohort study (CANTO). J Clin Oncol. 2020;38:734-743.
31. Rosenberg SM, Vaz-Luis I, Gong J, et al. Employment trends in young
women following a breast cancer diagnosis. Breast Cancer Res Treat.
2019;177:207-214.
32. Naughton MJ, Beverly Hery CM, Janse SA, et al. Prevalence and
correlates of job and insurance problems among young breast cancer
survivors within 18 months of diagnosis. BMC Cancer. 2020;20:432.
33. Tangka FKL, Subramanian S, Jones M, et al. Insurance coverage,
employment status, and nancial well-being of young women diag-
nosed with breast cancer. Cancer Epidemiol Biomarkers Prev. 2020;29:
616-624.
34. Boelhouwer IG, Vermeer W, van Vuuren T. The associations between
late effects of cancer treatment, work ability and job resources: a
systematic review. Int Arch Occup Environ Health. 2021;94:147-189.
35. Daly AA, Rolph R, Cutress RI, et al. A review of modiable risk factors
in young women for the prevention of breast cancer. Breast Cancer
(Dove Med Press). 2021;13:241-257.
36. Premenopausal Breast Cancer Collaborative GroupSchoemaker MJ,
Nichols HB, Wright LB, et al. Association of body mass index and age
with subsequent breast cancer risk in premenopausal women. JAMA
Oncol. 2018;4:e181771.
37. Mulder RL, Kremer LC, Hudson MM, et al. Recommendations for
breast cancer surveillance for female survivors of childhood, adoles-
cent, and young adult cancer given chest radiation: a report from the
International Late Effects of Childhood Cancer Guideline Harmoni-
zation Group. Lancet Oncol. 2013;14:e621-e629.
38. Evans A, Trimboli RM, Athanasiou A, et al. Breast ultrasound: recom-
mendations for information to women and referring physicians by the
European Society of Breast Imaging. Insights Imaging. 2018;9:449-461.
39. Conant EF, Barlow WE, Herschorn SD, et al. Association of digital breast
tomosynthesis vs digital mammography with cancer detection and
recall rates by age and breast density. JAMA Oncol. 2019;5:635-642.
40. Houssami N, Turner RM, Morrow M. Meta-analysis of pre-operative
magnetic resonance imaging (MRI) and surgical treatment for
breast cancer. Breast Cancer Res Treat. 2017;165:273-283.
41. Schunemann HJ, Lerda D, Quinn C, et al. Breast cancer screening and
diagnosis: a synopsis of the European Breast Guidelines. Ann Intern
Med. 2020;172:46-56.
42. Reig B, Lewin AA, Du L, et al. Breast MRI for evaluation of response to
neoadjuvant therapy. Radiographics. 2021;41:665-679.
43. Mann RM, Balleyguier C, Baltzer PA, et al. Breast MRI: EUSOBI rec-
ommendations for womens information. Eur Radiol. 2015;25:3669-
3678.
44. Baxter GC, Selamoglu A, Mackay JW, et al. A meta-analysis comparing
the diagnostic performance of abbreviated MRI and a full diagnostic
protocol in breast cancer. Clin Radiol. 2021;76:154.e23-154.e32.
45. Beitsch PD, Whitworth PW, Hughes K, et al. Underdiagnosis of he-
reditary breast cancer: are genetic testing guidelines a tool or an
obstacle? J Clin Oncol. 2019;37:453-460.
46. Hu C, Hart SN, Gnanaolivu R, et al. A population-based study of genes
previously implicated in breast cancer. N Engl J Med. 2021;384:440-
451.
47. Kemp Z, Turnbull A, Yost S, et al. Evaluation of cancer-based criteria
for use in mainstream BRCA1 and BRCA2 genetic testing in patients
with breast cancer. JAMA Netw Open. 2019;2:e194428.
48. Beard C, Monohan K, Cicciarelli L, et al. Mainstream genetic testing
for breast cancer patients: early experiences from the Parkville Fa-
milial Cancer Centre. Eur J Hum Genet. 2021;29:872-880.
49. Smith CE, Fullerton SM, Dookeran KA, et al. Using genetic technol-
ogies to reduce, rather than widen, health disparities. Health Aff
(Millwood). 2016;35:1367-1373.
50. Heymann S, Delaloge S, Rahal A, et al. Radio-induced malignancies
after breast cancer postoperative radiotherapy in patients with Li-
Fraumeni syndrome. Radiat Oncol. 2010;5:104.
51. Saya S, Killick E, Thomas S, et al. Baseline results from the UK SIGNIFY
study: a whole-body MRI screening study in TP53 mutation carriers
and matched controls. Fam Cancer. 2017;16:433-440.
52. Villani A, Shore A, Wasserman JD, et al. Biochemical and imaging
surveillance in germline TP53 mutation carriers with Li-Fraumeni
syndrome: 11 year follow-up of a prospective observational study.
Lancet Oncol. 2016;17:1295-1305.
53. Kratz CP, Achatz MI, Brugieres L, et al. Cancer screening recommen-
dations for individuals with Li-Fraumeni syndrome. Clin Cancer Res.
2017;23:e38-e45.
54. Sutherland N, Espinel W, Grotzke M, et al. Unanswered questions:
hereditary breast and gynecological cancer risk assessment in trans-
gender adolescents and young adults. JGenetCouns. 2020;29:625-633.
55. Findlay-Shirras L, Lima I, Smith G, et al. Canada follows the US in the
rise of bilateral mastectomies for unilateral breast cancer: a 23-year
population cohort study. Breast Cancer Res Treat. 2021;185:517-525.
56. Wong SM, Freedman RA, Sagara Y, et al. Growing use of contralateral
prophylactic mastectomy despite no improvement in long-term sur-
vival for invasive breast cancer. Ann Surg. 2017;265:581-589.
57. Sella T, Snow C, Freeman H, et al. Young, empowered and strong: a
web-based education and supportive care intervention for young
women with breast cancer across the care continuum. JCO Clin
Cancer Inform. 2021;5:933-943.
58. Galimberti V, Vicini E, Corso G, et al. Nipple-sparing and skin-sparing
mastectomy: review of aims, oncological safety and contraindica-
tions. Breast. 2017;34(suppl 1):S82-S84.
59. Tondu T, Tjalma WAA, Thiessen FEF. Breast reconstruction after
mastectomy. Eur J Obstet Gynecol Reprod Biol. 2018;230:228-232.
60. Filipe MD, Siesling S, Vriens MR, et al. Socioeconomic status signi-
cantly contributes to the likelihood of immediate postmastectomy
breast reconstruction in the Netherlands: a nationwide study. Eur J
Surg Oncol. 2021;47:245-250.
61. Frisell A, Lagergren J, Halle M, et al. Inuence of socioeconomic
status on immediate breast reconstruction rate, patient information
and involvement in surgical decision-making. BJS Open. 2020;4:232-
240.
62. Hart SE, Momoh AO. Breast reconstruction disparities in the United
States and internationally. Curr Breast Cancer Rep. 2020;12:132-139.
