CC BY-NC-ND 4.0 · Geburtshilfe Frauenheilkd 2022; 82(06): 580-589
DOI: 10.1055/a-1811-6106
GebFra Science
Review/Übersicht

Update Breast Cancer 2022 Part 1 – Early Stage Breast Cancer

Article in several languages: English | deutsch
Manfred Welslau
1   Onkologie Aschaffenburg, Aschaffenburg, Germany
,
Volkmar Müller
2   Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
,
Diana Lüftner
3   Charité University Hospital, Department of Hematology, Oncology and Tumour Immunology, University Medicine Berlin, Berlin, Germany
,
Florian Schütz
4   Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
,
Elmar Stickeler
5   Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Aachen, Germany
,
Peter A. Fasching
6   Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
,
Wolfgang Janni
7   Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
,
Christoph Thomssen
8   Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
,
Isabell Witzel
2   Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
,
Tanja N. Fehm
9   Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
,
Erik Belleville
10   ClinSol GmbH & Co KG, Würzburg, Germany
,
Simon Bader
6   Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
,
Katharina Seitz
6   Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
,
Michael Untch
11   Clinic for Gynecology and Obstetrics, Breast Cancer Center, Genecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
,
Marc Thill
12   Agaplesion Markus Krankenhaus, Department of Gynecology and Gynecological Oncology, Frankfurt am Main
,
Hans Tesch
13   Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
,
Nina Ditsch
14   Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
,
Michael P. Lux
15   Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Germany
,
Bahriye Aktas
16   Klinik und Poliklinik für Gynäkologie, Universitätsklinikum Leipzig, Leipzig, Germany
,
Maggie Banys-Paluchowski
17   Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
,
Andreas Schneeweiss
18   National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
,
Nadia Harbeck
19   Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
,
Rachel Würstlein
19   Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
,
Andreas D. Hartkopf
7   Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
,
Achim Wöckel
20   Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
,
Barbara Seliger
21   Martin-Luther-Universitat Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany
,
Chiara Massa
21   Martin-Luther-Universitat Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany
,
Hans-Christian Kolberg
22   Department of Gynecology and Obstetrics, Marienhospital Bottrop, Bottrop, Germany
› Author Affiliations
 

Abstract

Evidence relating to the treatment of breast cancer patients with early-stage disease has increased significantly in the past year. Abemaciclib, olaparib, and pembrolizumab are new drugs with good efficacy in the relevant patient groups. However, some questions remain unanswered. In particular, it remains unclear which premenopausal patients with hormone receptor-positive breast cancer should be spared unnecessary treatment. The question of the degree to which chemotherapy exerts a direct cytotoxic effect on the tumor or reduces ovarian function through chemotherapy could be of key importance. This group of patients could potentially be spared chemotherapy. New, previously experimental biomarker analysis methods, such as spatial analysis of gene expression (spatial transcriptomics) are gradually finding their way into large randomized phase III trials, such as the NeoTRIP trial. This in turn leads to a better understanding of the predictive factors of new therapies, for example immunotherapy. This review summarizes the scientific innovations from recent congresses such as the San Antonio Breast Cancer Symposium 2021 but also from recent publications.


#

Introduction

Recent scientific findings relating to the prevention and treatment of patients with early-stage breast cancer refer mainly to the recently introduced new drug classes of CDK4/6, PARP and immune checkpoint inhibitors. In addition, many studies address the question of whether treatment can be de-escalated without loss of efficacy. This includes both surgical trials, such as the INSEMA trial, and trials assessing the use of multigene testing of affected lymph nodes, as in the RxPONDER trial. Knowledge of prevention is improving, and about 40% of heritable familial risk can be explained by genetics. There is a need to develop definitive new prevention strategies, also in the context of non-genetic modifiable risk factors. These and other issues are discussed below.


#

Prevention, BRCA1/2 and Germline Testing

Prevention and germline gene variants

With the advent of PARP inhibitors in the treatment of patients with breast carcinoma in both early and metastatic disease [1], [2], [3], [4], [5], genetic testing for germline mutations in the BRCA1/2 genes is now considered necessary in most national and international treatment recommendations [6], [7], [8]. Studies in subgroups of patients with a lower mutation frequency indicate that testing is also useful in these patient groups [1], [9], [10], [11]. In unselected patients with advanced breast cancer, it was not possible to identify relevant subgroups with mutation frequencies below 3% [9]. Only patients over the age of 60 and patients with a G1 tumor had lower mutation frequencies, however these always exceeded 0.5%, which is still considered a threshold for meaningful testing in lung cancer and a BRAF mutation.

The PALB2 gene also has high penetrance and carries a lifetime risk of breast cancer similar to the risk of a BRCA2 mutation. PALB2 is among the panel genes that are commonly tested [12], [13], [14]. Preliminary evidence exists that olaparib would have clinically relevant activity in patients with a PALB2 mutation [15].


#

Prevention interventions

In recent years, more and more genetic causes associated with familial breast cancer risk have been discovered [16], [17], [18], [19], [20], [21], [22], and approximately 40% of the twofold increase in familial breast cancer risk can be explained [23]. However, the majority of breast cancer cases cannot be explained by genetic factors. In recent years, little has changed in the recommendations for the prevention of this disease. The relevant studies and their effects have recently been summarized [24], [25]. It is important to note that the prevention trials with tamoxifen, anastrozole, and exemestane did not demonstrate a reduction in mortality from breast cancer. Indeed, there were cumulatively more deaths (n = 58) in the anti-hormonal prevention groups than in the placebo arms (n = 49) ([Table 1]). Although the numbers of deaths is very low, no trend can be identified. By contrast, the Womens Health Initiative (WHI) trial showed that breast cancer mortality was reduced in the group of women who had undergone hysterectomy. In the WHI trial, hysterectomized women were randomized to an estrogen monotherapy (n = 5310) arm and a placebo (n = 5429) arm [26]. In the estrogen monotherapy group, the incidence of breast cancer was reduced (hazard ratio [HR] = 0.78, 95% CI: 0.65 – 0.93), as was breast cancer-related mortality (HR = 0.60; 95% CI: 0.37 – 0.97) [24], [26]. It must be emphasized that in the group of women who had not undergone hysterectomy, who were randomized to either an estrogen + progesterone replacement group or a placebo group, the incidence of breast cancer was increased in the estrogen + progesterone group. Furthermore, there was an increase in breast cancer-related mortality and the incidence of cardiovascular disease [27]. This led to a dramatic decrease in the use of hormone replacement therapies after publication in 2002.

Table 1 Results of chemoprevention trial endpoints for breast cancer.

Trial (n)

Intervention

Breast cancer incidence

Death from breast cancer

Reference

Placebo

Intervention

RR or HR1

Placebo

Intervention

HR2

1 Relative risk (RR) for the Royal Marsden trial and hazard ratio (HR) for all other studies.

2 Hazard ratios (HR) not reported. Note the low case numbers.

NSABP-P1 (n = 13 388)

Tamoxifen

250

145

0.57 (0.46 – 0.70)

11

12

NR

[67]

Royal Marsden (n = 2494)

Tamoxifen

104

82

0.78 (0.58 – 1.04)

9

12

NR

[68]

IBIS1 (n = 7154)

Tamoxifen

350

251

0.70 (0.60 – 0.83)

26

31

NR

[69]

IBIS2 (n = 3864)

Anastrozole

165

85

0.51 (0.39 – 0.66)

3

2

NR

[70]

MAP3 (n = 4560)

Exemestane

32

11

0.35 (0.18 – 0.70)

0

1

NR

[71]

Nonetheless, in light of these studies with long-term follow-up, it should be noted that antiestrogenic medication did not reduce breast cancer mortality, whereas a trial with estrogen therapy showed a possible reduction in mortality. The corresponding results cannot be transferred into practice; they represent the current state of knowledge for pharmacological prevention.


#

Vaccines as prevention

Another issue that is undoubtedly coming back into focus in light of vaccination in the COVID-19 pandemic is the attempt to prevent or treat breast cancer through vaccination. In this context, much of the discussion is based on antigens that are probably unique to breast cancer or on those that are measured individually (neo-antigens). These, in turn, are based on the genomic variants and aberrations that occur in the pathogenesis of the disease. It is known that a relevant proportion of breast cancers elicit an immune response that has an effect on treatment efficacy and prognosis [28], [29], [30]. Vaccines against SARS-CoV-2 have been developed extremely quickly. The platforms used for this purpose were initially developed, among other things, for the rapid production of cancer vaccines [31], [32], [33]. It is now hoped that the leap in development and the available data on the safety of such a vaccine can significantly advance this field of research. Indeed, several trials involving an mRNA approach are either in the planning stage or already under way [34]. Parallel efforts are being undertaken to identify tumor antigens or neo-antigens from cancers that may help identify targets for vaccination [35], [36], [37]. It remains to be seen how each type of cancer would respond to a modified immune system and whether such technology can be used for prevention.


#
#

De-escalation of Axillary Surgery

Axillary lymph node dissection is one of the main causes of long-term sequelae such as edema or functional limitation of the arm on the operative side. However, some trials have already demonstrated good local and regional tumor control in the absence of axillary lymph node dissection [38], [39]. In the INSEMA trial from GBG, AGO-B and ABCSG, this question was assessed using a randomized study design ([Fig. 1]). The quality of life data from this trial, which were collected using the EORTC-QLQ-C30 and BR23 questionnaires [38], have now been reported.

Zoom Image
Fig. 1 Study design of the INSEMA trial.

The INSEMA trial analyzed the consequences of omitting a surgical assessment of lymph node status by sentinel node biopsy (SLNB) in the cN0 group. Over 1000 patients had no axillary intervention and over 4000 patients underwent sentinel node biopsy (first randomization). The group of patients with a positive sentinel lymph node was subject to a second randomization (complete axillary dissection vs. no further treatment).

