breast cancer research and treatment impact factor 2023

Breast Cancer Research and Treatment Impact, Factor and Metrics, Impact Score, Ranking, h-index, SJR, Rating, Publisher, ISSN, and More

Impact score.

Note: The impact score or impact index shown here is equivalent to the average number of times documents published in a journal/conference in the past two years have been cited in the current year (i.e., Cites / Doc. (2 years)). It is based on Scopus data and can be a little higher or different compared to the impact factor (IF) produced by Journal Citation Report. Please refer to the Web of Science data source to check the exact journal impact factor ™ (Thomson Reuters) metric.

Important Metrics and Factor

Breast Cancer Research and Treatment
Breast Cancer Res. Treat.
Journal
Oncology (Q1); Cancer Research (Q2)
171
2281
1.442
3.49
Springer New York
United States
01676806, 15737217
Q1
1981-2022

Aim and Scope

Breast Cancer Research and Treatment provides the surgeon, radiotherapist, medical oncologist, endocrinologist, epidemiologist, immunologist or cell biologist investigating problems in breast cancer a single forum for communication. The journal creates a `market place for breast cancer topics which cuts across all the usual lines of disciplines, providing a site for presenting pertinent investigations and for discussing critical questions relevant to the entire field. It seeks to develop a new focus and new perspectives for all those concerned with breast cancer. Oncology is undoubtedly the most rapidly growing subspecialty in the field of medicine, and breast cancer is one of the most serious problems of oncology.

About Breast Cancer Research and Treatment

Breast Cancer Research and Treatment is a journal covering the technologies/fields/categories related to Oncology (Q1); Cancer Research (Q2) . It is published by Springer New York . The overall rank of Breast Cancer Research and Treatment is 2281 . According to SCImago Journal Rank (SJR) , this journal is ranked 1.442 . SCImago Journal Rank is an indicator, which measures the scientific influence of journals. It considers the number of citations received by a journal and the importance of the journals from where these citations come. SJR acts as an alternative to the Journal Impact Factor (or an average number of citations received in last 2 years). This journal has an h-index of 171 . The best quartile for this journal is Q1 .

The ISSN of Breast Cancer Research and Treatment journal is 01676806, 15737217 . An International Standard Serial Number (ISSN) is a unique code of 8 digits. It is used for the recognition of journals, newspapers, periodicals, and magazines in all kind of forms, be it print-media or electronic. Breast Cancer Research and Treatment is cited by a total of 5595 articles during the last 3 years (Preceding 2022).

Breast Cancer Research and Treatment Impact IF 2022-2023

The Impact IF 2022 of Breast Cancer Research and Treatment is 3.49 , which is computed in 2023 as per its definition. Breast Cancer Research and Treatment IF is decreased by a factor of 0.57 and approximate percentage change is -14.04% when compared to preceding year 2021, which shows a falling trend. The impact IF , also denoted as Journal impact score (JIS), of an academic journal is a measure of the yearly average number of citations to recent articles published in that journal. It is based on Scopus data.

Breast Cancer Research and Treatment Impact IF 2023 Prediction

Impact IF 2022 of Breast Cancer Research and Treatment is 3.49 . If the same downward trend persists, Impact IF may fall in 2023 as well.

Impact IF Trend

Year wise impact if of breast cancer research and treatment. based on scopus data..

Year Impact IF
2023/2024 Coming Soon
2022 3.49
2021 4.06
2020 4.09
2019 3.54
2018 3.32
2017 3.66
2016 3.85
2015 4.39
2014 4.45

Breast Cancer Research and Treatment h-index

Breast Cancer Research and Treatment has an h-index of 171 . It means 171 articles of this journal have more than 171 number of citations. The h-index is a way of measuring the productivity and citation impact of the publications. The h-index is defined as the maximum value of h such that the given journal/author has published h papers that have each been cited at least h number of times.

Breast Cancer Research and Treatment ISSN

The ISSN of Breast Cancer Research and Treatment is 01676806, 15737217 . ISSN stands for International Standard Serial Number.

An ISSN is a unique code of 8 digits. It is used for the recognition of journals, newspapers, periodicals, and magazines in all kind of forms, be it print-media or electronic.

Table Setting

Breast Cancer Research and Treatment Rank and SCImago Journal Rank (SJR)

The overall rank of Breast Cancer Research and Treatment is 2281 . According to SCImago Journal Rank (SJR), this journal is ranked 1.442 . SCImago Journal Rank is an indicator, which measures the scientific influence of journals. It considers the number of citations received by a journal and the importance of the journals from where these citations come.

SJR of Breast Cancer Research and Treatment by Year

Year SJR
2023/2024 Coming Soon
2022 1.442
2021 1.407
2020 1.908
2019 1.905
2018 1.899
2017 2.066
2016 2.181
2015 2.432
2014 2.397

Ranking of Breast Cancer Research and Treatment by Year

Year Ranking
2023/2024 Coming Soon
2022 2281
2021 2338
2020 1445
2019 1434
2018 1506
2017 1312
2016 1179
2015 989
2014 1003

Breast Cancer Research and Treatment Publisher

Breast Cancer Research and Treatment is published by Springer New York . It's publishing house is located in United States . Coverage history of this journal is as following: 1981-2022 . The organization or individual who handles the printing and distribution of printed or digital publications is known as Publisher.

Call For Papers

Visit the official website of the journal/conference to check the further details about the call for papers.

Abbreviation

The IS0 4 standard abbreviation of Breast Cancer Research and Treatment is Breast Cancer Res. Treat. . This abbreviation ('Breast Cancer Res. Treat.') is well recommended and approved for the purpose of indexing, abstraction, referencing and citing goals. It meets all the essential criteria of ISO 4 standard.

ISO 4 (International Organization for Standardization 4) is an international standard that defines a uniform and consistent system for abbreviating serial publication titles and journals.

How to publish in Breast Cancer Research and Treatment

If your research field is/are related to Oncology (Q1); Cancer Research (Q2) , then please visit the official website of this journal .

Acceptance Rate

  • The demand or interest of researchers/scientists in publishing in a specific Journal/Conference.
  • Peer review complexity and timeline.
  • The mix of unsolicited and invited submissions.
  • The time it takes from manuscript submission to final publication.
  • And Many More.

It is essential to understand that the acceptance rate/rejection rate of papers varies among journals. Some Journals considers all the manuscripts submissions as a basis of acceptance rate computation. On the other hand, few consider the only manuscripts sent for peer review or few even not bother about the accurate maintenance of total submissions. Hence, it can provide a rough estimation only.

The best way to find out the acceptance rate is to reach out to the associated editor or to check the official website of the Journal/Conference.

Frequently Asked Questions (FAQs)

What's the latest impact if of the breast cancer research and treatment.

Breast Cancer Research and Treatment latest impact IF is 3.49 . It's evaluated in the year 2022. The highest and the lowest impact IF or impact score of this journal are 4.45 (2014) and 3.32 (2018) , respectively, in the last 9 years. Moreover, its average IS is 3.87 in the previous 9 years.

What's the SCImago Journal Rank (SJR) of the Breast Cancer Research and Treatment?

The Breast Cancer Research and Treatment has an SJR (SCImago Journal Rank) of 1.442 , according to the latest data. It is computed in the year 2023. In the past 9 years, this journal has recorded a range of SJR, with the highest being 2.432 in 2015 and the lowest being 1.407 in 2021. Furthermore, the average SJR of the Breast Cancer Research and Treatment over the previous 9-year period stands at 3.87.

What's the latest h-index of the Breast Cancer Research and Treatment?

The latest h-index of the Breast Cancer Research and Treatment is 171 .

Who's the publisher of the Breast Cancer Research and Treatment?

The Breast Cancer Research and Treatment is published by the Springer New York , with its country of publication being the United States.

What's the current ranking of the Breast Cancer Research and Treatment?

The Breast Cancer Research and Treatment is currently ranked 2281 out of 27955 Journals, Conferences, and Book Series in the latest ranking. Over the course of the last 9 years, this journal has experienced varying rankings, reaching its highest position of 989 in 2015 and its lowest position of 2338 in 2021.

What's the abbreviation or short name for the Breast Cancer Research and Treatment?

The standard ISO4 abbreviation for the Breast Cancer Research and Treatment is Breast Cancer Res. Treat. .

Is the "Breast Cancer Research and Treatment" classified as a Journal, Conference and Proceedings, Trade Journal or Book Series?

Breast Cancer Research and Treatment is classified as a journal that the Springer New York publishes.

What's the scope or major areas of the Breast Cancer Research and Treatment?

  • Cancer Research

For a more comprehensive understanding of its scope, check the official website of this journal.

What's the ISSN of the Breast Cancer Research and Treatment?

The Breast Cancer Research and Treatment is assigned the following International Standard Serial Numbers (ISSN): 01676806, 15737217 .

What's the best quartile of the Breast Cancer Research and Treatment?

The best quartile for the Breast Cancer Research and Treatment is Q1 (2022).

What's the coverage history of the Breast Cancer Research and Treatment?

The Breast Cancer Research and Treatment coverage history can be summarized as follows: 1981-2022 .

Credits and Sources

  • Scimago Journal & Country Rank (SJR), https://www.scimagojr.com/
  • Journal Impact Factor, https://clarivate.com/

Impact Score, h-Index, and Other Important Details of These Journals, Conferences, and Books

Journal/Conference/Workshop/Book Title Type Ranking Publisher h-index Impact Score

Check complete list

Year wise Impact Score (IS) of Breast Cancer Research and Treatment

Year Impact Score (IS)
2023/2024 Coming Soon
2022 3.49
2021 4.06
2020 4.09
2019 3.54
2018 3.32
2017 3.66
2016 3.85
2015 4.39
2014 4.45

Top Journals/Conferences in Oncology

Top journals/conferences in cancer research.

Download Templates

IEEE Article Springer ACM Elsevier Wiley MITPress

Check out Quote of the Day

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • v.74; 2024 Apr
  • PMC10869942

Breast cancer highlights from 2023: Knowledge to guide practice and future research

Maria-joao cardoso.

a Champalimaud Foundation Breast Unit, Lisbon, Portugal

b University of Lisbon, Faculty of Medicine, Lisbon, Portugal

Philip Poortmans

c Department of Radiation Oncology, Iridium Netwerk, Wilrijk-Antwerp, Belgium

d Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk-Antwerp, Belgium

Elżbieta Senkus

e Department of Oncology and Radiotherapy, Medical University of Gdańsk, Poland

Oreste D. Gentilini

f Università Vita-Salute San Raffaele, Milano, Italy

g IRCCS Ospedale San Raffaele, Milano, Italy

Nehmat Houssami

h Sydney School of Public Health, The University of Sydney, Sydney, NSW, Australia

i The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, NSW, Australia

j Wiser Healthcare, The University of Sydney, Sydney, NSW, Australia

This narrative work highlights a selection of published work from 2023 with potential implications for breast cancer practice. We feature publications that have provided new knowledge immediately relevant to patient care or for future research. We also highlight guidelines that have reported evidence-based or consensus recommendations to support practice and evaluation in breast cancer diagnosis and treatment. The scope of selected highlights represents various domains and disciplines in cancer control, from prevention to treatment of early and advanced breast cancer.

  • • In 2023 the amount of publications including “Breast Cancer” in the title brings around 15 000 titles in PubMed.
  • • The ‘Breast Cancer Highlights from 2023’ offer our view on publications that deserve a special mention.
  • • The selection of featured work, attempts to highlight papers from the various disciplines that stood out amongst many others.

1. Introduction

The beginning of a new year is a time to reflect on the year that has passed, and from a professional perspective, to consider what knowledge 2023 has brought to help those involved in the care of breast cancer patients. While attempting to review and summarize the most interesting and informative scientific research published in 2023, we were challenged by the amount of publications in 2023 (in English language) and including “Breast Cancer” in the title, brings around 15 000 titles in PubMed. Attempting to filter the search by restricting to “trial” we attain around 500 new publications in that year.

It goes without saying that the majority of these will not translate into a global or immediate impact, and many will represent incremental knowledge laying ground for the next phase of research efforts.

In this paper, we present to readers of the journal the ‘Breast Cancer Highlights from 2023’ offering our view on publications that deserve a special mention from the vast amount of knowledge published in 2023, focusing predominantly on full-length papers (with brief mention of noteworthy studies reported as abstracts).

The selection of featured work, whilst not taking a systematic or exhaustive process, attempts to highlight papers from the various disciplines that stood out, for one of several reasons, including, but not exclusively, a clinical trial that led or can lead to practice change, an update or insight into a topic that introduces a new concept, or a consensus approach or new guideline that has the potential to be widely adopted and to improve patients’ care. Table 1 , Table 2 enumerate respectively the guidelines and consensus publications and the highlights (original studies).

Guidelines and consensus.

TITLEJOURNALDATEIF AUTHORSDOI
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 OncolNov 202351,77Curigliano G, Burstein HJ, Gnant M, Loibl S, Cameron D, Regan MM, Denkert C, Poortmans P, Weber WP, Thürlimann B; St Gallen Consensus Conference Panelists 202310.1016/j.annonc.2023.08.017
The Lucerne Toolbox 2 to optimise axillary management for early breast cancer: a multidisciplinary expert consensus [ ]EClinicalMedicineJul 202317,03Kaidar-Person O, Pfob A, Gentilini OD, Borisch B, Bosch A, Cardoso MJ, Curigliano G, De Boniface J, Denkert C, Hauser N, Heil J, Knauer M, Kühn T, Lee HB, Loibl S, Mannhart M, Meattini I, Montagna G, Pinker K, Poulakaki F, Rubio IT, Sager P, Steyerova P, Tausch C, Tramm T, Vrancken Peeters MJ, Wyld L, Yu JH, Weber WP, Poortmans P, Dubsky P10.1016/j.eclinm.2023.102085
ESMO expert consensus statements (ECS) on the definition, diagnosis, and management of HER2-low breast cancer [ ]Ann OncolAug 202351,77Tarantino P, Viale G, Press MF, Hu X, Penault-Llorca F, Bardia A, Batistatou A, Burstein HJ, Carey LA, Cortes J, Denkert C, Diéras V, Jacot W, Koutras AK, Lebeau A, Loibl S, Modi S, Mosele MF, Provenzano E, Pruneri G, Reis-Filho JS, Rojo F, Salgado R, Schmid P, Schnitt SJ, Tolaney SM, Trapani D, Vincent-Salomon A, Wolff AC, Pentheroudakis G, André F, Curigliano G10.1016/j.annonc.2023.05.008
ESMO Expert Consensus Statements on the management of breast cancer during pregnancy (PrBC) [ ]Ann OncolOct 202351,77Loibl S, Azim HA Jr, Bachelot T, Berveiller P, Bosch A, Cardonick E, Denkert C, Halaska MJ, Hoeltzenbein M, Johansson ALV, Maggen C, Markert UR, Peccatori F, Poortmans P, Saloustros E, Saura C, Schmid P, Stamatakis E, van den Heuvel-Eibrink M, van Gerwen M, Vandecaveye V, Pentheroudakis G, Curigliano G, Amant F.10.1016/j.annonc.2023.08.001
Early breast cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up [ ]Ann OncolDec 202351,77S. Loibl, F. André, T. Bachelot, C. H. Barrios, J. Bergh, H. J. Burstein, M. J. Cardoso, L. A. Carey, S. Dawood, L. Del Mastro, C. Denkert, E. M. Fallenberg, P. A. Francis, H. Gamal-Eldin, K. Gelmon, C. E. Geyer, M. Gnant, V. Guarneri, S. Gupta, S. B. Kim, D. Krug, M. Martin, I. Meattini, M. Morrow, W. Janni, S. Paluch-Shimon, A. Partridge, P. Poortmans, L. Pusztai, M. M. Regan, J. Sparano T. Spanic, S. Swain, S. Tjulandin, M. Toi, D. Trapani, A. Tutt, B. Xu, G. Curigliano & N. Harbeck, on behalf of the ESMO Guidelines Committee10.1016/j.annonc.2023.11.016
Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: ASCO-College of American Pathologists Guideline Update [ ]JCOAug 202350,7Wolff AC, Somerfield MR, Dowsett M, Hammond MEH, Hayes DF, McShane LM, Saphner TJ, Spears PA, Allison KH10.1200/JCO.22.02864
Designing clinical trials based on modern imaging and metastasis-directed treatments in patients with oligometastatic breast cancer: a consensus recommendation from the EORTC Imaging and Breast Cancer Groups [ ]Lancet OncologyAug 202351,1Pasquier D, Bidaut L, Oprea-Lager DE, deSouza NM, Krug D, Collette L, Kunz W, Belkacemi Y, Bau MG, Caramella C, De Geus-Oei LF, De Caluwé A, Deroose C, Gheysens O, Herrmann K, Kindts I, Kontos M, Kümmel S, Linderholm B, Lopci E, Meattini I, Smeets A, Kaidar-Person O, Poortmans P, Tsoutsou P, Hajjaji N, Russell N, Senkus E, Talbot JN, Umutlu L, Vandecaveye V, Verhoeff JJC, van Oordt WMH, Zacho HD, Cardoso F, Fournier L, Van Duijnhoven F, Lecouvet FE10.1016/S1470-2045(23)00286-3
European Society of Breast Cancer Specialists/Advanced Breast Cancer Global Alliance quality indicators for metastatic breast cancer care [ ]Eur J CancerJul 20238,4Cardoso F, McCartney A, Ponti A, Marotti L, Vrieling C, Eniu A, Sousa B, Ripamonti C, Travado L, Spitz S, Jolly E, Curigliano G, Penault-Llorca F, Lecouvet F, Rubio IT, Biganzoli L.10.1016/j.ejca.2023.03.028
EUSOMA quality indicators for non-metastatic breast cancer: An update [ ]Isabel T. Rubio, Lorenza Marotti, Laura Biganzoli, Cynthia Aristei, Alexandra Athanasiou, Christine Campbell, Fatima Cardoso, Maria Joao Cardoso, Charlotte E. Coles, Manuela Eicher, Nadia Harbeck, Andreas Karakatsanis, Birgitte V. Offersen, Ruud Pijnappel, Antonio Ponti, Peter Regitnig, Donatella Santini, Francesco Sardanelli, Tanja Spanic, Zsuzsanna Varga, Marie Jeanne T.F.D. Vrancken Peeters, Yvonne Wengström, Lynda Wyld, Giuseppe Curigliano10.1016/j.ejca.2023.113500

Highlights.

Table 2

Publications in Table 2 are listed in alphabetic order/Abstract only publications are shaded in grey; * IF – Impact Factor.

The guidelines and consensus publication ( Table 1 ) represent collaborative efforts aimed at improving the standard of diagnosis and treatment for breast cancer patients both in early and metastatic breast cancer. These could be integrated in the routine clinical practice of breast units and teams and can help to guide difficult decisions, in the case of expert consensus recommendations, mainly when level I or 2 evidence is not available, filling the gaps of evidence-based guidelines.

The highlights ( Table 2 ) are the main focus of the current paper, hence we comment briefly only on the most interesting papers or abstracts (based on our views), that were reported in 2023.

1.1. Guidelines and consensus ( Table 1 )

Although many countries discuss and formulate national (locally-relevant) guidelines and consensus or position statements, international collaborative efforts that aggregate recommendations according to levels of evidence, or expert consensus where high-level evidence is limited, represent important a2e that can translate into more standardized care and potentially better patient outcomes. In this regard, we note from 2023 the St Gallen consensus [ 1 ] and also the Lucerne Toolbox [ 2 ] concerning the challenges in locoregional axillary approaches.

The ESMO effort in publishing guidelines produced in 2023 a consensus paper on the definition of HER2-low Breast Cancer [ 3 ]; the management of Breast Cancer during Pregnancy [ 4 ] and the awaited update of the Early Breast Cancer guidelines [ 5 ].

ASCO and the American College of Pathologists also updated the important guidelines for Her2 receptor testing [ 6 ].

EORTC's Imaging and Breast Cancer Groups wrote an insightful article, in a topic where guidance was lacking, for the purpose of helping the design of trials for oligometastatic disease with the use of new imaging techniques [ 7 ].

We also mention the recent consensus papers from EUSOMA (the European Society of Medical Specialists) on the upgraded quality indicators for the accreditation of Breast Units in Early Breast Cancer [ 8 ] and the new quality indicators for Metastatic Breast Cancer [ 9 ]. Although several accreditation systems are available for early breast cancer the proposed standardized approach had not been attempted in metastatic breast cancer.

1.2. Highlights ( Table 2 )

The table aims at highlighting all those studies or trials that have contributed new knowledge to the breast cancer field. Some of these are reported as abstracts but are highlighted because their novelty or importance justifies their inclusion in our commentary. Table 2 lists more studies than what we comment on next – we opted to focus on studies that in our view are exemplary highlights of 2023.

We added to the highlights a list of promising Randomized Clinical trial protocols published in 2023 that we hope will, in the near future, contribute new knowledge to improve the care of breast cancer patients.

2. Early breast cancer

2.1. imaging, screening [ 10 , 11 ].

Artificial intelligence-supported screen reading versus standard double reading in the Mammography Screening with Artificial Intelligence trial (MASAI): a clinical safety analysis of a randomised, controlled, non-inferiority, single-blinded, screening accuracy study (Lång et al.) [ 10 ].

This RCT showed that amongst 80 033 women having population-based screening (in Swedish program), AI-supported screening detected 244 cancers (cancer detection rate, CDR 6·1/1000), while standard double-(human)-reading detected 203 (CDR 5·1/1000)– a CDR ratio of 1·2 (95 % CI: 1·0–1·5; p = 0·052). Recall rates were 2·2 % in the intervention group and 2·0 % in the control group. The screen-reading workload was reduced by 44·3 % using the AI-supported approach.

This pivotal trial represents the highest-level evidence on AI for early detection and is critical in supporting the planning of additional prospective trials in real-world screening practice. While current findings show the effectiveness and efficiency of AI-supported screen-reading, the RCT's primary endpoint, effect on interval cancer rates (which entails follow-up of participants), is eagerly awaited and will provide a key outcome on AI's effectiveness for population screening programs.

Comparison of Contrast-enhanced Mammography with MRI Utilizing an Enriched Reader Study: A Breast Cancer Study (CONTRRAST Trial) (Philips et al.) [ 11 ].

2.2. Prevention [ 12 ]

Randomized Placebo Controlled Trial of Low-Dose Tamoxifen to Prevent Recurrence in Breast Non-invasive Neoplasia: A 10-Year Follow-Up of TAM-01 Study (Lazzeroni et al.) [ 12 ].

This RCT reported long-term follow-up of women with atypical ductal hyperplasia, lobular carcinoma in situ, or hormone-sensitive or unknown DCIS (with the latter representing the largest group in the trial), treated with low-dose tamoxifen or administered placebo. After a median follow-up of 9.7 years, 66 breast cancers (15 in situ; 51 invasive) were diagnosed: 25 in the low-dose tamoxifen group and 41 in the placebo group (annual rate per 1,000 person-years, 11.3 with tamoxifen v 19.5 with placebo; hazard ratio = 0.58; 95 % CI: 0.35–0.95; log-rank P  = 0.03). Most recurrences were invasive (77 %) and ipsilateral (59 %).

This trial highlights the importance of primary and tertiary prevention, showing that low-dose Tamoxifen (5 mg per day) taken for 3 years has a sustained effect in preventing breast cancer and breast cancer recurrence in those with breast intraepithelial proliferations and non-invasive malignancy, respectively, without substantial long-term adverse events.

2.3. Genetics [ 13 ]

Contralateral Breast Cancer Risk Among Carriers of Germline Pathogenic Variants in ATM, BRCA1, BRCA2, CHEK2, and PALB2 (Yadav et al.) [ 13 ].

The study included 15 104 women from Cancer Risk Estimates Related to Susceptibility (CARRIERS) consortium treated with ipsilateral surgery for invasive breast cancer. Included prospective studies reported on contralateral breast cancer (CBC) after a breast cancer diagnosis in women with a germline pathogenic variant (BRCA1, BRCA2, PALB2, CHEK2 and ATM) and breast cancer survivors without a pathogenic variant matched for age of diagnosis, race/ethnicity, menopausal status, histology, estrogen receptor status of the first breast cancer, and use of endocrine therapy to estimate the risk of CBC.

Results showed that BRCA1, BRCA2, and CHEK2 PV carriers with breast cancer were at significantly elevated risk of CBC (HR 1.9) whereas among the PALB2 PV carriers only those with ER-negative breast cancer had elevated risks (HR, 2.9). By contrast, ATM PV carriers did not have significantly increased CBC risks. The 10-year cumulative incidence of CBC was substantially higher in pre-menopausal patients.

The study provides important information that can be applied in practice for surveillance and risk reducing strategies of these women. The work highlights that carriers of germline PVs in BRCA1/2, CHEK2, PALB2 are at substantially increased risk of CBC although age can have a striking impact on future risk. Information should be passed on to patients adding also what the authors of the paper stated, that the impact of risk reducing strategies on overall survival is still controversial.

2.4. Pathology [ 14 ]

Characterisation of luminal and triple-negative breast cancer with HER2 Low protein expression (Atallah et al.) [ 14 ].

2.5. Surgery [ [15] , [16] , [17] ]

Effect of Peritumoral Infiltration of Local Anaesthetic Before Surgery on Survival in Early Breast Cancer (Badwe et al.) [ 15 ].

This is a surprising trial using the recently explored concept that local anesthesia seems to block voltage-gated sodium channels, present in cancer cells, hindering the activation of pro-metastatic pathways.

The RCT tested the impact of presurgical, peritumoral infiltration of local anesthesia on disease-free survival (DFS) and overall survival (OS). Women with breast cancer planned for upfront surgery received either peritumoral injection of 0.5 % lidocaine, 7–10 min before surgery (local anesthetics [LA] arm) or surgery without lidocaine (no LA arm). 1,583 patients were included in this analysis (LA, 796; no LA, 804). 5-Year DFS rates were 86.6 % and 82.6 % (hazard ratio [HR], 0.74; 95 % CI, 0.58 to 0.95; P = 0.017) and 5-year OS rates were 90.1 % and 86.4 %, respectively (HR, 0.71; 95 % CI, 0.53 to 0.94; P = 0.019) in LA vs no LA. Using competing risk analyses, in LA and no LA arms, 5-year cumulative incidence rates of locoregional recurrence were 3.4 % and 4.5 % (HR, 0.68; 95 % CI, 0.41 to 1.11), and distant recurrence rates were 8.5 % and 11.6 %, respectively (HR, 0.73; 95 % CI, 0.53 to 0.99). Authors concluded that peritumoral injection of lidocaine before breast cancer surgery significantly increases DFS and OS at 5 years follow-up.

A word of caution needs to be said as the molecular mechanisms related to the observed benefit in the trial are not yet clear. Surprisingly the same benefit was observed in breast conservation and mastectomy patients questioning the vicinity of the surgical incision to the tumour as a possible vehicle of dissemination.

Local Recurrence After Breast-Conserving Therapy in Patients With Multiple Ipsilateral Breast Cancer: Results From ACOSOG Z11102 (Alliance). (Boughey et al.) [ 16 ].
Sentinel Lymph Node Biopsy vs No Axillary Surgery in Patients With Small Breast Cancer and Negative Results on Ultrasonography of Axillary Lymph Nodes: The SOUND Randomized Clinical Trial (Gentilini et al.) [ 17 ].

The SOUND (Sentinel node vs Observation after axillary ultra-soUND) trial was a prospective randomized non-inferiority study comparing sentinel node biopsy (SLNB) vs no axillary surgery in patients with breast cancer up to 2 cm receiving breast conserving therapy after a preoperative ultrasound with negative axillary lymph nodes. The primary outcome was distant disease-free survival (DDFS), analysed by intention to treat. Between February 6, 2012 and June 30, 2017, 1,463 women were enrolled: 727 were randomly assigned to receive SLNB and 736 were assigned to no axillary surgery. 19 and 39 patients were excluded in the SLNB group and in the no axillary surgery group, respectively. Overall, median tumor size was 1·1 cm (IQR 0.8–1.5), median age 60 years (IQR 52–68) and 1,234 (87·8 %) patients had estrogen receptor-positive HER2-non-overexpressing BC. In the SLNB group, 97 patients (13.7 %) had positive axillary nodes. The median follow-up was 5.7 years (IQR 5.0–6.7 years). Five-year DDFS was 97.7 % in the SLNB arm and 98.0 % in the no axillary surgery arm (Log-rank test P = 0.665; HR 0.84; 90 % CI 0.45–1.54; non-inferiority P = 0.024). 12 (1.7 %) loco-regional relapses, 13 (1.8 %) distant metastases and 21 (3.0 %) deaths were observed in the SLNB group, whereas 11 (1.6 %) loco-regional relapses, 14 (2.0 %) distant metastases and 18 (2.6 %) deaths were observed in the no axillary surgery group. In this trial the omission of axillary surgery was non-inferior to SLNB in patients with small breast cancer and a negative ultrasound of the axillary lymph nodes. The authors concluded that patients with these features can be safely spared any axillary surgery whenever the lack of pathologic information is not affecting the postoperative treatment plan.

2.6. Radiation therapy [ [18] , [19] , [20] , [21] ]

Dose-escalated simultaneous integrated boost radiotherapy in early breast cancer (IMPORT HIGH): a multicentre, phase 3, non-inferiority, open-label, randomised controlled trial (Coles et al.) [ 18 ].

