MMC 1
MMC 1
MMC 1
and follow-up
SUPPLEMENTARY MATERIAL
Breast cancer screening programmes have been initiated for women at average risk
of developing breast cancer in order to optimise outcomes through early detection.
Screening in women with a strong family history or known germline BRCA1/2
mutations (gBRCA1/2m) and other high-risk pathogenic variants (PVs) should follow
the ESMO Clinical Practice Guideline (CPG) for risk reduction and screening of
cancer in hereditary breast-ovarian cancer syndromes (HBOC).1
1
Section 2. Diagnosis and imaging
• Familial breast cancer associated with gBRCA1/2m and other high-risk PVs
• Lobular cancers (including detection of extensive multifocality, contralateral
breast cancer or to plan conservative surgery)
• Suspicion of multifocality and/or multicentricity
• Discrepancy between conventional imaging and clinical examination
• When the findings of conventional imaging are inconclusive (e.g. a positive
axillary lymph node with an occult primary tumour)
• Presence of breast implants
Genetic counselling, testing and patient management for carriers of gBRCA1/2m and
other high-risk PVs should follow the ESMO CPG for risk reduction and screening of
cancer in HBOC.1
Germline PVs in BRCA1/2 account for 5%-10% of all breast cancers and are
associated with an elevated lifetime risk of developing breast and ovarian cancers,
as well as other malignancies.18 The use of multigene panel tests is increasing.
These tests yield an 8%-9% detection rate of PVs in genes associated with
increased breast cancer risk, about half of which are a gBRCA1/2m.19 Other relevant
high- and moderate-penetrance germline PVs are found in TP53, PALB2, ATM and
CHEK2, although management recommendations vary according to the mutation
and the gene penetrance is generally lower than for BRCA1/2.1
2
Identification of germline mutations informs disease management and follow-up;
patients with gBRCA1/2m and other PVs may opt for different locoregional breast
cancer management and contralateral surgical prophylaxis, and warrant more
intensive risk-reducing measures for other malignancies such as ovarian cancer. 1
High-risk patients with gBRCA1/2m may be candidates for adjuvant therapy with a
poly (ADP-ribose) polymerase (PARP) inhibitor.
Germline testing for PVs in BRCA1/2 should be offered for patients who meet the
respective national criteria and for those who are candidates for adjuvant olaparib
therapy.20,21
Ki-67 expression levels are most informative for prognosis of luminal breast cancer
when the level is ≤5% (low proliferation, low risk) or ≥30% (high proliferation, high
risk) because technical reliability of distinguishing values between 5% and 30% is
limited. A decrease of Ki-67 (≤10%) after short term (4-6 weeks) preoperative
endocrine therapy (ET) in postmenopausal patients with HR-positive, HER2-negative
breast cancer is prognostic and may indicate endocrine responsiveness.29,30
HER2-negative tumours with 1%-9% ER and/or PgR expression (ER low/PgR low)
behave similarly to and have similar clinical outcomes to triple-negative breast
cancers (TNBCs). Many HR 1%-9% IHC-positive cancers show molecular features
of HR-negative cancer and are therefore unlikely to benefit from ET. However, an
uncertain proportion of these patients with HR-low cancers may have a sufficient
level of HR expression to benefit from ET. In the absence of randomised clinical trial
evidence to demonstrate benefit from adjuvant ET, treatment recommendations
should be individualised based on patient preferences, with careful consideration of
the risk versus benefit from long-term ET. Tumours with low HR expression
constitute a biologically mixed entity and are considered HR low, not HR negative.32
Therefore, patients with HR 1%-9% IHC-positive cancers should be included in trials
designed for HR-negative cancers to generate more evidence.
4
by IHC and is defined as ER <1%, PgR <1% and HER2 score 0, 1 or 2+ with
negative FISH or chromogenic ISH.
For prognostication and treatment decision making, tumours can be grouped into
surrogate intrinsic subtypes,34 defined by routine histology and IHC data, as luminal
A like, luminal B like, HER2 positive and triple negative. Luminal A-like tumours are
typically low grade, strongly ER positive/PgR positive and HER2 negative and have
low proliferation. Luminal B-like tumours are HR positive but may have variable
degrees of ER/PgR expression, are higher grade and have higher proliferation than
Luminal A-like tumours. Gene expression assays (e.g. EndoPredict, MammaPrint,
Oncotype DX, Prosigna and others) have been introduced in many countries for
prognostic assessment of HR-positive, HER2-negative breast cancer. The assays
differ in terms of genes as well as technology, but all can identify a low- to
intermediate-risk group that might not benefit from adjuvant chemotherapy (ChT).
Gene expression tests should only be used in a clinically intermediate-risk situation
(based on IHC biomarkers and clinical parameters). It is recommended to use only
one assay for any individual patient.
Prognosis in patients who received neoadjuvant ChT and did not obtain a
pathological complete response (pCR) can be estimated by the pretreatment clinical
stage and post-treatment pathological stage plus ER status and tumour grade35,
which also has been validated for luminal tumours.36
5
neoadjuvant therapy—cannot be demonstrated in FNA samples. Microcalcifications,
mainly only visible on mammogram, should be biopsied with a vacuum-assisted
device.8 If systemic neoadjuvant therapy is likely/expected, marking the primary
tumour with a clip is recommended. If suspicious locoregional LNs are detected, at
least one in each region should be biopsied and preferably also marked.