With respect to the first randomization, the omission of SLNB did not result in a clinically significant improvement in overall quality of life [38]. However, there were significant differences in terms of the arm symptom score on the EORTC-QLQ-BR23 questionnaire. In the long term, patients who did not undergo axillary surgery had a mean score of approximately 16. In contrast, patients who underwent surgery had a mean score of approximately 20 [38].

With respect to the second randomization, patients who underwent the less invasive procedure also fared significantly better in terms of their arm symptom score. Patients with SLNB alone had values of approximately 20 – 22, while patients who underwent complete axillary lymph node dissection had mean values of approximately 28 – 30 [38].

The benefits of a less invasive approach in terms of quality of life have thus been described. The extent to which this has an impact on prognosis will be determined by future analyses of the primary and secondary objectives of the INSEMA trial.


#

Patients with Hormone Receptor-Positive Breast Cancer in Early Stages of Disease

Aromatase inhibitors or tamoxifen with and without ovarian suppression in the context of adjuvant treatment in premenopausal patients

One issue that has puzzled scientists for more than a decade is which hormone receptor-positive premenopausal patients should receive ovarian function suppressor (OFS) treatment in the context of adjuvant treatment. Previous studies have shown that the addition of OFS to tamoxifen may improve disease-free survival and that treatment with aromatase inhibitors and OFS may have other advantages over treatment with tamoxifen and OFS. However, treatment escalation is associated with adverse effects and a reduction in quality of life. Therefore, OFS or treatment with aromatase inhibitors plus OFS is recommended only in patients at high risk of recurrence (e.g., after chemotherapy), whereas patients at lower risk of recurrence should preferably be treated with tamoxifen monotherapy. In patients at intermediate risk of recurrence, treatment with tamoxifen and OFS is recommended [6], [8]. However, it is often difficult to define risk categories with precision and it is up to the interdisciplinary tumor conference to determine the classification on an individual basis.

A meta-analysis by the Early Breast Cancer Trialists Collaborative Group (EBCTCG) was recently published, comparing patients who were all treated with OFS and then randomized to treatment with either tamoxifen or aromatase inhibitors [40]. The analysis included 7030 patients enrolled in the following studies: ABCSG-12, SOFT, TEXT, HOBOE. The median follow-up period was 8.0 years. It was shown that the recurrence rate with OFS plus aromatase inhibitors versus OFS plus tamoxifen was reduced from 17.5% to 14.7% at 10 years (RR = 0.79; 95% CI: 0.69 – 0.90). Although there was improvement in distant metastasis-free survival with a relative risk of 0.83 (95% CI: 0.71 – 0.97), there was no improvement in terms of overall survival. The mortality rate at 10 years was 7.2% with tamoxifen plus OFS and 6.8% with aromatase inhibitors plus OFS (RR = 1.01; 95% CI: 0.82 – 1.24).

Interestingly, the effect was observed only in years 2 – 4 after surgery and in patients with up to 3 affected lymph nodes. The effect was not detectable in patients with more than 4 affected lymph nodes (RR = 1.03; 95% CI: 0.73 – 1.46) [40]. However, only 729 patients were assigned to this cohort, so no firm conclusions regarding risk subgroups should be made on the basis of this analysis.

Similarly, in the context of premenopausal patients with hormone receptor-positive breast cancer, a new analysis of the SOFT and TEXT trials was published. This analysis had an even longer follow-up period than previous publications. Essentially, two questions were addressed by the analyses of the SOFT and TEXT trials, the first being the addition of OFS to tamoxifen and the second being the analysis of tamoxifen vs. aromatase inhibitors. The median follow-up period was 12 years for the SOFT trial and 13 years for the TEXT trial [41]. This analysis further demonstrated that overall survival was improved with the addition of OFS. Subgroup analyses showed that this effect was greatest in the cohort at highest risk of recurrence (e.g., status after [neo]adjuvant chemotherapy, age < 35 years, more than 3 affected lymph nodes, grading 3) [41]. No difference was detected in patients at low risk of recurrence. It should be noted that these subgroup analyses are based in part on very low case numbers. Only 103, 103, and 126 deaths occurred in the tamoxifen alone, tamoxifen plus OFS, and exemestane plus OFS groups, respectively [41].


#

Chemotherapy in premenopausal patients

Modern biomarker studies use multigene assays to investigate whether chemotherapy can be avoided in some patients with hormone receptor-positive breast cancer (MINDACT, TailorX, RxPONDER, ADAPT). They have all shown that in young patients < 50 years of age or in premenopausal patients, chemotherapy resulted in some improvement in prognosis even in terms of conventional cut-offs, whereas in postmenopausal patients with an appropriate risk score, there were no differences between patients who had received only antihormone treatment and those who had received additional chemotherapy [42], [43], [44].

The reasons for this finding are unknown. However, if it is due to the effect of chemotherapy on ovarian function (premature ovarian failure), another option would be to investigate whether ovarian suppression can be administered to premenopausal patients in order to spare them chemotherapy. Similarly, the results of the ADAPT trial showed that young women with up to 3 affected lymph nodes, a recurrence score ≤ 25, and a good response (Ki67post ≤ 10%) to a short course of preoperative endocrine treatment had excellent 5-year metastasis-free survival. This suggests that chemotherapy could be omitted if the efficacy of endocrine treatment is optimal [45].

In an analysis of the German SUCCESS trials, 39% of more than 1150 premenopausal patients under 45 years of age no longer had menstrual bleeding after chemotherapy [46]. The proportion of patients of approximately 40% corresponds to the prognostic effect in the multigene studies, but this is only a hypothesis and remains to be proven. In this context, analyses such as those presented at the San Antonio Breast Cancer Symposium are of particular importance. In this regard, data from the RxPONDER trial have been analyzed in detail as a post-hoc analysis of the subgroup of premenopausal patients (n = 1654) [47]. In the recently published study, which had a median follow-up of 6.1 years (previously 5.3 years), the chemotherapy benefit for premenopausal patients in terms of disease-free survival (HR = 0.64; 95% CI: 0.47 – 0.87) and distant metastasis-free survival (HR = 0.66; 95% CI: 0.45 – 0.97) confirmed the benefit of chemotherapy plus endocrine treatment compared with endocrine treatment alone [47]. After chemotherapy, approximately 75% of women in the chemotherapy arm and approximately 50% in the endocrine monotherapy arm had no menstrual bleeding in the first 6 months after randomization. Although no formal analysis was carried out on this question, there appears to be no difference between patients receiving endocrine treatment alone vs. chemotherapy, especially in patients in whom menstrual bleeding persisted. In patients whose menstrual bleeding had stopped, there appeared to be a modest benefit in the chemotherapy group ([Fig. 2]).

Zoom Image
Fig. 2 Invasive disease-free survival in the RxPONDER trial in the premenopausal patient group. ET: endocrine treatment, CT → ET: Chemotherapy and endocrine treatment (data from [47]).

The question for future studies is how to identify the group of premenopausal patients in whom chemotherapy can be avoided without worsening the prognosis. Until then, chemotherapy in this group must be considered on a case-by-case basis, depending on patient and disease characteristics. In this regard, it may be helpful to determine the endocrine response by means of a short course of preoperative endocrine treatment, similar to the approach used in the ADAPT trials.


#

CDK4/6 inhibitors in the adjuvant setting

Despite the negative outcome of the PALLAS trial, which investigated additional adjuvant therapy with 2 years of palbociclib [48], the final analysis of 469 events (previously 313) has now been reported [49]. There was no change with respect to the primary endpoint (invasive disease-free survival). The hazard ratio was 0.96 (95% CI: 0.81 – 1.14). The subgroup analysis also failed to identify any group that demonstrated particularly high efficacy. Previously, it had been discussed that the risk profile in the study population may not have been set high enough to show a difference. Again, this could not be confirmed by the current subgroup analysis. Moreover, it appeared that patients with negative lymph nodes derived greater benefit from taking palbociclib (HR: 0.63; 95% CI: 0.37 – 1.08) compared with patients with positive lymph nodes (hazard ratios ranging from 0.89 to 1.09) [49]. The adjuvant MonarchE trial had already yielded positive results, following which abemaciclib was approved for adjuvant treatment of high-risk patients in the United States [50]. The first, early analyses of the adjuvant ribociclib Natalee/TRIO-033 trial are expected in late 2022 [51]. In Germany, the ADAPTcycle and ADAPTlate trials on the significance of CDK 4/6i in early breast cancer are still ongoing.

In Europe, regulatory approval of abemaciclib is expected in the near future (as of March 2022), as the EMA has already made a recommendation [52]. The recommendation for the appropriate indication is the combination of abemaciclib with endocrine treatment in HER2-negative, hormone receptor-positive breast cancer with positive lymph node status and high risk of recurrence. Thus, widespread adoption of abemaciclib in patients with early breast cancer is anticipated in the near future. In particular, if this treatment becomes more widely used, issues such as treatment adherence and the management of toxicity will undoubtedly become the focus of adjuvant endocrine-based treatment once again. A recent publication described discontinuation rates due to side effects in addition to quality of life. In the abemaciclib arm, discontinuation rates due to adverse events for abemaciclib were 18.5% [53]. Aromatase inhibitors are also known to be discontinued at a similar frequency in the context of adjuvant treatment [54]. The extent to which abemaciclib or the aromatase inhibitor contributes to this is a subject for future research.


#

Selective estrogen receptor degraders

The selective estrogen receptor degraders (SERDs) are currently being investigated in many clinical trials in the metastatic setting and early disease stages [55], [56]. Since fulvestrant was not developed for adjuvant treatment [57], it is hoped that oral SERDs will further improve adjuvant endocrine treatment. Adjuvant trials such as liDERA/TRIO-045 or AMEERA-6 are already recruiting. Therefore, it is important to collect data to study the mode of action of SERDs in patients with early-stage disease. One such trial is the neoadjuvant coopERA trial, which compared the aromatase inhibitor anastrozole with the SERD giredestrant. Here, final analysis of Ki-67 reduction at 2 weeks showed that giredestrant resulted in a relative reduction of Ki-67 by 75% and anastrazole by 67%. This difference was statistically significant (p = 0.043) [58]. After the positive EMERALD trial [59] and the positive coopERA trial [58], there is a strong rationale for investigating SERDs in the early stages of the disease as well.