63. Meattini I, Lambertini M, Desideri I, et al. Radiation therapy for young
women with early breast cancer: current state of the art. Crit Rev
Oncol Hematol. 2019;137:143-153.
64. Ricci JA, Epstein S, Momoh AO, et al. A meta-analysis of implant-
based breast reconstruction and timing of adjuvant radiation ther-
apy. J Surg Res. 2017;218:108-116.
65. Arnautovic A, Olafsson S, Wong JS, et al. Optimizing breast recon-
struction through integration of plastic surgery and radiation
oncology. Plast Reconstr Surg Glob Open. 2021;9:e3577.
66. Duma MN, Baumann R, Budach W, et al. Heart-sparing radiotherapy
techniques in breast cancer patients: a recommendation of the breast
cancer expert panel of the German society of radiation oncology
(DEGRO). Strahlenther Onkol. 2019;195:861-871.
67. Brunt AM, Haviland JS, Sydenham M, et al. Ten-year results of FAST: a
randomized controlled trial of 5-fraction whole-breast radiotherapy
for early breast cancer. J Clin Oncol. 2020;38:3261-3272.
68. Murray Brunt A, Haviland JS, Wheatley DA, et al. Hypofractionated
breast radiotherapy for 1 week versus 3 weeks (FAST-Forward): 5-
year efcacy and late normal tissue effects results from a multi-
centre, non-inferiority, randomised, phase 3 trial. Lancet. 2020;395:
1613-1626.
69. Poortmans PMP, Kaidar-Person O. Contralateral breast irradiation in
BRCA carriers: the conundrum of prophylactic versus early treatment.
Ann Oncol. 2019;30:348-350.
70. Bernstein-Molho R, Laitman Y, Galper S, et al. Locoregional treat-
ments and ipsilateral breast cancer recurrence rates in BRCA1/2
mutation carriers. Int J Radiat Oncol Biol Phys. 2021;109:1332-
1340.
71. Reiner AS, Robson ME, Mellemkjaer L, et al. Radiation treatment,
ATM, BRCA1/2, and CHEK2*1100delC pathogenic variants and risk of
contralateral breast cancer. J Natl Cancer Inst. 2020;112:1275-1279.
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 19
72. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a
21-gene expression assay in breast cancer. N Engl J Med. 2015;373:
2005-2014.
73. Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy
guided by a 21-gene expression assay in breast cancer. N Engl J Med.
2018;379:111-121.
74. Cardoso F, vant Veer LJ, Bogaerts J, et al. 70-Gene signature as an aid
to treatment decisions in early-stage breast cancer. N Engl J Med.
2016;375:717-729.
75. Sparano JA, Gray RJ, Ravdin PM, et al. Clinical and genomic risk to
guide the use of adjuvant therapy for breast cancer. N Engl J Med.
2019;380:2395-2405.
76. Kalinsky K, Barlow WE, Gralow JR, et al. 21-Gene assay to inform
chemotherapy benet in node-positive breast cancer. N Engl J Med.
2021;385:2336-2347.
77. Nitz UA, Gluz O, Kummel S, et al. Endocrine therapy response and 21-
gene expression assay for therapy guidance in HRþ/HER2- early
breast cancer. J Clin Oncol. 2022. https://doi.org/10.1200/JCO.21.
02759.
78. Piccart M, van t Veer LJ, Poncet C, et al. 70-Gene signature as an aid
for treatment decisions in early breast cancer: updated results of the
phase 3 randomised MINDACT trial with an exploratory analysis by
age. Lancet Oncol. 2021;22:476-488.
79. Piccart MJ, Kalinsky K, Gray R, et al. Gene expression signatures for
tailoring adjuvant chemotherapy of luminal breast cancer: stronger
evidence, greater trust. Ann Oncol. 2021;32:1077-1082.
80. Francis PA, Pagani O, Fleming GF, et al. Tailoring adjuvant endocrine
therapy for premenopausal breast cancer. N Engl J Med. 2018;379:
122-137.
81. Gnant M, Mlineritsch B, Stoeger H, et al. Zoledronic acid combined
with adjuvant endocrine therapy of tamoxifen versus anastrozol plus
ovarian function suppression in premenopausal early breast cancer:
nal analysis of the Austrian Breast and Colorectal Cancer Study
Group Trial 12. Ann Oncol. 2015;26:313-320.
82. Saha P, Regan MM, Pagani O, et al. Treatment efcacy, adherence,
and quality of life among women younger than 35 years in the In-
ternational Breast Cancer Study Group TEXT and SOFT adjuvant
endocrine therapy trials. J Clin Oncol. 2017;35:3113-3122.
83. Visvanathan K, Fabian CJ, Bantug E, et al. Use of endocrine therapy for
breast cancer risk reduction: ASCO clinical practice guideline update.
J Clin Oncol. 2019;37:3152-3165.
84. Donnelly LS, Evans DG, Wiseman J, et al. Uptake of tamoxifen in
consecutive premenopausal women under surveillance in a high-risk
breast cancer clinic. Br J Cancer. 2014;110:1681-1687.
85. Kandagatla P, Rizk NN, Dokic D, et al. Patient and provider factors
associated with the noninitiation of tamoxifen for young women at
high-risk for the development of breast cancer. Breast J. 2020;26:464-
468.
86. DeCensi A, Puntoni M, Guerrieri-Gonzaga A, et al. Randomized pla-
cebo controlled trial of low-dose tamoxifen to prevent local and
contralateral recurrence in breast intraepithelial neoplasia. J Clin
Oncol. 2019;37:1629-1637.
87. DeCensi A, Puntoni M, Johansson H, et al. Effect modiers of low-
dose tamoxifen in a randomized trial in breast noninvasive disease.
Clin Cancer Res. 2021;27:3576-3583.
88. Korde LA, Somereld MR, Carey LA, et al. Neoadjuvant chemo-
therapy, endocrine therapy, and targeted therapy for breast cancer:
ASCO guideline. J Clin Oncol. 2021;39:1485-1505.
89. Harbeck N, Gluz O, Kuemmel S, et al. Abstract GS4-04: Endocrine
therapy alone in patients with intermediate or high-risk luminal early
breast cancer (0-3 lymph nodes), recurrence score <26 and Ki67
response after preoperative endocrine therapy: primary outcome
results from the WSG-ADAPT HRþ/HER2- trial. Cancer Res. 2021;81.
GS4-04-GS04-04.
90. Burstein HJ, Lacchetti C, Anderson H, et al. Adjuvant endocrine
therapy for women with hormone receptor-positive breast cancer:
American Society of Clinical Oncology clinical practice guideline up-
date on ovarian suppression. J Clin Oncol. 2016;34:1689-1701.
91. Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO
Clinical Practice Guidelines for diagnosis, treatment and follow-up.
Ann Oncol. 2019;30:1194-1220.
92. Pan H, Gray R, Braybrooke J, et al. 20-Year risks of breast-cancer
recurrence after stopping endocrine therapy at 5 years. N Engl J
Med. 2017;377:1836-1846.