When there is an intermediate risk after standard IHC and clinical assessment, a
gene expression test should be carried out in HR-positive, HER2-negative tumours
to identify low-risk tumours that would not benefit from treatment with ChT. Ki-67
analysis after short preoperative ET may add information about tumour endocrine
responsiveness; Ki-67 ≤10% post-ET is considered to indicate endocrine
responsiveness and low risk. If preoperative ET is used in HR-positive, HER2-
negative disease, gene expression analysis must be carried out in the diagnostic
pretreatment biopsy, not in the surgical specimen. PD-L1 testing in EBC is not
required for clinical decision making.
Disease stage and final pathological and clinical assessment of surgical specimens
should be made according to the WHO classification of tumours22 and the eighth
edition of the UICC TNM staging system.23 Minimum blood work-up (a full blood
count, liver and renal function tests, alkaline phosphatase and calcium levels) is
recommended before surgery and systemic (neo)adjuvant therapy. Chest, abdomen
and bone imaging is recommended for higher-risk patients (high tumour burden,
aggressive biology or clinical signs, symptoms or laboratory values suggesting the
presence of metastases). [18F]2-fluoro-2-deoxy-D-glucose (FDG)–positron emission
tomography (PET)–CT scanning may be useful for high-risk patients and when
conventional methods are inconclusive.
Validated gene expression profiles may complement pathology assessment and help
with adjuvant ChT decision making in clinically intermediate-risk situations.
6
Assessment of distant metastases (bone, liver and lung) is recommended only in
patients with stage IIb and higher disease (especially with extended LN
involvement), patients with a high risk of recurrence at first diagnosis and/or in
symptomatic patients.37,39 Brain imaging should not be routinely carried out in
asymptomatic patients. The minimum imaging work-up for staging includes
computed tomography (CT) of the chest and abdomen and bone scintigraphy. FDG–
PET–CT may be used instead of CT and bone scintigraphy.37
Particular attention must be paid to the treatment of EBC in special populations, e.g.
very young or elderly patients. However, age is a continuous variable and cut-offs in
studies are always arbitrarily chosen. For clinical decision making, biological age is
more important than chronological age.42
7
In younger premenopausal patients, fertility issues should be discussed and
guidance on fertility-preservation techniques should be provided before the initiation
of any systemic treatment.43-47 For details about fertility preservation, please refer to
the ESMO CPG for fertility preservation and post-treatment pregnancies in post-
pubertal cancer patients.48
As part of optimising breast cancer outcomes and survivorship, all patients should
have equitable access to the full range of reproductive care options that may be
necessary at different points in their breast cancer journey, including pregnancy
counselling, contraception and fertility preservation.
Elderly patients. Due to the limited data from randomised studies, strong
recommendations cannot be made regarding the use of (neo)adjuvant systemic
therapies in this population. Full doses of drugs should be used whenever feasible.
In patients suitable for standard ChT, single-agent capecitabine or docetaxel have
been demonstrated to be inferior to the standard multidrug regimen [doxorubicin–
cyclophosphamide [AC] or cyclophosphamide–methotrexate–fluorouracil (CMF)].49,50
Sequential, single-agent therapy (e.g. doxorubicin followed by paclitaxel and then
cyclophosphamide) is as effective as combination ChT (e.g. AC followed by
paclitaxel) and is associated with lower acute toxicity, making this approach an
option for any patient, especially older patients.51,52 A geriatric multidimensional
8
assessment should be carried out prior to treatment decisions; the G8 tool can be
used as a screening tool to select patients needing a full geriatric assessment.53
Male breast cancer. Most breast cancer cases in male patients are luminal-like
ductal invasive carcinoma and occur in older men. Tamoxifen is the standard
adjuvant ET; aromatase inhibitors (AIs) must be combined with gonadotropin
hormone-releasing hormone (GnRH) analogues.54 ChT and anti-HER2 therapy and
regimens using other targeted novel agents as indicated [e.g. immunotherapy,
cyclin-dependent kinase 4/6 (CDK4/6) or PARP inhibitors] should follow the same
recommendations as for female patients with breast cancer.55-57 Population-based
studies have indicated that ~20% of male patients with breast cancer will have an
inherited risk factor; therefore, genetic counselling and testing should be
considered.58
9
Supplementary Table S1. Summary of biomarkers used in standardised histopathological, immunohistochemical and
molecular pathology assessment in EBC
Classical Histological Pathological diagnosis and basic prognostic A TILs are not currently required for
H&E tumour type, characterisation of tumour therapy decisions but might become
pathology invasiveness, relevant in the future
grading, TILs
IHC and IHC panel: Standard IHC workup as a basis for major A HR-low tumours (ER/PgR 1%-9%) are
ISH ER, PgR, therapy decisions and prognostic assessment biologically TNBC and may behave as
HER2, Ki-67 such
Definition of intrinsic subtypes
ISH: for HER2 status should be reported as IHC
HER2 IHC 0, 1+ or 2+ (either negative or positive by
2+ ISH) and 3+
10
quality Ki-67 may also be evaluated after a short
assurance course of preoperative ET to indicate
programmes endocrine responsiveness (Ki-67 ≤10%)
NGS gBRCA1/2 Clinically important information for surgical A Testing is relevant for TNBC and HR-
testing management, follow-up and cancer screening positive, HER2-negative breast cancer
and has become a patient selection marker for
systemic adjuvant olaparib therapy in HER2-
negative tumours
EBC, early breast cancer; ER, estrogen receptor; ET, endocrine therapy; gBRCA1/2, germline BRCA1/2, GoR, grade of
recommendation; H&E, haematoxylin–eosin; HER2, human epidermal growth factor receptor 2; HR, hormone receptor; IHC,
immunohistochemistry; ISH, in situ hybridisation; MBC, metastatic breast cancer; NGS, next-generation sequencing; PgR,
progesterone receptor; TIL, tumour-infiltrating lymphocyte; TNBC, triple-negative breast cancer.