#
#

Immunotherapy in Early-Stage Disease

The latest biomarker analysis methods (spatial transcriptomics) have entered clinical trials

The NeoTRIP trial is a biomarker study that uses a methodological principle for biomarkers that will be used more and more in the years to come. For this reason, a brief description of the analysis is included here: When multiple genes are analyzed (as is the case in standard multigene assays), analytical methods have traditionally relied on lysing a portion of the tumor in its entirety and analyzing a sample of all the cells it contains. This can be optimized in terms of tumor content by dissecting different areas of the tumor, but ultimately it was not possible to exclude the presence of cells from the tumor environment in the analysis. With the development of new analytical methods, it is now possible to investigate a variety of markers in situ, in preserved tissue sections, or by spatial labeling of the biomaterial in a locally differentiated manner within the tumor [60], [61].

This makes it possible to assign expression signals to certain cell types, but also to investigate the spatial arrangement of certain cell types in relation to each other [60], [61]. These methods are referred to as “spatial genomics” or “spatial transcriptomics”. [Fig. 3] shows an example of how such techniques can be applied to a histological section in breast cancer.

Zoom Image
Fig. 3 Example of a multiplex analysis of several biomarkers and possible analysis methods for spatial arrangement (spatial proteomics, image courtesy of Barbara Seliger und Chiara Massa, Halle).

Such analyses have now been performed as part of the NeoTRIP trial, which compared a regimen of carboplatin and nab-paclitaxel with a regimen of carboplatin, nab-paclitaxel, and atezolizumab [62]. A total of 43 markers that can differentiate between immune cells and epithelial cells were assessed using an antibody- and mass spectroscopy-based system [63] in 237 of the 280 patients included. Indeed, two patterns could be identified that predicted a high pCR rate (approximately 65%) and a low pCR rate (approximately 25 – 35%) in patients treated with atezolizumab, whereas no predictive value was demonstrated for the spatial pattern in patients treated with chemotherapy alone [62].

This type of biomarker analysis will become more widely available in the years to come and will almost certainly provide new insights into tumor biology, particularly in terms of how certain cell types interact. In terms of understanding immune interactions between tumor cells and immune cells in the tumor and lymph nodes, these techniques will provide new, and hopefully clinically relevant, insights into tumor-microenvironment interactions.


#

Sensitivity analyses to confirm the effects of immunotherapy

Pembrolizumab has already been approved in the United States for neoadjuvant/adjuvant treatment of patients with triple-negative breast cancer in the early stages of disease based on an analysis of event-free survival [64], [65]. There was an absolute increase of 7.7% in event-free survival at 3 years (from 76.8% with platinum-containing chemotherapy without pembrolizumab to 84.5% with the addition of pembrolizumab) [64]. This corresponded to a hazard ratio of 0.63 (95% CI: 0.48 – 0.82) [64]. Various sensitivity analyses have now been performed to test the robustness of this analysis. In addition to events such as local recurrence, distant metastases or death, secondary cancer events, initiation of therapy for metastases or positive incision margins at surgery were included. These sensitivity analyses showed no evidence of higher or lower efficacy of pembrolizumab. All hazard ratios ranged from 0.63 to 0.65 [66].

Subgroup analyses by tumor stage or nodal status also showed no relevant differences in terms of the efficacy of pembrolizumab. The hazard ratio in nodal-negative patients was 0.58 (95% CI: 0.37 – 0.91) and in nodal-positive patients was 0.65 (95% CI: 0.46 – 0.91). Stratification by AJCC disease stage showed a hazard ratio of 0.60 (95% CI: 0.42 – 0.86) in stage II and 0.68 (95% CI: 0.45 – 1.03) in stage III [66].


#
#

Survival Analyses of the OlympiA Trial

Adjuvant therapy with olaparib confers an overall survival benefit

The OlympiA trial was designed to investigate whether 12 months of adjuvant therapy with olaparib could improve invasive disease-free survival (iDFS) in patients at high risk of recurrence in the early stages of disease. In the initial publication in 2021, it was shown that iDFS was improved with a HR of 0.58 (95% CI: 0.41 – 0.82), with a median follow-up of 2.5 years and 1836 randomized patients [1]. Regarding overall survival, the interim analysis showed no statistically significant difference (HR: 0.68; 95% CI: 0.44 – 1.05; p = 0.02) [1]. The next interim analysis has now been published, with a median follow-up of 3.5 years [5]. The median PFS is now 0.63 (95% CI: 0.50 – 0.78). In terms of overall survival, this interim analysis has now reported statistical significance with a hazard ratio of 0.68 (95% CI: 0.47 – 0.97, p = 0.009) [5]. On the basis of these data, adjuvant therapy with olaparib was approved in the United States on March 11, 2022. It is certain to become established as a new therapeutic standard in Europe following approval.


#
#

Future Perspectives

Some of the current issues are highly relevant because they have a direct effect on quality of life and the effectiveness of current treatments. There is an urgent need for clarity as to whether and which patients will benefit from chemotherapy, especially premenopausal patients with hormone receptor-positive breast cancer. This is where scientific efforts should be focused. In terms of new treatments, major adjuvant trials with SERDs are underway and other trials have been running for a long time. The question of whether trastuzumab-deruxtecan confers an advantage over T-DM1 in the HER2-positive post-neoadjuvant setting is currently being investigated as part of the DESTINY-B05/Trudy/AGO-B50 trial. The Astefania trial is investigating whether the effect of T-DM1 in this setting can be improved by immunotherapy with atezolizumab. Whether the anti-TROP2 ADC sacituzumab-govitecan is a good adjunct to the current regimen in triple-negative patients in the post-neoadjuvant setting is currently being investigated as part of the SASCIA trial. Individualized post-neoadjuvant therapies that are adapted to the genetic variants of resistant residual tumor tissue after neoadjuvant chemotherapy are also being investigated in innovative study designs with extensive translational support programs.

The next few years are expected to bring new discoveries that will improve treatment and, in some cases, reduce the need for unnecessary and toxic treatments that adversely impact quality of life.