93. Pagani O, Francis PA, Fleming GF, et al. Absolute improvements in
freedom from distant recurrence to tailor adjuvant endocrine ther-
apies for premenopausal women: results from TEXT and SOFT. J Clin
Oncol. 2020;38:1293-1303.
94. Regan MM, Walley BA, Francis PA, et al. Concurrent and sequential
initiation of ovarian function suppression with chemotherapy in
premenopausal women with endocrine-responsive early breast can-
cer: an exploratory analysis of TEXT and SOFT. Ann Oncol. 2017;28:
2225-2232.
95. Zhang Y, Ji Y, Li J, et al. Sequential versus simultaneous use of
chemotherapy and gonadotropin-releasing hormone agonist (GnRHa)
among estrogen receptor (ER)-positive premenopausal breast cancer
patients: effects on ovarian function, disease-free survival, and
overall survival. Breast Cancer Res Treat. 2018;168:679-686.
96. Lambertini M, Moore HCF, Leonard RCF, et al. Gonadotropin-
releasing hormone agonists during chemotherapy for preservation of
ovarian function and fertility in premenopausal patients with early
breast cancer: a systematic review and meta-analysis of individual
patient-level data. J Clin Oncol. 2018;36:1981-1990.
97. Kim HA, Lee JW, Nam SJ, et al. Adding ovarian suppression to
tamoxifen for premenopausal breast cancer: a randomized phase III
trial. J Clin Oncol. 2020;38:434-443.
98. Li JW, Liu GY, Ji YJ, et al. Switching to anastrozole plus goserelin vs
continued tamoxifen for adjuvant therapy of premenopausal early-
stage breast cancer: preliminary results from a randomized trial.
Cancer Manag Res. 2019;11:299-307.
99. Davies C, Pan H, Godwin J, et al. Long-term effects of continuing
adjuvant tamoxifen to 10 years versus stopping at 5 years after
diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a
randomised trial. Lancet. 2013;381:805-816.
100. 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(suppl
18):5.
101. van Hellemond IEG, Vriens IJH, Peer PGM, et al. Ovarian function
recovery during anastrozole in breast cancer patients with
chemotherapy-induced ovarian function failure. J Natl Cancer Inst.
2017;109.
102. van Hellemond IEG, Vriens IJH, Peer PGM, et al. Efcacy of anastrozole
after tamoxifen in early breast cancer patients with chemotherapy-
induced ovarian function failure. Int J Cancer. 2019;145:274-283.
103. Rosner W, Hankinson SE, Sluss PM, et al. Challenges to the mea-
surement of estradiol: an endocrine society position statement. J Clin
Endocrinol Metab. 2013;98:1376-1387.
104. Hillebrand JJ, Wickenhagen WV, Heijboer AC. Improving science by
overcoming laboratory pitfalls with hormone measurements. J Clin
Endocrinol Metab. 2021;106:e1504-e1512.
105. Bellet M, Gray K, Francis P, et al. Abstract P4-14-01: Estrogen levels in
premenopausal patients (pts) with hormone-receptor positive (HRþ)
early breast cancer (BC) receiving adjuvant triptorelin (Trip) plus
exemestane (E) or tamoxifen (T) in the SOFT trial: SOFT-EST substudy
nal analysis. Cancer Res. 2019;79:P4-P14.
106. Kurebayashi J, Toyama T, Sumino S, et al. Efcacy and safety of leu-
prorelin acetate 6-month depot, TAP-144-SR (6M), in combination
with tamoxifen in postoperative, premenopausal patients with hor-
mone receptor-positive breast cancer: a phase III, randomized, open-
label, parallel-group comparative study. Breast Cancer. 2017;24:161-
170.
107. Wu ZY, Lee YJ, Kim H, et al. Effectiveness of a 6-month 22.5-mg leu-
prolide acetate depot formulation with tamoxifen for postoperative
premenopausal estrogen suppression in hormone receptor-positive
breast cancer. Front Oncol. 2021;11:665426.
Annals of Oncology S. Paluch-Shimon et al.
20 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
108. Shiba E, Yamashita H, Kurebayashi J, et al. A randomized controlled
study evaluating safety and efcacy of leuprorelin acetate every-3-
months depot for 2 versus 3 or more years with tamoxifen for
5 years as adjuvant treatment in premenopausal patients with
endocrine-responsive breast cancer. Breast Cancer. 2016;23:499-
509.
109. Asiri MA, Tunio MA, Abdulmoniem R. Is radiation-induced ovarian
ablation in breast cancer an obsolete procedure? Results of a meta-
analysis. Breast Cancer (Dove Med Press). 2016;8:109-116.
110. Wassermann J, Gelber SI, Rosenberg SM, et al. Nonadherent behav-
iors among young women on adjuvant endocrine therapy for breast
cancer. Cancer. 2019;125:3266-3274.
111. Hershman DL, Shao T, Kushi LH, et al. Early discontinuation and non-
adherence to adjuvant hormonal therapy are associated with
increased mortality in women with breast cancer. Breast Cancer Res
Treat. 2011;126:529-537.
112. Montagna E, Pagan E, Bagnardi V, et al. Evaluation of endocrine
therapy and patients preferences in early breast cancer: results of
Elena study. Breast Cancer Res Treat. 2020;184:783-795.
113. Kuba S, Maeda S, Matsumoto M, et al. Adherence to adjuvant
endocrine therapy in women with breast cancer: a prospective
observational study in Japanese women. Clin Breast Cancer. 2018;18:
150-156.
114. Sheppard VB, Sutton AL, Hurtado-de-Mendoza A, et al. Race and
patient-reported symptoms in adherence to adjuvant endocrine
therapy: a report from the Womens Hormonal Initiation and
Persistence Study. Cancer Epidemiol Biomarkers Prev. 2021;30:699-
709.
115. Finitsis DJ, Vose BA, Mahalak JG, Salner AL. Interventions to promote
adherence to endocrine therapy among breast cancer survivors: a
meta-analysis. Psychooncology. 2019;28:255-263.
116. Toivonen KI, Williamson TM, Carlson LE, et al. Potentially modiable
factors associated with adherence to adjuvant endocrine therapy
among breast cancer survivors: a systematic review. Cancers (Basel).
2020;13:107.
117. Johnston SRD, Harbeck N, Hegg R, et al. Abemaciclib combined with
endocrine therapy for the adjuvant treatment of HRþ, HER2-, node-
positive, high-risk, early breast cancer (monarchE). J Clin Oncol.
2020;38:3987-3998.
118. Loibl S, Marme F, Martin M, et al. Palbociclib for residual high-risk
invasive HR-positive and HER2-negative early breast cancer-the
Penelope-B trial. J Clin Oncol. 2021;39:1518-1530.
119. Mayer EL, Dueck AC, Martin M, et al. Palbociclib with adjuvant
endocrine therapy in early breast cancer (PALLAS): interim analysis of
a multicentre, open-label, randomised, phase 3 study. Lancet Oncol.