11
Supplementary Table S2. Clinical classification of breast tumours according to
the UICC TNM eighth edition
T (primary tumour)
Tis (Paget) Paget disease of the nipple not associated with invasive carcinoma
and/or carcinoma in situ (DCIS and/or LCIS) in the underlying breast
parenchyma. Carcinomas in the breast parenchyma associated with
Paget disease are categorised based on the size and characteristics
of the parenchymal disease, although the presence of Paget disease
should still be noted
T4 Tumour of any size with direct extension to chest wall and/or to skin
(ulceration or skin nodules)b
T4a Extension to chest wall (does not include pectoralis muscle invasion
only)
12
T4b Ulceration, ipsilateral satellite skin nodules or skin oedema (including
peau d'orange)
N (regional LNs)
N0 No regional LN metastasis
M (distant metastasis)
M0 No distant metastasis
M1 Distant metastasis
13
Excisional biopsy of a LN or biopsy of a sentinel node, in the absence of assignment
of a pT, is classified as a clinical N, e.g. cN1. Pathological classification (pN) is used
for excision or sentinel LN biopsy only in conjunction with a pathological T
assignment.
ALN, axillary lymph node; DCIS, ductal carcinoma in situ; FNA, fine-needle
aspiration; LCIS, lobular carcinoma in situ; LN, lymph node; Tis, carcinoma in situ;
TNM, tumour–node–metastasis; UICC, Union for International Cancer Control.
aMicroinvasion is the extension of cancer cells beyond the basement membrane into
the adjacent tissues with no focus >0.1 cm in greatest dimension. When there are
multiple foci of microinvasion, the size of only the largest focus is used to classify the
microinvasion. Do not use the sum of all individual foci. The presence of multiple foci
of microinvasion should be noted, as it is with multiple larger invasive carcinomas.
bInvasion of the dermis alone does not qualify as T4. Chest wall includes ribs,
intercostal muscles and serratus anterior muscle but not pectoral muscle.
cInflammatory carcinoma of the breast is characterised by diffuse, brawny induration
of the skin with an erysipeloid edge, usually with no underlying mass. If the skin
biopsy is negative and there is no localised measurable primary cancer, the T
category is pTX when pathologically staging a clinical inflammatory carcinoma (T4d).
Dimpling of the skin, nipple retraction or other skin changes, except those in T4b and
T4d, may occur in T1, T2 or T3 without affecting the classification.
dClinically detected is defined as detected by clinical examination or by imaging
studies (excluding lymphoscintigraphy) and having characteristics highly suspicious
for malignancy or a presumed pathological macrometastasis based on FNA biopsy
with cytological examination. Confirmation of clinically detected metastatic disease
by FNA without excision biopsy is designated with a (f) suffix, e.g. cN3a(f).
14
Supplementary Table S3. Pathological classification of breast tumours
according to the UICC pTNM eighth edition
pN1mi Micrometastases (>0.2 mm and/or >200 cells, but none >2.0 mm)
pN1a Metastasis in 1-3 axillary LN(s), including at least one >2 mm in greatest
dimension
pN2a Metastasis in 4-9 axillary LNs, including at least one that is >2 mm
15
pN3a Metastasis in ≥10 ipsilateral ALNs (at least one >2 mm) or metastasis in
infraclavicular lymph nodes
Post‐treatment ypN
16
cClinically detected is defined as detected by imaging studies (excluding
lymphoscintigraphy) or by clinical examination and having characteristics highly
suspicious for malignancy or a presumed pathological macrometastasis based on
FNA biopsy with cytological examination. Not clinically detected is defined as not
detected by imaging studies (excluding lymphoscintigraphy) or not detected by
clinical examination.
17
Supplementary Table S4. Staging of breast tumours according to the UICC
TNM eighth edition
Stage T N M
Stage 0 Tis N0 M0
Stage IA T1a N0 M0
T2 N0 M0
Stage IIB T2 N1 M0
T3 N0 M0
T3 N1, N2 M0
18
Supplementary Table S5. Overview of adjuvant therapy for HR-positive, HER2-negative EBC
T1c Tamoxifen 5 May consider for Low riski: may Low riski: no No No No
or AI higher riskh,i consider
N0
I especially if not
receiving OFS
19
T2-3 Tamoxifen 7-10 Considerh Low riski: Low riski: no No No Yes
or AI consider
N0
especially if not
receiving OFS
Higher riski:
High riski: yes High riski: yes
yes
III
AI, aromatase inhibitor; ChT, chemotherapy; EBC, early breast cancer; ER, estrogen receptor; ET, endocrine therapy; gBRCA1/2,
germline BRCA1/2; HER2, human epidermal growth factor receptor 2; HR, hormone receptor; LN, lymph node; m, mutation; N,
20
node; OFS, ovarian function suppression; OS, overall survival; PgR, progesterone receptor; RS, recurrence score; T, tumour; TN,
tumour–node.
aRisk stratification factors for ET include tumour size, extent of nodal involvement, tumour grade, degree of ER/PgR expression,
high Ki-67 or other proliferation measures and adverse genomic signature results. For lower-risk cancers, either tamoxifen or an AI
for 5 years is standard. With increased risk, AI becomes preferred to tamoxifen, longer durations become appropriate and OFS is
progressively added for younger women, especially those <35 years of age.
bFor node-negative tumours, non-anthracycline regimens may suffice as adjuvant ChT; anthracycline-, taxane- and alkylator-based
regimens are preferred for higher-stage cancers warranting ChT.