An increased “boost” dose to the primary tumour bed following whole-breast irradiation (WBI) halves the risk for local recurrences. However, it increases the total number of treatment fractions and has a negative impact on the cosmetic outcome.

The IMPORT HIGH trial (ISRCTN47437448) compared 2 dose levels of a simultaneous integrated boost, adding no extra fractions, with a sequential boost consisting of extra fractions following WBI:

  • • Control group: 40 Gy in 15 fractions WBI, followed by a sequential boost consisting of 16 Gy in 8 fractions on the primary tumourbed.
  • • Test group 1: 36 Gy in 15 fractions WBI, with concomitant 40 Gy in 15 fractions to a partial breast volume and 48 Gy in 15 fractions to the primary tumourbed.
  • • Test group 2: 36 Gy in 15 fractions WBI, with concomitant 40 Gy in 15 fractions to a partial breast, volume and 53 Gy in 15 fractions to the primary tumourbed.

A total of 2617 patients after breast-conserving surgery for non-metastasised invasive breast carcinoma participated between March 2009 and September 2015, equally assigned over the 3 randomisation groups. The primary endpoint was ipsilateral breast tumour recurrence (IBTR), non-inferiority of the test arms if a less than 3 % increased risk for 5-years IBTR was seen above the estimated 5 % for the control group. Side events were assessed by clinicians, patients, and photographs.

Importantly, the median boost clinical target volume was only 13 cm3. After a median follow-up of 74 months, a total of 76 IBTR were reported, 20 for the control group, 21 for test group 1, and 35 for test group 2, resulting in a 5-year IBTR rate of 1.9 % (control), 2.0 % (test 1), and 3.2 % (test 2). These differences were not significant and didn't pass the pre-set estimations, confirming non-inferiority for test group 1, 36-40-48 Gy simultaneous boost technique. Also, the cumulative 5-year incidence of clinician-reported moderate or marked breast induration was similar for test group 1 (10.6 %) and the control group (11.5 %) for the control group, whereas it was higher for test group 2 (15.5 %) (p = 0·015 vs control group).

Conclusion: Local control rates were better in all groups than expected and side effects were limited, likely thanks to the small boost volumes. Increasing the (simultaneous) boost dose didn't result in improved control while it increased side effects. The scheme from test group 1 is safe, both in terms of local control as in side effects, and limits the total number of fractions from 23 to 15.

Note: a simultaneous integrated boost combined with the 1-weekly 26 Gy in 5 fractions schedule from the FAST-FORWARD trial is subject of a new study.

Radiotherapy to regional nodes in early breast cancer: an individual patient data meta-analysis of 14324 women in 16 trials (EBCTCG) [ 19 ].

Regional nodal irradiation (RNI) reduces risks of recurrence and death in patients with risk factors, but was associated with increased non-cancer related mortality.

In this meta-analysis, 16 trials evaluating RNI were evaluated, 8 (n = 2157) starting between 1961 and 1978, and 8 (n = 12167) between 1989 and 2008. In the older trials, RNI had no significant effect on breast cancer mortality (RR = 1.04, p = 0·55), while non-breast-cancer mortality was significantly increased (RR = 1.42, p = 0·00023), especially after longer follow-up, leading to an increased overall mortality (RR = 1.17, p = 0·0067). In sharp contrast to this, in the newer trials, RNI significantly reduced recurrence rates (RR = 0.88, p = 0·0008). This was especially seen for distant recurrence, and only few lymph node recurrences were reported. Moreover, RNI significantly reduced breast cancer-related mortality (RR = 0.87, p = 0·0010), and no increased non-breast-cancer-related mortality was seen (RR = 0.97, p = 0·63), finally resulting in a significantly reduced all-cause mortality (RR = 0.90, p = 0·0022). The estimated absolute reductions in 15-year breast cancer-related mortality were 1.6 % for pN0 disease, 2.7 % for pN1 stage, and 4.5 % for pN2-3 staged patients.

Conclusion: Regional nodal irradiation significantly reduced breast cancer-related mortality and overall mortality in trials which started after 1989, in contrast to older trials. This might be explained by the introduction of a combination of technical improvements in radiation therapy in the second half of the eighties.

Breast-Conserving Surgery with or without Irradiation in Early Breast Cancer (Kunkler et al.) (20)
Omitting Radiotherapy after Breast-Conserving Surgery in Luminal A Breast Cancer. LUMINA Trial (Whelan et al.) (21)

2.7. Systemic Treatment [ [22] , [23] , [24] , [25] , [26] ]

KEYNOTE-756: Phase III study of neoadjuvant pembrolizumab (pembro) or placebo (pbo) + chemotherapy(chemo), followed by adjuvant pembro or pbo + endocrine therapy (ET) for early-stage high-risk ER+/HER2e breast
Cancer (Cardoso et al.) [ 22 ].
Anthracycline-containing and taxane-containing chemotherapy for early-stage operable breast cancer: a patient-level meta-analysis of 100 000 women from 86 randomised trials by EBCTCG [ 23 ].

This is an individual patient-level meta-analysis of trials in early breast cancer, comparing efficacy of taxane regimens with versus without anthracycline, based on the data from 15 trials, which included 18103 patients and of 23 anthracycline regimens with versus without taxane, based on 35 trials of 52976 patients.

Across all trials assessing the effect of anthracyclines a 14 % relative reduction in recurrence rates was observed among patients treated with an anthracycline, albeit the benefit was limited to patients treated with concurrent docetaxel plus anthracycline versus same dose docetaxel plus cyclophosphamide, or taxane plus anthracycline versus higher cumulative dose taxane with or without capecitabine.

For the assessment of taxane effect, an overall 13 % relative reduction in the risk of recurrence was observed among patients administered a taxane. Larger recurrence reductions were seen from adding taxane to anthracycline regimens when the same cumulative dose of anthracycline was used than in trials with higher cumulative doses of non-taxane (mostly anthracycline) in the control group. Direct comparisons between anthracycline and taxane regimens demonstrated higher efficacy of higher cumulative dose and more dose-intense schedules. Importantly, the relative benefits from taxanes and anthracyclines were similar across estrogen receptor status, age, nodal status, or tumour size or grade.

Albeit the results of this meta-analysis support use of higher cumulative dose and more dose-intense schedules, potentially questioning the current tendency to omit anthracyclines and/or decrease the duration of chemotherapy, they should be interpreted with caution, given the years the contributing trials were conducted and the tremendous progress in the understanding of biology of breast cancer, resulting in better selection of patients and more active systemic therapies available today.

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
(Johnston SRB et al.) (24)

In this Phase 3 RCT 5637 patients were randomly assigned; 2808 received abemaciclib plus endocrine therapy and 2829 received endocrine therapy alone. At a median follow-up of 42 months, median invasive disease-free survival was not reached in either group, and the invasive disease-free survival benefit previously reported was sustained: HR 0·664 (nominal p < 0∙001). At 4 years, the absolute difference in invasive disease-free survival between the groups was 6.4 % (85.8 % in the abemaciclib plus endocrine therapy group vs 79.4 % in the endocrine therapy alone group). Similarly, a sustained benefit in distant relapse-free survival was observed, with absolute improvement in 4-year dRFS of 5.9 % (88.4 % vs 82.5 %, HR 0. 659, nominal p < 0.001). Overall survival data are immature, but significantly less patients have developed and are alive with metastatic disease in the abemaciclib arm (125 vs 249), raising hopes for overall survival benefit.

These results further support the use of abemaciclib in patients with high-risk hormone receptor-positive, HER2-negative early breast cancer, albeit further follow-up is needed to establish whether overall survival can be improved with abemaciclib plus endocrine therapy in these patients.

A randomized, double-blind trial of nivolumab (NIVO) vs placebo (PBO) with neoadjuvant chemotherapy (NACT) followed by adjuvant endocrine therapy (ET) ± NIVO in patients (pts) with high-risk, ER + HER2L primary breast cancer (BC) CheckMate 7FL (Loi et al.) [ 25 ].
Ribociclib and endocrine therapy as adjuvant treatment in patients with HR+/HER2− early breast cancer: Primary results from the phase III NATALEE trial (Slamon et al.) [ 26 ].

The results of the NATALEE trial were presented at ASCO 2023. NATALEE is a phase 3 RCT which assessed the efficacy and toxicity of CDK4/6 inhibitor ribociclib in patients receiving adjuvant endocrine therapy for intermediate/high risk disease with luminal HER2-negative non-metastatic breast cancer. The study enrolled 5101 patients who were randomly allocated to an Aromatase Inhibitor alone or combined with ribociclib 400 mg/d for 3 years. The dosing was reduced to 400 mg/day to improve tolerability and 3 years treatment duration was used with the intention to drive tumor cells into irreversible senescence. Patients were included with stage IIB and III, stage IIA node-positive and stage IIA node-negative disease with additional high-risk features. The primary endpoint of invasive disease-free survival (iDFS) was met with 3.3 % absolute reduction in the frequency of iDFS events (90.4 % vs 87.1 %, HR 0.748, p = 0.0014) at median follow up of 27.7 months. The study additionally demonstrated significant improvement in distant disease-free survival (90.8 % vs 88.6 %, HR 0.739, p = 0.0017). These results should be analysed with caution as at the data cutoff point only 20 % of patients have completed 3 years of ribociclib treatment. Additionally, the final manuscript is still awaited. Should these early data be confirmed, adjuvant ribociclib could become a new opportunity for improving outcomes for even a larger population of patients treated for early luminal HER2-negative breast cancer.

2.8. Quality of life [ [27] , [28] , [29] ]

Significantly longer time to deterioration of quality of life due to CANKADO PRO-React eHealth support in HR + HER2- metastatic breast cancer patients receiving palbociclib and endocrine therapy: primary outcome analysis of the multicenter randomized AGO-B WSG PreCycle trial (Harbeck et al.) [ 27 ].
Pregnancy After Breast Cancer in Young BRCA Carriers (Partridge et al.) [ 28 ].
Interrupting Endocrine Therapy to Attempt Pregnancy after Breast Cancer (Partridge et al.) [ 29 ].

POSITIVE was a prospective international single-group trial evaluating the temporary interruption of adjuvant endocrine therapy to attempt pregnancy in young (≤42 years) women, with previous ER-positive early breast cancer, who received adjuvant hormonal treatment for no less than 18 and no more than 30 months. The primary end point was the number of breast cancer events (local, regional, distant recurrence or new contralateral invasive breast cancer) during follow-up.

From 497 women who were followed for pregnancy 365 babies were born. At 1638 patient-years of follow-up (median 41 months), 44 patients had a breast cancer event, a result that did not exceed the safety threshold. The 3-year incidence of breast cancer events was 8.9 % (95 % confidence interval [CI], 6.3 to 11.6) in the treatment-interruption group and 9.2 % (95 % CI, 7.6 to 10.8) in the control cohort.

The POSITIVE trial represents an admirable joint effort that intends to give reassurance to women desiring a pregnancy after a breast cancer diagnosis. The main limitations of the trial are the single-arm trial design (with an external cohort used as control) and the short follow-up time. However, it still represents the best available evidence on this issue.

3. Advanced breast cancer [ [30] , [31] , [32] ]

Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): a randomised, open-label, multicentre, phase 3 trial (Rugo et al.) [ 30 ].

TROPiCS-02 was originally presented at ASCO 2022, demonstrating a 34 % reduction in the risk of progression or death (HR 0.66, p < 0.001) among 543 patients treated with sacituzumab govitecan (SG) vs treatment of physician's choice (capecitabine, vinorelbine, gemcitabine or eribulin) for advanced HR+/HER2-breast cancer that progressed after at least 2 lines of chemotherapy for metastatic disease. Last year we've seen mature overall survival results, demonstrating a prolongation in median OS from 11.2 months in the control arm to 14.4 months in patients treated with SG (HR 0.79, p = 0.02). Additionally, SG-treated patients experienced higher overall response rate (21 % vs 14 %; OR 1.63; p = 0·035), and prolongation of time to deterioration of global health status/quality of life (HR 0.75; p = 0.0059) and fatigue (HR 0.73; p = 0.0021).

Presented data confirm the efficacy of SG in this heavily pretreated population (median 3 prior chemotherapy lines, median time form MBC diagnosis of approximately 48 months), where treatment options are limited.

Primary outcome analysis of the phase 3 SONIA trial (BOOG 2017–03) on selecting the optimal position of cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors for patients with hormone receptor-positive (HR+), HER2-negative (HER2-) advanced breast cancer (ABC) (Sonke et al.) [ 31 ].

CDK4/6 inhibitors have greatly improved outcome for patients with advanced ER+/HER2-breast cancer. Studies have shown survival benefits when CDK4/6 inhibitors added to either first-line or second-line endocrine treatment. In the absence of direct comparisons, there was uncertainty whether a first-line or a second-line strategy has the optimal balance for patients between benefits and risks. The SONIA trial ( {"type":"clinical-trial","attrs":{"text":"NCT03425838","term_id":"NCT03425838"}} NCT03425838 ) included 1050 patients with advanced ER+/HER2-breast cancer who had not received treatment for advanced disease. All patients received first-line aromatase inhibitor follow by second-line fulvestrant upon disease progression. Patients were randomized to receive any of the three available CDK4/6 inhibitors either in first or in second line. Primary endpoint was progression-free survival after both protocol-defined treatment lines (PFS2). Overall survival, toxicity and quality-of-life were key secondary endpoints. PFS2 did not differ between the treatment arms (hazard ratio 0.87; 95 % CI 0.74–1.03). OS (HR 0.98; 95 % CI 0.80–1.20) and QoL (FACT-B total score p-value 0.4) were also similar. A first-line strategy, however, was associated with 16.5 months longer duration of CDK4/6 inhibitor use, leading to 74 % more grade 3-4 adverse events. Therefore, SONIA possibly challenges the need for CDK4/6 inhibitor use in first-line, albeit the clinical significance of this result in view of available targeted therapy options in 2nd line setting and use in the trial of possibly less biologically active CDK4/6 inhibitor seems rather questionable. Biomarkers should try to identify patients that could benefit from first-line use. Trials like SONIA can significantly reduce the toxicity of effective drugs and make them accessible to those who would otherwise find them unaffordable. These trials are self-funded as the costs are covered by the savings achieved through the less expensive treatments used in the trial.

Capivasertib in Hormone Receptor-Positive Advanced Breast Cancer (Turner et al.) [ 32 ].

CAPItello-291 was phase 3, randomized, double-blind trial conducted in patients with HR+/HER2–advanced breast cancer who developed disease progression during or after treatment with an aromatase inhibitor, with or without CDK4/6 inhibitor. 708 patients were randomly assigned to receive fulvestrant combined with capivasertib or placebo. PFS prolongation was found both in the overall population (median 7.2 vs 3.6 months, HR 0.6, p < 0.001) and in patients with AKT pathway–altered tumors (median 7.3 vs 3.1 months, HR 0.5, p < 0.001). Overall survival results are immature. The most frequent adverse events of grade ≥3 in patients treated with capivasertib were rash (in 12.1 % vs. in 0.3 %) and diarrhea (in 9.3 % vs. 0.3 %). 13 % of patients in the capivasertib arm (vs 2.3 % in the placebo arm) discontinued treatment due to toxicity.

Capivasterib creates a new treatment option in the difficult-to-treat patients failing endocrine therapy combined with a CDK4/6 inhibitor. Being active in both overall population and in AKT pathway–altered tumors it potentially creates an alternative to both everolimus and alpelisib and we need to learn, how to best select/sequence these agents, as well as how to manage their sometimes non-trivial toxicities.

4. Controlled clinical trial protocols [ [33] , [34] , [35] , [36] , [37] , [38] , [39] ] these are listed in Table 2

4.1. conclusion.

The above-tabulated and narrated work has discussed selected studies and recommendations relevant to breast cancer care from the year that has passed. Highlighted original work was dominated by RCTs, but also featured cohort studies and individual patient data meta-analyses leveraging the collective data from multiple trials. Published clinical trial protocols, while not discussed, provide an opportunity for early sharing of research concepts and methods. We look forward to findings from these studies and other research as we begin a new year of learning and practice in breast cancer.

The selection is the full responsibility of the authors.

Revision note

The change was the inclusion of an extra reference that altered the following reference sequence.

Along the text – nothing major was changed.

As such we opted to upload a new version without marking.

CRediT authorship contribution statement

Maria-Joao Cardoso: Writing – review & editing, Writing – original draft, Methodology, Conceptualization. Philip Poortmans: Writing – original draft. Elżbieta Senkus: Writing – review & editing, Writing – original draft. Oreste D. Gentilini: Writing – review & editing, Writing – original draft. Nehmat Houssami: Writing – review & editing, Writing – original draft, Supervision, Conceptualization.

Declaration of competing interest

N. Houssami receives funding via a NBCF Chair in Breast Cancer Prevention grant (EC-21-001) and a NHMRC Investigator (Leader) grant (1194410); MJ Cardoso and O Gentilini receive institutional funding as Leading Investigator and Principal Investigator of the Project: 101057389 — CINDERELLA — HORIZON-HLTH-2021-DISEASE-04.

Maria-Joao Cardoso honoraria: AstraZeneca, Eli Lilly, MSD, Roche, Novartis.

Oreste Gentilini honoraria: MSD, Astra-Zeneca, BD, Bayer, Eli-Lilly.

Elżbieta Senkus honoraria: AstraZeneca, Cancérodigest, Curio Science, Egis, Eli Lilly, Exact Sciences, Gilead, high5md, MSD, Novartis, Pfizer, Pierre Fabre, Roche; travel support: AstraZeneca, Egis, Gilead, Novartis, Pfizer, Roche; contracted research: Amgen, AstraZeneca, Daiichi Sankyo, Eli Lilly, Novartis, OBI Pharma, Pfizer, Roche, Samsung; grants: Pfizer (Stowarzyszenie Różowy Motyl); medical writing: Astellas, AstraZeneca, Eli Lilly; royalties: Springer; leadership or fiduciary role: Stowarzyszenie Różowy Motyl; stock: AstraZeneca, Eli Lilly, Pfizer.

Philip Poortmans no conflicts no disclose.

Acknowledgements

We thank the contribution and comments of Giuseppe Curigliano, Hope Rugo and Alberto Tagliafico for the selection of papers/abstracts listed. We also thank Gabe Sonke for the SONIA Trial comment.

BREAST CANCER RESEARCH AND TREATMENT - WoS Journal Info

Breast Cancer Research and Treatment

breast cancer research and treatment impact factor 2023

Subject Area and Category

  • Cancer Research

Springer New York

Publication type

01676806, 15737217

Information

How to publish in this journal

breast cancer research and treatment impact factor 2023

The set of journals have been ranked according to their SJR and divided into four equal groups, four quartiles. Q1 (green) comprises the quarter of the journals with the highest values, Q2 (yellow) the second highest values, Q3 (orange) the third highest values and Q4 (red) the lowest values.

CategoryYearQuartile
Cancer Research1999Q2
Cancer Research2000Q1
Cancer Research2001Q1
Cancer Research2002Q2
Cancer Research2003Q2
Cancer Research2004Q3
Cancer Research2005Q2
Cancer Research2006Q2
Cancer Research2007Q1
Cancer Research2008Q1
Cancer Research2009Q1
Cancer Research2010Q1
Cancer Research2011Q1
Cancer Research2012Q1
Cancer Research2013Q1
Cancer Research2014Q1
Cancer Research2015Q1
Cancer Research2016Q1
Cancer Research2017Q1
Cancer Research2018Q1
Cancer Research2019Q1
Cancer Research2020Q1
Cancer Research2021Q2
Cancer Research2022Q2
Cancer Research2023Q2
Oncology1999Q1
Oncology2000Q1
Oncology2001Q1
Oncology2002Q2
Oncology2003Q2
Oncology2004Q2
Oncology2005Q1
Oncology2006Q1
Oncology2007Q1
Oncology2008Q1
Oncology2009Q1
Oncology2010Q1
Oncology2011Q1
Oncology2012Q1
Oncology2013Q1
Oncology2014Q1
Oncology2015Q1
Oncology2016Q1
Oncology2017Q1
Oncology2018Q1
Oncology2019Q1
Oncology2020Q1
Oncology2021Q1
Oncology2022Q1
Oncology2023Q1

The SJR is a size-independent prestige indicator that ranks journals by their 'average prestige per article'. It is based on the idea that 'all citations are not created equal'. SJR is a measure of scientific influence of journals that accounts for both the number of citations received by a journal and the importance or prestige of the journals where such citations come from It measures the scientific influence of the average article in a journal, it expresses how central to the global scientific discussion an average article of the journal is.

YearSJR
19990.927
20001.151
20011.307
20020.645
20030.682
20040.749
20051.338
20061.540
20071.914
20082.004
20091.979
20102.292
20112.406
20122.485
20132.321
20142.397
20152.432
20162.181
20172.066
20181.899
20191.905
20201.908
20211.407
20221.442
20231.267

Evolution of the number of published documents. All types of documents are considered, including citable and non citable documents.

YearDocuments
1999166
2000151
2001585
2002180
2003226
2004179
2005230
2006241
2007251
2008352
2009429
2010582
2011655
2012664
2013457
2014401
2015426
2016345
2017440
2018465
2019466
2020466
2021466
2022365
2023349

This indicator counts the number of citations received by documents from a journal and divides them by the total number of documents published in that journal. The chart shows the evolution of the average number of times documents published in a journal in the past two, three and four years have been cited in the current year. The two years line is equivalent to journal impact factor ™ (Thomson Reuters) metric.

Cites per documentYearValue
Cites / Doc. (4 years)19992.438
Cites / Doc. (4 years)20002.776
Cites / Doc. (4 years)20012.942
Cites / Doc. (4 years)20022.044
Cites / Doc. (4 years)20031.849
Cites / Doc. (4 years)20041.946
Cites / Doc. (4 years)20052.182
Cites / Doc. (4 years)20063.551
Cites / Doc. (4 years)20073.542
Cites / Doc. (4 years)20084.038
Cites / Doc. (4 years)20094.051
Cites / Doc. (4 years)20104.551
Cites / Doc. (4 years)20114.547
Cites / Doc. (4 years)20124.612
Cites / Doc. (4 years)20134.481
Cites / Doc. (4 years)20144.377
Cites / Doc. (4 years)20154.439
Cites / Doc. (4 years)20164.419
Cites / Doc. (4 years)20174.163
Cites / Doc. (4 years)20183.843
Cites / Doc. (4 years)20193.766
Cites / Doc. (4 years)20204.262
Cites / Doc. (4 years)20214.563
Cites / Doc. (4 years)20224.265
Cites / Doc. (4 years)20233.721
Cites / Doc. (3 years)19992.438
Cites / Doc. (3 years)20002.678
Cites / Doc. (3 years)20012.992
Cites / Doc. (3 years)20021.661
Cites / Doc. (3 years)20031.675
Cites / Doc. (3 years)20041.779
Cites / Doc. (3 years)20053.258
Cites / Doc. (3 years)20063.649
Cites / Doc. (3 years)20073.666
Cites / Doc. (3 years)20084.078
Cites / Doc. (3 years)20094.097
Cites / Doc. (3 years)20104.622
Cites / Doc. (3 years)20114.542
Cites / Doc. (3 years)20124.577
Cites / Doc. (3 years)20134.492
Cites / Doc. (3 years)20144.343
Cites / Doc. (3 years)20154.627
Cites / Doc. (3 years)20164.262
Cites / Doc. (3 years)20174.007
Cites / Doc. (3 years)20183.601
Cites / Doc. (3 years)20193.770
Cites / Doc. (3 years)20204.210
Cites / Doc. (3 years)20214.510
Cites / Doc. (3 years)20224.074
Cites / Doc. (3 years)20233.359
Cites / Doc. (2 years)19992.139
Cites / Doc. (2 years)20002.482
Cites / Doc. (2 years)20012.552
Cites / Doc. (2 years)20021.304
Cites / Doc. (2 years)20031.356
Cites / Doc. (2 years)20042.618
Cites / Doc. (2 years)20053.202
Cites / Doc. (2 years)20063.606
Cites / Doc. (2 years)20073.497
Cites / Doc. (2 years)20083.913
Cites / Doc. (2 years)20094.068
Cites / Doc. (2 years)20104.625
Cites / Doc. (2 years)20114.376
Cites / Doc. (2 years)20124.395
Cites / Doc. (2 years)20134.377
Cites / Doc. (2 years)20144.450
Cites / Doc. (2 years)20154.394
Cites / Doc. (2 years)20163.856
Cites / Doc. (2 years)20173.658
Cites / Doc. (2 years)20183.329
Cites / Doc. (2 years)20193.585
Cites / Doc. (2 years)20204.184
Cites / Doc. (2 years)20214.198
Cites / Doc. (2 years)20223.553
Cites / Doc. (2 years)20233.046

Evolution of the total number of citations and journal's self-citations received by a journal's published documents during the three previous years. Journal Self-citation is defined as the number of citation from a journal citing article to articles published by the same journal.

CitesYearValue
Self Cites199992
Self Cites200069
Self Cites200180
Self Cites200244
Self Cites200365
Self Cites200457
Self Cites2005101
Self Cites2006101
Self Cites2007128
Self Cites2008146
Self Cites2009210
Self Cites2010473
Self Cites2011610
Self Cites2012632
Self Cites2013472
Self Cites2014391
Self Cites2015328
Self Cites2016227
Self Cites2017226
Self Cites2018226
Self Cites2019245
Self Cites2020236
Self Cites2021239
Self Cites2022191
Self Cites2023141
Total Cites19991131
Total Cites20001272
Total Cites20011469
Total Cites20021498
Total Cites20031534
Total Cites20041763
Total Cites20051906
Total Cites20062317
Total Cites20072383
Total Cites20082944
Total Cites20093458
Total Cites20104770
Total Cites20116191
Total Cites20127626
Total Cites20138539
Total Cites20147713
Total Cites20157042
Total Cites20165473
Total Cites20174696
Total Cites20184361
Total Cites20194712
Total Cites20205772
Total Cites20216300
Total Cites20225696
Total Cites20234357

Evolution of the number of total citation per document and external citation per document (i.e. journal self-citations removed) received by a journal's published documents during the three previous years. External citations are calculated by subtracting the number of self-citations from the total number of citations received by the journal’s documents.

CitesYearValue
External Cites per document19992.239
External Cites per document20002.533
External Cites per document20012.829
External Cites per document20021.612
External Cites per document20031.604
External Cites per document20041.721
External Cites per document20053.085
External Cites per document20063.490
External Cites per document20073.469
External Cites per document20083.875
External Cites per document20093.848
External Cites per document20104.164
External Cites per document20114.095
External Cites per document20124.198
External Cites per document20134.244
External Cites per document20144.123
External Cites per document20154.411
External Cites per document20164.086
External Cites per document20173.814
External Cites per document20183.415
External Cites per document20193.574
External Cites per document20204.038
External Cites per document20214.339
External Cites per document20223.938
External Cites per document20233.251
Cites per document19992.438
Cites per document20002.678
Cites per document20012.992
Cites per document20021.661
Cites per document20031.675
Cites per document20041.779
Cites per document20053.258
Cites per document20063.649
Cites per document20073.666
Cites per document20084.078
Cites per document20094.097
Cites per document20104.622
Cites per document20114.542
Cites per document20124.577
Cites per document20134.492
Cites per document20144.343
Cites per document20154.627
Cites per document20164.262
Cites per document20174.007
Cites per document20183.601
Cites per document20193.770
Cites per document20204.210
Cites per document20214.510
Cites per document20224.074
Cites per document20233.359

International Collaboration accounts for the articles that have been produced by researchers from several countries. The chart shows the ratio of a journal's documents signed by researchers from more than one country; that is including more than one country address.

YearInternational Collaboration
199915.06
200011.26
20016.84
200217.22
200317.26
200423.46
200520.87
200624.90
200728.69
200828.69
200926.11
201027.32
201124.73
201230.12
201327.57
201431.42
201527.46
201628.41
201722.05
201827.31
201922.53
202024.89
202125.32
202223.01
202319.48

Not every article in a journal is considered primary research and therefore "citable", this chart shows the ratio of a journal's articles including substantial research (research articles, conference papers and reviews) in three year windows vs. those documents other than research articles, reviews and conference papers.

DocumentsYearValue
Non-citable documents19999
Non-citable documents20008
Non-citable documents20018
Non-citable documents20028
Non-citable documents20038
Non-citable documents200412
Non-citable documents200510
Non-citable documents200619
Non-citable documents200726
Non-citable documents200832
Non-citable documents200946
Non-citable documents201075
Non-citable documents2011124
Non-citable documents2012203
Non-citable documents2013199
Non-citable documents2014163
Non-citable documents201576
Non-citable documents201656
Non-citable documents201745
Non-citable documents201861
Non-citable documents201988
Non-citable documents2020101
Non-citable documents2021101
Non-citable documents202290
Non-citable documents202380
Citable documents1999455
Citable documents2000467
Citable documents2001483
Citable documents2002894
Citable documents2003908
Citable documents2004979
Citable documents2005575
Citable documents2006616
Citable documents2007624
Citable documents2008690
Citable documents2009798
Citable documents2010957
Citable documents20111239
Citable documents20121463
Citable documents20131702
Citable documents20141613
Citable documents20151446
Citable documents20161228
Citable documents20171127
Citable documents20181150
Citable documents20191162
Citable documents20201270
Citable documents20211296
Citable documents20221308
Citable documents20231217

Ratio of a journal's items, grouped in three years windows, that have been cited at least once vs. those not cited during the following year.