2021;22:212-222.
120. Harbeck N, Rastogi P, Martin M, et al. Adjuvant abemaciclib combined
with endocrine therapy for high-risk early breast cancer: updated
efcacy and Ki-67 analysis from the monarchE study. Ann Oncol.
2021;32:1571-1581.
121. Oktay K, Harvey BE, Loren AW. Fertility preservation in patients with
cancer: ASCO clinical practice guideline update summary. J Oncol
Pract. 2018;14:381-385.
122. Lambertini M, Peccatori FA, Demeestere I, et al. Fertility preservation
and post-treatment pregnancies in post-pubertal cancer patients:
ESMO Clinical Practice Guidelines. Ann Oncol. 2020;31:1664-1678.
123. Leonard R, Yellowlees A, Mansi J, et al. The affect of goserelin on the
QoL of women having chemotherapy for EBC: results from the OP-
TION trial. Breast. 2020;52:122-131.
124. Shin JJ, Choi YM, Jun JK, et al. Effect of timing of gonadotropin-
releasing hormone agonist administration for ovarian protection in
patients with breast cancer. J Breast Cancer. 2020;23:268-278.
125. Marklund A, Eloranta S, Wikander I, et al. Efcacy and safety of
controlled ovarian stimulation using GnRH antagonist protocols for
emergency fertility preservation in young women with breast cancer-
a prospective nationwide Swedish multicenter study. Hum Reprod.
2020;35:929-938.
126. Marin L, Vitagliano A, Capobianco G, et al. Which is the optimal timing
for starting chemoprotection with gonadotropin-releasing hormone
agonists after oocyte cryopreservation? Reections on a critical case
of ovarian hyperstimulation syndrome. J Gynecol Obstet Hum Reprod.
2021;50:101815.
127. Anderson RA, Mansi J, Coleman RE, et al. The utility of anti-Mullerian
hormone in the diagnosis and prediction of loss of ovarian function
following chemotherapy for early breast cancer. Eur J Cancer. 2017;87:
58-64.
128. Leonard RCF, Adamson DJA, Bertelli G, et al. GnRH agonist for pro-
tection against ovarian toxicity during chemotherapy for early breast
cancer: the Anglo Celtic Group OPTION trial. Ann Oncol. 2017;28:
1811-1816.
129. Lee DY, Park YH, Lee JE, et al. Prediction of ovarian function recovery
in young breast cancer patients after protection with gonadotropin-
releasing hormone agonist during chemotherapy. Breast Cancer Res
Treat. 2018;171:649-656.
130. Peto R, Davies C, Godwin J, et al. Comparisons between different
polychemotherapy regimens for early breast cancer: meta-analyses of
long-term outcome among 100,000 women in 123 randomised trials.
Lancet. 2012;379:432-444.
131. Nitz U, Gluz O, Clemens M, et al. West German Study PlanB trial:
adjuvant four cycles of epirubicin and cyclophosphamide plus doce-
taxel versus six cycles of docetaxel and cyclophosphamide in HER2-
negative early breast cancer. J Clin Oncol. 2019;37:799-808.
132. van der Voort A, van Ramshorst MS, van Werkhoven ED, et al. Three-
year follow-up of neoadjuvant chemotherapy with or without
anthracyclines in the presence of dual ERBB2 blockade in patients
with ERBB2-positive breast cancer: a secondary analysis of the TRAIN-
2 randomized, phase 3 trial. JAMA Oncol. 2021;7:978-984.
133. Du F, Wang W, Wang Y, et al. Carboplatin plus taxanes are non-inferior
to epirubicin plus cyclophosphamide followed by taxanes as adjuvant
chemotherapy for early triple-negative breast cancer. Breast Cancer
Res Treat. 2020;182:67-77.
134. Tung N, Arun B, Hacker MR, et al. TBCRC 031: Randomized phase II
study of neoadjuvant cisplatin versus doxorubicin-cyclophosphamide
in germline BRCA carriers with HER2-negative breast cancer (the
INFORM trial). J Clin Oncol. 2020;38:1539-1548.
135. Loibl S, Sikov W, Huober J, et al. 119O Event-free survival (EFS),
overall survival (OS), and safety of adding veliparib (V) plus carbo-
platin (Cb) or carboplatin alone to neoadjuvant chemotherapy in
triple-negative breast cancer (TNBC) after 4 years of follow-up:
BrighTNess, a randomized phase III trial. Ann Oncol. 2021;32:S408.
136. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant capecitabine for breast
cancer after preoperative chemotherapy. N Engl J Med. 2017;376:
2147-2159.
137. Mayer IA, Zhao F, Arteaga CL, et al. Randomized phase III post-
operative trial of platinum-based chemotherapy versus capecitabine
in patients with residual triple-negative breast cancer following
neoadjuvant chemotherapy: ECOG-ACRIN EA1131. J Clin Oncol.
2021;39:2539-2551.
138. Schmid P, Cortes J, Dent R, et al. VP7-2021: KEYNOTE-522: Phase III
study of neoadjuvant pembrolizumab þchemotherapy vs. placebo þ
chemotherapy, followed by adjuvant pembrolizumab vs. placebo for
early-stage TNBC. Ann Oncol. 2021;32:1198-1200.
139. 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;396:1090-1100.
140. Loibl S, Schneeweiss A, Huober JB, et al. Durvalumab improves long-
term outcome in TNBC: results from the phase II randomized
GeparNUEVO study investigating neodjuvant durvalumab in addition
to an anthracycline/taxane based neoadjuvant chemotherapy in early
triple-negative breast cancer (TNBC). J Clin Oncol. 2021;39. 506-
506.
141. Bradley R, Braybrooke J, Gray R, et al. Trastuzumab for early-stage,
HER2-positive breast cancer: a meta-analysis of 13 864 women in
seven randomised trials. Lancet Oncol. 2021;22:1139-1150.
142. Perrone F, De Laurentiis M, De Placido S, et al. Adjuvant zoledronic
acid and letrozole plus ovarian function suppression in
S. Paluch-Shimon et al. Annals of Oncology
Volume xxx -Issue xxx -2022 https://doi.org/10.1016/j.annonc.2022.07.007 21
premenopausal breast cancer: HOBOE phase 3 randomised trial. Eur J
Cancer. 2019;118:178-186.
143. Jallouk AP, Paravastu S, Weilbaecher K, et al. Long-term outcome of
(neo)adjuvant zoledronic acid therapy in locally advanced breast
cancer. Breast Cancer Res Treat. 2021;187:135-144.
144. Awan A, Ng T, Conter H, et al. Feasibility outcomes of a randomised,
multicentre, pilot trial comparing standard 6-monthly dosing of
adjuvant zoledronate with a single one-time dose in patients with
early stage breast cancer. J Bone Oncol. 2021;26:100343.
145. Sokal A, Elefant E, Leturcq T, et al. Pregnancy and newborn outcomes
after exposure to bisphosphonates: a case-control study. Osteoporos
Int. 2019;30:221-229.