cAs adjuvant therapy for purposes of preventing distant metastatic recurrence.
dBenefits of ChT reflect tumour biology and menopausal status. Premenopausal women with lower-risk tumours who are not
advised/recommended to receive OFS may benefit more from ChT.
eThe role of ChT is largely determined by tumour pathobiology including high-risk genomic signature scores (preferred) or high
grade and/or high Ki-67 (>30%).
fLimited follow-up from a single study suggests that adjuvant abemaciclib may reduce recurrence in high-risk node-positive cancers
characterised by stage, grade and/or Ki-67 (>20%). In cases that are potential candidates for both abemaciclib and olaparib,
olaparib is preferred due to statistically significant OS benefit.
gAdjuvant olaparib is indicated in BRCA1 or BRCA2-associated cancers with high-risk features that would typically warrant ChT,
such as multiple positive LNs and/or residual disease after neoadjuvant ChT. In patients who are potential candidates for both
abemaciclib and olaparib, olaparib is preferred due to statistically significant OS benefit.
21
hUse of AI in premenopausal women demands OFS.
i‘Low risk’ implies low-risk genomic score (preferred; e.g. Endopredict 'Low'; MammaPrint ‘Low’ or 'Ultra Low'; Oncotype DX RS
≤15; Prosigna RS ≤60; luminal A on formal signature testing) and/or lower-risk features on traditional pathological analysis including
lower-grade histology, robust ER and PgR expression and lower measures of proliferation. ‘High risk’ implies high-risk genomic
score (Endopredict 'High'; MammaPrint ‘High’; Oncotype DX ≥26; Prosigna >60; luminal B) and/or higher-risk features on traditional
pathological analysis including higher-grade histology, lower ER and PgR expression and higher measures of proliferation.
jThere is no evidence of benefit for OFS beyond five years’ duration of therapy.
Table adapted with permission from St Gallen International Consensus Guidelines 2023.34 © 2023 European Society for Medical
Oncology. Published by Elsevier Ltd. All rights reserved.
22
Supplementary Table S6. Notable potential long-term side-effects related to adjuvant breast cancer treatmenta
Neuropathy
Myalgia
Exhaustion
Sleep disturbances
Ovarian dysfunction
Premature menopause
Sexual complaints
Lymphoedema
23
Others Bone density loss
Cardiac dysfunction
24
Supplementary Table S7. Biomarkers and molecular targets for precision medicines and corresponding ESCAT scores
25
HER2 (ERBB2) HER2 negative IHC (0, 1+ or 2+) with ET and concomitant I-A20,21,b
negative negative FISH/CISH adjuvant olaparib
26
Stage II-III HER2 positive IHC (3+) or Neoadjuvant systemic NA
ChT and anti-HER2
Positive FISH/CISH I-A65,c
therapy
N+ disease HER2 positive IHC (3+) or Adjuvant HP-ChT NA
27
ER negative IHC (<1%) Taxane–carboplatin in NA
sequence with AC or
PgR negative IHC (<1%)
EC followed by taxane
HER2 negative IHC (0, 1, or 2+) with
negative FISH/CISH
28
AC, doxorubicin–cyclophosphamide; AI, aromatase inhibitor; ChT, chemotherapy; CISH, chromogenic in situ hybridisation; EC,
epirubicin–cyclophosphamide; ER, estrogen receptor; ESCAT, ESMO Scale for Clinical Actionability of molecular Targets; ET,
endocrine therapy; gBRCA1/2, germline BRCA1/2; HER2, human epidermal growth factor receptor 2; HP, trastuzumab–
pertuzumab; HR, hormone receptor; IHC, immunohistochemistry; m, mutation; OFS, ovarian function suppression; N, node;
NA, not applicable; NGS, next-generation sequencing; pCR, pathological complete response; PgR, progesterone receptor; T-
DM1, trastuzumab–emtansine; TNBC, triple-negative breast cancer.
aESCAT scores apply to alterations from genomic-driven analyses only. These scores have been defined by the guideline authors
and assisted as needed by the ESMO Translational Research and Precision Medicine Working Group. ESCAT defines the
relevance of a genomic alteration to a matched targeted therapy and does not apply to prognostic use or to guide local or systemic
cytotoxic agents.
bI-A, alteration–drug match is associated with improved outcome with evidence from randomised clinical trials showing the
alteration–drug match in a specific tumour type results in a clinically meaningful improvement of a survival endpoint. 38
cESCAT applicable if HER2 gene amplification by FISH/CISH.