DocumentsYearValue
Uncited documents1999125
Uncited documents2000117
Uncited documents2001104
Uncited documents2002459
Uncited documents2003469
Uncited documents2004502
Uncited documents2005130
Uncited documents2006130
Uncited documents2007106
Uncited documents200889
Uncited documents2009117
Uncited documents2010139
Uncited documents2011171
Uncited documents2012259
Uncited documents2013295
Uncited documents2014298
Uncited documents2015193
Uncited documents2016169
Uncited documents2017163
Uncited documents2018193
Uncited documents2019219
Uncited documents2020225
Uncited documents2021246
Uncited documents2022245
Uncited documents2023262
Cited documents1999339
Cited documents2000358
Cited documents2001387
Cited documents2002443
Cited documents2003447
Cited documents2004489
Cited documents2005455
Cited documents2006505
Cited documents2007544
Cited documents2008633
Cited documents2009727
Cited documents2010893
Cited documents20111192
Cited documents20121407
Cited documents20131606
Cited documents20141478
Cited documents20151329
Cited documents20161115
Cited documents20171009
Cited documents20181018
Cited documents20191031
Cited documents20201146
Cited documents20211151
Cited documents20221153
Cited documents20231035

Evolution of the percentage of female authors.

YearFemale Percent
199935.94
200035.61
200138.52
200238.32
200339.74
200437.38
200541.15
200641.64
200744.88
200845.20
200945.37
201048.25
201147.25
201250.20
201349.98
201448.29
201549.56
201651.24
201752.10
201849.76
201948.82
202053.54
202153.28
202256.36
202352.63

Evolution of the number of documents cited by public policy documents according to Overton database.

DocumentsYearValue
Overton19996
Overton20000
Overton20010
Overton20020
Overton20030
Overton20047
Overton200539
Overton200652
Overton200747
Overton200866
Overton2009107
Overton2010149
Overton2011123
Overton2012136
Overton2013120
Overton201486
Overton201594
Overton201677
Overton201789
Overton201868
Overton201963
Overton202037
Overton202125
Overton202210
Overton20232

Evoution of the number of documents related to Sustainable Development Goals defined by United Nations. Available from 2018 onwards.

DocumentsYearValue
SDG2018454
SDG2019453
SDG2020450
SDG2021453
SDG2022362
SDG2023327

Scimago Journal & Country Rank

Leave a comment

Name * Required

Email (will not be published) * Required

* Required Cancel

The users of Scimago Journal & Country Rank have the possibility to dialogue through comments linked to a specific journal. The purpose is to have a forum in which general doubts about the processes of publication in the journal, experiences and other issues derived from the publication of papers are resolved. For topics on particular articles, maintain the dialogue through the usual channels with your editor.

Scimago Lab

Follow us on @ScimagoJR Scimago Lab , Copyright 2007-2024. Data Source: Scopus®

breast cancer research and treatment impact factor 2023

Cookie settings

Cookie Policy

Legal Notice

Privacy Policy

breast cancer research and treatment impact factor 2023

Search
2024 January 18
Purpose This study aimed to report the results from an early-phase study of rivoceranib, an oral tyrosine kinase inhibitor highly selective for vascular endothelial growth factor receptor 2, in patients with advanced solid tumors. Materials and Methods In this open-label, singl...
ORIGINAL ARTICLE 2024 January 8
Purpose There have been needs to improve the sensitivity of liquid biopsy. This report aims to report the analytical and clinical validation of a next-generation sequencing (NGS)–based circulating tumor DNA (ctDNA) assay. Materials and Methods Analytical validation was conduct...
ORIGINAL ARTICLE 2024 January 30
and Exon 20 Insertion Mutations in Non–Small Cell Lung Cancer: Single-Institution Experience
Purpose Exon 20 insertion mutations (E20ins) in epidermal growth factor receptor (EGFR) or human epidermal growth factor receptor 2 (HER2) in non–small cell lung cancer (NSCLC) patients has become more important with emergence of novel agents targeting E20ins. Materials and Me...
Editorial
                           
Review Article
Special Article
Original Articles
                                  
                                  
                                  
Case Report
Correspondence
-->
-->

Korean Cancer Association
Room 1824, Gwanghwamun Officia
92 Saemunan-ro, Jongno-gu, Seoul 03186, Korea
TEL: +82-2-3276-2410   FAX: +82-2-792-1410   E-mail:
 |   |   | 

Developed in M2PI

Submission guidelines

Our 3-step submission process, before you submit.

Now you’ve identified a journal to submit to, there are a few things you should be familiar with before you submit.

  • Make sure you are submitting to the most suitable journal - Aims and scope
  • Understand the costs and funding options - Fees and funding
  • Make sure your manuscript is accurate and readable - Language editing services
  • Understand the copyright agreement - Copyright

Ready to submit

To give your manuscript the best chance of publication, follow these policies and formatting guidelines.

  • Check the quality of your writing - Free Language check
  • General formatting rules for all article types - Preparing your manuscript
  • Make sure your submission is complete - Prepare supporting information
  • Copyright and license agreement - Conditions of publication
  • Read and agree to our Editorial Policies - Editorial policies

Submit and promote

After acceptance, we provide support so your article gains maximum impact in the scientific community and beyond.

Please note that manuscript can only be submitted by an author of the manuscript and may not be submitted by a third party. 

  • Who decides whether my work will be accepted? - Peer-review policy
  • Want to submit to a different journal? - Manuscript transfers
  • Spreading the word - Promoting your publication

Submit manuscript

  • Editorial Board
  • Manuscript editing services
  • Instructions for Editors
  • Sign up for article alerts and news from this journal
  • Collections
  • Follow us on Twitter

Annual Journal Metrics

Citation Impact 2023 Journal Impact Factor: 6.1 5-year Journal Impact Factor: 7.1 Source Normalized Impact per Paper (SNIP): 1.865 SCImago Journal Rank (SJR): 2.578 Speed 2023 Submission to first editorial decision (median days): 14 Submission to acceptance (median days): 129 Usage 2023 Downloads: 2,432,781 Altmetric mentions: 1,561

  • More about our metrics

Breast Cancer Research

ISSN: 1465-542X

Cancer Research and Treatment

Journal Abbreviation: CANCER RES TREAT Journal ISSN: 1598-2998

Year Impact Factor (IF) Total Articles Total Cites
2023 (2024 update) 4.1 - -
2022 - -
2021 5.036 - 4216
2020 4.679 119 3716
2019 3.761 159 2632
2018 3.363 140 2241
2017 3.230 119 1873
2016 3.772 159 1467
2015 4.245 115 993
2014 3.318 52 714
2013 2.975 46 584
2012 1.962 39 489
2011 - -
2010 - -

You may also be interested in the following journals

  • ► Anti-Cancer Drugs
  • ► Cancer Research
  • ► Journal of Clinical Oncology
  • ► Breast Cancer Research and Treatment
  • ► Cancer Biotherapy and Radiopharmaceuticals
  • ► Tumor Biology
  • ► Physical Review C
  • ► Mathematische Nachrichten
  • ► Molecular Biology and Evolution
  • ► Faseb Journal

Top Journals in other

  • Ca-A Cancer Journal For Clinicians
  • Chemical Reviews
  • Nature Materials
  • Reviews of Modern Physics
  • Nature Reviews Molecular Cell Biology
  • Annual Review of Astronomy and Astrophysics
  • Cancer Discovery
  • BMJ-British Medical Journal
  • Advanced Energy Materials
  • Advances in Optics and Photonics

Journal Impact

Picture

+1-866-353-3335

  • Custom Research
  • Research Partners
  • Enterprise Solution

PUBLISHER: TechSci Research | PRODUCT CODE:  1539527

Cover Image

North America Cancer Biopsy Market By Type (Liquid Biopsy, Tissue Biopsy), By Product Type (Instruments, Kits & Consumables, Others), By Application (Breast Cancer, Lung Cancer, Others), By Country, Competition, Forecast and Opportunities, 2019-2029F

Add to Cart

We offer 8 hour analyst time for an additional research. Please contact us for the details.

Description

Table of contents.

North America Cancer biopsy Market was valued at USD 10.71 Billion in 2023 and is anticipated to project impressive growth in the forecast period with a CAGR of 7.85% through 2029. The North America cancer biopsy market plays a vital role in the region's healthcare landscape, crucial for cancer diagnosis and treatment. Biopsies, involving tissue sample extraction for examination, are pivotal in identifying cancer presence, type, and extent, guiding personalized treatment plans for physicians. Recent years have seen significant market growth driven by several factors. Rising cancer incidence remains a major public health concern, with various types like breast, lung, colorectal, and prostate cancers steadily increasing. Early detection's importance fuels demand for precise diagnostic tools such as biopsies. Technological advancements, including liquid biopsy, offer less invasive alternatives, especially for challenging cancers. Advanced imaging and robotic-assisted biopsy techniques enhance accuracy and efficiency. Personalized medicine emphasis spurs molecular diagnostics adoption, tailoring treatment to tumor genetics, enhancing therapeutic precision and efficacy. Thus, biopsy samples serve not only as diagnostics but also critical genetic information sources, shaping targeted therapies and cancer care evolution.

Market Overview
Forecast Period2025-2029
Market Size 2023USD 10.71 Billion
Market Size 2029USD 16.78 Billion
CAGR 2024-20297.85%
Fastest Growing SegmentTissue biopsy
Largest MarketUnited States

Government initiatives and healthcare policies in North America have significantly influenced the cancer biopsy landscape. Regulatory frameworks and reimbursement policies facilitate the integration of advanced biopsy technologies into routine clinical practice. National cancer control programs prioritize preventive and early diagnostic measures, highlighting the importance of biopsy procedures in cancer care. The market is competitive, with key players driving innovation and expansion. Companies specializing in biopsy tools, imaging technologies, and laboratory services collaborate to enhance diagnostic capabilities. Strategic partnerships and mergers foster research and development, enabling the introduction of novel biopsy techniques.

Key Market Drivers

Rising cancer incidence drives demand for precise diagnostics

The North America cancer biopsy market is experiencing a significant surge in demand driven primarily by the escalating incidence of cancer across the region. With cancer rates on the rise, there is an urgent need for precise diagnostics to facilitate timely and accurate detection, thus enabling appropriate treatment strategies to be implemented. Biopsy procedures play a pivotal role in this scenario, serving as indispensable tools in confirming the presence of cancer, determining its specific type, and assessing the extent of its spread within the body. In the context of comprehensive cancer care, reliable diagnostic information is paramount. Biopsy techniques provide clinicians with crucial insights into the nature of the disease, guiding treatment decisions and ensuring that patients receive the most effective interventions tailored to their individual circumstances. From guiding surgical interventions to informing the selection of targeted therapies, biopsy results serve as the foundation upon which personalized treatment plans are built. The increasing demand for precise diagnostics underscores the critical importance of biopsy procedures within the cancer care continuum. As healthcare providers strive to meet the growing needs of patients, there is a corresponding emphasis on enhancing the accessibility, accuracy, and efficiency of biopsy techniques. This has led to ongoing advancements in biopsy technology, including the development of minimally invasive approaches such as liquid biopsies, which offer less discomfort and faster turnaround times compared to traditional tissue biopsies. The integration of advanced imaging modalities and molecular diagnostics has further expanded the diagnostic capabilities of biopsy procedures, enabling clinicians to obtain more detailed and comprehensive information about tumor characteristics and molecular profiles. As a result, the North America cancer biopsy market is poised for continued growth, driven by the imperative to meet the escalating demand for precise diagnostics in the fight against cancer.

Continuous technological advancements enhance biopsy accuracy

The North America cancer biopsy market is experiencing significant growth, largely driven by continuous technological advancements that have revolutionized cancer diagnostics. These innovations have ushered in a new era of precision and reliability in biopsy procedures, empowering healthcare professionals with more accurate and actionable insights into the nature of cancerous tumors. State-of-the-art imaging technologies, such as MRI-guided biopsies and robotic-assisted procedures, have emerged as cornerstones of modern biopsy techniques. These advanced imaging modalities allow for precise targeting of suspicious lesions, ensuring optimal tissue sample extraction with minimal invasiveness. As a result, clinicians can obtain high-quality biopsy specimens, thereby enhancing diagnostic accuracy and reducing the likelihood of sampling errors. The introduction of molecular diagnostics and liquid biopsy techniques has significantly expanded the diagnostic capabilities of cancer biopsies. Molecular profiling of tumor specimens enables oncologists to glean valuable information about specific genetic mutations and biomarkers associated with different cancer types. This deeper understanding of tumor biology not only facilitates more accurate cancer classification but also informs the selection of targeted therapies tailored to individual patients' molecular profiles. The integration of these advanced biopsy technologies into routine clinical practice has been transformative for cancer care. Early and accurate cancer diagnoses are now achievable, allowing for timely initiation of appropriate treatment strategies. The ability to tailor treatment plans based on molecular insights obtained from biopsy specimens has led to more personalized therapeutic interventions, resulting in improved patient outcomes and enhanced overall survival rates. The dynamic nature of the North America cancer biopsy market is sustained by a commitment to research and development. Industry leaders continue to invest in cutting-edge technologies aimed at further refining biopsy procedures and expanding diagnostic capabilities. As a result, the market remains at the forefront of delivering sophisticated and precise diagnostic solutions that are instrumental in the ongoing fight against cancer.

Key Market Challenges

Accuracy and Sensitivity

The Cancer Biopsy Market is confronted with the formidable challenge of upholding high levels of accuracy and sensitivity in diagnostic procedures. The precision and sensitivity of biopsy results are paramount in guiding oncologists toward making informed and effective treatment decisions for patients battling cancer. However, several factors pose hurdles to achieving consistent accuracy in biopsy outcomes. One significant challenge is the variability in sample quality obtained during biopsy procedures. Factors such as tissue preservation techniques, specimen handling, and processing methods can impact the integrity and quality of the tissue sample, potentially compromising the accuracy of diagnostic assessments. The inherent heterogeneity of tumors presents a formidable obstacle to achieving uniformity and reliability in biopsy results. Tumors may exhibit diverse cellular compositions and genetic mutations within different regions, leading to sampling errors and inaccuracies in diagnosis. Innovations in technology, including advanced imaging modalities and molecular diagnostics, hold promise for enhancing the sensitivity and accuracy of biopsy procedures. Advanced imaging techniques, such as MRI-guided biopsies and PET-CT scans, enable precise targeting of suspicious lesions, improving the likelihood of obtaining representative tissue samples. Likewise, molecular diagnostics offer insights into the genetic makeup of tumors, allowing for more refined and personalized treatment strategies based on individual tumor profiles. Despite these advancements, challenges persist in ensuring consistent accuracy and sensitivity in biopsy results. Addressing these challenges requires ongoing research and development efforts aimed at refining biopsy techniques, optimizing sample collection and processing protocols, and leveraging emerging technologies to overcome tumor heterogeneity and sampling errors. By surmounting these obstacles, the Cancer Biopsy Market can continue to provide reliable and actionable diagnostic information, ultimately contributing to improved patient outcomes in the dynamic landscape of cancer diagnostics.

Invasive Nature

The Cancer Biopsy Market faces a significant hurdle posed by the invasive nature of traditional biopsy procedures. These invasive techniques, while essential for obtaining tissue samples for diagnostic evaluation, present several drawbacks and challenges. One major concern is the discomfort and pain experienced by patients undergoing invasive biopsies, which can lead to increased anxiety and reluctance to undergo the procedure. The invasive biopsies carry the risk of complications such as bleeding, infection, and damage to surrounding tissues, particularly in sensitive or difficult-to-access anatomical locations. Another challenge associated with invasive biopsies is the potential for sampling errors, particularly in cases where obtaining adequate tissue specimens is challenging or where tumor heterogeneity complicates accurate diagnosis. These sampling errors can result in misdiagnosis or incomplete characterization of the tumor, leading to suboptimal treatment decisions and outcomes. In response to these challenges, there is growing interest in exploring non-invasive or minimally invasive biopsy alternatives in the Cancer Biopsy Market. These alternative approaches aim to reduce patient discomfort, minimize procedural risks, and improve the overall accessibility and acceptance of biopsy procedures in cancer diagnostics. Techniques such as liquid biopsies, which involve the analysis of circulating tumor cells or cell-free DNA in blood samples, offer a less invasive option for obtaining diagnostic information. Similarly, minimally invasive imaging-guided biopsy techniques, such as ultrasound-guided core needle biopsies or endoscopic biopsies, allow for targeted tissue sampling with reduced procedural invasiveness. Overcoming the invasiveness of traditional biopsy procedures remains a priority for advancing biopsy methodologies in the Cancer Biopsy Market. By developing and refining non-invasive and minimally invasive biopsy techniques, healthcare providers can improve patient experience, enhance diagnostic accuracy, and ultimately optimize treatment outcomes for individuals undergoing cancer diagnostics.

Key Market Trends

Liquid Biopsy Advancements

Bioprocessing automation is emerging as a transformative trend in the Cancer Biopsy Market, revolutionizing the way cell culture workflows are conducted. With automation, production processes are streamlined, leading to improved efficiency, reduced errors, and enhanced product quality. Advanced robotic systems, coupled with integrated sensors and data analytics, optimize every stage of cell culture workflows, from media preparation to cell harvesting. By minimizing human intervention and standardizing procedures, automation helps lower labor costs and ensures consistent results across batches. One of the significant advantages of bioprocessing automation is its ability to increase throughput and scalability. With automated systems, manufacturers can handle larger volumes of samples and meet the growing demand for cell-based products and therapies. This scalability is particularly crucial in the Cancer Biopsy Market, where the need for precise and timely diagnostic solutions continues to rise. The automation plays a pivotal role in improving reproducibility. By eliminating manual errors and variability, automated processes generate standardized results, enhancing the reliability of diagnostic tests and research outcomes. This consistency is essential for ensuring accurate diagnoses and treatment decisions in cancer care. As the Cancer Biopsy Market embraces Industry 4.0 principles, the integration of automation is poised to drive further growth and innovation. By leveraging data-driven insights and advanced technologies, automated systems accelerate research efforts, facilitate rapid prototyping of new diagnostic tools, and enable the development of cutting-edge therapies. Ultimately, bioprocessing automation is reshaping the future of cell culture technologies, paving the way for more efficient, reliable, and scalable solutions in cancer diagnostics and treatment.

Precision Oncology Integration

Precision oncology integration represents a significant advancement in the Cancer Biopsy Market, offering personalized treatment approaches tailored to individual patients' genetic makeup. This trend emphasizes the analysis of tumor tissue or liquid biopsy samples to identify specific genetic alterations, mutations, or biomarkers that drive cancer progression. By obtaining insights into the molecular characteristics of each patient's cancer, oncologists can make informed decisions regarding targeted therapies that precisely target the underlying drivers of tumor growth. The implementation of precision oncology aims to optimize treatment efficacy while minimizing the risk of adverse effects commonly associated with traditional chemotherapy and radiation therapy. By selecting therapies that specifically target the genetic vulnerabilities of the cancer cells, precision oncology holds the potential to improve patient outcomes and survival rates. Advancements in cancer biology and genomic technologies have been instrumental in facilitating the seamless integration of precision oncology into routine cancer biopsy practices. With increasingly sophisticated diagnostic tools and bioinformatics platforms, healthcare providers can efficiently analyze complex genomic data and identify actionable mutations or biomarkers that inform treatment decisions. As precision oncology continues to evolve, driven by ongoing research and technological innovation, it is expected to play an increasingly prominent role in guiding personalized cancer care. By harnessing the power of genomic information, precision oncology has the potential to revolutionize cancer treatment paradigms and improve outcomes for patients worldwide.

Segmental Insights

Type Insights

Based on the type, tissue biopsy remains a dominant method in the Cancer Biopsy Market, serving as a gold standard for diagnosing and characterizing various cancers. Tissue biopsy stands as a fundamental procedure in cancer diagnostics, involving the extraction of a minute sample of abnormal tissue for microscopic analysis. This approach offers invaluable insights into various aspects of cancer, including its type, stage, and molecular features. While innovative techniques like liquid biopsy have gained attention, tissue biopsy remains indispensable, particularly for solid tumours where obtaining tissue samples is essential for accurate diagnosis and characterization. The comprehensive data derived from tissue specimens play a pivotal role in guiding clinicians in devising personalized treatment strategies tailored to individual patients' unique cancer profiles. From selecting targeted therapies to assessing prognosis, tissue biopsy serves as a cornerstone in the holistic management of cancer patients, ensuring that they receive the most effective and appropriate care based on their specific disease characteristics and needs.

Product Type Insights

In the Cancer Biopsy Market, kits & consumables play a dominant role by providing essential tools for sample collection, preservation, and analysis. Biopsy kits play a pivotal role in modern cancer diagnostics, encompassing a range of essential components such as biopsy needles, containers, fixatives, and other consumables necessary for efficient tissue or liquid biopsy procedures. As biopsy techniques continue to advance, there is a corresponding increase in demand for specialized kits tailored to these evolving methodologies. These kits offer several advantages, including convenience, reliability, and compatibility, which are instrumental in ensuring the success of cancer biopsy procedures. Their dominance in the market underscores the growing emphasis on streamlined workflows and the critical importance of obtaining high-quality samples for precise molecular diagnostics and the development of personalized cancer treatment strategies. By providing clinicians with standardized tools and materials, biopsy kits facilitate consistent and accurate sample collection, thereby enhancing the reliability and effectiveness of biopsy-based diagnostic approaches in the realm of oncology.

Regional Insights

The United States holds a dominant position in the Cancer Biopsy Market, primarily due to its robust healthcare infrastructure, advanced diagnostic technologies, and high incidence of cancer cases. With a significant emphasis on healthcare research and development, coupled with substantial investments in oncology research, the U.S. remains at the forefront of cancer diagnostics and treatment globally. The country benefits from a proactive regulatory environment and strong industry collaborations, which foster continuous innovation in cancer biopsy techniques and technologies. The presence of leading biotech and pharmaceutical companies further contributes to the market's prominence, facilitating the development and commercialization of cutting-edge biopsy products and services. The well-established healthcare system in the U.S. ensures widespread adoption of biopsy techniques across various healthcare settings, including hospitals, clinics, and research institutions. As a result, the United States serves as a central hub for advancements in cancer biopsy practices, driving market growth and shaping the trajectory of cancer diagnostics on a global scale.

Key Market Players

Baxter International Inc

B. Braun Medical Inc.

Fresenius Medical Care North America

Medtronic, Inc

Asahi Kasei Homes North America, Inc.

Nipro Medical Corporation - North America Division

Becton Dickinson & Co

Nikkiso America, Inc.

Report Scope:

In this report, the North America Cancer Biopsy Market has been segmented into the following categories, in addition to the industry trends which have also been detailed below:

North America Cancer Biopsy Market, By Type:

North america cancer biopsy market, by product type:, north america cancer biopsy market, by application:, north america cancer biopsy market, by country:, competitive landscape.

Company Profiles: Detailed analysis of the major companies presents in the North America Cancer Biopsy Market.

Available Customizations:

North America Cancer Biopsy market report with the given market data, Tech Sci Research offers customizations according to a company's specific needs. The following customization options are available for the report:

Company Information

Detailed analysis and profiling of additional market players (up to five).

1. Product Overview

2. research methodology, 3. executive summary, 4. north america cancer biopsy market outlook, 5. united states cancer biopsy market outlook, 6. canada cancer biopsy market outlook, 7. mexico cancer biopsy market outlook, 8. market dynamics, 9. market trends & developments, 10. north america cancer biopsy market: swot analysis, 11. porter's five forces analysis, 12. competitive landscape, 12.10 angle plc, 13. strategic recommendations, 14. about us & disclaimer.

Picture

Jeroen Van Heghe

Manager - EMEA

+32-2-535-7543

Picture

Christine Sirois

Manager - Americas

+1-860-674-8796

  • Pharmaceuticals

breast cancer research and treatment impact factor 2023

traffic_analyzer/DigitalVision V

Evaluating the Breast Cancer Therapeutics Market

An analyst at citeline provided pharmanewsintelligence with insights into current trends, gaps, and predictions in the breast cancer therapeutics market..

Veronica Salib

  • Veronica Salib, Assistant Editor

As breast cancer awareness month continues, gathering a comprehensive understanding of the breast cancer therapeutics market is critical, including current trends and market players, gaps, and predictions for the coming year. Understanding the therapeutics market and market dynamics can help companies position themselves for prompt regulatory approval or successful market entry.

According to the World Health Organization (WHO), in 2020, there were approximately 2.3 million breast cancer diagnoses worldwide. Additionally, the global prevalence of breast cancer mortality was nearly 700,000.

The organization explained, "Breast cancer cells begin inside the milk ducts and/or the milk-producing lobules of the breast. The earliest form (in situ) is not life-threatening. Cancer cells can spread into nearby breast tissue (invasion). This creates tumors that cause lumps or thickening.”

Therapeutic interventions for breast cancer may include surgical tumor removal, radiation therapy, or medications, including hormonal therapies, chemotherapy, and targeted biologic therapies.

PharmaNewsIntelligence sat down with Millie Gray, Healthcare Analyst at Citeline, a Norstella company, to discuss the market for breast cancer therapies and treatments.

Breast Cancer Therapeutics Market

A recent market report published by Precedence Research, which analyzed from the base year 2022 to the forecast period of 2023–2032, revealed that in 2022 the breast cancer market size was estimated to be $28.8 billion (USD). Research projections anticipate the market will be valued at $73.68 billion (USD) by 2032 with a compound annual growth rate (CAGR) of 9.9%.

The report revealed that the largest breast cancer market is in North America (US, Canada, and Mexico), with Asia Pacific (Japan, China, Australia, India, South Korea, etc.) in second place. However, researchers also explored other regions, including Europe (Germany, France, United Kingdom, Italy, Spain, etc.), Latin America, the Middle East, and Africa (Brazil, South Africa, Saudi Arabia, etc.).

Analysts segmented the breast cancer market by distribution channel, cancer type, and therapy, evaluating trends in each segment.

According to the report, key players in the breast cancer market include Merck & Co., Bristol Myers Squibb, Kyowa Kirin, Eisai Co. Ltd, Sanofi, Pfizer Inc, AstraZeneca PLC, Novartis AG, Eli Lilly and Company, Genentech, Mylan Laboratories, Celltrion, Fresenius Kabi, Baxter Healthcare Corporation, Halozyme Inc., and GlaxoSmithKline.

In a 2021 Nature market research article , analysts divided breast cancer treatments depending on cancer subtypes, which included the following:

  • HER2-positive
  • HR-positive/HER2-negative
  • Triple-negative breast cancer

Key Market Focuses

PharmaNewsIntelligence asked Gray to provide insights into the key market focuses in breast cancer.

“Last year and the year before, the focus was more on the metastatic setting,” she revealed.

According to Gray, there have been significant advancements in metastatic breast cancer, especially triple-negative breast cancer (TNBC), over the past year.

“TNBC is a subtype of breast cancer, which, before these approvals, essentially was a hundred percent treated by chemotherapy. There was very little use of targeted therapy for TNBC. So that's a huge area of focus and development there,” explained Gray.

Considering the limited options, companies saw the potential for market growth and pursued treatments focused on this aggressive breast cancer subtype.

Early-Stage Focus

Beyond targeting TNBC, Gray noted increasing therapeutic interventions intended to treat breast cancer in its early stages.

“Pharma companies are now focusing on getting their drugs approved in the earlier stages of breast cancer,” she added. “Now, quite a few therapies are approved across all the subtypes of breast cancer. They're focusing on the early stage setting because it is the largest among all breast cancers. Approximately 90% of breast cancers get diagnosed at the early stages.”

Gray explains that targeting early-stage breast cancer allows pharmaceutical companies to reach the greatest portion of patients — meaning that they can make significant revenues even if they comprise a small market share .

Beyond the financial incentives to focus on early-stage breast cancers , Gray adds that early stages are the only stage where physicians could potentially cure the condition.

“In the metastatic setting, patients aren't treated with curative intent; they're treated with palliative attempts to manage the symptoms and potentially keep the patient alive longer,” she explained. “The development of better therapies, in the early-stage setting, is looking at being able to cure these patients or, at the very least, extend their life longer than the chemotherapies and endocrine therapies approved there before.”

Regulatory Approval and Competitive Landscape

In addition to describing therapeutic goals and target changes, Gray explored the competitive landscape and how increased competition and market saturation may impact regulatory approval. As more breast cancer therapies are researched, approved, and launched , market growth may disadvantage new researchers seeking regulatory approval. Companies and scientists are entering a saturated market.

“There can be issues with the approval of drugs. If there are a lot of therapies approved in one setting, there isn't a need for more,” noted Gray. “First of all, physicians probably aren't going to use it even if it does get improved.”

It is increasingly essential for companies to have an intelligent trial methodology to get approval. She explained that trials need a comparator arm showing clinically meaningful data that the experimental treatment is beneficial over the current standard of care. Researchers would submit trial data to the US Food and Drug Administration (FDA) in the US.

“There are already so many options; it's difficult for physicians to differentiate between them. If it's not addressing an unmet need — either by being better than what is currently approved or finding its niche, such as a strong benefit in a certain biomarker population — there's almost no point in getting it approved.”

Although there is approval potential for drugs that can be used down the line, Gray explains that the market for these therapies is slim.

“As patients progress down the lines, they will get sicker and sicker. Fewer patients are going to be treated. So the treatment rates in fourth or fifth lines will be minimal,” she explained.