146. Cardoso F, Paluch-Shimon S, Senkus E, et al. 5th ESO-ESMO inter-
national consensus guidelines for advanced breast cancer (ABC 5).
Ann Oncol. 2020;31:1623-1649.
147. Di Lascio S, Pagani O. Is it time to address survivorship in advanced
breast cancer? A review article. Breast. 2017;31:167-172.
148. Pilewskie M, Morrow M. Margins in breast cancer: how much is
enough? Cancer. 2018;124:1335-1341.
149. Wapnir IL, Price KN, Anderson SJ, et al. Efcacy of chemotherapy for
ER-negative and ER-positive isolated locoregional recurrence of
breast cancer: nal analysis of the CALOR trial. J Clin Oncol. 2018;36:
1073-1079.
150. Tutt ANJ, Garber JE, Kaufman B, et al. Adjuvant olaparib for patients
with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med.
2021;384:2394-2405.
151. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in patients with advanced
breast cancer and a germline BRCA mutation. N Engl J Med.
2018;379:753-763.
152. Cardoso F, Bartlett JMS, Slaets L, et al. Characterization of male breast
cancer: results of the EORTC 10085/TBCRC/BIG/NABCG International
Male Breast Cancer Program. Ann Oncol. 2018;29:405-417.
153. Li N, Wang X, Zhang H, et al. Young male breast cancer, a small crowd,
the survival, and prognosis?: a population-based study. Medicine
(Baltimore). 2018;97:e12686.
154. Hassett MJ, Somereld MR, Giordano SH. Management of male
breast cancer: ASCO guideline summary. JCO Oncol Pract. 2020;16:
e839-e843.
155. Silva SN, Gomes BC, Andre S, et al. Male and female breast cancer:
the two faces of the same genetic susceptibility coin. Breast Cancer
Res Treat. 2021;188:295-305.
156. Erices-Leclercq M, Lubig S, Forster F, et al. Prognostic relevance of
Ki67 expression in primary male breast cancer: determination of cut-
off points by different evaluation methods and statistical examina-
tions. J Cancer Res Clin Oncol. 2022;148:441-447.
157. Lyman GH, Greenlee H, Bohlke K, et al. Integrative therapies during
and after breast cancer treatment: ASCO endorsement of the SIO
clinical practice guideline. J Clin Oncol. 2018;36:2647-2655.
158. Hammersen F, Pursche T, Fischer D, et al. Use of complementary and
alternative medicine among young patients with breast cancer. Breast
Care (Basel). 2020;15:163-170.
159. Fernandes-Taylor S, Adesoye T, Bloom JR. Managing psychosocial is-
sues faced by young women with breast cancer at the time of diag-
nosis and during active treatment. Curr Opin Support Palliat Care.
2015;9:279-284.
160. Ahmad S, Fergus K, McCarthy M. Psychosocial issues experienced by
young women with breast cancer: the minority group with the ma-
jority of need. Curr Opin Support Palliat Care. 2015;9:271-278.
161. Vila MM, Barco Berron SD, Gil-Gil M, et al. Psychosocial aspects and life
project disruption in young women diagnosed with metastatic
hormone-sensitive HER2-negative breast cancer. Breast. 2020;53:44-50.
162. Purc-Stephenson R, Lyseng A. How are the kids holding up? A sys-
tematic review and meta-analysis on the psychosocial impact of
maternal breast cancer on children. Cancer Treat Rev. 2016;49:45-56.
163. Walczak A, McDonald F, Patterson P, et al. How does parental cancer
affect adolescent and young adult offspring? A systematic review. Int
J Nurs Stud. 2018;77:54-80.
164. Chan A, Lomma C, Chih H, et al. Psychosocial consequences in
offspring of women with breast cancer. Psychooncology. 2020;29:517-
524.
165. Cohee A, Storey S, Winger JG, et al. A cohort study of quality of life in
partners of young breast cancer survivors compared to partners of
healthy controls. J Patient Rep Outcomes. 2020;4:19.
166. Caparso C, Appel J, Benkert R. Dying concerns in young parents with
advanced cancer (PWAC): a scoping review. Palliat Support Care.
2021;19:93-102.
Annals of Oncology S. Paluch-Shimon et al.
22 https://doi.org/10.1016/j.annonc.2022.07.007 Volume xxx -Issue xxx -2022
... These included age and Body Mass Index (BMI) at the start of GnRHa therapy, type of GnRHa and endocrine therapy used, previous chemotherapy, and disease stage. Patients were categorized into age groups using 40 years as a cutoff, based on the European School of Oncology (ESO)-European Society of Medical Oncology (ESMO) fifth international consensus guidelines for breast cancer in young women (BCY5) guidelines [15]. The disease stage was classified according to the American Joint Committee on Cancer (AJCC) staging system, with advanced breast cancer when distant metastases were present, otherwise as early-stage breast cancer. ...
... Not coincidentally, Lee et al. and Wu et al. [27,28] chose this criterion when studying the effectiveness of 3-monthly and 6-monthly GnRHa formulations combined with tamoxifen therapy, respectively. These [13] SOFT-EST substudy final analysis [24] Burns [15] Van Houdt [14] This study Content courtesy of Springer Nature, terms of use apply. Rights reserved. ...
Article
Full-text available
Purpose This study evaluated the effectiveness of ovarian function suppression (OFS) of various gonadotropin-releasing hormone agonists (GnRHa) combined with aromatase inhibitors (AI) in premenopausal patients with hormone receptor-positive (HR-positive) breast cancer. Potential risk factors associated with insufficient OFS were analyzed. Patients and methods Premenopausal HR-positive breast cancer patients who had received AI with GnRHa were studied retrospectively. Patients were divided into different groups according to monthly or trimonthly GnRHa schedules they received, and the effectiveness of OFS was compared between groups. Insufficient OFS was defined as at least one instance of estradiol ≥ 30 pg/ml. Patient data was gathered from medical records for this comparison. Results Of the 264 patients enrolled in this study, 117 were administered 3.6 mg of goserelin monthly (goserelin 1 M group), 63 received 3.75 mg of leuprorelin monthly (leuprorelin 1 M group) and 84 were given 11.25 mg of leuprorelin every three months (leuprorelin 3 M group). Overall, 7.20% experienced insufficient OFS. The incidence rates in the three GnRHa depot groups were 7.69%, 6.35%, and 7.14%, respectively, without a significant statistical difference (P = 0.900). Notably, younger patients exhibited a higher likelihood of insufficient OFS [OR = 0.900, 95%CI (0.824–0.982), P = 0.018]. Conclusion Insufficient OFS remains a concern during GnRHa and AI treatment. The effectiveness of the three GnRHa depots commonly used in China seems comparable. Younger patients face a heightened risk of insufficient OFS.
... This significantly improves patients' event-free survival(EFS) and overall survival(OS) [5]. thus becoming one of the standard treatment approaches for the aforementioned breast cancer patients [6]. ...