29
Supplementary Table S8. ESMO-MCBS table for therapies/indications in EBC
Therapy Disease setting Trial Control Absolute Hazard ratio QoL/toxicity ESMO-MCBS
survival gain (95% CI) scorea
30
HER2- Phase III 5-year iDFS: 5-year iDFS iDFS: 0.73 (40% versus
overexpressed/amplified 87.7% gain: 2.5% (0.57-0.92) 2%)
breast cancer, to follow NCT00878709
adjuvant trastuzumab- 8-year OS: 8-year OS OS: 0.95
based therapy 90.2% gain: -0.1% (0.75-1.21)
ChT + 1 year Adjuvant treatment of APHINITY62,76,7 ChT + 1 year Increased A
of adult patients with 7 of placebo– grade 3 (Form 1)
pertuzumab– HER2-positive EBC at trastuzumab diarrhoea
trastuzumab high risk of recurrence Phase III during ChT
(node-positive or HR- 6-year iDFS: 6-year iDFS iDFS: 0.76 (9.8% versus
negative disease) NCT01358877 87.8% gain: 2.8% (0.64-0.91) 3.7%)
31
cancer at high risk of
recurrence
Trastuzumab Patients with HER2- HERA79,80 ChT A
(1 year) positive EBC following (Form 1)
surgery, ChT Phase III 2-year DFS: 2-year DFS DFS: 0.54
(neoadjuvant or 77.4% gain: 8.4% (0.43-0.67)
adjuvant) and RT (if NCT00045032
applicable)
T-DM1 Adjuvant treatment of KATHERINE63 Trastuzumab A
patients with HER2- (Form 1)
positive EBC who have Phase III 3-year iDFS: 3-year iDFS iDFS: 0.50
residual invasive 77.0% gain: 11.3% (0.39-0.64)
disease in the breast NCT01772472
and/or LNs nodes after
neoadjuvant taxane-
based and HER2-
targeted therapy
Pembrolizuma Neoadjuvant treatment, KEYNOTE- Placebo– A
b–paclitaxel– and then continued as 52281,82 paclitaxel– (Form 1)
carboplatin monotherapy as carboplatin
32
adjuvant treatment after Phase III 3-year EFS: 3-year EFS EFS: 0.63
surgery, for the 76.8% gain: 7.7% (0.48-0.82)
treatment of adults with NCT03036488
locally advanced, or pCR: 51.2% pCR gain:
early-stage TNBC at 13.6%
high risk of recurrence 3-year OS: 3-year OS OS: NS
86.9% gain: 2.8% (immature)
ChT, chemotherapy; CI, confidence interval; DFS, disease-free survival; EBC, early breast cancer; EFS, event-free survival; EMA,
European Medicines Agency; ESMO-MCBS, ESMO-Magnitude of Clinical Benefit Scale; ET, endocrine therapy; FDA, Food and
Drug Administration; gBRCA1/2, germline BRCA1/2; HER2, human epidermal growth factor receptor 2; HR, hormone receptor;
iDFS, invasive disease-free survival; ITT, intent to treat; LN, lymph node; NEB, no evaluable benefit; NS, not significant; OS, overall
survival; pCR, pathological complete response; QoL, quality of life; RT, radiotherapy; T-DM1, trastuzumab–emtansine; TNBC,
triple-negative breast cancer.
aESMO-MCBS v1.183 was used to calculate scores for therapies/indications approved by the EMA or FDA. The scores have been
calculated and validated by the ESMO-MCBS Working Group and reviewed by the authors (https://www.esmo.org/guidelines/esmo-
mcbs/esmo-mcbs-evaluation-forms).
bEMA approval was based on data from Cohort 1 of the monarchE study; however, analysis of Cohort 1 was not a prespecified
endpoint and the data are therefore not eligible for ESMO-MCBS scoring. Scoring is based on the ITT analysis.
c98.5% CI.
33
dEMA approval is restricted to patients with hormone receptor-positive HER2-positive breast cancer who completed adjuvant
trastuzumab-based therapy <1 year. This is based on an unplanned post hoc subgroup analysis and is not eligible for ESMO-
MCBS grading. Mature OS was not statistically significant.
34
Supplementary Table S9. Levels of evidence and grades of recommendation
(adapted from the Infectious Diseases Society of America-United States Public
Health Service Grading Systema)
Levels of evidence
Grades of recommendation
B Strong or moderate evidence for efficacy but with a limited clinical benefit,
generally recommended
C Insufficient evidence for efficacy or benefit does not outweigh the risk or the
disadvantages (adverse events, costs, etc.), optional
D Moderate evidence against efficacy or for adverse outcome, generally not
recommended
E Strong evidence against efficacy or for adverse outcome, never
recommended
aBy permission of Oxford University Press on behalf of the Infectious Diseases
Society of America.84
35
REFERENCES
1. Sessa C, Balmaña J, Bober SL, et al. Risk reduction and screening of cancer
in hereditary breast-ovarian cancer syndromes: ESMO Clinical Practice Guideline.
Ann Oncol. 2023;34(1):33-47.
2. European Commission. European Commission Initiatives on Breast and
Colorectal Cancer. Accessed March 02, 2023, https://healthcare-
quality.jrc.ec.europa.eu/
3. Duffy S, Vulkan D, Cuckle H, et al. Annual mammographic screening to
reduce breast cancer mortality in women from age 40 years: long-term follow-up of
the UK Age RCT. Health Technol Assess. 2020;24(55):1-24.
4. Duffy SW, Tabar L, Yen AM, et al. Mammography screening reduces rates of
advanced and fatal breast cancers: Results in 549,091 women. Cancer.
2020;126(13):2971-2979.
5. Duffy SW, Tabar L, Yen AM, et al. Beneficial Effect of Consecutive Screening
Mammography Examinations on Mortality from Breast Cancer: A Prospective Study.
Radiology. 2021;299(3):541-547.
6. Duffy SW, Vulkan D, Cuckle H, et al. Effect of mammographic screening from
age 40 years on breast cancer mortality (UK Age trial): final results of a randomised,
controlled trial. Lancet Oncol. 2020;21(9):1165-1172.
7. Houssami N, Hofvind S, Soerensen AL, et al. Interval breast cancer rates for
digital breast tomosynthesis versus digital mammography population screening: An
individual participant data meta-analysis. EClinicalMedicine. 2021;34:100804.
8. 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(1):46-56.
9. World Health Organization. WHO position paper on mammography screening.
WHO Press. Accessed July 4, 2023, https://www.who.int/publications/i/item/who-
position-paper-on-mammography-screening
10. Marmot MG, Altman DG, Cameron DA, Dewar JA, Thompson SG, Wilcox M.
The benefits and harms of breast cancer screening: an independent review. Br J
Cancer. 2013;108(11):2205-2240.