Companies generally do not intend to be fourth- or fifth-line treatments because of the small market size and limited revenue options. Late-stage breast cancer patients are most commonly treated with generic chemotherapies.

Beyond that, Gray adds, “Payers will have less of an issue treating these patients with these chemotherapies. The issue is that these patients are incredibly sick and would rather be treated with these cheaper, already-established chemotherapies this far down the line. The majority of them will eventually stop being treated.”

Gaps In the Market

Despite a relatively saturated market in some areas, Gray explained that the currently approved drugs do not meet all needs. She explored existing gaps in the market, identifying areas where new companies or research can address unmet needs.

“Triple-negative breast cancer is the area of breast cancer that has the biggest gaps in the market right now,” emphasized Gray.

So far, Keytruda (Merck & Co.) , Lynparza (AstraZeneca and Merck), and Talzenna (Pfizer Oncology) have been approved for triple-negative breast cancer.

“They are all approved for specific biomarker populations. So, Keytruda is only approved for PD-1-positive patients. Talzenna and Lynparza are approved for BRCA-mutated patients. There isn't a targeted therapy in triple-negative breast cancer that has applicability across the population that isn't biomarker-limited, except famously toxic chemotherapies,” Gray added.

According to the American Society of Clinical Oncology (ASCO), , including fatigue, nausea, diarrhea, mouth sores, hair loss, and anemia. The Centers for Disease Control and Prevention (CDC) also describes emotional and neurological impacts, including depression, anxiety, stress, sleep disturbances, and cognitive issues.

Gray identifies this as a critical unmet need in the breast cancer drug market: finding breast cancer treatments beyond chemotherapy that can address a broad range of triple-negative breast cancer cases.

“In the same vein, efficacy in TNBC is an issue,” she continued.

Although she notes that Keytruda and Lynparaza have generally strong efficacy data, Gray also emphasizes that TNBC is a highly aggressive form of breast cancer.

“Patients still don't live in the metastatic setting of TNBC longer than a year, which isn't necessarily the case in the other two breast cancer subtypes.”

HR-Positive/HER2-Negative

Additionally, Gray told PharmaNewsIntelligence that another unmet need is pre-treated metastatic hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer, another highly aggressive form. Beyond developing new and efficacious therapeutics , researchers should work toward developing less painful treatments.

Currently, researchers are exploring oral selective estrogen receptor degraders (SERDs), a hormonal therapy similar to Fulvestrant, as an alternative to the standard of care.

However, Gray explains, “Fulvestrant is off-patent; it's cheap, and it's been a standard of care for over 10 years. So, physicians’ familiarity is solid there. Oral SERDs must have impressive trial results to replace that.”

HER2-Positive

One of the most significant unmet needs in HER2-positive breast cancer is a therapeutic intervention to address brain metastasis, a substantial issue in this subtype. An article in Nature revealed that nearly 20% of all breast cancers are HER2-positive breast cancer.

Gray revealed that Herceptin (trastuzumab), manufactured by Roche, has been the standard of care across nearly every HER2-positive breast cancer setting. The drug was the first targeted treatment for this subtype, with the approval in 1998. However, the US patent expired in 2019 , introducing pricing pressures on the market.

“There are now far cheaper biosimilars . Herceptin's market share has dropped rapidly, meaning that it is going to be far harder to get approval for a novel therapy in this market, given that patients and insurers are going to prefer using this cheaper biosimilar that's available,” noted Gray, when asked to provide an example of how patent expiration may impact market entry.

Market Predictions

PharmaNewsIntelligence asked Gray to provide insights into the three biggest trends coming to the breast cancer market. She touched on multiple areas of focus, including a focus on early-stage therapeutics, increased segmentation of patients, and slowed development.

She explained that five years ago, few breast cancer therapies targeted early treatment stages, especially in HR-positive and triple-negative breast cancer.

“It’s a very lucrative area with so many patients. So, pharma companies are increasingly targeting this area to maximize profit.”

Another trend she predicted is increased market segmentation as research advances. For example, Keytruda targets PD-1-positive patients, while capivasertib and Piqray target patients with PI3K mutations.

“There are increasing numbers of trials looking at smaller and smaller subsets of patients with certain biomarkers. For example, in non-small cell lung cancer, [treatment] is heavily divided by the types of mutations a patient will have. That is going to happen in breast cancer,” predicted Gray. “All these different biomarkers are going to emerge, and it's going to be a far more segmented indication than it is now.”

Companies may do additional market research to understand segments with unmet needs and how new products could fit into the global market.

Finally, Gray emphasized that the saturated market will prompt slower development as companies navigate regulatory approval and product differentiation challenges.

“Particularly for HER2-positive and HR-positive, development will slow down over the next 10 years. They are relatively crowded at the moment. It will become increasingly difficult to show a strong efficacy benefit over what's currently approved. And, as patents start to expire, it'll be more difficult to justify the pricing of a novel therapeutic, which generally will be high,” she concluded.

Editor's Note: This article has been edited to correctly reference capivasertib.

  • Applying Artificial Intelligence, Machine Learning in Cancer Care
  • Nectin Therapeutics Collaborates with Merck on Cancer Treatment
  • From COVID to Cancer, Understanding the Applications of mRNA Vaccines

Related Resources

  • Pharmaceutical Service Provider Formulates Tomorrow's Medications –NetApp
  • Roche: Increasing Value For A Complex Data Platform –DataVid
  • Why insider threats in healthcare remain prevalent although security culture is... –XtelligentMedia Healthcare
  • How Pharmacy Benefit Managers and Payers Collaborate to Reduce Costs –XtelligentMedia Healthcare

Dig Deeper on Pharmaceuticals

breast cancer research and treatment impact factor 2023

AI Tool May Help Breast Cancer Patients Avoid Unnecessary Chemotherapy

ShaniaKennedy

FDA Approves Truqap, Faslodex for HR-Positive Breast Cancer

VeronicaSalib

The Impact of Biotechnology Breakthroughs in Healthcare

breast cancer research and treatment impact factor 2023

Understanding Implications of Pharmacogenomic on Prescribing

Related q&a from veronica salib, could roche’s malaria test help address the national blood shortage.

Strategies for ending the national blood shortage may include expanding the donor pool, rationing, and diagnostic tests.   Continue Reading

Effectively incorporating AI into drug research, development

A recent report from Accenture analyzes the value and impact AI can have on drug research and development.   Continue Reading

Accurately diagnosing T1D, identifying biomarkers

The ACCESS-T1D study works to improve T1D diagnoses among at-risk groups and identify biomarkers for more straightforward disease diagnosis.   Continue Reading

A national survey revealed that while many Americans are comfortable with artificial intelligence in healthcare, some have ...

AI revealed insights into the relationship between lung cancer therapy and heart complications, which could help minimize cardiac...

Houston Methodist has successfully piloted ambient clinical intelligence and AI tools to bolster efficiency and cost savings ...

Understanding the quality bonus payment program is critical to appreciating Medicare Advantage and discussing the calls for ...

Only 45% of individuals who received a surprise bill challenged it and only 43% of those who were denied coverage appealed the ...

Medicare includes four different segments that each insure a wide variety of services and supplies for enrollees.

Ambient AI technology is helping Ochsner Health improve patient-provider relationships by allowing clinicians to spend ...

Epic's goal to bring customers live on TEFCA by the end of 2025 complements Carequality's plan to align with TEFCA as the ...

Key considerations for selecting an EHR vendor include assessing practice needs, conducting a thorough market scan and evaluating...

NIST encouraged organizations to implement its three post-quantum cryptography standards to prepare for the emergence of powerful...

A healthcare data breach at Alabama Cardiovascular Group affected upwards of 280,000 individuals after an unauthorized party ...

Lawsuits often follow a healthcare data breach, but understanding what drives litigation trends can help healthcare organizations...

Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

cancers-logo

Article Menu

breast cancer research and treatment impact factor 2023

  • Subscribe SciFeed
  • Recommended Articles
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

The effects of radiotherapy on the sequence and eligibility of breast reconstruction: current evidence and controversy.

breast cancer research and treatment impact factor 2023

Simple Summary

1. introduction, 2. breast reconstruction techniques, 2.1. overview of immediate reconstruction options, 2.2. overview of delayed reconstruction options, 3. influence of post-mastectomy radiotherapy on choice of reconstruction, 4. complications.

AuthorStudy DesignPatients (n)Reconstruction
Modality
Complication Rates
Tran et al. [ ].Retrospective41Immediate autologousFat necrosis (34%) and asymmetry (78%)
Billig et al. [ ].Prospective108Immediate autologousFat necrosis (16.9%) and hematoma (5.6%)
Billig et al. [ ].Prospective67Delayed autologousFat necrosis (19.5%) and wound dehiscence (9%)
Albino et al. [ ].Retrospective76Immediate autologousFat necrosis (19%) and contracture or scarring (30%)
Dewael et al. [ ].Retrospective20Immediate autologousFat necrosis (60%), contracture (60%), and infection (20%)
Dewael et al. [ ].Retrospective40Delayed autologousFat necrosis (12%) and wound dehiscence (10%)
Jhaveri et al. [ ].Retrospective69Immediate autologousGrade 2–4 complications (55%)
Jhaveri et al. [ ].Retrospective23Delayed implantGrade 2–4 complications (8%)
Maalouf et al. [ ].Retrospective30Immediate autologousReoperation (40%)
Maalouf et al. [ ].Retrospective32Delayed autologousReoperation (12%)
McCarthy et al. [ ].Prospective10Delayed implantCapsular contracture (60%)
Cordeiro et al. [ ].Retrospective68Delayed implantCapsular contracture (68%)
Rella et al. [ ].Retrospective80Delayed implantCapsular contracture (15%) and seroma (10%)
Benediktsson et al. [ ].Prospective24Immediate implantCapsular contracture (41%)

5. Influence of Post-Mastectomy Radiotherapy on Survival Outcomes

Impact of nodal and molecular subtypes on locoregional recurrence (lrr) and survival, 6. influence of post-mastectomy radiotherapy on cosmetic outcomes and patient satisfaction, 7. future directions and premrt, 8. conclusions, author contributions, conflicts of interest.

  • Veronesi, P.; Ballardini, B.; De Lorenzi, F.; Magnoni, F.; Lissidini, G.; Caldarella, P.; Galimberti, V. Immediate breast reconstruction after mastectomy. Breast 2011 , 20 (Suppl. S3), S104–S107. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Galimberti, V.; Vicini, E.; Corso, G.; Morigi, C.; Fontana, S.; Sacchini, V.; Veronesi, P. Nipple-sparing and skin-sparing mastectomy: Review of aims, oncological safety and contraindications. Breast 2017 , 34 (Suppl. S1), S82–S84. [ Google Scholar ] [ CrossRef ]
  • Scardina, L.; Di Leone, A.; Biondi, E.; Carnassale, B.; Sanchez, A.M.; D’Archi, S.; Franco, A.; Moschella, F.; Magno, S.; Terribile, D.; et al. Prepectoral vs. Submuscular Immediate Breast Reconstruction in Patients Undergoing Mastectomy after Neoadjuvant Chemotherapy: Our Early Experience. J. Pers. Med. 2022 , 12 , 1533. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Du, F.; Liu, R.; Zhang, H.; Xiao, Y.; Long, X. Post-mastectomy adjuvant radiotherapy for direct-to-implant and two-stage implant-based breast reconstruction: A meta-analysis. J. Plast. Reconstr. Aesthet. Surg. 2022 , 75 , 3030–3040. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Schmauss, D.; Machens, H.G.; Harder, Y. Breast Reconstruction after Mastectomy. Front. Surg. 2015 , 2 , 71. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Teymouri, H.; Stergioula, S.; Eder, M.; Kovacs, L.; Biemer, E.; Papadopulos, N. Breast reconstruction with autologous tissue following mastectomy. Hippokratia 2006 , 10 , 153–162. [ Google Scholar ]
  • Rusby, J.E.; Waters, R.A.; Nightingale, P.G.; England, D.W. Immediate breast reconstruction after mastectomy: What are the long-term prospects? Ann. R. Coll. Surg. Engl. 2010 , 92 , 193–197. [ Google Scholar ] [ CrossRef ]
  • Alderman, A.K.; Wilkins, E.G.; Kim, H.M.; Lowery, J.C. Complications in postmastectomy breast reconstruction: Two-year results of the Michigan Breast Reconstruction Outcome Study. Plast. Reconstr. Surg. 2002 , 109 , 2265–2274. [ Google Scholar ] [ CrossRef ]
  • Von Glinski, M.; Holler, N.; Kummel, S.; Reinisch, M.; Wallner, C.; Wagner, J.M.; Dadras, M.; Sogorski, A.; Lehnhardt, M.; Behr, B. Autologous vs. implant-based breast reconstruction after skin- and nipple-sparing mastectomy-A deeper insight considering surgical and patient-reported outcomes. Front. Surg. 2022 , 9 , 903734. [ Google Scholar ] [ CrossRef ]
  • Kronowitz, S.J.; Hunt, K.K.; Kuerer, H.M.; Babiera, G.; McNeese, M.D.; Buchholz, T.A.; Strom, E.A.; Robb, G.L. Delayed-immediate breast reconstruction. Plast. Reconstr. Surg. 2004 , 113 , 1617–1628. [ Google Scholar ] [ CrossRef ]
  • Terao, Y.; Taniguchi, K.; Fujii, M.; Moriyama, S. Postmastectomy radiation therapy and breast reconstruction with autologous tissue. Breast Cancer 2017 , 24 , 505–510. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Shumway, D.A.; Momoh, A.O.; Sabel, M.S.; Jagsi, R. Integration of Breast Reconstruction and Postmastectomy Radiotherapy. J. Clin. Oncol. 2020 , 38 , 2329–2340. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Cordeiro, P.G.; Albornoz, C.R.; McCormick, B.; Hudis, C.A.; Hu, Q.; Heerdt, A.; Matros, E. What Is the Optimum Timing of Postmastectomy Radiotherapy in Two-Stage Prosthetic Reconstruction: Radiation to the Tissue Expander or Permanent Implant? Plast. Reconstr. Surg. 2015 , 135 , 1509–1517. [ Google Scholar ] [ CrossRef ]
  • Baschnagel, A.M.; Shah, C.; Wilkinson, J.B.; Dekhne, N.; Arthur, D.W.; Vicini, F.A. Failure rate and cosmesis of immediate tissue expander/implant breast reconstruction after postmastectomy irradiation. Clin. Breast Cancer 2012 , 12 , 428–432. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Motwani, S.B.; Strom, E.A.; Schechter, N.R.; Butler, C.E.; Lee, G.K.; Langstein, H.N.; Kronowitz, S.J.; Meric-Bernstam, F.; Ibrahim, N.K.; Buchholz, T.A. The impact of immediate breast reconstruction on the technical delivery of postmastectomy radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2006 , 66 , 76–82. [ Google Scholar ] [ CrossRef ]
  • Heine, N.; Eigenberger, A.; Brebant, V.; Kempa, S.; Seitz, S.; Prantl, L.; Kuehlmann, B. The effect of radiotherapy on fat engraftment for complete breast reconstruction using lipofilling only. Arch Gynecol. Obstet. 2023 , 307 , 549–555. [ Google Scholar ] [ CrossRef ]
  • Debald, M.; Pech, T.; Kaiser, C.; Keyver-Paik, M.D.; Walgenbach-Bruenagel, G.; Kalff, J.C.; Kuhn, W.; Walgenbach, K.J. Lipofilling effects after breast cancer surgery in post-radiation patients: An analysis of results and algorithm proposal. Eur. J. Plast. Surg. 2017 , 40 , 447–454. [ Google Scholar ] [ CrossRef ]
  • Tran, N.V.; Evans, G.R.; Kroll, S.S.; Baldwin, B.J.; Miller, M.J.; Reece, G.P.; Robb, G.L. Postoperative adjuvant irradiation: Effects on tranverse rectus abdominis muscle flap breast reconstruction. Plast. Reconstr. Surg. 2000 , 106 , 313–317; discussion 318–320. [ Google Scholar ] [ CrossRef ]
  • Billig, J.; Jagsi, R.; Qi, J.; Hamill, J.B.; Kim, H.M.; Pusic, A.L.; Buchel, E.; Wilkins, E.G.; Momoh, A.O. Should Immediate Autologous Breast Reconstruction Be Considered in Women Who Require Postmastectomy Radiation Therapy? A Prospective Analysis of Outcomes. Plast. Reconstr. Surg. 2017 , 139 , 1279–1288. [ Google Scholar ] [ CrossRef ]
  • Albino, F.P.; Koltz, P.F.; Ling, M.N.; Langstein, H.N. Irradiated autologous breast reconstructions: Effects of patient factors and treatment variables. Plast. Reconstr. Surg. 2010 , 126 , 12–16. [ Google Scholar ] [ CrossRef ]
  • Dewael, S.; Vandevoort, M.; Fabre, G.; Nanhekhan, L. Immediate versus delayed autologous breast reconstruction: A retrospective matched cohort study of irradiated patients. J. Plast. Reconstr. Aesthet. Surg. 2019 , 72 , 1769–1775. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Jhaveri, J.D.; Rush, S.C.; Kostroff, K.; Derisi, D.; Farber, L.A.; Maurer, V.E.; Bosworth, J.L. Clinical outcomes of postmastectomy radiation therapy after immediate breast reconstruction. Int. J. Radiat. Oncol. Biol. Phys. 2008 , 72 , 859–865. [ Google Scholar ] [ CrossRef ]
  • Maalouf, C.; Bou-Merhi, J.; Karam, E.; Patocskai, E.; Danino, A.M. The impact of autologous breast reconstruction using DIEP flap on the oncologic efficacy of radiation therapy. Ann. Chir. Plast. Esthet. 2017 , 62 , 630–636. [ Google Scholar ] [ CrossRef ]
  • McCarthy, C.M.; Pusic, A.L.; Disa, J.J.; McCormick, B.L.; Montgomery, L.L.; Cordeiro, P.G. Unilateral postoperative chest wall radiotherapy in bilateral tissue expander/implant reconstruction patients: A prospective outcomes analysis. Plast. Reconstr. Surg. 2005 , 116 , 1642–1647. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Cordeiro, P.G.; Pusic, A.L.; Disa, J.J.; McCormick, B.; VanZee, K. Irradiation after immediate tissue expander/implant breast reconstruction: Outcomes, complications, aesthetic results, and satisfaction among 156 patients. Plast. Reconstr. Surg. 2004 , 113 , 877–881. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Rella, L.; Telegrafo, M.; Nardone, A.; Milella, A.; Ianora, A.A.S.; Lioce, M.; Angelelli, G.; Moschetta, M. MRI evaluation of post-mastectomy irradiated breast implants: Prevalence and analysis of complications. Clin. Radiol. 2015 , 70 , 948–953. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Benediktsson, K.; Perbeck, L. Capsular contracture around saline-filled and textured subcutaneously-placed implants in irradiated and non-irradiated breast cancer patients: Five years of monitoring of a prospective trial. J. Plast. Reconstr. Aesthet. Surg. 2006 , 59 , 27–34. [ Google Scholar ] [ CrossRef ]
  • Ogita, M.; Nagura, N.; Kawamori, J.; In, R.; Yoshida, A.; Yamauchi, H.; Takei, J.; Hayashi, N.; Iwahira, Y.; Ohde, S.; et al. Risk factors for complications among breast cancer patients treated with post-mastectomy radiotherapy and immediate tissue-expander/permanent implant reconstruction: A retrospective cohort study. Breast Cancer 2018 , 25 , 167–175. [ Google Scholar ] [ CrossRef ]
  • Fan, S.; Chen, H.; Grant, A.; DeLyzer, T. Outcomes of Immediate Alloplastic Breast Reconstruction in Patients Receiving Post-Mastectomy Radiotherapy. Plast. Surg. 2022 , 30 , 136–143. [ Google Scholar ] [ CrossRef ]
  • Wu Young, M.Y.; Garza, R.M.; Chang, D.W. Immediate versus delayed autologous breast reconstruction in patients undergoing post-mastectomy radiation therapy: A paradigm shift. J. Surg. Oncol. 2022 , 126 , 949–955. [ Google Scholar ] [ CrossRef ]
  • Matar, D.Y.; Wu, M.; Haug, V.; Orgill, D.P.; Panayi, A.C. Surgical complications in immediate and delayed breast reconstruction: A systematic review and meta-analysis. J. Plast. Reconstr. Aesthet. Surg. 2022 , 75 , 4085–4095. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Hershenhouse, K.S.; Bick, K.; Shauly, O.; Kondra, K.; Ye, J.; Gould, D.J.; Patel, K.M. Systematic review and meta-analysis of immediate versus delayed autologous breast reconstruction in the setting of post-mastectomy adjuvant radiation therapy. J. Plast. Reconstr. Aesthet. Surg. 2021 , 74 , 931–944. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • O’Donnell, J.P.M.; Murphy, D.; Ryan, E.J.; Gasior, S.A.; Sugrue, R.; O’Neill, B.L.; Boland, M.R.; Lowery, A.J.; Kerin, M.J.; McInerney, N.M. Optimal reconstructive strategies in the setting of post-mastectomy radiotherapy—A systematic review and network meta-analysis. Eur. J. Surg. Oncol. 2021 , 47 , 2797–2806. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Magill, L.J.; Robertson, F.P.; Jell, G.; Mosahebi, A.; Keshtgar, M. Determining the outcomes of post-mastectomy radiation therapy delivered to the definitive implant in patients undergoing one- and two-stage implant-based breast reconstruction: A systematic review and meta-analysis. J. Plast. Reconstr. Aesthet. Surg. 2017 , 70 , 1329–1335. [ Google Scholar ] [ CrossRef ]
  • Overgaard, M.; Hansen, P.S.; Overgaard, J.; Rose, C.; Andersson, M.; Bach, F.; Kjaer, M.; Gadeberg, C.C.; Mouridsen, H.T.; Jensen, M.B.; et al. Postoperative radiotherapy in high-risk premenopausal women with breast cancer who receive adjuvant chemotherapy. Danish Breast Cancer Cooperative Group 82b Trial. N. Engl. J. Med. 1997 , 337 , 949–955. [ Google Scholar ] [ CrossRef ]
  • Overgaard, M.; Jensen, M.B.; Overgaard, J.; Hansen, P.S.; Rose, C.; Andersson, M.; Kamby, C.; Kjaer, M.; Gadeberg, C.C.; Rasmussen, B.B.; et al. Postoperative radiotherapy in high-risk postmenopausal breast-cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet 1999 , 353 , 1641–1648. [ Google Scholar ] [ CrossRef ]
  • Kayali, M.; Abi Jaoude, J.; Mohammed, M.; Khabsa, J.; Tfayli, A.; Poortmans, P.; Zeidan, Y.H. Post-mastectomy Radiation Therapy in Triple-Negative Breast Cancer Patients: Analysis of the BEATRICE Trial. Ann. Surg. Oncol. 2022 , 29 , 460–466. [ Google Scholar ] [ CrossRef ]
  • Abi Jaoude, J.; de Azambuja, E.; Makki, M.; Tamim, H.; Tfayli, A.; Geara, F.; Piccart, M.; Poortmans, P.; Zeidan, Y.H. Post-Mastectomy Radiation Therapy in Human Epidermal Growth Factor Receptor 2 Positive Breast Cancer Patients: Analysis of the HERA Trial. Int. J. Radiat. Oncol. Biol. Phys. 2020 , 106 , 503–510. [ Google Scholar ] [ CrossRef ]
  • Libson, S.; Perez, E.; Takita, C.; Avisar, E. Post Mastectomy Radiation for Stage II Breast Cancer Patients with T1/T2 Lesions. Eur. J. Breast Health 2019 , 15 , 71–75. [ Google Scholar ] [ CrossRef ]
  • Zhang, J.Q.; Lu, C.Y.; Qin, L.; Chen, H.M.; Wu, S.Y. Outcome of post-mastectomy radiotherapy after primary systemic treatment in patients with different clinical tumor and nodal stages of breast cancer: A cohort study. Am. J. Cancer Res. 2020 , 10 , 2185–2198. [ Google Scholar ]
  • Scheer, A.S.; Zih, F.S.; Maki, E.; Koch, C.A.; McCready, D.R. Post-mastectomy Radiation: Should Subtype Factor into the Decision? Ann. Surg. Oncol. 2016 , 23 , 2462–2470. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kyndi, M.; Sorensen, F.B.; Knudsen, H.; Overgaard, M.; Nielsen, H.M.; Overgaard, J.; Danish Breast Cancer Cooperative Group. Estrogen receptor, progesterone receptor, HER-2, and response to postmastectomy radiotherapy in high-risk breast cancer: The Danish Breast Cancer Cooperative Group. J. Clin. Oncol. 2008 , 26 , 1419–1426. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wu, S.P.; Tam, M.; Shaikh, F.; Lee, A.; Chun, J.; Schnabel, F.; Guth, A.; Adams, S.; Schreiber, D.; Oh, C.; et al. Post-mastectomy Radiation Therapy in Breast Cancer Patients with Nodal Micrometastases. Ann. Surg. Oncol. 2018 , 25 , 2620–2631. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pusic, A.L.; Klassen, A.F.; Scott, A.M.; Klok, J.A.; Cordeiro, P.G.; Cano, S.J. Development of a new patient-reported outcome measure for breast surgery: The BREAST-Q. Plast. Reconstr. Surg. 2009 , 124 , 345–353. [ Google Scholar ] [ CrossRef ]
  • Kuhlefelt, C.; Repo, J.P.; Jahkola, T.; Kauhanen, S.; Homsy, P. Immediate versus delayed breast reconstruction: Long-term follow-up on health-related quality of life and satisfaction with breasts. J. Plast. Reconstr. Aesthet. Surg. 2024 , 88 , 478–486. [ Google Scholar ] [ CrossRef ]
  • Pacaric, S.; Orkic, Z.; Babic, M.; Farcic, N.; Milostic-Srb, A.; Lovric, R.; Barac, I.; Miksic, S.; Vujanic, J.; Turk, T.; et al. Impact of Immediate and Delayed Breast Reconstruction on Quality of Life of Breast Cancer Patients. Int. J. Environ. Res. Public Health 2022 , 19 , 8546. [ Google Scholar ] [ CrossRef ]
  • Yun, J.H.; Diaz, R.; Orman, A.G. Breast Reconstruction and Radiation Therapy. Cancer Control 2018 , 25 , 1073274818795489. [ Google Scholar ] [ CrossRef ]
  • Kim, S.H.; Kim, J.M.; Park, S.H.; Lee, S.Y. Analysis of the effects of breast reconstruction in breast cancer patients receiving radiotherapy after mastectomy. Arch. Plast. Surg. 2012 , 39 , 222–226. [ Google Scholar ] [ CrossRef ]
  • Reinders, F.C.J.; Young-Afat, D.A.; Batenburg, M.C.T.; Bruekers, S.E.; van Amerongen, E.A.; Macare van Maurik, J.F.M.; Braakenburg, A.; Zonnevylle, E.; Hoefkens, M.; Teunis, T.; et al. Higher reconstruction failure and less patient-reported satisfaction after post mastectomy radiotherapy with immediate implant-based breast reconstruction compared to immediate autologous breast reconstruction. Breast Cancer 2020 , 27 , 435–444. [ Google Scholar ] [ CrossRef ]
  • Giunta, S.; Laidsaar-Powell, R.; Huang, L.; Hatcher, N.; Dhillon, H.; Muscat, D.M.; Carroll, S.; McNeil, C.; Burke, L.; Howson, P.; et al. Considering the type and timing of breast reconstruction after mastectomy: Qualitative insights into women’s decision-making. Eur. J. Oncol. Nurs. 2021 , 54 , 102024. [ Google Scholar ] [ CrossRef ]
  • Flitcroft, K.; Brennan, M.; Spillane, A. Making decisions about breast reconstruction: A systematic review of patient-reported factors influencing choice. Qual. Life Res. 2017 , 26 , 2287–2319. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Holland, F.; Archer, S.; Montague, J. Younger women’s experiences of deciding against delayed breast reconstruction post-mastectomy following breast cancer: An interpretative phenomenological analysis. J. Health Psychol. 2016 , 21 , 1688–1699. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Schaverien, M.V.; Singh, P.; Smith, B.D.; Qiao, W.; Akay, C.L.; Bloom, E.S.; Chavez-MacGregor, M.; Chu, C.K.; Clemens, M.W.; Colen, J.S.; et al. Premastectomy Radiotherapy and Immediate Breast Reconstruction: A Randomized Clinical Trial. JAMA Netw. Open 2024 , 7 , e245217. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Facer, B.D.; Wang, T.; Weed, C.; Pariser, A.; Cherian, M.; Johnson, K.C.; Quiroga, D.; Stover, D.; Obeng-Gyasi, S.; Agnese, D.; et al. Radiation Management for Breast Cancer after Neoadjuvant Therapy. Curr. Breast Cancer Rep. 2023 , 15 , 371–378. [ Google Scholar ] [ CrossRef ]
  • Sivina, E.; Blumberga, L.; Purkalne, G.; Irmejs, A. Pathological complete response to neoadjuvant chemotherapy in triple negative breast cancer—Single hospital experience. Hered. Cancer Clin. Pract. 2023 , 21 , 4. [ Google Scholar ] [ CrossRef ] [ PubMed ]