#
#

Conflict of Interest/Interessenkonflikt

B. A. received honoria and travel grants from AstraZeneca, Gilead, Genomic Health, Roche, Novartis, Celgene, Lilly, MSD, Eisai, Teva, Tesaro, Daiichi Sankyo and Pfizer. M. B.-P. received honoria for lectures and advisory role from AstraZeneca, Gilead, Roche, Novartis, Eli Lilly, MSD, Eisai, pfm, Amgen, Seagen, Daiichi-Sankyo and Pfizer, and study support from Mammotome, Endomag and Merit Medical. E. B. received honoraria from Novartis, Hexal, BMS, Lilly, Pfizer, Roche, MSD, BBraun and onkowissen.de for consulting, clinical research management or medical education activities. S. B. has no conflict of interest. N. D. has received honoraria from MSD, Roche, AstraZeneca, Teva, Pfizer, Novartis, Seagen,Gilead, MCI Healthcare. P. A. F. reports personal fees from Novartis, grants from Biontech, personal fees from Pfizer, personal fees from Daiichi Sankyo, personal fees from AstraZeneca, personal fees from Eisai, personal fees from Merck Sharp & Dohme, grants from Cepheid, personal fees from Lilly, personal fees from Pierre Fabre, personal fees from SeaGen, personal fees from Roche, personal fees from Hexal, personal fees from Agendia, personal fees from Gilead. T. N. F. has participated on advisory boards for Amgen, Daiichi Sankyo, Novartis, Pfizer, and Roche and has received honoraria for lectures from Amgen, Celgene, Daiichi Sankyo, Roche, Novartis and Pfizer. A. D. H. received speaker and consultancy honoraria from AstraZeneca, Genomic Health, Roche, Novartis, Celgene, Lilly, MSD, Eisai, Teva, Tesaro, Daiichi Sankyo, Hexal and Pfizer. N. H. received honoraria for lectures and/or consulting from Amgen, AstraZeneca, Daiichi Sankyo, Exact Sciences, Gilead, Lilly, MSD, Mylan, Novartis, Pierre-Fabre, Pfizer, Roche, Sandoz, Seagen. W. J. has received research Grants and/or honoraria from Sanofi-Aventis, Daiichi Sankyo, Novartis, Roche, Pfizer, Lilly, AstraZeneca, Chugai, GSK, Eisai, Cellgene and Johnson & Johnson. H.-C. K. has received honoraria from Pfizer, Novartis, Roche, Genomic Health/Exact Sciences, Amgen, AstraZeneca, Riemser, Carl Zeiss Meditec, Teva, Theraclion, Janssen-Cilag, GSK, LIV Pharma, Lily, SurgVision, Onkowissen, Gilead, Daiichi Sankyo and MSD, travel support from Carl Zeiss Meditec, LIV Pharma, Novartis, Amgen, Pfizer, Daiichi Sankyo, Tesaro and owns stock of Theraclion SA and Phaon Scientific GmbH. D. L. received honoraria from Amgen, AstraZeneca, Eli Lilly, Gilead, GSK, Loreal, MSD, Novartis, Onkowissen, Pfizer, Seagen, Teva. M. P. L. has participated on advisory boards for AstraZeneca, Lilly, MSD, Novartis, Pfizer, Eisai, Gilead, Exact Sciences, Pierre Fabre, Grünenthal, Daiichi Sankyo, PharmaMar and Roche and has received honoraria for lectures from MSD, Lilly, Roche, Novartis, Pfizer, Exact Sciences, Daiichi Sankyo, Grünenthal, Gilead, AstraZeneca, and Eisai. He is editorial board member of medactuell from medac. V. M. received speaker honoraria from Amgen, AstraZeneca, Daiichi Sankyo, Eisai, GSK, Pfizer, MSD, Medac, Novartis, Roche, Teva, Seagen, Onkowissen, high5 Oncology, Medscape, Gilead. Consultancy honoraria from Hexal, Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi Sankyo, Eisai, Lilly, Sanofi, Seagen, Gilead. Institutional research support from Novartis, Roche, Seagen, Genentech. Travel grants: Roche, Pfizer, Daiichi Sankyo. E. S. received honoraria from Roche, Celgene, AstraZeneca, Novartis, Pfizer, Tesaro, Aurikamed GmbH, MCI Deutschland GmbH, bsh medical communications GmbH, Onkowissen TV. A. S. received research grants from Celgene, Roche, honoraria from Amgen, AstraZeneca, Aurikamed, Bayer, Celgene, Clinsol, Connectmedica, Gilead, GSK, I-MED, Lilly, MCI Deutschland, Metaplan, MSD, Nanostring, Novartis, Onkowissen.de, Promedicis, Pfizer, Pierre Fabre, Roche, Seagen, Streamedup, Teva, Tesaro, Thieme and travel support from Celgene, Pfizer, Roche. F. S. participated on advisory boards for Novartis, Lilly, Amgen and Roche and received honoraria for lectures from Roche, AstraZeneca, MSD, Novartis and Pfizer. K. S. has no conflict of interest. H. T. received honoraria from Novartis, Roche, Celgene, Teva, Pfizer, AstraZeneca and travel support from Roche, Celgene and Pfizer. C. T. received honoraria for advisory boards and lectures from Amgen, AstraZeneca, Celgene, Daiichi Sankyo, Eisai, Gilead, Lilly, MSD, Mylan, Nanostring, Novartis, Pfizer, Pierre Fabre, Puma, Roche, Seagen, Vifor. M. T. has participated on advisory boards for AstraZeneca, Clovis, Daiichi Sanyo, Eisai, Gilead Science, GSK, Lilly, MSD, Novartis, Organon, Pfizer, Exact Sciences, Pierre-Fabre, Seagen and Roche and has received honoraria for lectures from Amgen, Clovis, Daiichi Sankyo, Eisai, GSK, Lilly, MSD, Roche, Novartis, Organon, Pfizer, Seagen, Exact Sciences, Viatris, Vifor and AstraZeneca and has received trial funding by Exact Sciences and Endomag Manuscript support was done by Amgen, ClearCut, pfm medical, Roche, Servier, Vifor. M. U. all honoraria went to the institution/employer: Abbvie, Amgen, AstraZeneca, Celgene, Daiichi Sankyo, Eisai, Lilly, MSD Merck, Mundipharma, Myriad Genetics, Pfizer, PUMA Biotechnology, Roche, Sanofi Aventis, Novartis, Pierre Fabre. M. W. has participated on advisory boards for AstraZeneca, Lilly, MSD, Novartis, Pfizer and Roche. I. W. has participated on advisory boards for Novartis, Daichii Sankyo, Lilly, Pfizer and received speaker honoraria from Astra Zeneca, Daichii Sankyo, MSD, Novartis, Pfizer, Roche. A. W. participated on advisory boards for Novartis, Lilly, Amgen, Pfizer, Roche, Tesaro, Eisai and received honoraria for lectures from Novartis, Pfizer, Aurikamed, Roche, Celgene. R. W. has received honoraria, travel support from Agendia, Amgen, Aristo, AstraZeneca, Boehringer Ingelheim, Carl Zeiss, Celgene, Daiichi Sankyo, Eisai, Exact Sciences, Genomic Health, Gilead, GlaxoSmithKline, Hexal, Lilly, Medstrom Medical, MSD, Mundipharma, Mylan, Nanostring, Novartis, Odonate, Paxman, Palleos, Pfizer, Pierre Fabre, PumaBiotechnology, Riemser, Roche, Sandoz/Hexal, Sanofi Genzyme, Seattle Genetics/Seagen, Tesaro Bio, Teva, Veracyte, Viatris./
B. A. hat von AstraZeneca, Gilead, Genomic Health, Roche, Novartis, Celgene, Lilly, MSD, Eisai, Teva, Tesaro, Daiichi Sankyo und Pfizer Honorare und Reisekostenzuschüsse erhalten. M. B.-P. hat von AstraZeneca, Gilead, Roche, Novartis, Eli Lilly, MSD, Eisai, pfm, Amgen, Seagen, Daiichi Sankyo und Pfizer Honorare für Vorlesungen und die Ausübung einer beratenden Funktion sowie von Mammotome, Endomag und Merit Medical Studienbeihilfen erhalten. E. B. hat von Novartis, Hexal, BMS, Lilly, Pfizer, Roche, MSD, BBraun und onkowissen.de für Beratung, klinisches Forschungsmanagement oder medizinische Aus- und Weiterbildung Honorare erhalten. S. B. hat keinen Interessenkonflikt. N. D. hat von MSD, Roche, AstraZeneca, Teva, Pfizer, Novartis, Seagen, Gilead und MCI Healthcare Honorare erhalten. P. A. F. weist Folgendes aus: Honorare von Novartis, Zuwendungen von Biontech, Honorare von Pfizer, Honorare von Daiichi Sankyo, Honorare von AstraZeneca, Honorare von Eisai, Honorare von Merck Sharp & Dohme, Zuwendungen von Cepheid, Honorare von Lilly, Honorare von Pierre Fabre, Honorare von SeaGen, Honorare von Roche, Honorare von Hexal, Honorare von Agendia und Honorare von Gilead. T. N. F. hat in beratenden Gremien bei Amgen, Daiichi Sankyo, Novartis, Pfizer und Roche mitgewirkt und von Amgen, Celgene, Daiichi Sankyo, Roche, Novartis und Pfizer Honorare für Vorlesungen erhalten. A. D. H. hat als Referent und Berater von AstraZeneca, Genomic Health, Roche, Novartis, Celgene, Lilly, MSD, Eisai, Teva, Tesaro, Daiichi Sankyo, Hexal und Pfizer Honorare erhalten. N. H. hat für Vorlesungen und/oder Beratung von Amgen, AstraZeneca, Daiichi Sankyo, Exact Sciences, Gilead, Lilly, MSD, Mylan, Novartis, Pierre-Fabre, Pfizer, Roche, Sandoz und Seagen Honorare erhalten. W. J. hat von Sanofi-Aventis, Daiichi Sankyo, Novartis, Roche, Pfizer, Lilly, AstraZeneca, Chugai, GSK, Eisai, Cellgene und Johnson & Johnson Forschungsbeihilfen und/oder Honorare erhalten. H.-C. K. hat von Pfizer, Novartis, Roche, Genomic Health/Exact Sciences, Amgen, AstraZeneca, Riemser, Carl Zeiss Meditec, Teva, Theraclion, Janssen-Cilag, GSK, LIV Pharma, Lily, SurgVision, Onkowissen, Gilead, Daiichi Sankyo und MSD Honorare sowie von Carl Zeiss Meditec, LIV Pharma, Novartis, Amgen, Pfizer, Daiichi Sankyo und Tesaro Reisekostenzuschüsse erhalten und besitzt Aktien von Theraclion SA und Phaon Scientific GmbH. D. L. hat von Amgen, AstraZeneca, Eli Lilly, Gilead, GSK, Loreal, MSD, Novartis, Onkowissen, Pfizer, Seagen und Teva Honorare erhalten. M. P. L. hat in beratenden Gremien bei AstraZeneca, Lilly, MSD, Novartis, Pfizer, Eisai, Gilead, Exact Sciences, Pierre Fabre, Grünenthal, Daiichi Sankyo, PharmaMar und Roche mitgewirkt und von MSD, Lilly, Roche, Novartis, Pfizer, Exact Sciences, Daiichi Sankyo, Grünenthal, Gilead, AstraZeneca und Eisai Honorare für Vorlesungen erhalten. Er ist Mitglied der Redaktion von medactuell von medac. V. M. hat von Amgen, AstraZeneca, Daiichi Sankyo, Eisai, GSK, Pfizer, MSD, Medac, Novartis, Roche, Teva, Seagen, Onkowissen, high5 Oncology, Medscape und Gilead Honorare als Referent erhalten. Beraterhonorare von Hexal, Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi Sankyo, Eisai, Lilly, Sanofi, Seagen, Gilead. Institutionelle Forschungsunterstützung von Novartis, Roche, Seagen, Genentech. Reisekostenzuschüsse von: Roche, Pfizer, Daiichi Sankyo. E. S. hat von Roche, Celgene, AstraZeneca, Novartis, Pfizer, Tesaro, Aurikamed GmbH, MCI Deutschland GmbH, bsh medical communications GmbH und Onkowissen TV-Honorare erhalten. A. S. hat von Celgene und Roche Forschungszuschüsse, von Amgen, AstraZeneca, Aurikamed, Bayer, Celgene, Clinsol, Connectmedica, Gilead, GSK, I-MED, Lilly, MCI Deutschland, Metaplan, MSD, Nanostring, Novartis, Onkowissen.de, Promedicis, Pfizer, Pierre Fabre, Roche, Seagen, Streamedup, Teva, Tesaro und Thieme Honorare sowie von Celgene, Pfizer und Roche Reisekostenzuschüsse erhalten. F. S. hat in beratenden Gremien bei Novartis, Lilly, Amgen und Roche mitgewirkt und von Roche, AstraZeneca, MSD, Novartis und Pfizer Honorare für Vorlesungen erhalten. K. S. hat keinen Interessenkonflikt. H. T. hat von Novartis, Roche, Celgene, Teva, Pfizer und AstraZeneca Honorare sowie von Roche, Celgene und Pfizer Reisekostenzuschüsse erhalten. C. T. hat für die Mitwirkung in beratenden Gremien und für Vorlesungen von Amgen, AstraZeneca, Celgene, Daiichi Sankyo, Eisai, Gilead, Lilly, MSD, Mylan, Nanostring, Novartis, Pfizer, Pierre Fabre, Puma, Roche, Seagen und Vifor Honorare erhalten. M. T. hat in beratenden Gremien bei AstraZeneca, Clovis, Daiichi Sankyo, Eisai, Gilead Science, GSK, Lilly, MSD, Novartis, Organon, Pfizer, Exact Sciences, Pierre-Fabre, Seagen und Roche mitgewirkt und von Amgen, Clovis, Daiichi Sankyo, Eisai, GSK, Lilly, MSD, Roche, Novartis, Organon, Pfizer, Seagen, Exact Sciences, Viatris, Vifor und AstraZeneca Honorare für Vorlesungen sowie von Exact Sciences und Endomag finanzielle Mittel für Versuche erhalten. Manuskriptzuschüsse wurden von Amgen, ClearCut, pfm medical, Roche, Servier und Vifor geleistet. M. U. Alle Honorare gingen an die Institution/den Arbeitgeber: Abbvie, Amgen, AstraZeneca, Celgene, Daiichi Sankyo, Eisai, Lilly, MSD Merck, Mundipharma, Myriad Genetics, Pfizer, PUMA Biotechnology, Roche, Sanofi Aventis, Novartis, Pierre Fabre. M. W. hat in beratenden Gremien bei AstraZeneca, Lilly, MSD, Novartis, Pfizer und Roche mitgewirkt. I. W. hat in beratenden Gremien bei Novartis, Daichii Sankyo, Lilly und Pfizer mitgewirkt und von AstraZeneca, Daichii Sankyo, MSD, Novartis, Pfizer und Roche Honorare als Referent erhalten. A. W. hat in beratenden Gremien bei Novartis, Lilly, Amgen, Pfizer, Roche, Tesaro und Eisai mitgewirkt und von Novartis, Pfizer, Aurikamed, Roche und Celgene Honorare für Vorlesungen erhalten. R. W. hat von Agendia, Amgen, Aristo, AstraZeneca, Boehringer Ingelheim, Carl Zeiss, Celgene, Daiichi Sankyo, Eisai, Exact Sciences, Genomic Health, Gilead, GlaxoSmithKline, Hexal, Lilly, Medstrom Medical, MSD, Mundipharma, Mylan, Nanostring, Novartis, Odonate, Paxman, Palleos, Pfizer, Pierre Fabre, PumaBiotechnology, Riemser, Roche, Sandoz/Hexal, Sanofi Genzyme, Seattle Genetics/Seagen, Tesaro Bio, Teva, Veracyte und Viatris Honorare und Reisekostenzuschüsse erhalten.