36
11. Siu AL, U. S. Preventive Services Task Force. Screening for Breast Cancer:
U.S. Preventive Services Task Force Recommendation Statement. Ann Intern Med.
2016;164(4):279-296.
12. Duggan C, Trapani D, Ilbawi AM, et al. National health system characteristics,
breast cancer stage at diagnosis, and breast cancer mortality: a population-based
analysis. Lancet Oncol. 2021;22(11):1632-1642.
13. Bakker MF, de Lange SV, Pijnappel RM, et al. Supplemental MRI Screening
for Women with Extremely Dense Breast Tissue. N Engl J Med. 2019;381(22):2091-
2102.
14. Comstock CE, Gatsonis C, Newstead GM, et al. Comparison of Abbreviated
Breast MRI vs Digital Breast Tomosynthesis for Breast Cancer Detection Among
Women With Dense Breasts Undergoing Screening. JAMA. 2020;323(8):746-756.
15. Veenhuizen SGA, de Lange SV, Bakker MF, et al. Supplemental Breast MRI
for Women with Extremely Dense Breasts: Results of the Second Screening Round
of the DENSE Trial. Radiology. 2021;299(2):278-286.
16. Perry N, Broeders M, De Wolf C, Törnberg S, Holland R, Von Karsa L.
European Commission: European guidelines for quality assurance in breast cancer
screening and diagnosis. Luxembourg: Office of Official Publications of the European
Communities. 2006;
17. Ditsch N, Kolberg-Liedtke C, Friedrich M, et al. AGO Recommendations for
the Diagnosis and Treatment of Patients with Early Breast Cancer: Update 2021.
Breast Care (Basel). 2021;16(3):214-227.
18. Breast Cancer Association Consortium, Dorling L, Carvalho S, et al. Breast
Cancer Risk Genes - Association Analysis in More than 113,000 Women. N Engl J
Med. 2021;384(5):428-439.
19. Kurian AW, Ward KC, Abrahamse P, et al. Time Trends in Receipt of
Germline Genetic Testing and Results for Women Diagnosed With Breast Cancer or
Ovarian Cancer, 2012-2019. J Clin Oncol. 2021;39(15):1631-1640.
20. 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(25):2394-2405.
21. Geyer CE, Jr., Garber JE, Gelber RD, et al. Overall survival in the OlympiA
phase III trial of adjuvant olaparib in patients with germline pathogenic variants in
BRCA1/2 and high-risk, early breast cancer. Ann Oncol. 2022;33(12):1250-1268.
37
22. International Agency for Research on Cancer. Breast Tumours, WHO
Classification of Tumours. 5th ed. vol 9. IARC Press; 2019.
23. Brierley JD, Gospodarowicz MK, Wittekind C. Union for International Cancer
Control. TNM Classification of Malignant Tumours. 8th ed. John Wiley & Sons, Inc.;
2016.
24. Loi S, Michiels S, Adams S, et al. The journey of tumor-infiltrating
lymphocytes as a biomarker in breast cancer: clinical utility in an era of checkpoint
inhibition. Ann Oncol. 2021;32(10):1236-1244.
25. Allison KH, Hammond MEH, Dowsett M, et al. Estrogen and Progesterone
Receptor Testing in Breast Cancer: American Society of Clinical Oncology/College of
American Pathologists Guideline Update. Arch Pathol Lab Med. 2020;144(5):545-
563.
26. Wolff AC, Hammond MEH, Allison KH, et al. Human Epidermal Growth Factor
Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College
of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol.
2018;36(20):2105-2122.
27. Nielsen TO, Leung SCY, Rimm DL, et al. Assessment of Ki67 in Breast
Cancer: Updated Recommendations From the International Ki67 in Breast Cancer
Working Group. J Natl Cancer Inst. 2021;113(7):808-819.
28. Salgado R, Denkert C, Demaria S, et al. The evaluation of tumor-infiltrating
lymphocytes (TILs) in breast cancer: recommendations by an International TILs
Working Group 2014. Ann Oncol. 2015;26(2):259-271.
29. Nitz UA, Gluz O, Kümmel S, et al. Endocrine Therapy Response and 21-Gene
Expression Assay for Therapy Guidance in HR+/HER2– Early Breast Cancer. J Clin
Oncol. 2022;40(23):2557-2567.
30. Smith I, Robertson J, Kilburn L, et al. Long-term outcome and prognostic
value of Ki67 after perioperative endocrine therapy in postmenopausal women with
hormone-sensitive early breast cancer (POETIC): an open-label, multicentre,
parallel-group, randomised, phase 3 trial. Lancet Oncol. 2020;21(11):1443-1454.
31. Denkert C, Seither F, Schneeweiss A, et al. Clinical and molecular
characteristics of HER2-low-positive breast cancer: pooled analysis of individual
patient data from four prospective, neoadjuvant clinical trials. Lancet Oncol.
2021;22(8):1151-1161.
38
32. Villegas SL, Nekljudova V, Pfarr N, et al. Therapy response and prognosis of
patients with early breast cancer with low positivity for hormone receptors - An
analysis of 2765 patients from neoadjuvant clinical trials. Eur J Cancer.
2021;148:159-170.
33. Boman C, Zerdes I, Martensson K, et al. Discordance of PD-L1 status
between primary and metastatic breast cancer: A systematic review and meta-
analysis. Cancer Treat Rev. 2021;99:102257.
34. Curigliano G, Burstein HJ, Gnant M, et al. Understanding breast cancer
complexity to improve patient outcomes: The St Gallen International Consensus
Conference for the Primary Therapy of Individuals with Early Breast Cancer 2023.