Article
Introduction Breast cancer has become the most prevalent malignant tumor among women globally, posing a serious threat to women's life and health. Neoadjuvant therapy (NAT) has emerged as one of the standard treatment approaches for breast cancer patients. However, due to varying responses to NAT among different patients, significant differences in treatment effectiveness occur, impacting the timely alteration of treatment strategies for patients. Material and methods Based on clinical and pathological characteristics along with the RCB scoring, we utilized a support vector machine (SVM) algorithm to construct a Breast Cancer Pathological Characteristics(PBCS) prediction model. We thoroughly validated the PBCS and compared it to a Pathomics Signatures (PS) prediction model. Results we developed a pathological prediction label, named PS. Subsequently, through univariate and multivariate analysis, we discovered a significant correlation between HER2 and the patients' RCB scores. Integrating HER2 into PS, we constructed a breast cancer pathological prediction model, named PBCS.PBCS exhibits good performance in predicting the effectiveness of postoperative therapy(RCB 0~Ⅰ) in both the training sets(AUC0.86[95%CI0.7988-0.9173])and validation sets(AUC0.83[95%CI0.7219-0.9382 ]). In the validation set, PBCS significantly outperforms the PS(AUC0.65[95%CI0.5121-0.7886 ]).Calibration curves and clinical decision curves also strongly support PBCS's ability to effectively predict the efficacy of therapy(RCB 0~Ⅰ). Conclusions Conclusions PBCS can assist clinical and pathological physicians in accurately predicting patients' post-treatment RCB grading before initiating NAT. This offers a new approach to forecast breast cancer patients' responsiveness to NAT, aiding in devising personalized treatment strategies for patients.
Article
Background Despite the wide use of a 3-month gonadotropin-releasing hormone (GnRH) agonist for ovarian function suppression in premenopausal breast cancer patients, it remains unclear whether it is as effective and safe as a 1-month GnRH agonist regimen when combined with selective estrogen receptor modulators or aromatase inhibitors, especially in younger patients. Methods This retrospective cohort study included 1109 premenopausal hormone receptor–positive breast cancer patients treated with GnRH agonist plus selective estrogen receptor modulator or aromatase inhibitor. The estradiol (E2) inhibition rate within 1-24 months after treatment with 1-month or 3-month GnRH agonist in cohorts and different subgroups was analyzed. Results Following 1:1 propensity score matching, 950 patients with a mean age of 39 years and a median follow-up of 46 months were included. Both the 1-month and 3-month groups achieved more than 90% E2 inhibition within 24 months (94.53% vs 92.84%, with a 95% confidence interval for the difference ranging from −4.78% to 1.41%), confirming the noninferiority of 3-month GnRH agonist. Both 1-month and 3-month GnRH agonist rapidly and consistently reduced E2 levels. Of the patients, 60 (6.3%) experienced incomplete ovarian function suppression, with similar rates in the 1-month and 3-month groups (5.5% vs 7.2%). Incomplete ovarian function suppression mainly occurred within the first 12 months, with age younger than 40 years and no prior chemotherapy being the risk factors. Similar disease-free survival and overall survival were found in the 1-month and 3-month groups and in patients with complete and incomplete ovarian function suppression (P > .05). Conclusions The ovarian function suppression with 3-month GnRH agonist was not inferior to that with 1-month GnRH agonist, regardless of age or combination with a selective estrogen receptor modulator or an aromatase inhibitor.
Article
PURPOSE We investigated time to pregnancy, efficacy and safety of fertility preservation, and assisted reproductive technologies (ARTs) in women with early hormone receptor–positive breast cancer (BC) desiring future pregnancy. PATIENTS AND METHODS POSITIVE is an international, single-arm, prospective trial, in which 518 women temporarily interrupted adjuvant endocrine therapy to attempt pregnancy. We evaluated menstruation recovery and factors associated with time to pregnancy and investigated if ART use was associated with achieving pregnancy. The cumulative incidence of BC-free interval (BCFI) events was estimated according to the use of ovarian stimulation at diagnosis. The median follow-up was 41 months. RESULTS Two hundred seventy-three patients (53%) reported amenorrhea at enrollment, of whom 94% resumed menses within 12 months. Among 497 patients evaluable for pregnancy, 368 (74%) reported at least one pregnancy. Young age was the main factor associated with shorter time to pregnancy with cumulative incidences of pregnancy by 1 year of 63.5%, 54.3%, and 37.7% for patients age <35, 35-39, and 40-42 years, respectively. One hundred and seventy-nine patients (36%) had embryo/oocyte cryopreservation at diagnosis, of whom 68 reported embryo transfer after enrollment. Cryopreserved embryo transfer was the only ART associated with higher chance of pregnancy (odds ratio, 2.41 [95% CI, 1.75 to 4.95]). The cumulative incidence of BCFI events at 3 years was similar for women who underwent ovarian stimulation for cryopreservation at diagnosis, 9.7% (95% CI, 6.0 to 15.4), compared with those who did not, 8.7% (95% CI, 6.0 to 12.5). CONCLUSION In POSITIVE, fertility preservation using ovarian stimulation was not associated with short-term detrimental impact on cancer prognosis. Pregnancy rates were highest among those who underwent embryo/oocyte cryopreservation followed by embryo transfer.
Article
Full-text available
Background Adjuvant abemaciclib combined with endocrine therapy (ET) previously demonstrated clinically meaningful improvement in invasive disease-free survival (IDFS) and distant relapse-free survival (DRFS) in hormone receptor-positive, human epidermal growth factor receptor 2-negative, node-positive, high-risk early breast cancer at the second interim analysis, however follow-up was limited. Here, we present results of the prespecified primary outcome analysis and an additional follow-up analysis. Patients and methods This global, phase III, open-label trial randomized (1 : 1) 5637 patients to adjuvant ET for ≥5 years ± abemaciclib for 2 years. Cohort 1 enrolled patients with ≥4 positive axillary lymph nodes (ALNs), or 1-3 positive ALNs and either grade 3 disease or tumor ≥5 cm. Cohort 2 enrolled patients with 1-3 positive ALNs and centrally determined high Ki-67 index (≥20%). The primary endpoint was IDFS in the intent-to-treat population (cohorts 1 and 2). Secondary endpoints were IDFS in patients with high Ki-67, DRFS, overall survival, and safety. Results At the primary outcome analysis, with 19 months median follow-up time, abemaciclib + ET resulted in a 29% reduction in the risk of developing an IDFS event [hazard ratio (HR) = 0.71, 95% confidence interval (CI) 0.58-0.87; nominal P = 0.0009]. At the additional follow-up analysis, with 27 months median follow-up and 90% of patients off treatment, IDFS (HR = 0.70, 95% CI 0.59-0.82; nominal P < 0.0001) and DRFS (HR = 0.69, 95% CI 0.57-0.83; nominal P < 0.0001) benefit was maintained. The absolute improvements in 3-year IDFS and DRFS rates were 5.4% and 4.2%, respectively. Whereas Ki-67 index was prognostic, abemaciclib benefit was consistent regardless of Ki-67 index. Safety data were consistent with the known abemaciclib risk profile. Conclusion Abemaciclib + ET significantly improved IDFS in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative, node-positive, high-risk early breast cancer, with an acceptable safety profile. Ki-67 index was prognostic, but abemaciclib benefit was observed regardless of Ki-67 index. Overall, the robust treatment benefit of abemaciclib extended beyond the 2-year treatment period.