Click here to enlarge figure

 AdvantagesDisadvantages
Immediate AutologousLower chance of capsular contracture
Lower morbidity compared with implant
One surgery
Better QOL vs. delayed options
Often contraindicated due to patient comorbidities or anatomy
More invasive than implant
Longer recovery time
More expensive in the short term
Immediate ImplantOne surgery
Less complex
Shorter recovery time
More cost-effective in the short term
Better QOL vs. delayed options
Increased risk of complications, including capsular contracture, infection, and skin breakdown
Delayed AutologousLower complication and failure rates compared with immediate autologous technique
More invasive
More cost-effective in the long term
Lower overall QOL
Multiple surgeries
Less cost-effective in the short term
Longer time to achieve cosmetic results
Increased risk of capsular contracture
Delayed Expander/ImplantLower complication and failure rates compared with immediate implant
Option to revise implants after TE removal
Faster recovery time than delayed autologous options
Lower QOL
Multiple surgeries
Increased risk of capsular contracture compared with immediate implant
NCTPhaseNumber of Patients
Enrolled (Estimated *)
Primary EndpointCurrently
Enrolling
NCT0599355931314Evaluates 5-year survival rate in breast cancer patients receiving a mastectomy and neoadjuvant chemotherapy without PMRT compared to with PMRTNo
NCT05512286N/A80Evaluates patient-reported outcomes for pre-operative and post-mastectomy radiotherapy regarding patients with DIEP flapsNo
NCT0544014931106Evaluates 7-year survival receiving PMRT/whole breast irradiation versus those not receiving itYes
NCT052531703622Evaluates non-inferiority of complication rates in patients with breast reconstructions between hypofractionated vs. conventional fraction radiotherapyNo
NCT05045287257Evaluates failure rate of hypofractionated PMRT in patients with two-stage expander/implant reconstructionsYes
NCT04992650N/A50Evaluates breast skin blood supply after fat grafting in patients with PMRTBy invitation
NCT03523078N/A500Evaluates cosmetic/patient-reported outcomes and complications in patients with and without PMRTUnknown
NCT034149703897Evaluates non-inferiority of hypofractionated PMRT complication rates and reoccurrence rates in patients with stage IIa-IIIa breast cancerNo
NCT03319069360Evaluates efficacy/toxicities of hypofractionated vs. conventional PMRT in high-risk breast cancer patientsUnknown
NCT03072316N/A300Evaluates effects of PMRT on breast cancer patients who received DIEP reconstructionsUnknown
NCT02992574N/A1022Evaluates efficacy/reoccurrence in patients with early high-risk, but node-negative, breast cancer treated with PMRTYes
NCT026790402101Evaluates histological response of patients receiving neoadjuvant chemotherapy and radiation after a mastectomy and immediate reconstructionNo
NCT01925651N/A58Evaluated if bolus usage during PMRT increased the treatment time or decreased efficacyComplete
NCT01666899N/A10Evaluates effect of PMRT on skin and blood vessels after treatmentComplete
NCT014526723600Evaluates the necessity of chest wall irradiation alone vs. chest wall and supraclavicular fossa irradiation in PMRTUnknown
NCT01417286269Evaluates efficacy and toxicities of accelerated radiotherapy in post-mastectomy patientsComplete
NCT01292772N/A12Evaluates effects of PMRT on patients who had immediate breast reconstructionsComplete
NCT0096688833500Evaluates efficacy of PMRT in patients with stage II breast cancer compared to observations alone after a mastectomyUnknown
NCT000055883Not listedEvaluates different radiation regimens in patients with early-stage breast cancer after a mastectomyComplete
NCT05483712N/A20Evaluates efficacy of brass mesh bolus compared to the current standard of care in PMRT patientsYes
The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

Campbell, A.R.; Didier, A.J.; Sheikh, T.M.; Ansari, S.; Watkins, D.E.; Fahoury, A.M.; Nandwani, S.V.; Rashid, M. The Effects of Radiotherapy on the Sequence and Eligibility of Breast Reconstruction: Current Evidence and Controversy. Cancers 2024 , 16 , 2939. https://doi.org/10.3390/cancers16172939

Campbell AR, Didier AJ, Sheikh TM, Ansari S, Watkins DE, Fahoury AM, Nandwani SV, Rashid M. The Effects of Radiotherapy on the Sequence and Eligibility of Breast Reconstruction: Current Evidence and Controversy. Cancers . 2024; 16(17):2939. https://doi.org/10.3390/cancers16172939

Campbell, Andrew R., Alexander J. Didier, Taha M. Sheikh, Sami Ansari, Dean E. Watkins, Alan M. Fahoury, Swamroop V. Nandwani, and Mohammad Rashid. 2024. "The Effects of Radiotherapy on the Sequence and Eligibility of Breast Reconstruction: Current Evidence and Controversy" Cancers 16, no. 17: 2939. https://doi.org/10.3390/cancers16172939

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

  • Alzheimer's disease & dementia
  • Arthritis & Rheumatism
  • Attention deficit disorders
  • Autism spectrum disorders
  • Biomedical technology
  • Diseases, Conditions, Syndromes
  • Endocrinology & Metabolism
  • Gastroenterology
  • Gerontology & Geriatrics
  • Health informatics
  • Inflammatory disorders
  • Medical economics
  • Medical research
  • Medications
  • Neuroscience
  • Obstetrics & gynaecology
  • Oncology & Cancer
  • Ophthalmology
  • Overweight & Obesity
  • Parkinson's & Movement disorders
  • Psychology & Psychiatry
  • Radiology & Imaging
  • Sleep disorders
  • Sports medicine & Kinesiology
  • Vaccination
  • Breast cancer
  • Cardiovascular disease
  • Chronic obstructive pulmonary disease
  • Colon cancer
  • Coronary artery disease
  • Heart attack
  • Heart disease
  • High blood pressure
  • Kidney disease
  • Lung cancer
  • Multiple sclerosis
  • Myocardial infarction
  • Ovarian cancer
  • Post traumatic stress disorder
  • Rheumatoid arthritis
  • Schizophrenia
  • Skin cancer
  • Type 2 diabetes
  • Full List »

share this!

August 23, 2024

This article has been reviewed according to Science X's editorial process and policies . Editors have highlighted the following attributes while ensuring the content's credibility:

fact-checked

trusted source

Breast cancer survival improved in Gaza between 2017–2020, finds study

by King's College London

breast cancer

A recent study shines light on the treatment and survival outcomes for women with breast cancer in Gaza before the current conflict. While over half of reported diagnoses were made at advanced stages, a high short-term survival rate was discovered, highlighting the positive impact of international investment and collaboration in recent years before the conflict.

The paper, published in JCO Oncology and authored by Dr. Shaymaa Alwaheidi and Professors Richard Sullivan and Elizabeth Davies, looks at women in Gaza who were diagnosed with breast cancer between 2017 and 2018. It describes the careful follow up of women using paper medical records and interview data and the calculation of their survival until the end of 2020. This detailed analysis was needed due to the significant gaps in the available research on breast cancer outcomes in Gaza.

Results showed that women in Gaza with breast cancer had a 95.4% chance of survival after the first year, and 86.6% chance of survival after two years. The short-term survival rates (as of 2020) had improved from previous reports, which were lower than some of their neighboring countries in the Middle East including Lebanon and Jordan.

However, 52% of diagnoses were at the advanced stages III or IV. The authors suggest that this high proportion of advanced stage diagnoses could be due to a combination of women who delay seeking a diagnosis, and delays from the health care system , both of which need to be resolved by creating clearer referral guidelines.

The increase in short-term survival could be attributed to increased awareness of breast cancer symptoms among women during this period, leading to their earlier presentation, quicker diagnoses and better treatment. The authors also note that it could be due to changes to the treatment offered and the reduction of modified radical mastectomy.

These changes coincided with investment into the cancer care system, most notably the development of a multidisciplinary breast cancer team at Al-Shifa Hospital and collaborative efforts from the international community as shown by the support of visiting surgeons from the UK.

These positive results highlight how such investments and collaborations had significant benefits for women with breast cancer in Gaza, at least prior to the current conflict. The authors hope that it can encourage more investments and collaborations in breast cancer in Gaza, and more broadly encourage efforts in places throughout the world where women need greater medical support with breast cancer.

"Our results show how much had been achieved for Gazan women with breast cancer before the current conflict. Sadly, they now emphasize how the international community must find more robust ways of stepping up to protect vital cancer services during conflict ," says Davies.

Explore further

Feedback to editors

breast cancer research and treatment impact factor 2023

Self-deployable, biodegradable electrode offers minimally invasive brain signal monitoring

9 hours ago

breast cancer research and treatment impact factor 2023

Study identifies metabolic switch essential for generation of memory T cells and anti-tumor immunity

Aug 24, 2024

breast cancer research and treatment impact factor 2023

Multiple sclerosis appears to protect against Alzheimer's disease

Aug 23, 2024

breast cancer research and treatment impact factor 2023

Good sleep habits important for overweight adults, study suggests

breast cancer research and treatment impact factor 2023

Mediterranean diet supplement can affect epigenetics associated with healthy aging

breast cancer research and treatment impact factor 2023

New method for quantifying boredom in the body during temporary stress

breast cancer research and treatment impact factor 2023

Cancer researchers develop new method that uses internal clock inside tumor cells to optimize therapies

breast cancer research and treatment impact factor 2023

Strength training activates cellular waste disposal, interdisciplinary research reveals

breast cancer research and treatment impact factor 2023

Being a 'weekend warrior' could be as good for brain health as exercising throughout the week

breast cancer research and treatment impact factor 2023

Simple blood test for Alzheimer's disease could change how the disease is detected and diagnosed

Related stories.

breast cancer research and treatment impact factor 2023

American Indian/Alaska Native patients less likely to undergo breast reconstruction

Jul 2, 2024

breast cancer research and treatment impact factor 2023

High insulin levels contribute to worse outcomes for Black women with aggressive form of breast cancer, research finds

Jun 3, 2024

breast cancer research and treatment impact factor 2023

1975 to 2019 saw 58% decrease in breast cancer mortality

Jan 17, 2024

breast cancer research and treatment impact factor 2023

Women with benign breast disease face higher risk of breast cancer in the long term, according to large-scale study

Nov 17, 2022

breast cancer research and treatment impact factor 2023

Consumer Health: Are you due for a mammogram?

Oct 18, 2021

breast cancer research and treatment impact factor 2023

Motherhood after breast cancer doesn't lower survival chances

Jul 5, 2022

Recommended for you

breast cancer research and treatment impact factor 2023

AI platform enhances lung cancer diagnosis accuracy

breast cancer research and treatment impact factor 2023

A modified peptide shows promise for fighting tumors

Aug 22, 2024

breast cancer research and treatment impact factor 2023

Promising treatment for rectal cancer confirmed in major study

breast cancer research and treatment impact factor 2023

Study reveals RNA's role in regulating gene expression in cancer cells

breast cancer research and treatment impact factor 2023

New work validates targets for personalized cancer immunotherapy

Let us know if there is a problem with our content.

Use this form if you have come across a typo, inaccuracy or would like to send an edit request for the content on this page. For general inquiries, please use our contact form . For general feedback, use the public comments section below (please adhere to guidelines ).

Please select the most appropriate category to facilitate processing of your request

Thank you for taking time to provide your feedback to the editors.

Your feedback is important to us. However, we do not guarantee individual replies due to the high volume of messages.

E-mail the story

Your email address is used only to let the recipient know who sent the email. Neither your address nor the recipient's address will be used for any other purpose. The information you enter will appear in your e-mail message and is not retained by Medical Xpress in any form.

Newsletter sign up

Get weekly and/or daily updates delivered to your inbox. You can unsubscribe at any time and we'll never share your details to third parties.

More information Privacy policy

Donate and enjoy an ad-free experience

We keep our content available to everyone. Consider supporting Science X's mission by getting a premium account.

E-mail newsletter

breast cancer research and treatment impact factor 2023

Volume 201, Issue 1

No more disparities among regions in italy: recent approval of genomic test reimbursability for early breast cancer patients in the country.

  • Sara Bravaccini
  • Massimiliano Mazza
  • Roberta Maltoni

Identifying miRNA biomarkers for breast cancer and ovarian cancer: a text mining perspective

breast cancer research and treatment impact factor 2023

Advances of neuroimaging in chemotherapy related cognitive impairment (CRCI) of patients with breast cancer

  • Senbang Yao
  • Qianqian Zhang
  • Huaidong Cheng

breast cancer research and treatment impact factor 2023

Heterogeneous circulating tumor cells correlate with responses to neoadjuvant chemotherapy and prognosis in patients with locally advanced breast cancer

  • Jingyi Wang

breast cancer research and treatment impact factor 2023

Modeling the novel SERD elacestrant in cultured fulvestrant-refractory HR-positive breast circulating tumor cells

  • Taronish D. Dubash
  • Aditya Bardia
  • Shyamala Maheswaran

breast cancer research and treatment impact factor 2023

Conventional specimen radiography in breast-conserving therapy: a useful tool for intraoperative margin assessment after neoadjuvant therapy?

  • Benedikt Schäfgen
  • Annabelle Haller
  • Michael Golatta

breast cancer research and treatment impact factor 2023

Radiofrequency localization of nonpalpable breast cancer in a multicentre prospective cohort study: feasibility, clinical acceptability, and safety

  • Anke Christenhusz
  • Bianca M. den Dekker
  • Anneriet E. Dassen

breast cancer research and treatment impact factor 2023

Impact of non-adherence to endocrine therapy on recurrence risk in older women with stage I breast cancer after breast-conserving surgery

  • Danielle Rodin
  • Rinku Sutradhar
  • Eileen Rakovitch

breast cancer research and treatment impact factor 2023

Survival benefits associated with being adherent and having longer persistence to adjuvant hormone therapy across up to five years among U.S. Medicare population with breast cancer

  • Dandan Zheng
  • Joseph Thomas III

Treatment patterns and outcomes associated with sequential and non-sequential use of CDK4 & 6 inhibitors in patients with HR+, HER2− MBC in the real world

  • Megan Kruse
  • Emily Nash Smyth
  • Kevin Kalinsky

breast cancer research and treatment impact factor 2023

Development of cardiometabolic risk factors following endocrine therapy in women with breast cancer

  • Eileen Rillamas-Sun
  • Marilyn L. Kwan
  • Heather Greenlee

Surviving the COVID-19 pandemic: navigating the recovery of breast imaging services in a safety-net hospital

  • Jessica H. Porembka
  • Stephen J. Seiler
  • Basak E. Dogan

breast cancer research and treatment impact factor 2023

Validating the PROMIS cognitive function short form in cancer survivors

  • Ashley M. Henneghan
  • Kathleen Van Dyk
  • Patricia A. Ganz

Is there any association between declined creatinine clearence and higher endoxifen levels in older patients with non-metastatic breast cancer?

  • Kadri Altundag

It is the impact of racism, not race, that causes breast cancer inequities

  • Francois G. Rollin
  • Shub S. Agrawal
  • Mackenzie L. W. Garcia
  • Find a journal
  • Publish with us
  • Track your research
  • Introduction
  • Conclusions
  • Article Information

Tornado diagram shows results of one-way sensitivity analysis for key model inputs. The gray portion of each bar represents the incremental cost-effectiveness ratio (ICER) range when the specified model input is lower than its base-case value. The black portion of each bar represents the ICER range when the specified model input is higher than its base-case value. Low and high values for all parameters are presented in Table 1. The first vertical line reflects the base-case value of each input, where the ICER is $110 962 per quality-adjusted life-year (QALY) gained comparing adjuvant olaparib with no adjuvant olaparib. The second vertical line reflects the value of each input that would yield an ICER of $150 000 per QALY, holding all other inputs constant. Threshold values above or below which the ICER exceeded $150 000 per QALY were 0.694 for the hazard ratio for recurrence with olaparib; 0.048 for the discount rate; and 0.743 for the utility of no recurrence/no olaparib, holding all other parameters constant at their base-case values. EV indicates expected value; WTP, willingness to pay.

Incremental cost-effectiveness ratios (ICERs) from 10 000 simulations in probabilistic sensitivity analysis. Quality-adjusted life-year (QALY) threshold indicated by dashed line. In all simulations, adjuvant olaparib was both more effective and more costly than no adjuvant olaparib. The oval depicts a 95% confidence ellipse. WTP indicates willingness to pay.

Curves indicate the proportion of simulations (n = 10 000) in probabilistic sensitivity analysis for which each strategy is preferred, as a function of the threshold willingness to pay for an additional quality-adjusted life-year.

Data Sharing Statement

See More About

Sign up for emails based on your interests, select your interests.

Customize your JAMA Network experience by selecting one or more topics from the list below.

  • Academic Medicine
  • Acid Base, Electrolytes, Fluids
  • Allergy and Clinical Immunology
  • American Indian or Alaska Natives
  • Anesthesiology
  • Anticoagulation
  • Art and Images in Psychiatry
  • Artificial Intelligence
  • Assisted Reproduction
  • Bleeding and Transfusion
  • Caring for the Critically Ill Patient
  • Challenges in Clinical Electrocardiography
  • Climate and Health
  • Climate Change
  • Clinical Challenge
  • Clinical Decision Support
  • Clinical Implications of Basic Neuroscience
  • Clinical Pharmacy and Pharmacology
  • Complementary and Alternative Medicine
  • Consensus Statements
  • Coronavirus (COVID-19)
  • Critical Care Medicine
  • Cultural Competency
  • Dental Medicine
  • Dermatology
  • Diabetes and Endocrinology
  • Diagnostic Test Interpretation
  • Drug Development
  • Electronic Health Records
  • Emergency Medicine
  • End of Life, Hospice, Palliative Care
  • Environmental Health
  • Equity, Diversity, and Inclusion
  • Facial Plastic Surgery
  • Gastroenterology and Hepatology
  • Genetics and Genomics
  • Genomics and Precision Health
  • Global Health
  • Guide to Statistics and Methods
  • Hair Disorders
  • Health Care Delivery Models
  • Health Care Economics, Insurance, Payment
  • Health Care Quality
  • Health Care Reform
  • Health Care Safety
  • Health Care Workforce
  • Health Disparities
  • Health Inequities
  • Health Policy
  • Health Systems Science
  • History of Medicine
  • Hypertension
  • Images in Neurology
  • Implementation Science
  • Infectious Diseases
  • Innovations in Health Care Delivery
  • JAMA Infographic
  • Law and Medicine
  • Leading Change
  • Less is More
  • LGBTQIA Medicine
  • Lifestyle Behaviors
  • Medical Coding
  • Medical Devices and Equipment
  • Medical Education
  • Medical Education and Training
  • Medical Journals and Publishing
  • Mobile Health and Telemedicine
  • Narrative Medicine
  • Neuroscience and Psychiatry
  • Notable Notes
  • Nutrition, Obesity, Exercise
  • Obstetrics and Gynecology
  • Occupational Health
  • Ophthalmology
  • Orthopedics
  • Otolaryngology
  • Pain Medicine
  • Palliative Care
  • Pathology and Laboratory Medicine
  • Patient Care
  • Patient Information
  • Performance Improvement
  • Performance Measures
  • Perioperative Care and Consultation
  • Pharmacoeconomics
  • Pharmacoepidemiology
  • Pharmacogenetics
  • Pharmacy and Clinical Pharmacology
  • Physical Medicine and Rehabilitation
  • Physical Therapy
  • Physician Leadership
  • Population Health
  • Primary Care
  • Professional Well-being
  • Professionalism
  • Psychiatry and Behavioral Health
  • Public Health
  • Pulmonary Medicine
  • Regulatory Agencies
  • Reproductive Health
  • Research, Methods, Statistics
  • Resuscitation
  • Rheumatology
  • Risk Management
  • Scientific Discovery and the Future of Medicine
  • Shared Decision Making and Communication
  • Sleep Medicine
  • Sports Medicine
  • Stem Cell Transplantation
  • Substance Use and Addiction Medicine
  • Surgical Innovation
  • Surgical Pearls
  • Teachable Moment
  • Technology and Finance
  • The Art of JAMA
  • The Arts and Medicine
  • The Rational Clinical Examination
  • Tobacco and e-Cigarettes
  • Translational Medicine
  • Trauma and Injury
  • Treatment Adherence
  • Ultrasonography
  • Users' Guide to the Medical Literature
  • Vaccination
  • Venous Thromboembolism
  • Veterans Health
  • Women's Health
  • Workflow and Process
  • Wound Care, Infection, Healing

Get the latest research based on your areas of interest.

Others also liked.

  • Download PDF
  • X Facebook More LinkedIn

Zettler CM , De Silva DL , Blinder VS , Robson ME , Elkin EB. Cost-Effectiveness of Adjuvant Olaparib for Patients With Breast Cancer and Germline BRCA1/2 Mutations. JAMA Netw Open. 2024;7(1):e2350067. doi:10.1001/jamanetworkopen.2023.50067

Manage citations:

© 2024

  • Permissions

Cost-Effectiveness of Adjuvant Olaparib for Patients With Breast Cancer and Germline BRCA1/2 Mutations

  • 1 COTA Healthcare, New York, New York
  • 2 Peter MacCallum Cancer Centre, Parkville Familial Cancer Centre, Melbourne, Victoria, Australia
  • 3 Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
  • 4 Breast Medicine Service and Immigrant Health and Cancer Disparities Service, Memorial Sloan Kettering Cancer Center, New York, New York
  • 5 Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, New York
  • 6 Department of Health Policy and Management, Columbia University Mailman School of Public Health, New York, New York

Question   Is adjuvant olaparib cost-effective in patients with early-stage breast cancer with high-risk disease and germline BRCA1/2 mutations?

Findings   In an economic evaluation using a Markov model, adjuvant olaparib therapy was associated with a 1.25-year increase in life expectancy and a 1.20-quality-adjusted life-year (QALY) increase at an incremental cost of $133 133 compared with no olaparib, yielding a discounted incremental cost-effectiveness ratio of approximately $111 000 per QALY gained. Results were sensitive to assumptions about the effectiveness of olaparib and quality of life for patients with no disease recurrence.

Meaning   The findings of this study suggest that, at a willingness-to-pay threshold of $150 000 per QALY, adjuvant olaparib may be cost-effective at its 2021 price.

Importance   The OlympiA trial found that 1 year of adjuvant olaparib therapy can improve distant disease-free survival and overall survival from early-stage breast cancer in patients with a germline BRCA1/2 mutation. However, olaparib, an oral poly-adenosine diphosphate ribose polymerase inhibitor, is estimated to cost approximately $14 000 per month in the US.

Objective   To estimate the incremental cost-effectiveness of adjuvant olaparib compared with no olaparib in eligible patients.

Design, Setting, and Participants   In an economic evaluation from a health care system perspective, the cost-effectiveness of adjuvant olaparib was analyzed using a Markov state-transition model. The model simulated costs and lifetime health outcomes of 42-year-old women with high-risk early-stage breast cancer and a known BRCA1/2 mutation who completed definitive primary therapy and neoadjuvant or adjuvant systemic therapy. The study was conducted from August 2021 to July 2023. The effectiveness of olaparib was based on the findings of the OlympiA randomized clinical trial, and other model parameters were identified from the literature. The model was calibrated to the 1-, 2-, 3-, and 4-year distant disease-free and overall survival observed in the OlympiA trial, and olaparib was assumed to reduce the risk of distant recurrence only in the first 4 years.

Exposure   One year of adjuvant olaparib or no adjuvant olaparib.

Main Outcome and Measure   Incremental cost-effectiveness ratio (ICER) in 2021 US dollars per quality-adjusted life-year (QALY) gained. All outcomes were discounted by 3% annually.

Results   In the base case, adjuvant olaparib was associated with a 1.25-year increase in life expectancy and a 1.20-QALY increase at an incremental cost of $133 133 compared with no olaparib. The resulting ICER was approximately $111 000 per QALY gained. At a willingness-to-pay threshold of $150 000 per QALY, olaparib was cost-effective at its 2021 price and in more than 92% of simulations in probabilistic sensitivity analysis. The results were sensitive to assumptions about the effectiveness of olaparib and quality of life for patients with no disease recurrence.

Conclusions and Relevance   In this study, from a US health care system perspective, adjuvant olaparib was a cost-effective option for patients with high-risk, early-stage breast cancer and a germline BRCA1/2 mutation.

Olaparib, a poly-adenosine diphosphate ribose polymerase (PARP) inhibitor, was first approved by the US Food and Drug Administration in 2014 for the treatment of advanced ovarian cancer in patients with a deleterious or suspected deleterious germline BRCA1/2 mutation (gBRCAm). In 2018, it was approved in the US for ERBB2 (previously HER-2 )-negative metastatic breast cancer associated with a gBRCAm in patients who previously received chemotherapy. 1

In June 2021, interim results of the randomized, double-blind OlympiA clinical trial were published, demonstrating the efficacy of olaparib for treating breast cancer in the adjuvant setting. 2 Patients with pathogenic or likely pathogenic gBRCAm and nonmetastatic, ERBB2 -negative primary breast cancer were randomized to receive twice-daily oral olaparib or placebo for 1 year following completion of definitive local treatment and neoadjuvant or adjuvant chemotherapy. In March 2022, an updated interim analysis 3 with 1836 patients, 330 invasive-disease-free survival events, and a median of 3.5 years of follow-up reported that patients receiving olaparib had superior 4-year distant disease-free survival (DDFS) (86.5% vs 79.1% for placebo) and overall survival (OS) (89.8% vs 86.4% for placebo). The hazard ratio (HR) for distant disease or death at 4 years was 0.61 (95% CI, 0.48-0.77). Olaparib was subsequently approved for use in the adjuvant setting among patients similar to those in the OlympiA trial.

While approximately 3% of all breast cancers in the US are associated with a gBRCAm, pathogenic variants of these genes are prevalent in more than 10% of patients with triple-negative breast cancer (TNBC)—tumors that lack estrogen and progesterone receptors and do not overexpress ERBB2 protein. 4 - 6 Triple-negative breast cancer generally has more limited therapeutic options and poorer prognosis than other types of breast cancer. 7 , 8 In the OlympiA trial, more than 80% of patients in both arms had TNBC.

The results of the OlympiA trial indicate that adjuvant olaparib provides a clinically and statistically significant health benefit for patients with early-stage breast cancer and a known gBRCAm. Our goal was to assess the value of these improvements relative to olaparib’s cost.

In an economic evaluation conducted from the health care system perspective, we created a Markov model to estimate the lifetime costs, life-years, and quality-adjusted life-years (QALYs) associated with adjuvant olaparib or no olaparib for early-stage breast cancer with known gBRCAm. The model included 4 health states: no recurrence, metastatic recurrence, death due to breast cancer, and death from other causes. All patients started in the no recurrence health state after completion of definitive primary therapy and neoadjuvant or adjuvant chemotherapy. From no recurrence, patients could transition to metastatic recurrence or die from other causes. Patients could only die from breast cancer in the metastatic recurrence health state. Local recurrences and other primary cancers were not modeled. The model used 1-month cycles and simulated the DDFS and OS observed in the OlympiA trial for the first 48 months. We extrapolated beyond this period by applying monthly estimates of metastatic recurrence from studies of patients with breast cancer with similar tumor features. 9 , 10 The model was created in TreeAge Pro 2020 software (TreeAge Software LLC). The study was conducted from August 2021 to July 2023. The study was conducted using information from published reports in the peer-reviewed literature and did not involve new collection or analysis of data from humans; therefore, the study did not require institutional review board review or exemption according to the Common Rule (45 CFR §46). We followed the Consolidated Health Economic Evaluation Reporting Standards ( CHEERS ) reporting guideline.

Consistent with the OlympiA trial, 2 we assumed that patients received either oral olaparib, 300 mg, twice daily or no olaparib. The starting age was 42 years, which was the median age of patients in the olaparib arm of the trial. We also examined starting ages of 50 and 60 years.

The probability of metastatic recurrence without olaparib was derived from the DDFS results observed in the placebo arm of the OlympiA study. We estimated the annual rate of metastatic recurrence in each of the first 4 years and translated these year-specific rates into constant monthly probabilities. 11 We modeled the association between olaparib and risk of metastatic recurrence in the first 4 years using the stratified HR for distant disease or death observed in the trial (HR, 0.61; 95% CI, 0.48-0.77). We assumed that olaparib did not reduce the risk of metastatic recurrence after 4 years. Consistent with the literature 9 , 10 on recurrence risks for patients with TNBC, we assumed that the risk of metastatic recurrence decreased after the first 4 years and remained constant after 10 years.

Monthly rates of metastatic recurrence in the model were iteratively adjusted to recreate the 12-, 24-, 36-, and 48-month DDFS and OS observed in the OlympiA trial. We converted the reported annual recurrence probabilities to constant monthly rates within each year, then increased or decreased each monthly rate by increments of 0.0001 until the rates converted to monthly probabilities yielded the arm- and year-specific DDFS and OS estimates from the trial. The DDFS rates for the olaparib arm in the OlympiA trial were 94.4% at 12 months, 90.6% at 24 months, 88.0% at 36 months, and 86.5% at 48 months. 3 For the no olaparib arm, the DDFS rates were 90.3% at 12 months, 84.0% at 24 months, 81.0% at 36 months, and 79.1% at 48 months. The OS based on this interim analysis for the olaparib arm was 98.0% at 12 months, 95.0% at 24 months, 92.8% at 36 months, and 89.8% at 48 months. For the no olaparib arm, the OS was 96.9% at 12 months, 92.8% at 24 months, 89.1% at 36 months, and 86.4% at 48 months.