Ann Oncol. 2023;In press. 10.1016/j.annonc.2023.08.017.
35. Mittendorf EA, Jeruss JS, Tucker SL, et al. Validation of a novel staging
system for disease-specific survival in patients with breast cancer treated with
neoadjuvant chemotherapy. J Clin Oncol. 2011;29(15):1956-1962.
36. Marmé F, Lederer B, Blohmer JU, et al. Utility of the CPS+EG staging system
in hormone receptor-positive, human epidermal growth factor receptor 2-negative
breast cancer treated with neoadjuvant chemotherapy. Eur J Cancer. 2016;53:65-74.
37. Gennari A, André F, Barrios CH, et al. ESMO Clinical Practice Guideline for
the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann
Oncol. 2021;32(12):1475-1495.
38. Mateo J, Chakravarty D, Dienstmann R, et al. A framework to rank genomic
alterations as targets for cancer precision medicine: the ESMO Scale for Clinical
Actionability of molecular Targets (ESCAT). Ann Oncol. 2018;29(9):1895-1902.
39. Gerber B, Seitz E, Müller H, et al. Perioperative screening for metastatic
disease is not indicated in patients with primary breast cancer and no clinical signs of
tumor spread. Breast Cancer Res Treat. 2003;82(1):29-37.
40. Cardoso F, Cataliotti L, Costa A, et al. European Breast Cancer Conference
manifesto on breast centres/units. Eur J Cancer. 2017;72:244-250.
41. Wilson AR, Marotti L, Bianchi S, et al. The requirements of a specialist Breast
Centre. Eur J Cancer. 2013;49(17):3579-3587.
42. Paluch-Shimon S, Cardoso F, Partridge AH, et al. ESO-ESMO 4th
International Consensus Guidelines for Breast Cancer in Young Women (BCY4).
Ann Oncol. 2020;31(6):674-696.
39
43. Senkus E, Gomez H, Dirix L, et al. Attitudes of young patients with breast
cancer toward fertility loss related to adjuvant systemic therapies. EORTC study
10002 BIG 3-98. Psychooncology. 2014;23(2):173-182.
44. Lee SJ, Schover LR, Partridge AH, et al. American Society of Clinical
Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol.
2006;24(18):2917-2931.
45. Paluch-Shimon S, Pagani O, Partridge AH, et al. ESO-ESMO 3rd international
consensus guidelines for breast cancer in young women (BCY3). Breast.
2017;35:203-217.
46. 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(18):3355-3377.
47. Peccatori FA, Azim HA, Jr., Orecchia R, et al. Cancer, pregnancy and fertility:
ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann
Oncol. 2013;24 Suppl 6:vi160-170.
48. 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(12):1664-1678.
49. Muss HB, Berry DA, Cirrincione CT, et al. Adjuvant chemotherapy in older
women with early-stage breast cancer. N Engl J Med. 2009;360(20):2055-2065.
50. Perrone F, Nuzzo F, Di Rella F, et al. Weekly docetaxel versus CMF as
adjuvant chemotherapy for older women with early breast cancer: final results of the
randomized phase III ELDA trial. Ann Oncol. 2015;26(4):675-682.
51. Crivellari D, Gray KP, Dellapasqua S, et al. Adjuvant pegylated liposomal
doxorubicin for older women with endocrine nonresponsive breast cancer who are
NOT suitable for a "standard chemotherapy regimen": the CASA randomized trial.
Breast. 2013;22(2):130-137.
52. Citron ML, Berry DA, Cirrincione C, et al. Randomized trial of dose-dense
versus conventionally scheduled and sequential versus concurrent combination
chemotherapy as postoperative adjuvant treatment of node-positive primary breast
cancer: first report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial
9741. J Clin Oncol. 2003;21(8):1431-1439.
40
53. Biganzoli L, Battisti NML, Wildiers H, et al. Updated recommendations
regarding the management of older patients with breast cancer: a joint paper from
the European Society of Breast Cancer Specialists (EUSOMA) and the International
Society of Geriatric Oncology (SIOG). Lancet Oncol. 2021;22(7):e327-e340.
54. Reinisch M, Seiler S, Hauzenberger T, et al. Efficacy of Endocrine Therapy for
the Treatment of Breast Cancer in Men: Results from the MALE Phase 2
Randomized Clinical Trial. JAMA Oncol. 2021;7(4):565-572.
55. 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(2):405-417.
56. Korde LA, Zujewski JA, Kamin L, et al. Multidisciplinary meeting on male
breast cancer: summary and research recommendations. J Clin Oncol.
2010;28(12):2114-2122.
57. Sousa B, Moser E, Cardoso F. An update on male breast cancer and future
directions for research and treatment. Eur J Pharmacol. 2013;717(1-3):71-83.
58. Pensabene M, Von Arx C, De Laurentiis M. Male Breast Cancer: From
Molecular Genetics to Clinical Management. Cancers. 2022;14(8):2006.
59. Gronchi A, Miah AB, Dei Tos AP, et al. Soft tissue and visceral sarcomas:
ESMO-EURACAN-GENTURIS Clinical Practice Guidelines for diagnosis, treatment
and follow-up. Ann Oncol. 2021;32(11):1348-1365.
60. Raj SD, Shurafa M, Shah Z, Raj KM, Fishman MDC, Dialani VM. Primary and
Secondary Breast Lymphoma: Clinical, Pathologic, and Multimodality Imaging
Review. Radiographics. 2019;39(3):610-625.
61. Eichenauer DA, Aleman BMP, André M, et al. Hodgkin lymphoma: ESMO
Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol.