Acknowledgements

This review was developed in part as a result of grants from onkowissen.de, Lilly, Pfizer, MSD, Gilead, Daiichi Sankyo, Sanofi, Hexal, and Novartis. None of the companies had any part in the preparation and recommendations contained in this manuscript. The authors are solely responsible for the content of the manuscript.

  • References/Literatur

  • 1 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 DOI: 10.1056/NEJMoa2105215.
  • 2 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 DOI: 10.1056/NEJMoa1802905.
  • 3 Robson M, Im S-A, Senkus E. et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med 2017; 377: 523-533 DOI: 10.1056/NEJMoa1706450.
  • 4 Turner NC, Telli ML, Rugo HS. et al. A Phase II Study of Talazoparib after Platinum or Cytotoxic Nonplatinum Regimens in Patients with Advanced Breast Cancer and Germline BRCA1/2 Mutations (ABRAZO). Clin Cancer Res 2019; 25: 2717-2724 DOI: 10.1158/1078-0432.CCR-18-1891.
  • 5 Tutt ANJ, Garber J, Gelber RD. et al. Pre-specified event driven analysis of Overall Survival (OS) in the OlympiA phase III trial of adjuvant olaparib (OL) in germline BRCA1/2 mutation (gBRCAm) associated breast cancer. Ann Oncol 2022; ESMO Virtual Plenary VP1 – 2022.
  • 6 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: 214-227 DOI: 10.1159/000516419.
  • 7 Ditsch N, Untch M, Kolberg-Liedtke C. et al. AGO Recommendations for the Diagnosis and Treatment of Patients with Locally Advanced and Metastatic Breast Cancer: Update 2020. Breast Care (Basel) 2020; 15: 294-309 DOI: 10.1159/000508736.
  • 8 Ditsch N, Untch M, Thill M. et al. AGO Recommendations for the Diagnosis and Treatment of Patients with Early Breast Cancer: Update 2019. Breast Care (Basel) 2019; 14: 224-245 DOI: 10.1159/000501000.
  • 9 Fasching PA, Yadav S, Hu C. et al. Mutations in BRCA1/2 and Other Panel Genes in Patients With Metastatic Breast Cancer-Association With Patient and Disease Characteristics and Effect on Prognosis. J Clin Oncol 2021; 39: 1619-1630 DOI: 10.1200/JCO.20.01200.
  • 10 Couch FJ, Hart SN, Sharma P. et al. Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol 2015; 33: 304-311 DOI: 10.1200/JCO.2014.57.1414.
  • 11 Shimelis H, LaDuca H, Hu C. et al. Triple-Negative Breast Cancer Risk Genes Identified by Multigene Hereditary Cancer Panel Testing. J Natl Cancer Inst 2018; DOI: 10.1093/jnci/djy106.
  • 12 Foulkes WD. The ten genes for breast (and ovarian) cancer susceptibility. Nat Rev Clin Oncol 2021; 18: 259-260 DOI: 10.1038/s41571-021-00491-3.
  • 13 Müller V, Welslau M, Lüftner D. et al. Update Breast Cancer 2022 Part 2 – Advanced Stage Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82: 590-600 DOI: 10.1055/a-1811-6148.
  • 14 Kraus C, Hoyer J, Vasileiou G. et al. Gene panel sequencing in familial breast/ovarian cancer patients identifies multiple novel mutations also in genes others than BRCA1/2. Int J Cancer 2017; 140: 95-102 DOI: 10.1002/ijc.30428.
  • 15 Tung NM, Robson ME, Ventz S. et al. TBCRC048: Phase II Study of Olaparib for Metastatic Breast Cancer and Mutations in Homologous Recombination-Related Genes. J Clin Oncol 2020; 38: 4274-4282 DOI: 10.1200/JCO.20.02151.
  • 16 Kar SP, Beesley J, Amin Al Olama A. et al. Genome-Wide Meta-Analyses of Breast, Ovarian, and Prostate Cancer Association Studies Identify Multiple New Susceptibility Loci Shared by at Least Two Cancer Types. Cancer Discov 2016; 6: 1052-1067 DOI: 10.1158/2159-8290.CD-15-1227.
  • 17 Stevens KN, Vachon CM, Lee AM. et al. Common breast cancer susceptibility loci are associated with triple-negative breast cancer. Cancer Res 2011; 71: 6240-6249 DOI: 10.1158/0008-5472.CAN-11-1266.
  • 18 Schmidt MK, Hogervorst F, van Hien R. et al. Age- and Tumor Subtype-Specific Breast Cancer Risk Estimates for CHEK2*1100delC Carriers. J Clin Oncol 2016; 34: 2750-2760 DOI: 10.1200/JCO.2016.66.5844.
  • 19 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; DOI: 10.1056/NEJMoa1913948.
  • 20 Dunning AM, Michailidou K, Kuchenbaecker KB. et al. Breast cancer risk variants at 6q25 display different phenotype associations and regulate ESR1, RMND1 and CCDC170. Nat Genet 2016; 48: 374-386 DOI: 10.1038/ng.3521.
  • 21 Wu L, Shi W, Long J. et al. A transcriptome-wide association study of 229,000 women identifies new candidate susceptibility genes for breast cancer. Nat Genet 2018; DOI: 10.1038/s41588-018-0132-x.
  • 22 Couch FJ, Kuchenbaecker KB, Michailidou K. et al. Identification of four novel susceptibility loci for oestrogen receptor negative breast cancer. Nat Commun 2016; 7: 11375 DOI: 10.1038/ncomms11375.
  • 23 Wunderle M, Olmes G, Nabieva N. et al. Risk, Prediction and Prevention of Hereditary Breast Cancer – Large-Scale Genomic Studies in Times of Big and Smart Data. Geburtshilfe Frauenheilkd 2018; 78: 481-492 DOI: 10.1055/a-0603-4350.
  • 24 Chlebowski RT, Aragaki AK, Pan K. Breast Cancer Prevention: Time for Change. JCO Oncol Pract 2021; 17: 709-716 DOI: 10.1200/OP.21.00343.
  • 25 Nangia JR, Rimawi MF. Breast Cancer Prevention: Where Are We?. JCO Oncol Pract 2021; 17: 720-722 DOI: 10.1200/OP.21.00605.
  • 26 Chlebowski RT, Anderson GL, Aragaki AK. et al. Association of Menopausal Hormone Therapy With Breast Cancer Incidence and Mortality During Long-term Follow-up of the Womenʼs Health Initiative Randomized Clinical Trials. JAMA 2020; 324: 369-380 DOI: 10.1001/jama.2020.9482.
  • 27 Rossouw JE, Anderson GL, Prentice RL. et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Womenʼs Health Initiative randomized controlled trial. JAMA 2002; 288: 321-333 DOI: 10.1001/jama.288.3.321.
  • 28 Denkert C, von Minckwitz G, Darb-Esfahani S. et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 2018; 19: 40-50 DOI: 10.1016/S1470-2045(17)30904-X.
  • 29 Ingold Heppner B, Untch M, Denkert C. et al. Tumor-Infiltrating Lymphocytes: A Predictive and Prognostic Biomarker in Neoadjuvant-Treated HER2-Positive Breast Cancer. Clin Cancer Res 2016; 22: 5747-5754 DOI: 10.1158/1078-0432.CCR-15-2338.
  • 30 Wurfel F, Erber R, Huebner H. et al. TILGen: A Program to Investigate Immune Targets in Breast Cancer Patients – First Results on the Influence of Tumor-Infiltrating Lymphocytes. Breast Care (Basel) 2018; 13: 8-14 DOI: 10.1159/000486949.
  • 31 Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 2019; 4: 7 DOI: 10.1038/s41541-019-0103-y.
  • 32 Antonarelli G, Corti C, Tarantino P. et al. Therapeutic cancer vaccines revamping: technology advancements and pitfalls. Ann Oncol 2021; 32: 1537-1551 DOI: 10.1016/j.annonc.2021.08.2153.
  • 33 Corti C, Giachetti P, Eggermont AMM. et al. Therapeutic vaccines for breast cancer: Has the time finally come?. Eur J Cancer 2022; 160: 150-174 DOI: 10.1016/j.ejca.2021.10.027.
  • 34 Van Hoecke L, Verbeke R, Dewitte H. et al. mRNA in cancer immunotherapy: beyond a source of antigen. Mol Cancer 2021; 20: 48 DOI: 10.1186/s12943-021-01329-3.
  • 35 Hashimoto S, Noguchi E, Bando H. et al. Neoantigen prediction in human breast cancer using RNA sequencing data. Cancer Sci 2021; 112: 465-475 DOI: 10.1111/cas.14720.
  • 36 Li W, Amei A, Bui F. et al. Impact of Neoantigen Expression and T-Cell Activation on Breast Cancer Survival. Cancers (Basel) 2021; DOI: 10.3390/cancers13122879.
  • 37 Reimann H, Nguyen A, Sanborn JZ. et al. Identification and validation of expressed HLA-binding breast cancer neoepitopes for potential use in individualized cancer therapy. J Immunother Cancer 2021; DOI: 10.1136/jitc-2021-002605.