Article
Full-text available
Background Trastuzumab targets the extracellular domain of the HER2 protein. Adding trastuzumab to chemotherapy for patients with early-stage, HER2-positive breast cancer reduces the risk of recurrence and death, but is associated with cardiac toxicity. We investigated the long-term benefits and risks of adjuvant trastuzumab on breast cancer recurrence and cause-specific mortality. Methods We did a collaborative meta-analysis of individual patient data from randomised trials assessing chemotherapy plus trastuzumab versus the same chemotherapy alone. Randomised trials that enrolled women with node-negative or node-positive, operable breast cancer were included. We collected individual patient-level data on baseline characteristics, dates and sites of first distant breast cancer recurrence and any previous local recurrence or second primary cancer, and the date and underlying cause of death. Primary outcomes were breast cancer recurrence, breast cancer mortality, death without recurrence, and all-cause mortality. Standard intention-to-treat log-rank analyses, stratified by age, nodal status, oestrogen receptor (ER) status, and trial yielded first-event rate ratios (RRs). Findings Seven randomised trials met the inclusion criteria, and included 13 864 patients enrolled between February, 2000, and December, 2005. Mean scheduled treatment duration was 14·4 months and median follow-up was 10·7 years (IQR 9·5 to 11·9). The risks of breast cancer recurrence (RR 0·66, 95% CI 0·62 to 0·71; p<0·0001) and death from breast cancer (0·67, 0·61 to 0·73; p<0·0001) were lower with trastuzumab plus chemotherapy than with chemotherapy alone. Absolute 10-year recurrence risk was reduced by 9·0% (95% CI 7·4 to 10·7; p<0·0001) and 10-year breast cancer mortality was reduced by 6·4% (4·9 to 7·8; p<0·0001), with a 6·5% reduction (5·0 to 8·0; p<0·0001) in all-cause mortality, and no increase in death without recurrence (0·4%, –0·3 to 1·1; p=0·35). The proportional reduction in recurrence was largest in years 0–1 after randomisation (0·53, 99% CI 0·46 to 0·61), with benefits persisting through years 2–4 (0·73, 0·62 to 0·85) and 5–9 (0·80, 0·64 to 1·01), and little follow-up beyond year 10. Proportional recurrence reductions were similar irrespective of recorded patient and tumour characteristics, including ER status. The more high risk the tumour, the larger the absolute reductions in 5-year recurrence (eg, 5·7% [95% CI 3·1 to 8·3], 6·8% [4·7 to 9·0], and 10·7% [7·7 to 13·6] in N0, N1–3, and N4+ disease). Interpretation Adding trastuzumab to chemotherapy for early-stage, HER2-positive breast cancer reduces recurrence of, and mortality from, breast cancer by a third, with worthwhile proportional reductions irrespective of recorded patient and tumour characteristics. Funding Cancer Research UK, UK Medical Research Council.
Article
Full-text available
Background Poly(adenosine diphosphate–ribose) polymerase inhibitors target cancers with defects in homologous recombination repair by synthetic lethality. New therapies are needed to reduce recurrence in patients with BRCA1 or BRCA2 germline mutation–associated early breast cancer. Methods We conducted a phase 3, double-blind, randomized trial involving patients with human epidermal growth factor receptor 2 (HER2)–negative early breast cancer with BRCA1 or BRCA2 germline pathogenic or likely pathogenic variants and high-risk clinicopathological factors who had received local treatment and neoadjuvant or adjuvant chemotherapy. Patients were randomly assigned (in a 1:1 ratio) to 1 year of oral olaparib or placebo. The primary end point was invasive disease–free survival. Results A total of 1836 patients underwent randomization. At a prespecified event-driven interim analysis with a median follow-up of 2.5 years, the 3-year invasive disease–free survival was 85.9% in the olaparib group and 77.1% in the placebo group (difference, 8.8 percentage points; 95% confidence interval [CI], 4.5 to 13.0; hazard ratio for invasive disease or death, 0.58; 99.5% CI, 0.41 to 0.82; P<0.001). The 3-year distant disease–free survival was 87.5% in the olaparib group and 80.4% in the placebo group (difference, 7.1 percentage points; 95% CI, 3.0 to 11.1; hazard ratio for distant disease or death, 0.57; 99.5% CI, 0.39 to 0.83; P<0.001). Olaparib was associated with fewer deaths than placebo (59 and 86, respectively) (hazard ratio, 0.68; 99% CI, 0.44 to 1.05; P=0.02); however, the between-group difference was not significant at an interim-analysis boundary of a P value of less than 0.01. Safety data were consistent with known side effects of olaparib, with no excess serious adverse events or adverse events of special interest. Conclusions Among patients with high-risk, HER2-negative early breast cancer and germline BRCA1 or BRCA2 pathogenic or likely pathogenic variants, adjuvant olaparib after completion of local treatment and neoadjuvant or adjuvant chemotherapy was associated with significantly longer survival free of invasive or distant disease than was placebo. Olaparib had limited effects on global patient-reported quality of life. (Funded by the National Cancer Institute and AstraZeneca; OlympiA ClinicalTrials.gov number, NCT02032823.)
Article
PURPOSE To our knowledge, WSG-ADAPT-HR+/HER2– ( NCT01779206 ; n = 5,625 registered) is the first trial combining the 21-gene expression assay (recurrence score [RS]) and response to 3-week preoperative endocrine therapy (ET) to guide systemic therapy in early breast cancer. MATERIALS AND METHODS Baseline and postendocrine Ki67 (Ki67 post ) were evaluated centrally. In the endocrine trial, all patients received exclusively ET: patients with pathologic regional lymph node status (pN) 0-1 (ie, 0-3 involved lymph nodes) entered control arm if RS ≤ 11 and experimental arm if RS12-25 with ET response (Ki67 post ≤ 10%). All other patients (including N0-1 RS12-25 without ET response) received dose-dense chemotherapy (CT) followed by ET in the CT trial. Primary end point of the endocrine trial was noninferiority of 5-year invasive disease-free survival (5y-iDFS) in experimental ( v control) arm; secondary end points included distant DFS, overall survival, and translational research. RESULTS Intention-to-treat population comprised 2,290 patients (n = 1,422 experimental v n = 868 control): 26.3% versus 34.6% premenopausal and 27.4% versus 24.0% pN1. One-sided 95% lower confidence limit of the 5y-iDFS difference was –3.3%, establishing prespecified noninferiority ( P = .05). 5y-iDFS was 92.6% (95% CI, 90.8 to 94.0) in experimental versus 93.9% (95% CI, 91.8 to 95.4) in control arm; 5-year distant DFS was 95.6% versus 96.3%, and 5-year overall survival 97.3% versus 98.0%, respectively. Differences were similar in age and nodal subgroups. In N0-1 RS12-25, outcome of ET responders (ET alone) was comparable with that of ET nonresponders (CT) for age > 50 years and superior for age ≤ 50 years. ET response was more likely with aromatase inhibitors (mostly postmenopausal) than with tamoxifen (mostly premenopausal): 78.1% versus 41.1% ( P < .001). ET response was 78.8% in RS0-11, 62.2% in RS12-25, and 32.7% in RS > 25 (n = 4,203, P < .001). CONCLUSION WSG-ADAPT-HR+/HER2– demonstrates that guiding systemic treatment by both RS and ET response is feasible in clinical routine and spares CT in pre- and postmenopausal patients with ≤ 3 involved lymph nodes.