Age-specific non–breast cancer mortality was estimated from 2018 US life tables. 12 The monthly probability of death after metastatic recurrence was based on a published estimate of 13.3 months median survival in patients with TNBC. 13

Using a health care system perspective, we estimated direct medical costs related to breast cancer, including olaparib, routine oncology visits, annual surveillance imaging, and the cost of care for metastatic recurrence. Olaparib was discontinued after the first of metastatic recurrence or 1 year of treatment. We assumed that patients receiving olaparib would see their oncologist monthly during the year of treatment, then every 6 months for 5 years after completing systemic therapy, and annually thereafter. Patients not receiving olaparib were assumed to have an oncologist visit every 6 months through year 5 and annually thereafter. We assumed that patients who had at least 1 breast (53.5% of patients in the OlympiA trial) would receive a diagnostic mammogram and breast magnetic resonance imaging with contrast annually. The costs of managing serious adverse events were not considered, given the tolerability profile of olaparib found in the OlympiA trial. Treatment costs for metastatic recurrence did not differ by receipt of adjuvant olaparib.

The costs of olaparib, routine oncology visits, breast imaging, and care for metastatic recurrence were identified from previously published studies, inflated to 2021 US dollars, and from the 2021 Medicare Fee Schedule ( Table 1 ). 15 - 18 Because most patients in the OlympiA trial had TNBC in both the olaparib (81.5%) and placebo (82.8%) arms, estimates for this specific patient population were used when available. All costs and health outcomes were discounted by 3% annually. 19

The utility weight, which reflects health-related quality of life on a scale of 0 (worst imaginable health) to 1 (best imaginable health), for patients not receiving olaparib was assumed to be equivalent to the utility associated with no recurrence after completion of adjuvant treatment. This utility was 0.98, based on published estimates. 14 The utility weight for patients receiving olaparib was estimated as a multiplicative function of the utility for patients not receiving olaparib and a factor less than 1. The utility for metastatic recurrence was estimated from published studies. 14

We performed 1-way sensitivity analysis to evaluate the uncertainty around model inputs. Ranges of values were obtained from published studies or were varied ±25% from their base-case value ( Table 1 ). We also performed probabilistic sensitivity analysis, assigning probability distributions to model inputs and conducting 10 000-second-order Monte Carlo simulations. Costs were characterized by γ distributions and utilities by β distributions. The HR for the association between olaparib and metastatic recurrence was modeled using a log-normal distribution, and the monthly rate of death following metastatic recurrence was modeled with a β distribution. Probabilistic sensitivity analysis results were plotted in a scatterplot of ICERs and a cost-effectiveness acceptability curve, varying the willingness-to-pay threshold from 0 to $300 000 per QALY.

In the base-case analysis, treatment with olaparib yielded 17.94 life-years and 17.40 QALYs at an average cost of $314 789 compared with no olaparib, which yielded 16.69 life-years and 16.20 QALYs at a cost of $181 655 ( Table 2 ). Adjuvant olaparib was associated with a 1.25-year increase in life expectancy and a 1.20-QALY increase at an incremental cost of $133 133. The resulting ICERs were $106 506 per life-year and $110 962 per QALY gained. At a willingness-to-pay threshold of $150 000 per QALY gained, olaparib would be considered cost-effective at its 2021 price. Varying the age of the cohort resulted in a modest change in the results. Holding constant assumptions about disease recurrence and drug efficacy, adjuvant olaparib was associated with ICERs of $124 897 per QALY in patients aged 50 years and $154 921 per QALY in those aged 60 years.

The ICER was most sensitive to assumptions about the effectiveness of olaparib, the discount rate, and the utility of the no recurrence health state ( Figure 1 ). At the upper bound of the 95% CI for the HR for distant disease or death estimated in the OlympiA trial (0.77), adjuvant olaparib was associated with a cost of $212 303 per QALY gained. At the lower bound of the 95% CI (0.48), the ICER was $75 999 per QALY gained. When olaparib cost 25% more than the base case value ($18 154), the ICER was $145 868 per QALY gained. At the lower bound of the cost of olaparib ($10 892), the ICER was $76 056 per QALY gained. Results were least sensitive to the utility weight for metastatic recurrence and costs of routine oncology visits and surveillance mammograms.

In probabilistic sensitivity analysis, we found that, in all 10 000 simulations, adjuvant olaparib was both more costly and more effective than no olaparib ( Figure 2 ). In more than 92% of the simulations, olaparib was preferred to no olaparib at a willingness-to-pay threshold of $150 000 per QALY gained. The probability that adjuvant olaparib was cost-effective, compared with no olaparib, exceeded 50% at a willingness-to-pay threshold of approximately $111 000 per QALY and exceeded 80% at a threshold of approximately $130 000 per QALY ( Figure 3 ). At a threshold of $100 000 per QALY, the probability of adjuvant olaparib being cost-effective was approximately 30%.

Since the approval of trastuzumab for ERBB2 -positive tumors in 1998, targeted systemic therapy remains a paramount goal of drug discovery and clinical care for breast cancer treatment. 20 PARP inhibitors represent one of the newer classes of targeted therapies for breast and other solid tumors. The OlympiA trial indicates that adjuvant olaparib improves both DDFS and OS in patients with high-risk, early-stage breast cancer with gBRCAm. Our economic evaluation suggests that olaparib is also cost-effective in this setting.

In our base-case analysis from a health care system perspective, adjuvant olaparib was associated with an ICER of approximately $111 000 per QALY gained. This estimate compares favorably with currently accepted thresholds of willingness to pay for health gains in the US and with estimates of the cost-effectiveness of olaparib in other tumor types and settings. 21 In its original indication for recurrent ovarian cancer in women with gBRCAm, estimates of the cost-effectiveness of olaparib in the US have varied from approximately $193 000 to $287 000 per progression-free year of life saved. 22 - 24 For newly diagnosed advanced ovarian cancer, the ICER could be as low as $51 000 per QALY. 25 In metastatic prostate cancer, the estimated cost-effectiveness of olaparib has varied from approximately $117 000 to just under $250 000 per QALY. 26 , 27

It is notable that more than 80% of patients in each arm of the OlympiA trial had TNBC. Given the relatively poor prognosis for patients with this tumor profile, adjuvant therapies that can reduce recurrence risk in TNBC are especially meaningful. Not only did olaparib have clinical benefit in this patient population, our results suggest that, despite its relatively high cost, it also represents a good value.

Our results were sensitive to assumptions about the effectiveness of olaparib. Within the 95% CI of the HR for DDFS found in the OlympiA trial, the ICER for adjuvant olaparib varied nearly 3-fold, from as low as $76 000 per QALY gained to as high as $212 000 per QALY gained. Now that olaparib is approved for adjuvant breast cancer treatment in patients with gBRCAm, it will be important to monitor its effectiveness and cost-effectiveness in the clinical practice setting. 1

Our model was agnostic to specific therapies received in the metastatic setting. We used an average monthly cost estimate for treating metastatic TNBC based on a mix of agents and regimens, which may have included a PARP inhibitor, and our estimate of breast cancer mortality following distant recurrence was based on evidence from cohorts with TNBC who were treated before the advent of PARP inhibitors. It is not yet clear whether oncologists will routinely administer PARP inhibitors in the metastatic setting following disease recurrence in patients who received adjuvant olaparib. As physicians begin incorporating adjuvant olaparib into their treatment protocols, we will learn more about the subsequent use and effectiveness of PARP inhibitors for treating metastatic disease. It is also not clear how the effectiveness and cost-effectiveness of adjuvant olaparib might be affected by the use of other agents, including pembrolizumab, capecitabine, and abemaciclib. Some of these medications have become standard of care in the neoadjuvant or adjuvant settings for patients with TNBC or other tumor features. 28 As newer data on costs and outcomes of different treatment combinations and sequences become available, estimates of olaparib effectiveness and cost-effectiveness can be updated to consider the impact of these other systemic therapies.

This study has several limitations. We did not explicitly model local and regional recurrences or subsequent primary cancers. Although these events were uncommon during the available follow-up, and the OlympiA trial was not powered to test differences in these end points, observed rates favored the adjuvant olaparib arm. Similarly, we did not explicitly model adverse events or treatment discontinuation. The rate of serious adverse events—events causing death, hospitalization, or major disability—was similar in the 2 study arms. While the frequency of adverse events leading to treatment discontinuation was greater among patients receiving olaparib (10.8% vs 4.6% of those receiving placebo), the absolute difference was relatively small. Given the substantial impact of olaparib on DDFS and OS, explicitly incorporating treatment discontinuation would not likely alter our conclusions. Moreover, our base-case analysis assumed that adjuvant olaparib was, on average, associated with slightly diminished quality of life compared with no olaparib. Although there was no significant difference between treatment arms in quality of life during the first 24 months following randomization in the OlympiA trial, more patients in the olaparib arm experienced a grade 3 or 4 adverse event, particularly anemia. Even when the utility weight for olaparib was assumed to be as low as 80% of the utility of no olaparib (ie, olaparib utility, 0.784; no olaparib utility, 0.98), the ICER for adjuvant olaparib was less than $150 000 per QALY gained.

In some instances, we used Medicare reimbursement rates as a proxy for health care costs, although most patients in the model would not be eligible for Medicare, since we assumed a starting age of 42 years, consistent with the OlympiA trial. 2 However, sensitivity analysis on these model inputs—costs of routine oncology visits and surveillance mammograms and breast magnetic resonance imaging—suggested that they had almost no impact on the estimated ICER of adjuvant olaparib.

In extrapolating results beyond the OlympiA trial, we conservatively assumed that olaparib had no association with DDFS and OS after 4 years. If olaparib has longer-term benefit, then our analysis would yield an overestimate of the ICER, and therefore an underestimate of the cost-effectiveness of the drug in this setting.

In addition, our base-case analysis was conducted from the health care system perspective, and thus did not include costs associated with informal caregiving or lost productivity. Patients receiving olaparib might incur greater costs associated with lost workdays due to a greater number of physician visits during the year of treatment. However, these costs would be insubstantial compared with the costs of informal caregiving in patients at the end of life and productivity losses associated with morbidity and premature mortality due to metastatic cancer. Such productivity losses could be considerable given the age distribution of these patients with breast cancer. 29 However, since adjuvant olaparib should decrease these costs on average by reducing the risk of metastatic recurrence, an analysis conducted from a societal perspective would likely find a more favorable ICER.

We assumed that gBRCAm status is known for every patient entering the model. Despite calls for universal germline genetic testing in all patients with newly diagnosed breast cancer, National Comprehensive Cancer Network guidelines still recommend testing based on risk factors associated with clinical presentation, personal health history, and family history. 30 - 32 The availability and effectiveness of adjuvant olaparib for patients with gBRCAm is likely to prompt increased testing among patients with newly diagnosed early-stage breast cancer. A recent study of strategies to identify patients with breast cancer eligible for olaparib treatment reported that universal testing in patients with TNBC was cost-effective (ICER<$60 000 per QALY) compared with a selective testing strategy, and expanding testing to all patients with ERBB2-negative disease had a favorable ICER as well. 33 Universal germline testing might have even greater value when cascade testing in family members is considered. However, ensuring equitable access to adjuvant olaparib may require dedicated efforts to reduce racial disparities in the use of germline genetic testing. 5 , 34 , 35

Expensive new cancer drugs such as olaparib can pose a challenge for both payers and patients. 36 , 37 Ensuring access to this effective and cost-effective agent is critical if we are to attenuate, rather than exacerbate, disparities in breast cancer outcomes. This is especially important in the context of TNBC, which is disproportionately common and lethal in Black women. 38 , 39 The findings of this economic evaluation suggest that with an ICER less than $150 000 per QALY gained, adjuvant olaparib in eligible patients represents a good value at its 2021 price.

Accepted for Publication: November 6, 2023.

Published: January 3, 2024. doi:10.1001/jamanetworkopen.2023.50067

Open Access: This is an open access article distributed under the terms of the CC-BY License . © 2024 Zettler CM et al. JAMA Network Open .

Corresponding Author: Elena B. Elkin, PhD, MPA, Department of Health Policy and Management, Mailman School of Public Health, 722 W 168th St, New York, NY 10032 ( [email protected] ).

Author Contributions: Ms Zettler and Dr Elkin had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Concept and design: All authors.

Acquisition, analysis, or interpretation of data: Zettler, De Silva, Elkin.

Drafting of the manuscript: Zettler, De Silva, Robson, Elkin.

Critical review of the manuscript for important intellectual content: De Silva, Blinder, Robson, Elkin.

Statistical analysis: Zettler, De Silva, Elkin.

Obtained funding: Elkin.

Administrative, technical, or material support: De Silva, Elkin.

Supervision: Elkin.

Conflict of Interest Disclosures: Ms Zettler reported receiving personal fees from Columbia University Mailman School of Public Health during the conduct of the study and current employment by and equity ownership in COTA Healthcare Inc. Dr Robson reported receiving grants and nonfinancial support from AstraZeneca and Pfizer, and grants from Merck outside the submitted work. Dr Elkin reported receiving institutional research funding from Pfizer. No other disclosures were reported.

Funding/Support: Ms Zettler’s work on this study was supported by research funds from the Department of Health Policy and Management at the Mailman School of Public Health, Columbia University.

Role of the Funder/Sponsor: Columbia University had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.

Meeting Presentation: A preliminary version of this study was presented at the Annual Meeting of the American Society of Clinical Oncology; June 6, 2022; Chicago, Illinois.

Data Sharing Statement: See the Supplement .

Additional Contributions: Ling Chen provided statistical assistance to parameterize distributions for probabilistic sensitivity analysis.

Additional Information: Ms Zettler was a full-time student at the Mailman School of Public Health at the time this research was conducted; COTA Healthcare was not involved in this study.

  • Register for email alerts with links to free full-text articles
  • Access PDFs of free articles
  • Manage your interests
  • Save searches and receive search alerts

Log in using your username and password

  • Search More Search for this keyword Advanced search
  • Latest content
  • For authors
  • Browse by collection
  • BMJ Journals

You are here

  • Volume 14, Issue 8
  • Qualitative exploration of patients’ experiences with Intrabeam TARGeted Intraoperative radioTherapy (TARGIT-IORT) and External-Beam RadioTherapy Treatment (EBRT) for breast cancer
  • Article Text
  • Article info
  • Citation Tools
  • Rapid Responses
  • Article metrics

Download PDF

  • http://orcid.org/0000-0002-7573-6712 Sandeep Kumar Bagga 1 ,
  • Natalie Swiderska 2 ,
  • Charlotte Hooker 1 ,
  • Jennifer Royle 3 ,
  • Marie Ennis-O'Connor 4 ,
  • Siobhan Freeney 5 ,
  • Dympna Watson 4 ,
  • Robin Woolcock 6 ,
  • George Lodge 7 ,
  • Siobhan Laws 7 ,
  • http://orcid.org/0000-0003-1760-1278 Jayant S Vaidya 8
  • 1 Research , MediPaCe , London , UK
  • 2 Patient Engagement , MediPaCe , London , UK
  • 3 Strategy , MediPaCe , London , UK
  • 4 Independent Patient Advocate , Dublin , Ireland
  • 5 Lobular (Breast Cancer) Ireland , Dublin , Ireland
  • 6 Triple Negative Breast Cancer Foundation Inc , London , UK
  • 7 Royal Hampshire County Hospital , Winchester , UK
  • 8 Division of Surgery and Interventional Science , University College London , London , UK
  • Correspondence to Dr Sandeep Kumar Bagga; sandeep{at}medipace.com

Objective To gather a deep qualitative understanding of the perceived benefits and impacts of External-Beam RadioTherapy (EBRT) and TARGeted Intraoperative radioTherapy (TARGIT-IORT) using Intrabeam to assess how the treatments affected patient/care partner experiences during their cancer treatment and beyond.

Design and participants A patient-led working group was established to guide study design and to help validate findings. Patients with experience of receiving EBRT or TARGIT-IORT were purposively sampled by Hampshire Hospitals NHS Foundation Trust. These patients had been offered both regimens as per their clinical features and eligibility. Semistructured interviews were conducted with 29 patients and care partners with lived experience of either EBRT (n=12, 5-day FAST-Forward regimen and n=3, 3-week regimen) or TARGIT-IORT (n=14). Thematic analysis was then carried out by two coders generating 11 themes related to EBRT or TARGIT-IORT.

Setting Semistructured interviews were conducted virtually via Zoom during February and March 2023.

Results A number of procedural grievances were noted among EBRT patients. EBRT was perceived as being disruptive to normal routines (work, home and travel) and caused discomfort from side effects. TARGIT-IORT was perceived by patients and care partners as the safer option and efficient with minimal if any disruptions to quality of life. The need for timely accessible information to reduce anxieties was noted in both cohorts.

Conclusions This qualitative study found that patients perceived EBRT as being greatly disruptive to their lives. In contrast, the one-off feature of TARGIT-IORT given while they are asleep during surgery gives them the feeling of stamping out the cancer without conscious awareness. These insights can help healthcare staff and policy-makers further justify the incorporation of the treatment favoured by these patient perceptions (TARGIT-IORT) more widely in routine practice. Further research is planned to explore TARGIT-IORT in more diverse populations and in the 35 countries where it is an established treatment option.

  • breast cancer
  • radiotherapy
  • qualitative research
  • breast surgery
  • quality of life

Data availability statement

Data are available upon reasonable request. Raw data such as interview transcripts are not publicly available due to participant confidentiality and risk of compromising privacy but can be made available to researchers if appropriate confidentiality, ethics, regulatory and consent processes can be put in place.

This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See:  http://creativecommons.org/licenses/by-nc/4.0/ .

https://doi.org/10.1136/bmjopen-2023-081222

Statistics from Altmetric.com

Request permissions.

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

STRENGTHS AND LIMITATIONS OF THIS STUDY

This qualitative study included the two routinely offered radiotherapy treatment options (External-Beam RadioTherapy and TARgeted Intraoperative radioTherapy using Intrabeam) allowing for assessment of patients’ perceptions and experiences in each.

Methodological strengths include measures to prevent researcher bias, such as producing reflexive accounts, independent coding, and exploring both patient and care partner perspectives .

Extensive involvement of a patient-led working group ensures the study design and delivery is robust yet sensitive and respectful.

A limitation is the lack of diversity in the study population, being predominantly white and of higher socioeconomic status from a single English location because of which we are planning to explore these concepts in more diverse populations and in the 35 other countries where TARGIT-IORT is already a well-established treatment option.

The COVID-19 pandemic during which the patients were treated may have introduced some confounding factors, but they did provide useful insights into patient isolation issues.

Introduction

Conventionally, radiotherapy treatment for breast cancer has involved patients undergoing External-Beam Radiotherapy (EBRT) several weeks or months after their surgical removal (lumpectomy). EBRT is usually delivered postoperatively to the whole breast. For external beam radiotherapy, patients are required to attend 15 treatment sessions, each lasting about 15 min, 5 days a week over 3–6 weeks. 1 , 2 In 2020, the FAST-Forward protocol, administering radiotherapy over five sessions, was adopted in some parts of UK, partly as a response to the COVID-19 pandemic and even before the results of the FAST-Forward trial were published. 3 An additional 5–8 days of tumour bed boost is given in about a quarter of cases who are found to have higher-risk disease. 3

Targeted Intraoperative Radiotherapy (TARGIT-IORT) using Intrabeam offers an alternative to women with early breast cancer that is currently being used in a small number of hospitals across England. This approach, first used in 1998, delivers a single dose of radiotherapy directly to the breast tissue surrounding the tumour immediately after the tumour has been removed and the patient is still under the same anaesthetic in the operating theatre. The long-term results of the international randomised TARGIT-A trial (n=2298) in which TARGIT-IORT was compared with EBRT found TARGIT-IORT to be effective as whole breast radiotherapy, reduced non-breast cancer deaths and improved overall survival in those with grade 1 and grade 2 cancers. 4–12

To date, several studies have investigated patients’ experiences with TARGIT-IORT quantitatively. 13–18 These studies gathered information about patients’ quality of life (QoL) during and after treatment via questionnaires and have concluded that patients receiving TARGIT-IORT report high QoL scores 13 and better emotional well-being, less pain, fewer breast and arm symptoms compared with patients receiving EBRT. 14 19 The social impact of reducing the repeated journeys to the radiotherapy centre for both the patient and their care partner has been established. 20 Patient preferences have also been explored in studies based in the USA and Australia. 21–24 However, qualitative insights can give researchers and practitioners an in-depth understanding of patient perceptions that can help explain, with confidence, the reasoning behind the difference in QoL experienced by patients having these treatments.

Much of the literature on patients’ experiences of receiving radiotherapy has focused on EBRT alone where qualitative studies have used various methods such as workshops, interviews and diary entry analysis. Recurring themes include the need for adequate information provision, healthcare professionals’ knowledge of breast or arm lymphoedema (sluggish drainage of lymph fluid), perceived lack of choice, experiences of being naked and feelings of disempowerment, 25 psychological burdens of impact (and the resources required to support patients), 26 impact of side effects such as skin toxicity on patients’ QoL, life and health after radiotherapy and feeling mystified by radiotherapy and how it works. 27 28 While there are other studies investigating breast cancer patients’ lived experiences of receiving the diagnosis, treatment perceptions, experiences of survivorship and symptoms from radiotherapy, 29–34 they do not focus on lived experiences of receiving EBRT specifically.

In addition, the literature review has highlighted that no qualitative comparison of patients’ experiences of TARGIT-IORT and EBRT has been conducted although one qualitative study, exploring overdiagnosis of breast cancer, did briefly describe the experiences of patients having TARGIT-IORT and EBRT. 35 Rich descriptions of authentic experience can help to place the treatment pathway in the context of patients’ everyday world and to truly understand the perceived barriers, benefits and personal consequences of treatment. Therefore, this study is designed to gather a deep understanding of how patients define the benefits and impacts of each therapeutic regimen and how this qualitatively affects patients’ and/or care partners’ experiences. As a secondary aim, the study will also identify where there have been unnecessary treatment-related impacts on QoL and areas of potential improvement.

Methodology

Study design.

This study used a qualitative research design with semistructured interviews as the primary research instrument. Researchers adopted a phenomenological approach which encourages a bracketing off of researchers’ own preconceptions and opinions to help mitigate bias and promotes a special importance to individual human experience where multiple realities exist (based on participants’ own subjective experiences).

At the inception of the study, a patient-led working group was established to ensure the research was designed and conducted in a respectful and sensitive manner. Figure 1 outlines the research process. The authors have used the Consolidated criteria for Reporting Qualitative research to report the study 36 (see online supplemental file 1 ).

Supplemental material

  • Download figure
  • Open in new tab
  • Download powerpoint

Overview of research process. EBRT, External-Beam RadioTherapy; TARGIT-IORT, TARGeted intraoperative radiotherapy; REC, Research Ethics Committee.

Working group

Four patient advocates (three patients who had been treated for breast cancer and one care partner) with lived experiences of radiotherapy were invited to participate in a working group with the researchers. An initial meeting was held on 19 August 2022. In this meeting, the research design was discussed which included reviewing the study aims, the need for a comparison group, data collection method and participant recruitment channels. The second meeting, on 30 May 2023, focused on validating the emerging themes from the analysis. Between these meetings, the researchers shared early drafts of the core research material (eg, participant information sheet and consent form) to obtain members’ feedback and suggestions for amendments. The working group has also coauthored this paper.

Recruitment of participants and consent

A key outcome of the first working group meeting was to ensure the study design had a comparison group. This meant recruiting patients or care partners with lived experience of receiving EBRT to enable a comparison to those who had received TARGIT-IORT. The eligibility criteria are based on criteria used previously in TARGIT-IORT clinical trials ( table 1 ). All patients were, at the time of their cancer diagnosis, eligible for both TARGIT-IORT and EBRT.

  • View inline

Eligibility criteria for interview participants

Participants were first identified by SL and GL at Hampshire Hospitals NHS Foundation Trust in accordance with the eligibility criteria, using purposive sampling (NHS stands for National Health Service, which is provided free for cost to patients in the United Kingdom). This Trust recruited patients to the randomised TARGIT-A trial between 2000 and 2012. Since the National Institute of Health and Care Excellence (NICE) recommendation to offer TARGIT-IORT to suitable patients, they have been offering the procedure to their patients. For this study, GL compiled a list of all patients who received either TARGIT-IORT or EBRT. Prospective participants were stratified first into rural and urban subgroups and then by age (50–60 and 60–70). A randomiser was then applied to these subgroups to ensure the final selection process was free from bias from clinicians who had treated patients. Cover letters and recruitment advertisements (approved by the ethics committee) were posted by GL to 58 eligible patients. Those interested in participating in the study contacted researchers voluntarily. Subsequently, the researchers shared a participant information sheet, consent form and provided further information during a short introductory call where participants also had the opportunity to ask further questions about the purpose and conduct of the research. In total 29 participants responded, and all were successfully recruited to the study. Participant characteristics can be found in table 2 .

Sample characteristics of interview participants

Semistructured interviews

Working group members agreed semistructured interviews should be used to gain rich descriptive accounts of experiences with EBRT and TARGIT-IORT. Members felt discussing sensitive and privileged information, namely people’s experiences of receiving the cancer diagnosis and treatment, would be more uncomfortable in, for example, a focus group environment. Therefore, two discussion guides (one for each type of radiotherapy) were developed and refined with the help of working group members (see online supplemental files 2 and 3 ). Interviews were conducted between 9 February 2023 and 2 March 2023 by researchers SKB, NS and JR who are experienced in using qualitative research methods as part of their professional roles. Each interview lasted approximately 60 min and was conducted virtually through Zoom and either digitally video recorded or audio recorded depending on participants’ wishes. The identity of the interviewer can positively or negatively affect the interviewer–interviewee relationship. Participants were, therefore, asked whether they would prefer an interviewer of the same sex, with the default position being that a female interviewer (NS or JR) will conduct the interview with patients and similarly, a male interviewer (SKB) for male care partners. During the interviews, informal member checking took place by interviewers routinely summarising what participants had said to check for accuracy and understanding.

Recordings were transcribed verbatim, with potentially identifying details anonymised and assigned a unique identifier.

In keeping with a phenomenological approach to analysis, researchers began by writing reflexive accounts. This involved reflecting on their own experiences, preconceptions and assumptions that have the potential to influence interpretations of participants’ accounts. This process helps to create the self-awareness required when attempting to consciously bracket out thoughts and opinions that could lead to bias.

Reflexive thematic analysis based on Braun and Clarke’s 37–39 six-step approach was used to analyse the qualitative data and to identify recurring themes related to patient and care partner experience ( figure 2 ). The process of data familiarisation took place during data collection, postinterview reflections, transcribing and re-reading the transcriptions and interview field notes. Initial transcripts were individually coded (identifying units of meaning) by two researchers (SKB and NS) who then reviewed the other’s codes. Through subsequent discussion and reflection and agreement that theoretical saturation was sufficiently achieved, codes were finalised and applied throughout the remaining transcripts. Through an iterative process, descriptive and interpretative codes were categorised to form 28 subthemes and 11 major themes. Microsoft Word and Excel were used to facilitate coding, grouping and text retrieval to identify illustrative quotes.

Reflexive thematic analysis (adapted from Braun and Clarke, 2020). 37

Patient and public involvement

Patients were involved in refining the research question, study design and outcome measures. Their contributions during these discussions were informed by their lived experiences, priorities and preferences during the first working group meeting (described above in the ‘Methodology, Working group’ section). Participant identification and recruitment channels were also discussed with the patient-led working group though they were not directly involved in recruitment into the study nor in the conduct of the study. The results of the study will be disseminated to the study participants once the peer-reviewed paper is published. The burden of the intervention was assessed by the patients themselves for this qualitative study.

The following section presents findings from the thematic analysis which looked at EBRT and TARGIT-IORT separately and the outputs from discussions of the second working group meeting ( table 3 ). Participants’ quotes have been labelled with identifiers (eg, P1, P2) not known to anyone other than the researchers (ie, not the hospital staff, participants themselves or anyone else).

Themes and subthemes arising from the interviews

Themes coming out of interviews with patients who had EBRT

Dissatisfaction with unalterable elements of ebrt procedure.

The majority of EBRT participants expressed discontent with many of the standard elements of the EBRT procedure. Some participants felt intimidated by the size of the room being ‘disturbing’ (P22) and the radiotherapy machine being ‘scary’ (P19):

…the room that you go into where the machine is, is cold…it could be a bit warmer. Now, some of that could be psychological because you're in a big white room with a big, huge machine… (P3) …the 2 nurses go into another side room, so, you feel so alone, and you know, and this machine sort of moving around you. It’s, it is quite scary to deal with. (P19)

Four participants also described the challenge associated with needing to hold one’s breath during sessions. This is done with the hope that the heart may receive less radiation by pushing the chest wall and the breast away from it. Participants described it as saying, ‘ that was the worst bit’ (P12), ‘ it’s going to be difficult’ (P15), ‘ I don’t want to be zapped on my heart’ (P21) and another felt it was ‘ really claustrophobic’ (P19) or causing ‘ panic’ (P19). The planning appointment required for EBRT was met with similar dissatisfaction. While there is a clear appreciation for healthcare staff and their workload, participants were unhappy with the dehumanising nature of these appointments:

You become another face… you do feel like a slab of meat while they're trying to get you in the right position and it’s not a pleasant experience. (P19)

These experiences resonated with working group members’ recollections: ‘silent’ and ‘cold, dark room’ and finding it difficult, a ‘physical challenge’ to maintain position after surgery. Another member felt that while healthcare staff were pleasant, the experience of receiving radiotherapy itself is ‘quite traumatic’ and emotional, ‘I remember lying there and tears came from nowhere…’.