  • 38 Gerber B, Heintze K, Stubert J. et al. Axillary lymph node dissection in early-stage invasive breast cancer: is it still standard today?. Breast Cancer Res Treat 2011; 128: 613-624 DOI: 10.1007/s10549-011-1532-0.
  • 39 Mittendorf EA, Bellon JR, King TA. Regional Nodal Management in Patients With Clinically Node-Negative Breast Cancer Undergoing Upfront Surgery. J Clin Oncol 2020; 38: 2273-2280 DOI: 10.1200/JCO.19.02891.
  • 40 Bradley R, Braybrooke J, Gray R. et al. Aromatase inhibitors versus tamoxifen in pre-menopausal women with estrogen receptor positive early stage breast cancer treated with ovarian suppression: A patient level meta-analysis of 7,030 women in four randomised trials. San Antonio Breast Cancer Symposium 2021; 2021: GS2-04
  • 41 Regan MM, Walley BA, Fleming GF. et al. Randomized comparison of adjuvant aromatase inhibitor exemestane (E) plus ovarian function suppression (OFS) vs. tamoxifen (T) plus OFS in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): update of the combined TEXT and SOFT trials. San Antonio Breast Cancer Symposium 2021; 2021: GS2-05
  • 42 Kalinsky K, Barlow WE, Gralow JR. et al. 21-Gene Assay to Inform Chemotherapy Benefit in Node-Positive Breast Cancer. N Engl J Med 2021; 385: 2336-2347 DOI: 10.1056/NEJMoa2108873.
  • 43 Piccart M, vanʼt 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 DOI: 10.1016/S1470-2045(21)00007-3.
  • 44 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 DOI: 10.1056/NEJMoa1804710.
  • 45 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; DOI: 10.1200/JCO.21.02759. in press
  • 46 Ruddy KJ, Schaid DJ, Partridge AH. et al. Genetic predictors of chemotherapy-related amenorrhea in women with breast cancer. Fertil Steril 2019; 112: 731-739.e1 DOI: 10.1016/j.fertnstert.2019.05.018.
  • 47 Kalinsky KM, Barlow WE, Gralow JR. et al. Distant-disease free interval in participants (pts) with 1–3 positive lymph nodes (LN), hormone receptor-positive (HR+) and her2-negative (HER2-) breast cancer (BC) with recurrence score (RS) < or = 25 randomized to endocrine therapy (ET) ± chemotherapy (CT): SWOG s1007 (RxPONDER). San Antonio Breast Cancer Symposium 2021; 2021: GS2-07
  • 48 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 DOI: 10.1016/S1470-2045(20)30642-2.
  • 49 Gnant M, Dueck AC, Frantal S. et al. Adjuvant Palbociclib for Early Breast Cancer: The PALLAS Trial Results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol 2022; 40: 282-293 DOI: 10.1200/JCO.21.02554.
  • 50 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: 1571-1581 DOI: 10.1016/j.annonc.2021.09.015.
  • 51 Precisiononcologynews.com. Novartis Anticipating Radioligands, New Precision Therapies to Continue Oncology Segment Growth. 2022 Accessed February 02, 2022 at: https://www.precisiononcologynews.com/cancer/novartis-anticipating-radioligands-new-precision-therapies-continue-oncology-segment-growth%23Yf6ALvgxkuU
  • 52 European Medicines Agency. Verzenios. 2022 Accessed May 19, 2022 at: https://www.ema.europa.eu/en/documents/product-information/verzenios-epar-product-information_en.pdf
  • 53 Rugo HS, OʼShaughnessy 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; DOI: 10.1016/j.annonc.2022.03.006.
  • 54 Nabieva N, Kellner S, Fehm T. et al. Influence of patient and tumor characteristics on early therapy persistence with letrozole in postmenopausal women with early breast cancer: results of the prospective Evaluate-TM study with 3941 patients. Ann Oncol 2018; 29: 186-192 DOI: 10.1093/annonc/mdx630.
  • 55 Lüftner D, Schütz F, Stickeler E. et al. Update Breast Cancer 2021 Part 5 – Advanced Breast Cancer. Geburtshilfe Frauenheilkd 2022; DOI: 10.1055/a-1724-9569.
  • 56 Thomssen C, Fehm TN, Stickeler E. et al. Update Breast Cancer 2021 Part 4 – Prevention and Early Stages. Geburtshilfe Frauenheilkd 2022; DOI: 10.1055/a-1724-9639.
  • 57 Ruiz-Borrego M, Guerrero-Zotano A, Bermejo B. et al. Phase III evaluating the addition of fulvestrant (F) to anastrozole (A) as adjuvant therapy in postmenopausal women with hormone receptor-positive HER2-negative (HR+/HER2-) early breast cancer (EBC): results from the GEICAM/2006–10 study. Breast Cancer Res Treat 2019; 177: 115-125 DOI: 10.1007/s10549-019-05296-8.
  • 58 Hurvitz SA, Quiroga V, Park YH. et al. Neoadjuvant giredestrant (GDC-9545) + palbociclib versus anastrozole + palbociclib in postmenopausal women with estrogen receptor-positive, HER2-negative, untreated early breast cancer: Primary analysis of the randomized, open-label, phase II coopERA breast cancer study. San Antonio Breast Cancer Symposium 2021; 2021: GS1-07
  • 59 Bardia A, Neven P, Streich G. et al. Elacestrant, an oral selective estrogen receptor degrader (SERD), vs. investigatorʼs choice of endocrine monotherapy for ER+/HER2- advanced/metastatic breast cancer (mBC) following progression on prior endocrine and CDK4/6 inhibitor therapy: Results of EMERALD phase 3 trial. San Antonio Breast Cancer Symposium 2021; 2021: GS2-02
  • 60 Wang N, Li X, Wang R. et al. Spatial transcriptomics and proteomics technologies for deconvoluting the tumor microenvironment. Biotechnol J 2021; 16: e2100041 DOI: 10.1002/biot.202100041.
  • 61 Longo SK, Guo MG, Ji AL. et al. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet 2021; 22: 627-644 DOI: 10.1038/s41576-021-00370-8.
  • 62 Bianchini G, Wang XQ, Danenberg E. et al. Single-cell spatial analysis by imaging mass cytometry and immunotherapy response in triple-negative breast cancer (TNBC) in the NeoTRIPaPDL1 trial. San Antonio Breast Cancer Symposium 2021; 2021: GS1-00
  • 63 Keating SM, Taylor DL, Plant AL. et al. Opportunities and Challenges in Implementation of Multiparameter Single Cell Analysis Platforms for Clinical Translation. Clin Transl Sci 2018; 11: 267-276 DOI: 10.1111/cts.12536.
  • 64 Schmid P, Cortes J, Dent R. et al. Event-free Survival with Pembrolizumab in Early Triple-Negative Breast Cancer. N Engl J Med 2022; DOI: 10.1056/NEJMoa2112651.
  • 65 Schmid P, Cortes J, Dent R. et al. KEYNOTE-522: Phase III study of neoadjuvant pembrolizumab + chemotherapy vs. placebo + chemotherapy, followed by adjuvant pembrolizumab vs. placebo for early-stage TNBC. Ann Oncol 2021; 32: VP7-2021 DOI: 10.1016/j.annonc.2021.06.014.
  • 66 Schmid P, Cortes J, Dent R. et al. KEYNOTE-522: Phase 3 study of pembrolizumab + chemotherapy vs. placebo + chemotherapy as neoadjuvant treatment, followed by pembrolizumab vs. placebo as adjuvant treatment for early-stage high-risk triple-negative breast cancer (TNBC). San Antonio Breast Cancer Symposium 2021; 2021: GS1-01
  • 67 Fisher B, Costantino JP, Wickerham DL. et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst 2005; 97: 1652-1662 DOI: 10.1093/jnci/dji372.
  • 68 Powles TJ, Ashley S, Tidy A. et al. Twenty-year follow-up of the Royal Marsden randomized, double-blinded tamoxifen breast cancer prevention trial. J Natl Cancer Inst 2007; 99: 283-290 DOI: 10.1093/jnci/djk050.
  • 69 Cuzick J, Sestak I, Cawthorn S. et al. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 2015; 16: 67-75 DOI: 10.1016/S1470-2045(14)71171-4.
  • 70 Cuzick J, Sestak I, Forbes JF. et al. Use of anastrozole for breast cancer prevention (IBIS-II): long-term results of a randomised controlled trial. Lancet 2020; 395: 117-122 DOI: 10.1016/S0140-6736(19)32955-1.
  • 71 Goss PE, Ingle JN, Ales-Martinez JE. et al. Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 2011; 364: 2381-2391 DOI: 10.1056/NEJMoa1103507.