Article
Background The recurrence score based on the 21-gene breast-cancer assay has been clinically useful in predicting a chemotherapy benefit in hormone-receptor–positive, human epidermal growth factor receptor 2 (HER2)–negative, axillary lymph-node–negative breast cancer. In women with positive lymph-node disease, the role of the recurrence score with respect to predicting a benefit of adjuvant chemotherapy is unclear. Methods In a prospective trial, we randomly assigned women with hormone-receptor–positive, HER2-negative breast cancer, one to three positive axillary lymph nodes, and a recurrence score of 25 or lower (scores range from 0 to 100, with higher scores indicating a worse prognosis) to endocrine therapy only or to chemotherapy plus endocrine (chemoendocrine) therapy. The primary objective was to determine the effect of chemotherapy on invasive disease–free survival and whether the effect was influenced by the recurrence score. Secondary end points included distant relapse–free survival. Results A total of 5083 women (33.2% premenopausal and 66.8% postmenopausal) underwent randomization, and 5018 participated in the trial. At the prespecified third interim analysis, the chemotherapy benefit with respect to increasing invasive disease–free survival differed according to menopausal status (P=0.008 for the comparison of chemotherapy benefit in premenopausal and postmenopausal participants), and separate prespecified analyses were conducted. Among postmenopausal women, invasive disease–free survival at 5 years was 91.9% in the endocrine-only group and 91.3% in the chemoendocrine group, with no chemotherapy benefit (hazard ratio for invasive disease recurrence, new primary cancer [breast cancer or another type], or death, 1.02; 95% confidence interval [CI], 0.82 to 1.26; P=0.89). Among premenopausal women, invasive disease–free survival at 5 years was 89.0% with endocrine-only therapy and 93.9% with chemoendocrine therapy (hazard ratio, 0.60; 95% CI, 0.43 to 0.83; P=0.002), with a similar increase in distant relapse–free survival (hazard ratio, 0.58; 95% CI, 0.39 to 0.87; P=0.009). The relative chemotherapy benefit did not increase as the recurrence score increased. Conclusions Among premenopausal women with one to three positive lymph nodes and a recurrence score of 25 or lower, those who received chemoendocrine therapy had longer invasive disease–free survival and distant relapse–free survival than those who received endocrine-only therapy, whereas postmenopausal women with similar characteristics did not benefit from adjuvant chemotherapy. (Funded by the National Cancer Institute and others; RxPONDER ClinicalTrials.gov number, NCT01272037.)
Article
PURPOSE Patient-centered digital interventions may help empower young women to self-manage symptoms and psychosocial concerns and support informational needs often unaddressed in clinic. METHODS Young, Empowered and Strong (YES) is an interactive web-based intervention designed to engage young women with personalized education and symptom self-management resources on the basis of responses to patient-reported outcome–based questionnaires. We piloted YES among young women (< 45 years) with newly diagnosed early breast cancer (EBC) or metastatic breast cancer (MBC) and breast cancer survivors (BCSs). Assessments were deployed weekly (EBC and MBC) or every 4 weeks (BCSs) over 12 weeks. At study completion, use, feasibility, and acceptability of YES were evaluated via a survey and semistructured interview. RESULTS Thirty women were enrolled between April and June 2019: 10 EBC, 10 BCSs, and 10 MBC. The mean age at diagnosis and enrollment was 36 (range 25-44) and 39 (range 31-44) years, respectively. Most participants were actively treated (96%, 27 of 28) with endocrine therapy (54%, 15 of 28) or chemotherapy (43%, 12 of 28). Overall, 61% (180 of 296) of assessments were completed (EBC: 70%, BCSs: 63%, and MBC: 52%). Of 37 patient-reported outcome and need domains, the most frequently triggered were sexual health (EBC: 90%, BCSs: 90%, and MBC: 90%), anxiety (EBC: 80%, BCSs: 90%, and MBC: 90%), stress and mindfulness (EBC: 80%, BCSs: 90%, and MBC: 90%), and fatigue (EBC: 90%, BCSs: 80%, and MBC: 90%). On postpilot survey, participants reported that YES helped them to learn (50%, 7 of 14), monitor (43%, 6 of 14), and manage (57%, 8 of 14) their symptoms. CONCLUSION YES is a feasible and acceptable digital intervention to support young women across the breast cancer care continuum. The nearly universal triggering of sexual and mental health needs suggests suboptimal management in the clinical setting and the potential for self-management through a digital platform.
Article
PURPOSE Patients with triple-negative breast cancer (TNBC) and residual invasive disease (RD) after completion of neoadjuvant chemotherapy (NAC) have a high-risk for recurrence, which is reduced by adjuvant capecitabine. Preclinical models support the use of platinum agents in the TNBC basal subtype. The EA1131 trial hypothesized that invasive disease-free survival (iDFS) would not be inferior but improved in patients with basal subtype TNBC treated with adjuvant platinum compared with capecitabine. PATIENTS AND METHODS Patients with clinical stage II or III TNBC with ≥ 1 cm RD in the breast post-NAC were randomly assigned to receive platinum (carboplatin or cisplatin) once every 3 weeks for four cycles or capecitabine 14 out of 21 days every 3 weeks for six cycles. TNBC subtype (basal v nonbasal) was determined by PAM50 in the residual disease. A noninferiority design with superiority alternative was chosen, assuming a 4-year iDFS of 67% with capecitabine. RESULTS Four hundred ten of planned 775 participants were randomly assigned to platinum or capecitabine between 2015 and 2021. After median follow-up of 20 months and 120 iDFS events (61% of full information) in the 308 (78%) patients with basal subtype TNBC, the 3-year iDFS for platinum was 42% (95% CI, 30 to 53) versus 49% (95% CI, 39 to 59) for capecitabine. Grade 3 and 4 toxicities were more common with platinum agents. The Data and Safety Monitoring Committee recommended stopping the trial as it was unlikely that further follow-up would show noninferiority or superiority of platinum. CONCLUSION Platinum agents do not improve outcomes in patients with basal subtype TNBC RD post-NAC and are associated with more severe toxicity when compared with capecitabine. Participants had a lower than expected 3-year iDFS regardless of study treatment, highlighting the need for better therapies in this high-risk population.