2018;29(Suppl 4):iv19-iv29.
62. von Minckwitz G, Procter M, de Azambuja E, et al. Adjuvant Pertuzumab and
Trastuzumab in Early HER2-Positive Breast Cancer. N Engl J Med.
2017;377(2):122-131.
63. von Minckwitz G, Huang CS, Mano MS, et al. Trastuzumab Emtansine for
Residual Invasive HER2-Positive Breast Cancer. N Engl J Med. 2019;380(7):617-
628.
41
64. Tolaney SM, Tarantino P, Graham N, et al. Adjuvant paclitaxel and
trastuzumab for node-negative, HER2-positive breast cancer: final 10-year analysis
of the open-label, single-arm, phase 2 APT trial. Lancet Oncol. 2023;24(3):273-285.
65. Gianni L, Pienkowski T, Im YH, et al. 5-year analysis of neoadjuvant
pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or
early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label,
phase 2 randomised trial. Lancet Oncol. 2016;17(6):791-800.
66. Loibl S, Jassem J, Sonnenblick A, et al. VP6-2022: Adjuvant pertuzumab and
trastuzumab in patients with early HER-2 positive breast cancer in APHINITY: 8.4
years' follow-up. Ann Oncol. 2022;33(9):986-987.
67. Martin M, Holmes FA, Ejlertsen B, et al. Neratinib after trastuzumab-based
adjuvant therapy in HER2-positive breast cancer (ExteNET): 5-year analysis of a
randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol.
2017;18(12):1688-1700.
68. Chan A, Moy B, Mansi J, et al. Final Efficacy Results of Neratinib in HER2-
positive Hormone Receptor-positive Early-stage Breast Cancer From the Phase III
ExteNET Trial. Clin Breast Cancer. 2021;21(1):80-91 e87.
69. Johnston SRD, Harbeck N, Hegg R, et al. Abemaciclib Combined With
Endocrine Therapy for the Adjuvant Treatment of HR+, HER2-, Node-Positive, High-
Risk, Early Breast Cancer (monarchE). J Clin Oncol. 2020;38(34):3987-3998.
70. Rugo HS, O'Shaughnessy J, Boyle F, et al. Adjuvant abemaciclib combined
with endocrine therapy for high-risk early breast cancer: safety and patient-reported
outcomes from the monarchE study. Ann Oncol. 2022;33(6):616-627.
71. Harbeck N, Rastogi P, Martin M, et al. Adjuvant abemaciclib combined with
endocrine therapy for high-risk early breast cancer: updated efficacy and Ki-67
analysis from the monarchE study. Ann Oncol. 2021;32(12):1571-1581.
72. Johnston SR, Toi M, O'Shaughnessy J, et al. Abemaciclib plus endocrine
therapy for hormone receptor-positive, HER2-negative, node-positive, high-risk early
breast cancer (monarchE): results from a preplanned interim analysis of a
randomised, open-label, phase 3 trial. The Lancet Oncology. 2023;24(1):77-90.
73. Chan A, Delaloge S, Holmes FA, et al. Neratinib after trastuzumab-based
adjuvant therapy in patients with HER2-positive breast cancer (ExteNET): a
42
multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet
Oncol. 2016;17(3):367-377.
74. Delaloge S, Cella D, Ye Y, et al. Effects of neratinib on health-related quality
of life in women with HER2-positive early-stage breast cancer: longitudinal analyses
from the randomized phase III ExteNET trial. Ann Oncol. 2019;30(4):567-574.
75. Holmes FA, Moy B, Delaloge S, et al. Overall survival with neratinib after
trastuzumab-based adjuvant therapy in HER2-positive breast cancer (ExteNET): A
randomised, double-blind, placebo-controlled, phase 3 trial. Eur J Cancer.
2023;184:48-59.
76. Piccart M, Procter M, Fumagalli D, et al. Adjuvant Pertuzumab and
Trastuzumab in Early HER2-Positive Breast Cancer in the APHINITY Trial: 6 Years'
Follow-Up. J Clin Oncol. 2021;39(13):1448-1457.
77. Bines J, Clark E, Barton C, et al. Patient-reported function, health-related
quality of life, and symptoms in APHINITY: pertuzumab plus trastuzumab and
chemotherapy in HER2-positive early breast cancer. Br J Cancer. 2021;125(1):38-
47.
78. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant
pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early
HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label,
phase 2 trial. Lancet Oncol. 2012;13(1):25-32.
79. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after
adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med.
2005;353(16):1659-1672.
80. Cameron D, Piccart-Gebhart MJ, Gelber RD, et al. 11 years' follow-up of
trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final
analysis of the HERceptin Adjuvant (HERA) trial. Lancet. 2017;389(10075):1195-
1205.
81. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for Early Triple-Negative
Breast Cancer. N Engl J Med. 2020;382(9):810-821.
82. Schmid P, Cortes J, Dent R, et al. Event-free Survival with Pembrolizumab in
Early Triple-Negative Breast Cancer. N Engl J Med. 2022;386(6):556-567.
83. Cherny NI, Dafni U, Bogaerts J, et al. ESMO-Magnitude of Clinical Benefit
Scale version 1.1. Ann Oncol. 2017;28(10):2340-2366.
43
84. Dykewicz CA. Summary of the Guidelines for Preventing Opportunistic
Infections among Hematopoietic Stem Cell Transplant Recipients. Clin Infect Dis.
2001;33(2):139-144 [adapted from: Gross PA, Barrett TL, Dellinger EP, et al.
Purpose of quality standards for infectious diseases. Clin Infect Dis. 1994;18(3):421].
44