As with study participants, working group members acknowledged that while healthcare staff themselves are not at fault, the ‘system’ causes the dehumanising elements described by participants referring to poor staffing levels and a high-pressure work environment within healthcare.

In contrast, one participant positively describes the EBRT sessions, ‘ there’s music on, and I didn’t find any cause to worry at all’ (P15). The relaxing effect of music was echoed by a working group member who also recalled how music helped in which she felt was ‘ brilliant’ and stated ‘ it helped my head’ .

Finally, one-third of participants expressed a strong objection to being tattooed which was required to ensure radiation is delivered to the right location.

What really did wind me up actually, I had to have the 3 dots tattooed on me and I didn’t want tattoos. (P3)

Participants were frustrated by the fact that it is permanent, the colour used and two participants felt it affected their confidence in wearing certain clothes, ‘ I can still see that now, if I wear a swimsuit or something’ (19 ). One working group member felt that patients are often uninformed about radiotherapy and that patients’ preferences are not listened to. She concluded that this was a good example of an area that required adequate information sharing at the right time.

Unanticipated disruptions cause helplessness

One-third of EBRT participants experienced either delays on the day of the EBRT session or extensions to their course owing to either machine failure or staff absences. The impacts on patients and care partners include stress, aggravation and disappointment with knock-on consequences proving to be burdensome:

…the machine broke down…but I couldn't find [the new hospital], and I got really tired and upset. I was trying to find where I was supposed to go, and nobody seemed to know, and I just managed to grab the team before they went home. I was like, ‘Give me my last radiotherapy now!’ It was down in some basement I mean. Location S is a maze. So, that was a bit stressy. (P1)

Patients who received their EBRT during the COVID-19 pandemic were unable to have their care partners (in most cases husbands) with them at key treatment stages. This isolation caused additional anxieties in the EBRT cohort as one care partner stated, ‘ it’s disappointing and it would have been nicer for me to be able to support her more…’ (P23). Similarly, a patient participant states:

…it’s anxiety level of just having that, that security blanket of having somebody there with you. (P19)

The working group discussed isolation and the emotional impact during EBRT sessions. Since EBRT continues for days and indeed weeks in some cases (5–6 weeks for all members), patients truly feel alone during this phase. They recalled overwhelming feelings of sadness during the sessions with thoughts such as, ‘ how did I get here’ . One member expressed empathy with study participants who would have had to endure further isolation during the COVID-19 pandemic period.

Straightforward sessions were met with surprise but travelling for EBRT was still a concern

Four out of the 15 participants interviewed from the EBRT cohort, stated that no side effects or other complications were experienced with one participant saying she felt like ‘ one of the lucky ones’ (P20). In the absence of side effects such as pain, burning or tiredness, patients experience a sense of surprise and relief after having received prior warnings from either clinical staff or hearing stories from friends and family.

…they said to put a couple of tubes of aloe vera, and keep in the fridge, and put it on religiously. But I was fine. I was fine. I literally had no burning. No rash, no nothing. I’d heard about friends having burns, but I just didn’t, I didn’t. (P12)

Similarly, three participants were appreciative of the fact that individual sessions of EBRT were in fact ‘ very straightforward’ (P1) and quick:

I did find the time, actually, went quite quick. It wasn’t very long; it might have been an hour. Yeah, it wasn’t too long. (P1)

Although these four participants did not experience radiation-related side effects, it should be noted that two of the four participants did, express frustrations about the burden of travelling during EBRT sessions and that the overall radiotherapy course was ‘ time-consuming’ (P2).

Disruption to normal life routines

A few participants were either employed or self-employed and described how EBRT impacted their own work performance (eg, tiredness, weakness in arm) with one person concluding, ‘ I’m an office worker but if I’d be doing a manual job, I think it might have impacted more.’ (P3). There is also the realisation that patients would likely need to adjust work sessions to accommodate for the EBRT sessions and the related side effects: ‘ I’ve worked out a part time basis to get back into work.’ (P19).

Participants shared concerns about the impact on work colleagues. One participant states: ‘ I’m the only person that does my job. So, I was acutely aware that when I’m not at work other people are picking up my job’ (P19). Similarly:

…we were a short-staffed team, I was aware that when I wasn’t there, it was putting work onto other people, and I felt I should have been there…. (P3)

The EBRT cohort also revealed the emotional toll of work-related worries and impacts, and the work guilt associated with the impact on colleagues. Similarly, the potential impact on the care partner’s work is also, clearly, a significant consideration for patients:

The first night after my first session I was in so much pain, I mean I didn’t sleep a wink the first night. It was absolute agony…it was my self-confidence, and everything was destroyed…and I didn’t dare, I didn’t want to wake my husband up. He had to go to work early, so then he could take me to radiotherapy in the afternoon. (P19)

These impacts on employment were corroborated during working group discussions. One member, commenting in particular on self-employed people, described the impact on financial standing and home life as ‘catastrophic’.

In contrast, those who had flexible hours of working were less affected, as one patient participant describes her care partner: ‘ luckily he was doing a job where could sort of pick and choose what he did and his hours, so it was all right.’ (P3).

The repetitive nature of the EBRT sessions, the travel involved and the side effects experienced all also impacted on participants’ normal daily routines. For instance, home activities such as shopping, gardening and caring responsibilities were impacted:

I think we might have cut down [caring for elderly parents] to once a week instead of two or three times… (P3)

One participant mentioned a close family member taking a week off from work to support looking after her, her husband, family pets and ‘ doing a few household chores like pushing the Hoover around’ (P22).

Severe pain brought on by EBRT was described as ‘ agony’ (P19) both during sessions and after sessions (P7). Ongoing pain months and years after the radiotherapy means patients need to settle into a new norm, now constantly having to be aware of their own ‘ limitations’ (P6):

I’m still aching like mad from [radiotherapy], that’s two years later and I’m still achy and in pain. (P6)

For working group members, these experiences were familiar. One member clarified that although there were no specific side effects from the radiotherapy the disruption to life was ‘ hugely problematic’ .

Many of the patients who received the FAST-Forward protocol included sessions on either side of a weekend resulting in a total of 7 days to complete the course. No participants complained specifically about the weekend being involved in this way however many described dedicated activities for the weekend, for example, food shopping, family commitments and short trips away which would undoubtedly be affected while in active treatment.

Conversations with participants revealed personalities play a role in patients’ cancer journey. One participant felt the emotional impact of radiotherapy treatment (and the subsequent withdrawal of routine contact with healthcare staff) more than all the others. The anxieties associated with this perceived void and the mental health impact itself are disruptive to re-engaging with normal life and routines:

…you’ve got people checking on you as in consultants or the breast care specialist nurses, your GP, the radiographer, people ringing you, checking on you, you’re seeing them all the time. After the radiotherapy, I’m suddenly though on my own now. I didn’t realise it was a real security blanket…it was so reassuring to seeing the consultant and seeing this nurses every day. That was the bit where I took a bit of a dive…I had two or three days where I couldn’t stop crying, I thought, ‘Oh I’m on my own now’… (P12)

There were also those who demonstrated a pragmatic mindset viewing any discomfort and impactful delays associated with EBRT sessions as realities to, in effect, take in their stride, ‘ any inconvenience, you just get on with it.’ (P20 )

Travel is clearly an uncomfortable reality and a demanding aspect of receiving EBRT. Many participants complained about the repeated journeys required for EBRT sessions. The burden has been described as ‘ dreadful’ (P7) with people feeling ‘ exhausted’ particularly where the effects of radiotherapy (tiredness and pain) are felt. The burden of travel also manifests as experiencing a longer day overall as well as the sheer cost of public transport used to make the trips independently:

It’s a pain having to go to Location S, there’s a hospital there where the bus doesn’t even go. So, from Location O, I have to get a bus from here to Location H, then wait half an hour, then from Location H to Location W, bus station, then a taxi to the hospital. That all costs 30 quid and is time-consuming, and when you’re sore, it’s not ideal. (P2)

In the majority of cases, there is a reliance on others (the husband but on occasion friends or children) to drive participants to the EBRT sessions. While there are no direct statements indicating a burden to care partners, it is important to note that in these cases both the patient and the care partner endure the repeated journeys:

My husband drove…I’m not a good driver…I certainly can’t park so it was good that he went with me because in case, if something went wrong or something because he’s like my rock he is. (P3)

The location of the EBRT sessions is critical to the quality of patient experience and when participants were presented with two hospitals to choose from it was clearly valued:

…it’s the same surgeons, all the same team, who were in Hospital W or B. I opted for Hospital B, it’s nearer to me and I could get to Hospital B easily, whereas Hospital W was an ordeal for me. (P21)

Two participants who were retired and for who the location of the hospital was particularly close indicated that travel was not a burden and considered themselves ‘ very lucky’ (P22), though of note, one of these participants experienced no EBRT side effects which may have contributed to relatively positive experience.

In contrast to many of the experiences described above, one working group member shared that she was relieved to have regained her independence since she was able to travel to her EBRT sessions herself. However, the working group notes that all their experiences included a difficult period of receiving chemotherapy first where radiotherapy was viewed as the ‘lesser of evils’.

Experience characterised by discomfort from side effects

A wide range of side effects, clearly attributed to EBRT, were reported by the vast majority of participants and this characterises an important part of the EBRT experience. These included varying intensities of tiredness; burning (from warm sensation to blistered and sore); skin-related conditions (dryness, itchiness, rash); pain; and breast size and density changes. The most common complaint was tiredness:

…there were 3 or 4 days the following week when I just had to go to bed, or just have to, you know, lie on the sofa in the afternoon, or you know I was just really bombed out, and I’m not someone who goes to bed and afternoon normally, I’m always busy, and but I had to. I was knackered. (P12)

EBRT sessions can be very uncomfortable. Severe pain, described as ‘ agony’ (P7, P19) was experienced by two participants. One individual, unable to withstand the pain and the tiredness experienced, made an independent decision to stop attending the sessions for a few days:

I was very tired…I think I might have missed a few days, because I couldn’t make it in between…I thought I’ve done so much now, I’m not going to go anymore because it was really, really hurting…I just wanted it to end and go away? And not think about it anymore basically. (P7)

Patients experienced some side effects such pain in the breast or weakness in the arm for a prolonged period—months and years after the EBRT sessions. There were also reports of new symptoms requiring follow-up that were attributed to EBRT. One participant (P8), who felt uninformed about the ‘long-term, lasting and late effects of radiotherapy’ had developed a new pain under her ribs—she reflected:

I would not have had radiotherapy, and I would not be beating myself up about having had it now had I been given the full information about the long-term effects. You know, it’s life-shortening, radiotherapy is life shortening in itself like chemotherapy. (P8)

During discussions at the second working group meeting, members were not surprised by the insights captured from study participants and felt strongly that there were ‘no positives’ from EBRT (particularly when compared with TARGIT-IORT). One member reflecting on their experience with EBRT stated they came away thinking ‘almost anything is better than this.’ (WG member)

Specific anxieties about receiving EBRT

Discussions with the EBRT cohort revealed three main anxieties associated with receiving radiotherapy. First, while there is evidence that more information early on (particularly from consultants) helps to reduce worry, stories of radiotherapy experiences from friends and family members can raise concerns and anxiety levels:

I only knew what I knew as a lay person, you know, various friends have had radiotherapy, unfortunately, people know a lot of people who have all these sort of things… I’d heard about friends having burns… (P12) …and actually, talking to another friend, she said she would do chemotherapy any day over radiotherapy because of how the radiotherapy, the pushing around and making you feel like a piece of meat, how it how it made her feel. (P19)

The second concern was the potential for radiation to cause harm: (a) so soon after surgery and (b) to healthy tissue and organs: ‘ You’ve got to heal a bit; you can’t go straight into radiotherapy because obviously you’re as raw as hell’ (P6). Two participants in particular experienced discomfort in their arms during their EBRT sessions—one participant had a number of lymph nodes removed from the armpit and one participant developed a seroma which is an abnormal accumulation of fluid following surgery. A working group member shared a similar experience where the development of seroma delayed the start of her radiotherapy course.

Another participant who was particularly concerned about unnecessary radiation exposure requested that only half the breast be irradiated because she wanted ‘ the absolute minimum’ (P8). Similarly, another care partner described his wife’s concerns:

One thing is that my wife was worried about was the radiotherapy because obviously there is this thing with radiotherapy, particularly on the breast, of potential damage to the lungs and she was very concerned about that. (P23)

Third, since many of the participants received their EBRT during the COVID-19 pandemic, a few expressed worries about potential delays either due to staff shortages, protocol-driven cancellations (ie, limiting patient numbers) or themselves contracting COVID-19 (since multiple visits are required with EBRT) and thereby being unable to attend hospital:

But COVID was going on and I remember being so scared that my appointments would be cancelled. (P12)

Targeted Intraoperative radiotherapy using Intrabeam

Perception that targit-iort is efficient and aggravation-free.

Of the 14 TARGIT-IORT participants interviewed, 11 indicated the one-off feature of the procedure was appealing. There are many references to how quickly the procedure was completed ‘ it’s lovely to get it all done and finished with on the day’ (P26). Similarly:

…having it done and dusted, and then then waving goodbye at the hospital gates, it was like why, why would I say, ‘No thank you’? (P16)

As a consequence of this efficiency, there is relief that the procedure permitted radiotherapy to be administered without any COVID-19-related delays, or exposure to the COVID-19 virus during travel or hospital during the multiple visits for EBRT, or complications for which three participants had expressed initial concerns:

I was just delighted that it was dealt with really, really quickly, because back at that time the news was full of things where, you know, because of COVID-19, you know, everything has been delayed, and people not getting cancer treatment and that was one of my, I remember having that conversation with the consultant and said, ‘Look are we going to be delayed’. (P25)

There is a similar relief detected in participants discussing the potential positive impact TARGIT-IORT can have on patient’s mental health as one care partner states:

…the alternative would have been [EBRT]… her symptoms of depression are she gets very, very tired… so intuitively our reaction to [TARGIT-IORT] was… actually quite a good idea. (P24)

Going through a cancer diagnosis and receiving treatment was clearly an emotional time. One participant was impressed with TARGIT-IORT precisely because the efficient delivery of radiotherapy facilitates her moving on quickly:

…the beauty of intraoperative radiotherapy is that I could say ‘OK, been there, done that, move on. (P9)

Convenience of performing TARGIT-IORT during surgery is valued

Most participants from the TARGIT-IORT cohort shared why they preferred to receive radiotherapy at the same time as the surgery. There is a recognition of the convenience that TARGIT-IORT brings as a result of not having to attend hospital on multiple occasions, for example, less travel and car parking and supporting independence (particularly for retired individuals):

It’s my choice to have [TARGIT-IORT] because I thought that it was a better option for me particularly because I live on my own and it would allow me to be more independent. (P18)

While the majority of participants were retired, those who did have young children felt TARGIT-IORT supports their caring responsibilities: ‘ I’ve got a [child] and I’ve got to look after him… This is a better way to go…’ (P14). Additionally, one retired participant who valued the independence TARGIT-IORT facilitated concluded it would also suit younger, busy, women well:

…particularly for younger women this would be an extremely good thing, if they're working, it allows them to get back to work without that constant interruption and if they've got a young family. (P18)

Many participants were able to draw from stories and experiences they had heard from friends and families. The apparent inconvenience and impact of daily radiotherapy doses discouraged patients from EBRT when TARGIT-IORT was presented as an option. One participant whose father received daily doses for prostate cancer felt she would ‘ rather get it all over in one go’ (P10). Similarly:

[TARGIT-IORT] was perfect, because it just meant I didn't have to queue up in the car park with the other poor people having radiotherapy, and I did have friends who had serious cancers who were having radiotherapy at the time, and it was just miserable. (P9)

There is also a perception that with TARGIT-IORT recovery times are likely to be faster since it would signify the end of their cancer treatment: ‘ I’m going to get [TARGIT-IORT] and it’s done’ (P16) and ‘ I can just then get on and recover’ (P4). Another participant summarises her main reasons for opting to receive TARGIT-IORT:

So, there were probably 3 reasons I went for [TARGIT-IORT]. You know, COVID, convenience, and the fact that I thought, you know, ultimately, I’d probably recover quicker. (P9)

Only one participant from the TARGIT-IORT cohort, a care partner, described a significant logistical impact due to his wife’s cancer treatment in general:

…created quite a challenge really for me, I mean, I was never going to moan about it, I wasn’t the one who just had cancer surgery! But you know, it meant the days suddenly got very challenging… (P25)

Perception TARGIT-IORT is a safer alternative to standard practice

Five participants felt that they did not experience any complications as a result of TARGIT-IORT and were able to resume their normal activities quickly. While there are a few cases of soreness and itchiness that participants specifically attributed to TARGIT-IORT, most participants did not report the range of side effects seen in the EBRT cohort. As a result, participants gave their endorsements for TARGIT-IORT, respectively:

I moved around, I got up, got changed, got dressed. It was surprising actually, this is why I’ve decided to do this, if this is what it gives you then everyone should have it. You know you don't need to feel debilitated, and you can carry on with your life. I've got a [child], and I've got to look after him. So, if you can, why not. This is a better way to go if the prognosis allows it. (P14) There were no, no after-effects, no problems. It all healed up very well, because it was quite a small incision anyway and very, very successful. (P28)

The majority of participants felt the procedure prevented healthy tissue and organs from being unnecessarily exposed to radiation because ‘ the radiotherapy is directed immediately where the lump [is]’ (P17).

I confess I heard that and thought ‘God, that’s a bloody good idea, why don’t they do that more often?’. Because obviously if you don’t have to beam through loads of flesh and muscle to get at what you're aiming for then that’s got to be better to be honest. (P24)

A few participants described side effects (soreness, tiredness), precautions (new bra needed, seatbelt cushion) and restrictions (no pressure, sport, lifting), however, they were unable to clearly attribute whether these were related to the surgery or the TARGIT-IORT procedure since both occur at the same time.

…yeah, my arm was a little bit sore…I’m sure it must have been the radiotherapy or the operation, I don’t know. (P29) …a special seat belt cushion that protects your breast from the seat belt and I had one another cushion under my breast supporting it… (P11)

Novel nature of TARGIT-IORT impresses while prompting early caution

Although it has been in use for the last 25 years since the first case was done in 1998 TARGIT-IORT is seen as novel and innovative with advantages acknowledged over EBRT. The decision to proceed with TARGIT-IORT is widely considered ‘ easy’ (P28) or ‘ intuitive’ (P24) or a ‘ no brainer’ (P4):

…well, you’re in there, so you might as well get on and do it and that would surely save the need for me having to come back, I can then just get on and recover basically…it was a no brainer for me, an absolute no brainer. (P4)

However, a few participants described their initial concerns since TARGIT-IORT was introduced by the consultants as a clinical trial and was largely unheard or ‘ unknown’ (P9). Care partners, often husbands and sometimes participants’ children wanted to carry out their own research to help making an informed decision about TARGIT-IORT. One participant had already felt she was convinced by the consultant’s explanation and the advantages over EBRT, however, her daughter, who worked in healthcare, stated ‘ …‘hold on a minute, we need to look at the statistics and the recovery times, side effects’…’ (P10). Similarly, another participant’s husband wanted an opportunity to ask the consultant more questions to help feel more reassured:

…but [care partner] just wanted to have the conversation around the intraoperative radiotherapy because it was an unknown really. (P9)

It should be noted that many of the participants were either themselves or their close family (eg, husband) highly educated, often with a science-based background and were able to explore clinical study papers and statistics: ‘ I’ve got a little statistical training…so I looked at the stats and what the mean variation was…what the levels of certitude at either end of the scale were…’ (P24).

TARGIT-IORT patients have high information needs

As mentioned above, due to the relative novelty of TARGIT-IORT and in the absence of experiences of TARGIT-IORT among participants’ friends and family, reliable information from trustworthy sources is critical. The majority of participants (in both EBRT and TARGIT-IORT cohorts) displayed high levels of trust in their consultant. Receiving adequate information from them about TARGIT-IORT, particularly due to its initial availability via a clinical trial, was appreciated:

I think what was good was the way that it was explained in the first place and what the pros and cons were, or if in fact, there weren't any cons really at all…So, you know, we were told that the treatment, doing it during the operation, is just as effective but it would mean that you would have no subsequent radiotherapy and, you know, of course I’m young and foolish, I assume that to be true, we trust the doctor… (P25) [The consultant] said’ ‘This is this, that is that…pluses and minus’…gone through pros and cons and I had made up my mind that that was a good way to go. (P14)

Working group members could relate closely to this subtheme of trust. They explained that the retrospective perception of TARGIT-IORT was always likely to be a ‘no brainer’, however, for a patient going through the highly emotionally charged process of receiving their diagnosis and treatment, at a time when they are already overwhelmed with new information, the relationship with the doctor is important:

…if it’s being offered to you, it’s important how it’s being offered to you. We put out trust in, so much, our doctors. (WG member)

Two participants described receiving explanations from radiation oncologists during their presurgery appointment, however, these discussions were not influential in helping to decide which type of radiotherapy they would receive. A few participants were wary of using the Internet to search for information related to their treatment options: ‘ I’m very cautious of what information I take in from Google’ (P4). However, the majority did conduct their own Internet searches to bolster their understanding of TARGIT-IORT:

I then went away and looked the bugger up, and then you could learn for yourself a little bit, reading between the technical stuff, what it’s all about, the success rate is there or there about the same, it’s not wonderful but for me, it was a no brainer. (P16)

The provision of information was discussed on a number of occasions by working group members. Simple and clear language is particularly important at a time when patients are already in a vulnerable, stressed and emotional state:

…you are so blindsided…the normal way you operate doesn’t necessarily apply. (WG member)

Working group members pointed to the need for information sources to be created adequately in the first place, for example, being written by patients/care partners who possess the lived experiences and so are able to elaborate on the areas that matter.

…there should never be a need for a patient to go home and want to Google, you should go home with the information in hand or go home with reputable evidence-based sources of information. (WG member)

The primary finding of this study is that the subjective experiences of patients and care partners receiving EBRT or TARGIT-IORT differ significantly. Strong recurring themes of appreciation and recognition of innovation, convenience, absence of side effects and lack of disruption to life have emerged from the TARGIT-IORT cohort while in the EBRT cohort, we have largely heard about discomfort and disruption to life. These themes—centring around (a) treatment procedure itself; (b) impact on QoL and (c) information needs—were presented to and were validated by a patient-led working group.

Patients and care partners involved in this study described numerous challenges, concerns and dissatisfaction with elements of the EBRT procedure while processing a difficult and emotional diagnosis. These findings are consistent with the existing literature on EBRT experience. 25 Probst et al 25 also identified procedural grievances, for instance, patients described the radiotherapy sessions as ‘dehumanising’, ‘emotionally draining’ and complained about the tattoos being a permanent reminder of the cancer. Previous studies exploring patient-perceived barriers to radiotherapy include patients’ fear surrounding radiation toxicity which can result in non-compliance and insufficient treatment. 13 40 In fact, research has identified that fears and anxiety regarding the EBRT experience can influence a patient’s decision to opt for a mastectomy over EBRT, despite the latter having equivalent if not non-inferior survival rates. 30 Several studies have demonstrated that as the distance from radiotherapy centre increases, the rate of mastectomy also increases. 41–45 Indeed, this was the primary patient-centric reason that the TARGIT-IORT procedure was originally conceived. 10–12 46

Our study demonstrates the need for improvements in the way EBRT is delivered and has implications for practice that extend to cases where patients are not eligible for TARGIT-IORT. In stark contrast, those receiving TARGIT-IORT have no awareness or recollection of the procedure since radiation is administered during surgery. Patients and care partners found this feature particularly appealing which contributed to their decision to opt for TARGIT-IORT. Indeed, TARGIT-IORT has been widely adopted elsewhere and treated 45 000 patients across 38 countries. 6

A high proportion of our EBRT cohort (12/15) received the FAST-Forward regimen. This regimen of highly compressed higher-dose-per-fraction radiotherapy was adopted in the UK even before the results of the FAST-Forward trial were published with the aim of reducing waiting time pressures during the early part of the COVID-19 pandemic. We recognise this 5-day regimen is indeed not adopted elsewhere in the world—it has much higher toxicity—19 times higher fibrosis and a quarter of women reporting hardened breasts 3 and this toxicity is seen even with the short follow-up of the FAST-Forward trial. 47 48 It is noteworthy that a large part of the patient’s perceived benefit came from the immediacy of TARGIT-IORT due to its administration during the same anaesthetic as their lumpectomy, the resulting convenience and the absence of additional hospital visits for radiotherapy that would be otherwise required for EBRT. In our study, this benefit of TARGIT-IORT was perceived by patients even though the majority of the comparator group received EBRT over just 5 days rather than the international standard of 3 weeks. It is, therefore, likely that the contrasting experience of patients may have even higher significance and the perceived benefit may be greater when TARGIT-IORT is compared with 3 weeks of EBRT.

Patients in the study who received TARGIT-IORT had been given the option to have it because they fulfilled the eligibility criteria ( table 1 ). Since patients made a conscious choice, it is plausible that the results of this study could be biased favouring TARGIT-IORT. However, the authors of this study submit that patients should be given a choice. Our study shows that those who choose TARGIT-IORT have a positive perception of treatment and the overall experience is better than those who opted for EBRT. Others have shown that if given a choice between no radiotherapy, mastectomy, EBRT and TARGIT-IORT, 75% of patients preferentially choose TARGIT-IORT. 24

It is evident that QoL-related benefits and impacts are a central component of radiotherapy lived experiences. Compared with TARGIT-IORT, EBRT has a prolonged impact on patients and perhaps a compounded impact on QoL where patients live alone (lack emotional or practical support), do not drive (reliance on others or public transport with additional costs and travel time) or have caring responsibilities (partners, parents, children and pets). Travel and mobility issues have been recognised as barriers already 49 as has the inconvenience of a prolonged treatment plan which can affect those living in remote areas even more. 13 Our findings demonstrate the advantages TARGIT-IORT offers to those who are eligible. All participants in our study acknowledged the efficiency of the procedure with many drawn to the option (over EBRT) because it was considered ‘straightforward’ and ‘over-and-done’ during surgery. The benefits of TARGIT-IORT to patients in terms of cost, travel time and distance have been demonstrated, in principle, elsewhere. 49 50 Furthermore, the environmental and social impact of the substantially more travel required for EBRT, and a huge reduction in carbon footprint from cancer treatment by use of TARGIT-IORT has also been well documented. 20

In our study, inconveniences and logistical complications were exacerbated by EBRT side effects which were recognised as a key characteristic of the EBRT patient experience and have implications on QoL. Stanton et al 51 investigated factors affecting QoL during and after radiotherapy and found that functional impacts of treatment, particularly breast-specific pain (eg, mobility) are important correlates of QoL. In addition, Schnur et al 27 showed that key patient concerns include the timing of side effects and the impact of side effects on self-esteem affecting patients’ perception of being attractive, good workers, patients and parents. Another study supports these findings and also shares one case of such extreme physical discomfort (pain, burning, etc) that the patient admitted she had considered ending treatment and another saying she would never choose to have radiotherapy again due to the burning sensation. 28 Our study has captured similar cases. This study also underscores the emotional toll, anxieties and stresses that disruptions to life (eg, work-related) cause and have been heard at a NICE Committee meeting. 52 There were fewer reports of side effects directly attributed to TARGIT-IORT in our findings. This is consistent with a study comparing TARGIT-IORT with EBRT (quantitatively) in which patients receiving TARGIT-IORT also reported less pain, fewer breast, and arm symptoms, and better everyday functioning when compared with patients receiving EBRT. 14 We recognise that our study has found stark differences in patient experience and perception between TARGIT-IORT and EBRT. This can seem obvious because patients with TARGIT-IORT have almost no poor experience in relation to actually receiving the treatment (mainly because they are under general anaesthetic when it is given). Our findings resonate with others who also report the negative patient experiences with EBRT and have suggested interventions to improve them. 25 The important qualitative patient benefits identified in this study are of course in addition to the quantitatively proven significant reduction in non-breast cancer deaths, and an improved overall survival in patients with grade 1 and grade 2 cancers within the randomised TARGIT-A trial. 7 However, our study does detect apparent patient obscurity between surgery-linked or TARGIT-IORT-linked side effects—this clearly needs to be addressed through appropriate education and adequate information provision.