Correspondence/Korrespondenzadresse

Peter A. Fasching, MD
Erlangen University Hospital, Department of Gynecology and Obstetrics
Comprehensive Cancer Center Erlangen EMN
Friedrich Alexander University of Erlangen-Nuremberg
Universitätsstraße 21 – 23
91054 Erlangen
Germany   

Publication History

Received: 21 March 2022

Accepted after revision: 26 March 2022

Article published online:
03 June 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commecial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References/Literatur

  • 1 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 DOI: 10.1056/NEJMoa2105215.
  • 2 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 DOI: 10.1056/NEJMoa1802905.
  • 3 Robson M, Im S-A, Senkus E. et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med 2017; 377: 523-533 DOI: 10.1056/NEJMoa1706450.
  • 4 Turner NC, Telli ML, Rugo HS. et al. A Phase II Study of Talazoparib after Platinum or Cytotoxic Nonplatinum Regimens in Patients with Advanced Breast Cancer and Germline BRCA1/2 Mutations (ABRAZO). Clin Cancer Res 2019; 25: 2717-2724 DOI: 10.1158/1078-0432.CCR-18-1891.
  • 5 Tutt ANJ, Garber J, Gelber RD. et al. Pre-specified event driven analysis of Overall Survival (OS) in the OlympiA phase III trial of adjuvant olaparib (OL) in germline BRCA1/2 mutation (gBRCAm) associated breast cancer. Ann Oncol 2022; ESMO Virtual Plenary VP1 – 2022.
  • 6 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: 214-227 DOI: 10.1159/000516419.
  • 7 Ditsch N, Untch M, Kolberg-Liedtke C. et al. AGO Recommendations for the Diagnosis and Treatment of Patients with Locally Advanced and Metastatic Breast Cancer: Update 2020. Breast Care (Basel) 2020; 15: 294-309 DOI: 10.1159/000508736.
  • 8 Ditsch N, Untch M, Thill M. et al. AGO Recommendations for the Diagnosis and Treatment of Patients with Early Breast Cancer: Update 2019. Breast Care (Basel) 2019; 14: 224-245 DOI: 10.1159/000501000.
  • 9 Fasching PA, Yadav S, Hu C. et al. Mutations in BRCA1/2 and Other Panel Genes in Patients With Metastatic Breast Cancer-Association With Patient and Disease Characteristics and Effect on Prognosis. J Clin Oncol 2021; 39: 1619-1630 DOI: 10.1200/JCO.20.01200.
  • 10 Couch FJ, Hart SN, Sharma P. et al. Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol 2015; 33: 304-311 DOI: 10.1200/JCO.2014.57.1414.
  • 11 Shimelis H, LaDuca H, Hu C. et al. Triple-Negative Breast Cancer Risk Genes Identified by Multigene Hereditary Cancer Panel Testing. J Natl Cancer Inst 2018; DOI: 10.1093/jnci/djy106.
  • 12 Foulkes WD. The ten genes for breast (and ovarian) cancer susceptibility. Nat Rev Clin Oncol 2021; 18: 259-260 DOI: 10.1038/s41571-021-00491-3.
  • 13 Müller V, Welslau M, Lüftner D. et al. Update Breast Cancer 2022 Part 2 – Advanced Stage Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82: 590-600 DOI: 10.1055/a-1811-6148.
  • 14 Kraus C, Hoyer J, Vasileiou G. et al. Gene panel sequencing in familial breast/ovarian cancer patients identifies multiple novel mutations also in genes others than BRCA1/2. Int J Cancer 2017; 140: 95-102 DOI: 10.1002/ijc.30428.
  • 15 Tung NM, Robson ME, Ventz S. et al. TBCRC048: Phase II Study of Olaparib for Metastatic Breast Cancer and Mutations in Homologous Recombination-Related Genes. J Clin Oncol 2020; 38: 4274-4282 DOI: 10.1200/JCO.20.02151.
  • 16 Kar SP, Beesley J, Amin Al Olama A. et al. Genome-Wide Meta-Analyses of Breast, Ovarian, and Prostate Cancer Association Studies Identify Multiple New Susceptibility Loci Shared by at Least Two Cancer Types. Cancer Discov 2016; 6: 1052-1067 DOI: 10.1158/2159-8290.CD-15-1227.
  • 17 Stevens KN, Vachon CM, Lee AM. et al. Common breast cancer susceptibility loci are associated with triple-negative breast cancer. Cancer Res 2011; 71: 6240-6249 DOI: 10.1158/0008-5472.CAN-11-1266.
  • 18 Schmidt MK, Hogervorst F, van Hien R. et al. Age- and Tumor Subtype-Specific Breast Cancer Risk Estimates for CHEK2*1100delC Carriers. J Clin Oncol 2016; 34: 2750-2760 DOI: 10.1200/JCO.2016.66.5844.
  • 19 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; DOI: 10.1056/NEJMoa1913948.
  • 20 Dunning AM, Michailidou K, Kuchenbaecker KB. et al. Breast cancer risk variants at 6q25 display different phenotype associations and regulate ESR1, RMND1 and CCDC170. Nat Genet 2016; 48: 374-386 DOI: 10.1038/ng.3521.
  • 21 Wu L, Shi W, Long J. et al. A transcriptome-wide association study of 229,000 women identifies new candidate susceptibility genes for breast cancer. Nat Genet 2018; DOI: 10.1038/s41588-018-0132-x.
  • 22 Couch FJ, Kuchenbaecker KB, Michailidou K. et al. Identification of four novel susceptibility loci for oestrogen receptor negative breast cancer. Nat Commun 2016; 7: 11375 DOI: 10.1038/ncomms11375.
  • 23 Wunderle M, Olmes G, Nabieva N. et al. Risk, Prediction and Prevention of Hereditary Breast Cancer – Large-Scale Genomic Studies in Times of Big and Smart Data. Geburtshilfe Frauenheilkd 2018; 78: 481-492 DOI: 10.1055/a-0603-4350.
  • 24 Chlebowski RT, Aragaki AK, Pan K. Breast Cancer Prevention: Time for Change. JCO Oncol Pract 2021; 17: 709-716 DOI: 10.1200/OP.21.00343.
  • 25 Nangia JR, Rimawi MF. Breast Cancer Prevention: Where Are We?. JCO Oncol Pract 2021; 17: 720-722 DOI: 10.1200/OP.21.00605.
  • 26 Chlebowski RT, Anderson GL, Aragaki AK. et al. Association of Menopausal Hormone Therapy With Breast Cancer Incidence and Mortality During Long-term Follow-up of the Womenʼs Health Initiative Randomized Clinical Trials. JAMA 2020; 324: 369-380 DOI: 10.1001/jama.2020.9482.
  • 27 Rossouw JE, Anderson GL, Prentice RL. et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Womenʼs Health Initiative randomized controlled trial. JAMA 2002; 288: 321-333 DOI: 10.1001/jama.288.3.321.
  • 28 Denkert C, von Minckwitz G, Darb-Esfahani S. et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 2018; 19: 40-50 DOI: 10.1016/S1470-2045(17)30904-X.
  • 29 Ingold Heppner B, Untch M, Denkert C. et al. Tumor-Infiltrating Lymphocytes: A Predictive and Prognostic Biomarker in Neoadjuvant-Treated HER2-Positive Breast Cancer. Clin Cancer Res 2016; 22: 5747-5754 DOI: 10.1158/1078-0432.CCR-15-2338.
  • 30 Wurfel F, Erber R, Huebner H. et al. TILGen: A Program to Investigate Immune Targets in Breast Cancer Patients – First Results on the Influence of Tumor-Infiltrating Lymphocytes. Breast Care (Basel) 2018; 13: 8-14 DOI: 10.1159/000486949.
  • 31 Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 2019; 4: 7 DOI: 10.1038/s41541-019-0103-y.
  • 32 Antonarelli G, Corti C, Tarantino P. et al. Therapeutic cancer vaccines revamping: technology advancements and pitfalls. Ann Oncol 2021; 32: 1537-1551 DOI: 10.1016/j.annonc.2021.08.2153.
  • 33 Corti C, Giachetti P, Eggermont AMM. et al. Therapeutic vaccines for breast cancer: Has the time finally come?. Eur J Cancer 2022; 160: 150-174 DOI: 10.1016/j.ejca.2021.10.027.
  • 34 Van Hoecke L, Verbeke R, Dewitte H. et al. mRNA in cancer immunotherapy: beyond a source of antigen. Mol Cancer 2021; 20: 48 DOI: 10.1186/s12943-021-01329-3.
  • 35 Hashimoto S, Noguchi E, Bando H. et al. Neoantigen prediction in human breast cancer using RNA sequencing data. Cancer Sci 2021; 112: 465-475 DOI: 10.1111/cas.14720.
  • 36 Li W, Amei A, Bui F. et al. Impact of Neoantigen Expression and T-Cell Activation on Breast Cancer Survival. Cancers (Basel) 2021; DOI: 10.3390/cancers13122879.
  • 37 Reimann H, Nguyen A, Sanborn JZ. et al. Identification and validation of expressed HLA-binding breast cancer neoepitopes for potential use in individualized cancer therapy. J Immunother Cancer 2021; DOI: 10.1136/jitc-2021-002605.
  • 38 Gerber B, Heintze K, Stubert J. et al. Axillary lymph node dissection in early-stage invasive breast cancer: is it still standard today?. Breast Cancer Res Treat 2011; 128: 613-624 DOI: 10.1007/s10549-011-1532-0.
  • 39 Mittendorf EA, Bellon JR, King TA. Regional Nodal Management in Patients With Clinically Node-Negative Breast Cancer Undergoing Upfront Surgery. J Clin Oncol 2020; 38: 2273-2280 DOI: 10.1200/JCO.19.02891.
  • 40 Bradley R, Braybrooke J, Gray R. et al. Aromatase inhibitors versus tamoxifen in pre-menopausal women with estrogen receptor positive early stage breast cancer treated with ovarian suppression: A patient level meta-analysis of 7,030 women in four randomised trials. San Antonio Breast Cancer Symposium 2021; 2021: GS2-04
  • 41 Regan MM, Walley BA, Fleming GF. et al. Randomized comparison of adjuvant aromatase inhibitor exemestane (E) plus ovarian function suppression (OFS) vs. tamoxifen (T) plus OFS in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): update of the combined TEXT and SOFT trials. San Antonio Breast Cancer Symposium 2021; 2021: GS2-05
  • 42 Kalinsky K, Barlow WE, Gralow JR. et al. 21-Gene Assay to Inform Chemotherapy Benefit in Node-Positive Breast Cancer. N Engl J Med 2021; 385: 2336-2347 DOI: 10.1056/NEJMoa2108873.
  • 43 Piccart M, vanʼt 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 DOI: 10.1016/S1470-2045(21)00007-3.
  • 44 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 DOI: 10.1056/NEJMoa1804710.
  • 45 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; DOI: 10.1200/JCO.21.02759. in press
  • 46 Ruddy KJ, Schaid DJ, Partridge AH. et al. Genetic predictors of chemotherapy-related amenorrhea in women with breast cancer. Fertil Steril 2019; 112: 731-739.e1 DOI: 10.1016/j.fertnstert.2019.05.018.
  • 47 Kalinsky KM, Barlow WE, Gralow JR. et al. Distant-disease free interval in participants (pts) with 1–3 positive lymph nodes (LN), hormone receptor-positive (HR+) and her2-negative (HER2-) breast cancer (BC) with recurrence score (RS) < or = 25 randomized to endocrine therapy (ET) ± chemotherapy (CT): SWOG s1007 (RxPONDER). San Antonio Breast Cancer Symposium 2021; 2021: GS2-07
  • 48 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 DOI: 10.1016/S1470-2045(20)30642-2.
  • 49 Gnant M, Dueck AC, Frantal S. et al. Adjuvant Palbociclib for Early Breast Cancer: The PALLAS Trial Results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol 2022; 40: 282-293 DOI: 10.1200/JCO.21.02554.
  • 50 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: 1571-1581 DOI: 10.1016/j.annonc.2021.09.015.
  • 51 Precisiononcologynews.com. Novartis Anticipating Radioligands, New Precision Therapies to Continue Oncology Segment Growth. 2022 Accessed February 02, 2022 at: https://www.precisiononcologynews.com/cancer/novartis-anticipating-radioligands-new-precision-therapies-continue-oncology-segment-growth%23Yf6ALvgxkuU
  • 52 European Medicines Agency. Verzenios. 2022 Accessed May 19, 2022 at: https://www.ema.europa.eu/en/documents/product-information/verzenios-epar-product-information_en.pdf
  • 53 Rugo HS, OʼShaughnessy 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; DOI: 10.1016/j.annonc.2022.03.006.
  • 54 Nabieva N, Kellner S, Fehm T. et al. Influence of patient and tumor characteristics on early therapy persistence with letrozole in postmenopausal women with early breast cancer: results of the prospective Evaluate-TM study with 3941 patients. Ann Oncol 2018; 29: 186-192 DOI: 10.1093/annonc/mdx630.
  • 55 Lüftner D, Schütz F, Stickeler E. et al. Update Breast Cancer 2021 Part 5 – Advanced Breast Cancer. Geburtshilfe Frauenheilkd 2022; DOI: 10.1055/a-1724-9569.
  • 56 Thomssen C, Fehm TN, Stickeler E. et al. Update Breast Cancer 2021 Part 4 – Prevention and Early Stages. Geburtshilfe Frauenheilkd 2022; DOI: 10.1055/a-1724-9639.
  • 57 Ruiz-Borrego M, Guerrero-Zotano A, Bermejo B. et al. Phase III evaluating the addition of fulvestrant (F) to anastrozole (A) as adjuvant therapy in postmenopausal women with hormone receptor-positive HER2-negative (HR+/HER2-) early breast cancer (EBC): results from the GEICAM/2006–10 study. Breast Cancer Res Treat 2019; 177: 115-125 DOI: 10.1007/s10549-019-05296-8.
  • 58 Hurvitz SA, Quiroga V, Park YH. et al. Neoadjuvant giredestrant (GDC-9545) + palbociclib versus anastrozole + palbociclib in postmenopausal women with estrogen receptor-positive, HER2-negative, untreated early breast cancer: Primary analysis of the randomized, open-label, phase II coopERA breast cancer study. San Antonio Breast Cancer Symposium 2021; 2021: GS1-07
  • 59 Bardia A, Neven P, Streich G. et al. Elacestrant, an oral selective estrogen receptor degrader (SERD), vs. investigatorʼs choice of endocrine monotherapy for ER+/HER2- advanced/metastatic breast cancer (mBC) following progression on prior endocrine and CDK4/6 inhibitor therapy: Results of EMERALD phase 3 trial. San Antonio Breast Cancer Symposium 2021; 2021: GS2-02
  • 60 Wang N, Li X, Wang R. et al. Spatial transcriptomics and proteomics technologies for deconvoluting the tumor microenvironment. Biotechnol J 2021; 16: e2100041 DOI: 10.1002/biot.202100041.
  • 61 Longo SK, Guo MG, Ji AL. et al. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet 2021; 22: 627-644 DOI: 10.1038/s41576-021-00370-8.
  • 62 Bianchini G, Wang XQ, Danenberg E. et al. Single-cell spatial analysis by imaging mass cytometry and immunotherapy response in triple-negative breast cancer (TNBC) in the NeoTRIPaPDL1 trial. San Antonio Breast Cancer Symposium 2021; 2021: GS1-00
  • 63 Keating SM, Taylor DL, Plant AL. et al. Opportunities and Challenges in Implementation of Multiparameter Single Cell Analysis Platforms for Clinical Translation. Clin Transl Sci 2018; 11: 267-276 DOI: 10.1111/cts.12536.
  • 64 Schmid P, Cortes J, Dent R. et al. Event-free Survival with Pembrolizumab in Early Triple-Negative Breast Cancer. N Engl J Med 2022; DOI: 10.1056/NEJMoa2112651.
  • 65 Schmid P, Cortes J, Dent R. et al. KEYNOTE-522: Phase III study of neoadjuvant pembrolizumab + chemotherapy vs. placebo + chemotherapy, followed by adjuvant pembrolizumab vs. placebo for early-stage TNBC. Ann Oncol 2021; 32: VP7-2021 DOI: 10.1016/j.annonc.2021.06.014.
  • 66 Schmid P, Cortes J, Dent R. et al. KEYNOTE-522: Phase 3 study of pembrolizumab + chemotherapy vs. placebo + chemotherapy as neoadjuvant treatment, followed by pembrolizumab vs. placebo as adjuvant treatment for early-stage high-risk triple-negative breast cancer (TNBC). San Antonio Breast Cancer Symposium 2021; 2021: GS1-01
  • 67 Fisher B, Costantino JP, Wickerham DL. et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst 2005; 97: 1652-1662 DOI: 10.1093/jnci/dji372.
  • 68 Powles TJ, Ashley S, Tidy A. et al. Twenty-year follow-up of the Royal Marsden randomized, double-blinded tamoxifen breast cancer prevention trial. J Natl Cancer Inst 2007; 99: 283-290 DOI: 10.1093/jnci/djk050.
  • 69 Cuzick J, Sestak I, Cawthorn S. et al. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 2015; 16: 67-75 DOI: 10.1016/S1470-2045(14)71171-4.
  • 70 Cuzick J, Sestak I, Forbes JF. et al. Use of anastrozole for breast cancer prevention (IBIS-II): long-term results of a randomised controlled trial. Lancet 2020; 395: 117-122 DOI: 10.1016/S0140-6736(19)32955-1.
  • 71 Goss PE, Ingle JN, Ales-Martinez JE. et al. Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 2011; 364: 2381-2391 DOI: 10.1056/NEJMoa1103507.

Zoom Image
Fig. 1 Study design of the INSEMA trial.
Zoom Image
Fig. 2 Invasive disease-free survival in the RxPONDER trial in the premenopausal patient group. ET: endocrine treatment, CT → ET: Chemotherapy and endocrine treatment (data from [47]).
Zoom Image
Fig. 3 Example of a multiplex analysis of several biomarkers and possible analysis methods for spatial arrangement (spatial proteomics, image courtesy of Barbara Seliger und Chiara Massa, Halle).
Zoom Image
Abb. 1 Studiendesign der INSEMA-Studie.
Zoom Image
Abb. 2 Invasives rückfallfreies Überleben in der RxPONDER-Studie in der Gruppe der prämenopausalen Patientinnen. ET: endokrine Therapie, CT → ET: Chemotherapie und endokrine Therapie (Daten aus [47]).
Zoom Image
Abb. 3 Beispiel für eine Multiplex-Analyse mehrerer Biomarker und mögliche Analysemethoden der räumlichen Anordnung (Spatial Proteomics, Bild mit freundlicher Genehmigung von Barbara Seliger und Chiara Massa, Halle).