Results from our study cohorts point to the need for improvements in communication and information provision. The role of high-quality communication by healthcare staff and access to emotional support services, particularly when radiotherapy treatment ends has been highlighted already. 26 Previous research has also identified that patients can often feel mystified by radiotherapy (EBRT), how it works and will have anxieties about life and health after radiotherapy 27 or feel disempowered and lacking the ability to make an informed choice. 25 In our study, working group members emphasised the importance of trust in connection with information provision, particularly during an emotional cancer diagnosis. Members felt a number of study findings could be addressed adequately by effectively communicating the right information at the right time. Examples include letting participants know clearly that tattoos will be permanent; what the immediate and long-term side effects of both radiotherapy types are; understanding the side effects of surgery thereby avoiding confusion with TARGIT-IORT; ensuring TARGIT-IORT explanations are always supplemented with lay language overviews of the efficacy and safety profile compared with EBRT. One study showed that more than 90% of patients felt that if they were more informed about radiotherapy, they would be less scared about it. 30 The working group advocated for any shared information, such as leaflets, to be written by patients, that is, those who have experiences of receiving radiotherapy, and therefore, have an awareness of where there are likely to be challenges in understanding treatments and their impacts clearly. Similarly, considerations also ought to be given to ensuring people with learning disabilities and communication difficulties are able to make an informed choice by developing accessible information. To satisfy ‘valid consent’, doctors in the UK are now obliged to follow the new GMC guidelines underlining the essential nature of adequate patient information, 53 about all proven treatment options, even if they are not available at their own centre. In the UK, this powerful principle is now fully enshrined in law (Montgomery vs Lancashire Health Board, 2015). 54 55 The substantially better patient perception and experience documented in this study need to be included during consultations with patients when discussing treatment options before they have their surgery for breast cancer.

This qualitative study, co-led by patients, uncovered detailed lived experiences of receiving either EBRT or TARGIT-IORT from patients treated for early breast cancer, as well as those of their care partners. The research demonstrated a patient-perceived superiority of TARGIT-IORT over EBRT—it is considered more efficient with less disruption to life routines. The paper also illustrates the importance of provision of accessible information about all radiotherapy treatment options from trusted sources, at the right time (before breast cancer surgery), to reduce initial anxieties and help patients make informed choices. These new insights need to be taken together with the established quantitative survival and QoL benefits of TARGIT-IORT over EBRT. We believe that these deep insights into the patient’s perspective will substantially improve our understanding of the lived experiences of patients with breast cancer and will help clinicians, patients and policy-makers to comprehensively consider how access to better treatments can improve patients’ lives.

Ethics statements

Patient consent for publication.

Not applicable.

Ethics approval

This study involves human participants and was approved by Health and Social Care Research Ethics Committee B (HSC REC B), Office for Research Ethics Committees Northern Ireland (ORECNI). IRAS Project ID number: 320976. Participants gave informed consent to participate in the study before taking part.

Acknowledgments

The authors thank the patients and care partners who volunteered to help in development of the research question, helping design the study and outcome measures. The authors thank patients and care partners (the study participants) who volunteered to participate in this study. We are grateful to them for giving up their time and sharing their treatment experiences and valuable insights during a difficult period in their lives for the benefit of science and clinical research.

  • Bentzen SM ,
  • Agrawal RK ,
  • Aird EGA , et al
  • Correa C , et al
  • Murray Brunt A ,
  • Haviland JS ,
  • Wheatley DA , et al
  • Vaidya JS ,
  • Bulsara M ,
  • Wenz F , et al
  • Vaidya UJ ,
  • Baum M , et al
  • Bulsara M , et al
  • Joseph DJ ,
  • Tobias JS , et al
  • Wang JS , et al
  • Sperk E , et al
  • Saunders CM , et al
  • Keshtgar MRS ,
  • Williams NR ,
  • Andersen KG ,
  • Gärtner R ,
  • Kroman N , et al
  • Hofmann F ,
  • Blank E , et al
  • Coombs NJ ,
  • Coombs JM ,
  • Vaidya UJ , et al
  • Alvarado MD ,
  • Conolly J ,
  • Park C , et al
  • Saunders C , et al
  • Saunders CM ,
  • Bulsara MK , et al
  • Beattie G , et al
  • Rosbottom K ,
  • Crank H , et al
  • Llewellyn A ,
  • Schnur JB ,
  • Ouellette SC ,
  • Bovbjerg DH , et al
  • Dilorenzo TA , et al
  • Williams F ,
  • Jeanetta SC
  • Shaverdian N ,
  • Kishan AU ,
  • Veruttipong D , et al
  • Murchison SC ,
  • DeVries KJ ,
  • Hickok JT ,
  • Morrow GR ,
  • Roscoe JA , et al
  • Hopwood P ,
  • Mills J , et al
  • Pickles K ,
  • Nickel B , et al
  • Sainsbury P ,
  • Guidolin K ,
  • Brackstone M
  • Nattinger AB
  • Nattinger AB ,
  • Kneusel RT ,
  • Hoffmann RG , et al
  • Chandwani S ,
  • Haffty BG , et al
  • Adams-Cameron M ,
  • Hunt WC , et al
  • Celaya MO ,
  • Gibson JJ , et al
  • Chinoy RF , et al
  • Vogt K , et al
  • Bargallo-Rocha JE ,
  • Soto-Perez-de-Celis E ,
  • Picó-Guzmán FJ , et al
  • Stanton AL ,
  • Krishnan L ,
  • (NICE) NIfHaCE
  • General Medical Council U
  • Mansell J ,
  • ↵ Montgomery v Lanarkshire health board , 2015 . Available : https://www.supremecourt.uk/cases/docs/uksc-2013-0136-judgment.pdf

X @jsvaidya

Contributors SKB, NS, CH, ME-O'C, SF, DW and RW were responsible for the study concept and design. SL and GL identified patients who met the eligibility criteria. SL, GL and JSV contributed to the study design. GL posted cover letters and recruitment adverts to all identified patients. SKB, NS and JR collected the data. SKB and NS analysed and conducted the thematic analysis from the data. ME-O'C, SF, DW and RW approved the initial report. SKB and CH wrote the first draft of this manuscript. JSV and all other authors were involved in interpreting the data and made substantial contributions to the intellectual content of the manuscript and approved the final version. The authors took full responsibility for the manuscript. SKB is responsible for the overall content (as guarantor).

Funding The study was sponsored by MediPaCe. Unrestricted funding was provided to MediPaCe by Carl Zeiss Medtech AG. The manufacturers of the Intrabeam device (Carl Zeiss Medtech AG) did not have any part in concept, design, or management of the study, or in data analysis, data interpretation, or writing of the report. A grant/award number was not issued for the funder.

Competing interests This qualitative study was initiated by MediPaCe, a patient engagement and patient research company. The manufacturers of the TARGIT-IORT device (Carl Zeiss Medtech AG) did not have any part in concept, design, or management of the study, or in data analysis, data interpretation, or writing of the report. Authors SKB, NS, CH and JR are employed at MediPaCe. MediPaCe received payment to independently plan, coordinate and conduct this study. JSV declares Support from University College London Hospitals (UCLH)/ UCL Comprehensive Biomedical Research Centre, UCLH Charities, HTA, NIHR, National Institute for Health Research (NIHR) Health Technology Assessment (HTA) programme, Department of Health and Social Care, UK Ninewells Cancer Campaign and Cancer Research Campaign (now Cancer Research UK); Research grant from Photoelectron Corp (1996–1999) and for supporting data management at the University of Dundee (Dundee, UK, 2004–2008) and travel reimbursements and honorariums from Carl Zeiss. SL and GL declare no conflicts of interest.

Patient and public involvement Patients and/or the public were involved in the design, or conduct, or reporting, or dissemination plans of this research. Refer to the Methods section for further details.

Provenance and peer review Not commissioned; externally peer reviewed.

Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.

Read the full text or download the PDF:

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Open access
  • Published: 21 August 2024

Proteomic insights into breast cancer response to brain cell-secreted factors

  • Shreya Ahuja 1 &
  • Iulia M. Lazar 1 , 2 , 3 , 4  

Scientific Reports volume  14 , Article number:  19351 ( 2024 ) Cite this article

322 Accesses

1 Altmetric

Metrics details

  • Breast cancer

The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2 + and TN breast cancers frequently metastasize to the brain and stay potentially dormant for years until favorable conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the early response of HER2 + breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment was simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., endothelial cells, astrocytes, and microglia. Cytokine microarrays were used to investigate the secretome mediators of intercellular communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The cytokines detected in the brain secretomes were supportive of inflammatory conditions, while the SKBR3 cells secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell cultures, indicating that upon exposure the SKBR3 cells may have been deprived of favorable conditions for optimal growth. Altogether, the results suggest that the exposure of SKBR3 cells to the brain cell-secreted factors altered their growth potential and drove them toward a state of quiescence, with broader overall outcomes that affected cellular metabolism, adhesion and immune response processes. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, provide insights into the cellular cross-talk that may lead cancer cells into dormancy, and highlight novel opportunities for the development of metastatic breast cancer therapeutic strategies.

Similar content being viewed by others

breast cancer research and treatment impact factor 2023

Interaction of tumor cells and astrocytes promotes breast cancer brain metastases through TGF-β2/ANGPTL4 axes

breast cancer research and treatment impact factor 2023

Breast cancer cell-secreted miR-199b-5p hijacks neurometabolic coupling to promote brain metastasis

breast cancer research and treatment impact factor 2023

Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis

Introduction.

Brain metastasis accounts for more than half of the intracranial tumors in adults, making it one of the primary causes of brain cancer in patients 1 . Tumors of the lung, breast, and kidney, in addition to melanoma and colorectal cancer, have the highest propensity to spread to the brain, with approximately 80% of cancers metastasizing to the cerebral hemispheres due to favorable blood flow patterns 1 . Moreover, recent statistics indicate that among the breast cancer patient cohort, the triple negative and HER2 + breast cancer subtypes are the most common to metastasize to the brain with a median incidence rate of 31% and 32%, respectively 2 . In order to colonize the brain, the breast cancer cells must breach the blood brain barrier (BBB), invade the neural matrix, and engage in a cross-communication of signaling molecules with the brain cells to evade the eliminatory signals from the host matrix. Crossing the BBB is a challenge in itself, as the endothelial cells of the BBB are linked together by tight junctions that restrict the paracellular movement of most large molecules and cells. Once the BBB has been breached, the tumor cells attach themselves to the brain microvasculature, a necessary process for extracting nourishment and proliferation cues 3 . In this stage, the cancer cells can stay dormant for years 4 . To create favorable growth conditions and further enable successful colonization, the cancer cells engage with the brain cells through direct physical interactions or via paracrine signaling. The neurons and astrocytes produce large amounts of extracellular matrix which accounts for ~ 20% of the CNS volume, alongside a broad spectrum of neural stimulants like trophic factors, neurotransmitters, cytokines, etc., that render a fertile ground for tumor outgrowth 5 . For example, the gap junction connexin Cx43 that interconnects the network of astrocytes has been found to also instigate cell survival and proliferative signaling in cancer cells 6 . Nevertheless, the brain also contains its own population of macrophages, the microglia, that survey the CNS for pathogenic insults and initiate immune responses. In this unfamiliar environment, the metastatic cancer cells face unique challenges, and, in response, express anti-inflammatory mediators that can suppress the immune activation of microglia and reprogram them into a tumor-supportive type. Several studies have underscored the complex interactions that occur between tumor cells and their microenvironment, and that lead to the activation of processes and pathways that favor the development of metastases 7 , 8 . Very little is known, however, about the early molecular responses which are activated in cancer cells when they encounter the foreign neural niche. To gain insights into this process, the objective of this study was to explore the response of SKBR3/HER2 + breast cancer cells to factors expressed by three main cell types present in the neurovascular unit, i.e., microglia, astrocytes, and brain endothelial cells, with which the cancer cells come in contact shortly after entering the brain parenchyma. The selection of cells was driven by their most relevant functions in the brain, i.e.: (a) endothelial cells, due to their role in angiogenic processes that provide nutrients to cancer cells; (b) microglia, due to their role as “immune barrier” cells essential to triggering immune responses; and (c) astrocytes, due to their role in maintaining homeostasis and providing biochemical support to brain cells such as neurons and endothelial BBB cells.

The “neural niche” conditions were replicated by treating the cancer cells with conditioned media (CM) collected from in vitro cultured cells that contain secreted factors such as proteins, cytokines, growth factors, as well as extracellular vesicles (EVs) released by the cultured cells 9 . These components provide the ground for establishing the intercellular communication and promote processes that can accelerate the malignant growth of cells. In one such study, for example, the authors reported the presence of angiogenic factors FGF2 and VEGF in EVs shed by astrocytes in culture 10 . Astrocytes have been also found to interact with the invading tumor cells in early stages of brain metastases by secreting factors such as MMP9 and SDF1 that in turn support cancer progression 11 . Another group demonstrated that the astrocyte-conditioned medium contained exosomes with high levels of microRNAs which inhibit the expression of tumor suppressor PTEN in metastatic breast cancer cells, leading to aggressive tumor formation in the brain 12 . Together with astrocytes, microglia too secrete a variety of cytokines and factors that contribute to immunosuppression, increased invasiveness, angiogenesis, and cancer cell proliferation 13 , 14 . Furthermore, microglial EVs have been found to induce survival and provide metabolic support to other cells 15 . Owing to their role in promoting neurogenesis, maintenance of neuronal circuitry and blood brain barrier functions, microglia and astrocytes secrete neurotrophic factors to carry out neuroprotective and developmental functions in the brain 16 , 17 , 18 . Previous studies have highlighted the brain-derived neurotrophic factor (BDNF) released by astrocytes, which activates the tropomyosin-related kinase B (TrkB) receptor on metastasized breast cancer cells to promote cell proliferation and colonization of the brain 19 . Therefore, we hypothesized that the factors that are present in the CM collected from brain cells would alter the proliferative capacity of breast cancer cells and induce changes in the proteome that are suggestive of increased metastatic potential.

Despite the fact that the SKBR3 cells were not harvested from a brain-metastatic site, there are several features that made this cell line attractive for this study. It is a HER2 + cell line, and HER2 + breast cancers metastasize frequently to the brain. Previous studies in mice that used SKBR3/HER2 + cells as a brain metastatic model were able to demonstrate that metabolic diversity and plasticity can determine metastatic fitness 20 (doi.org/ https://doi.org/10.1016/j.cmet.2021.12.001 ). SKBR3 is also a useful preclinical model system to screen for the effectiveness of therapeutic agents that target HER2 cancers and to investigate resistance mechanisms that evolve in response to HER2-targeted therapies 21 . Last, in contrast to other brain metastasized breast cancers, the SKBR3 cells were not exposed prior to brain factors, and we speculated that the observable impact would be highest in this case.

To test our aforementioned hypothesis, we first explored the cytokine and growth factor profile of the conditioned medium collected from the three brain cell cultures by using a panel of capture antibodies spotted on a membrane-based cytokine array. Next, we used high-resolution mass spectrometry (MS)-based proteomics to measure the change in protein abundances between the CM-treated and non-treated SKBR3 cells. Finally, we mapped these changes to biological processes that are reflective of cancer growth and progression. This study provides preliminary, yet novel, insights into the early response of breast cancer cells exposed to in vitro conditions that mimic the neural niche. Our results suggest that the exposure of SKBR3 cells to neural niche factors may usher the cells into a more advanced state of quiescence, which is a possible result of inflammatory mediators and reduced levels of certain growth factors in the CM from brain cells.

Experimental methods

Human HER2 + breast cancer cells (SKBR3), microglia (HMC3), brain endothelial cells (HBEC5i), and penicillin–streptomycin (PenStrep) solution were purchased from ATCC (Manassas, VA, USA). Normal human astrocytes (NHA), Clonetics AGM™ Astrocyte Growth Medium BulletKit™ (comprised of ABM-astrocyte basal medium and SingleQuots™ growth factors, cytokines, and supplements pack), and astrocyte ReagentPack™ subculture reagents were procured from Lonza (Morristown, NJ). Minimum essential medium (MEM), McCoy’s 5A (Modified) medium, DMEM/F-12, DMEM high glucose (HG), Dulbecco’s phosphate buffered saline (DPBS) and trypsin–EDTA were obtained from Gibco (Gaithersburg, MD). BenchMark Fetal Bovine Serum (FBS) was purchased from Gemini Bio Products (West Sacramento, CA). Endothelial cell growth supplements (ECGS) and all commonly used reagents (Na 2 VO 3 , NaF, DTT, NH 4 HCO 3 , TFA, urea) were from Sigma Aldrich (St. Louis, MO). Acetonitrile, HPLC grade, was from Fischer Scientific (Fair Lawn, NJ), and high purity water was prepared from DI water, in-house, by distillation.

Cell culture

The cells were initially propagated in the manufacturer’s suggested growth media, i.e., SKBR3 in McCoy 5A, HMC3 in EMEM, HBEC5i in DMEM/F-12/ECGS, and NHA in ABM with SingleQuots growth supplements (FBS, EGF, insulin, L-glutamine, ascorbic acid, and gentamycin). After the third passage, 50% of the culture media content was replaced with DMEM-HG, and after yet another 48 h the cell cultures were continued in 100% DMEM-HG ( Fig.  1 A ) . All culture media were supplemented with FBS (10%) and PenStrep (0.5%) at all times, the astrocytes with the additional growth supplements, and the endothelial cells with ECGS. At ~ 80–100% confluence, the cell cultures were serum-starved for 24 h by growing them in DMEM-HG/PenStrep. Serum-free conditioned media (CM) collected from HMC3, HBEC5i and NHA were mixed and spun down by centrifugation at 500 × g for 5 min to remove the floating cells. Clarified CM was added to 24 h serum-starved SKBR3 cells which were grown for another 24 h in the presence of the secreted factors. The SKBR3 cells that were used as control were left under serum-starved conditions for a total of 48 h. Morphological differences between treated and non-treated SKBR3 cells were visualized by using phase-contrast and immunostaining microscopy and a Nikon Eclipse TE2000 U epi-fluorescence inverted microscope (Nikon Instruments Inc., Melville, NY) equipped with the NIS-Elements Advanced Research imaging platform. Mouse monoclonal anti-HER2 primary antibody (Santa Cruz sc-08) and mouse IgG-kappa binding proteins conjugated to CruzFluor488, as well as all Western blot antibodies, were purchased from Santa Cruz Biotechnology (Dallas, TX). The cells were harvested by trypsinization and spun down to pellets which were flash frozen at − 80 °C until further use. DNA content analysis for cell cycle stage assessment was performed by FACS using a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA). Three independent biological replicates of control and treated cells were performed [control: 69–72% in G1 (CV = 9%), 8–13% in S, 2–7% in G2; treatment: 70–90% in G1 (CV = 13%), 11–14% in S and 0.3–4% in G2].

figure 1

Overview of the experimental setup. ( A ) Schematic outline of the cell culture. Three biological replicates were established for HMC3, HBEC5i and NHA. All cells were grown in DMEM-HG medium under serum-deprived conditions for 24 h after which the media was collected and pooled from the three brain cell types to treat the SKBR3 cells for another 24 h. SKBR3-control cells were maintained in serum-free media for 48 h. ( B ) Phase contrast microscopy image of non-treated SKBR3 cells under 20X magnification. ( C ) Immunofluorescence microscopy of non-treated SKBR3 cells labeled with anti-HER2 antibody and detected using an anti-mouse IgG secondary antibody conjugated to CruzFluor488. ( D ) Phase contrast microscopy image of CM-treated SKBR3 cells under 20X magnification shows development of filopodia/lamellipodia type protrusions from the surface of the cells. ( E ) Immunofluorescence microscopy of treated SKBR3 cells labeled with anti-HER2 antibody and detected using an anti-mouse IgG secondary antibody conjugated to CruzFluor488.

Cytokine array analysis

Protein profiling of the secreted factors from each cell culture was performed using the Proteome Profiler Human XL Cytokine array kit (R&D systems, Minneapolis, MN) according to the manufacturer’s recommended protocol. CM from HMC3, NHA and HBEC5i, at 90% confluence, was collected after 24 h of serum starvation. CM from SKBR3 cells was collected after 24 h arrest + 24 h of stimulation with CM from brain cells (treatment), and after 48 h of serum starvation alone (control). After collection, the conditioned media were centrifuged at 500 × g for removal of floating cells, and 2 mL of the supernatant solution was added to the blocked membrane arrays and left to incubate overnight at 4 °C. After rinsing three times with wash buffer, the membranes were incubated with the antibody cocktail for 1 h at room temperature followed by a 30 min incubation with Streptavidin-HRP solution. The membranes were once again rinsed with the wash buffer and the signal was developed using the ChemiReagent mix provided in the kit. The chemiluminescent signal was captured using a ChemiDoc™ Imaging System (BioRad, Hercules, CA) and the raw images were analyzed for pixel intensities using the EMPIRIA Studio software ( https://www.licor.com/bio/empiria-studio/ ). Background subtraction was performed by the software’s adaptive background subtraction method to minimize variability and improve the consistency of data from each array. The signal intensities from the duplicate cytokine spots on each array were averaged and normalized to the average of reference spots from all arrays that were exposed to the secretome of serum-starved cells. Two independent microarray measurements were acquired from two biological replicates of each cell line and further averaged for measuring differences in protein abundance. Only spots with averaged normalized pixel intensities > 2000 were used for comparative purposes.

Cellular fractionation into nuclear and cytoplasmic proteins

The cells were separated into nuclear and cytoplasmic fractions prior to MS analysis 22 , 23 . The isolation of nuclear proteins was achieved with the CelLytic™ NuCLEAR™ extraction kit (Sigma Aldrich, St. Louis, MO) using the manufacturer’s recommended protocol. In brief, the cells were disrupted under ice-cold conditions with a hypotonic buffer in the presence of protease inhibitors (Sigma product P8340), Na 2 VO 3 /NaF phosphatase inhibitors (1 mM), DTT (1 mM), and Igepal. The lysed cells were centrifuged at 4 °C for 1 min at 15,000 × g. The clarified supernatant containing the cytoplasmic components was set aside until further use. The nuclear pellet was further disrupted with a high-salt containing buffer, supplemented with the same inhibitors, by applying constant agitation using a vortex mixer for 30 min at 4 °C. The lysate was sonicated and centrifuged at 4 °C for 10 min at 15,000 × g to generate the nuclear protein fraction. The total protein concentration in the cell extracts was measured using the Bradford assay.

Protein digestion and sample preparation for MS analysis

For MS analysis, the protein extracts were denatured for 1 h at 57 °C, pH ~ 8, in the presence of urea (8 M) and DTT (5 mM). The samples were diluted tenfold with 50 mM NH 4 HCO 3 prior to tryptic digestion. In-solution digestion was performed overnight on a shaker set at 37 °C using sequencing grade trypsin (Promega, Madison, WI) at ~ 1:25 w/w enzyme/protein ratio. The reaction was quenched with TFA and the peptides were isolated and desalted using reverse phase C18 and SCX cartridges (Agilent technologies, Santa Clara, CA). Tryptic peptide mixtures were evaporated with speed vacuum and dissolved in 2% acetonitrile solution acidified with 1% TFA at a final concentration of 2 μg/uL.

LC–MS analysis

The peptide samples (1–1.5 μL) were analyzed with a Q-Exactive hybrid quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific) equipped with a nano-electrospray source operating at 2 kV at a capillary temperature of 320 °C and coupled to an EASY-nLC 1200 UHPLC system (Thermo Fisher Scientific) 23 . The nano-LC separation was performed on a 75 μm i.d. reverse-phase EASY-Spray column ES802A (250 mm long) packed with 2 μm C18/silica particles (Thermo Fisher Scientific). Column equilibration was performed with buffer A (H 2 O 96%, CH 3 CN 4%, TFA 0.01%) and the peptides were eluted in a 2 h long gradient of buffer B (H 2 O 10%, CH 3 CN 90%, TFA 0.01%) at a 250 nL/min flow rate. The MS was operated in a data-dependent acquisition mode with a full-scan range of 400–1600 m/z, resolution 70,000, AGC target 3E6, and maximum IT 100 ms. Tandem MS/HCD spectral acquisition was performed with a quadrupole isolation width of 2.4 m/z, 30% normalized collision energy, resolution of 17,500, AGC target 1E5, maximum IT 50 ms, loop count of 20, and dynamic exclusion 10 s. To improve the consistency and reproducibility of MS measurements, each sample was injected three times. Relevant quantitative results were validated via parallel reaction monitoring (PRM) accomplished with quadrupole isolation width of 2 m/z, resolution of 35,000, AGC target (1–2)E5, and maximum IT of 110 ms.

Database searching and protein identifications

Tandem mass spectra raw files were processed with the Proteome Discoverer 2.5 software package (Thermo Fisher Scientific, Waltham, MA), using the Sequest HT search engine, a minimally redundant/reviewed Homo sapiens database (UniProtKB, 2019 download), and two processing workflows that utilized either (a) the Percolator or (b) the Target/Decoy peptide spectrum match (PSM) validator nodes to discriminate between correct and incorrect PSMs and calculate the associated scores and statistical features (e.g., q-values, FDRs). The Sequest search parameters were set for fully tryptic precursor peptides with a maximum of 2 missed cleavages, min of 6 and max of 144 amino acids, 400–5000 m/z, dynamic modifications allowed on Met (+ 15.995 Da/oxidation) and on the protein N-terminal (+ 42.011 Da/acetylation), and precursor mass tolerance of 15 ppm and fragment ion tolerance of 0.02 Da. The Percolator node was run by using a concatenated Target/Decoy strategy and default parameters, i.e., q-value based validation, max delta Cn of 0.05, and strict/relaxed peptide FDRs of 0.01/0.05. Chromatographic peak identification across the different samples was performed by adding the Minora Feature Detector node in the processing workflow. Parameters for this node were set to min trace length of 5, S/N threshold of 1, max deltaRT 0.2, and PSM confidence set to at least high (FDR < 0.01) for feature to ID mapping. The Target/Decoy PSM Validator node also used a concatenated Target/Decoy selection strategy, but the PSM and peptide strict/relaxed FDRs were set to 0.01/0.03. In the consensus workflows, the PSMs with Xcorr < 1 were filtered out, the peptide group modification site probability threshold was 75, only rank 1 peptides were matched to parent proteins, and protein grouping was performed by enabling the strict parsimony principle. The Protein Validator nodes had the confidence thresholds set to FDRs of 0.01/0.05 when using the Percolator, and to 0.01/0.03 when using the Target/Decoy Validator nodes, respectively, in the two processing workflows.

Label-free quantification and statistical analysis

Six datasets, i.e., three biological replicates of the treated and three of the non-treated cells, were considered for quantitation. Nuclear and cytoplasmic datasets were treated separately at the processing stage but combined later to enable a better understanding of the biological context. Protein abundances were calculated based on the summed peptide peak areas assigned to a particular protein, when using the Percolator node, and based on the count of correct PSMs, when using the Target/Decoy node. For peak area measurements, the chromatographic peaks were aligned with the Feature Mapper node, allowing for a max RT shift of 15 min and a peptide mass tolerance of 15 ppm. Global drifts in peptide abundance measurements were adjusted by normalizing the data based on total peptide abundance in a given sample (all peptides were used for normalization). Missing values were handled by enabling the imputation mode in the Precursor Ions Quantifier node with the low abundance resampling parameter (lower 5% of detected values). Log 2 fold changes (FC) of protein abundances in SKBR3 treated vs. non-treated datasets were calculated by using a pairwise approach, i.e. , by calculating the protein ratios as the median of all possible pairwise peptide ratios (unique and razor, excluding modified peptides). Statistically significant changes were derived by applying a t-test (background based), with multiple testing being performed based on the Benjamini–Hochberg correction method. For PSM count-based abundance measurements, the datasets were normalized based on the average of total PSMs identified in each of the six datasets, one PSM was added to each protein to account for the missing values, and a t-test was performed for assessing significance. For both scenarios, only proteins that were matched by at least two unique peptide sequences, and for which the Log 2 (FC) was either ≥ 1 or ≤ (− 1) with p -value < 0.05, were included in downstream analysis for biological interpretation. The PRM data were analyzed with Skyline ( https://skyline.ms ) 24 , using mass spectral libraries created from the SKBR3 MS raw files. Peptides having a minimum of 5 transitions with a dot product ( dotp ) score > 0.9 were considered for validation.

Biological data interpretation

The biological relevance and visualization of proteins that displayed statistically significant differences in peak areas or PSM counts was performed by using publicly available bioinformatics tools provided by STRING, GeneCards, and Cytoscape. GO biological process enrichment was explored with the STRING web interface. Biological schematics were created with tools provided by Biorender.com.

To explore the early response of HER2 + breast cancer cells to the biological effects of the brain microenvironment, the SKBR3 cells were cultured using serum-deprived conditions in the presence (T-treated) and absence (C-control) of conditioned medium combined from serum-deprived astrocytes (NHA), microglia (HMC3) and brain endothelial (HBEC5i) cells ( Fig.  1 A ) . It was anticipated that the combined CM from all three brain cells would mimic the microenvironment of the metastasized tumor cells in the perivascular niche. All brain cells were cultured to high confluence to ensure the secretion of factors in sufficient concentration for the stimulation experiments. The study was conducted in the absence of fetal bovine serum for multiple reasons. First, we intended to explore the behavior of cancer cells under conditions that induce cell cycle arrest—a state which is believed to be a major reason for metastatic latency of brain metastasized breast cancer cells, relapse being enabled by re-entering of the cancer cells in the cell cycle 25 , 26 . Under such conditions, it has been shown that adherence to endothelial cells is critical for the survival of single cancer cells that extravasate in the perivascular niche and encounter a foreign environment deprived of typical nutrients present in the blood stream. It has been also shown that after therapy, metastatic growth evolves from preexisting dormant disseminated cancer cells. Second, we intended to avoid the interference of the broad range of components of undefined concentration present in the serum (e.g., growth factors, hormones, enzymes, proteins, carbohydrates, etc.), as well as the variable batch-to-batch composition that can affect the reproducibility of findings 27 . The SKBR3 cells were depleted of serum for 24 h prior to, not just during, treatment to ensure that the impact of the treatment on SKBR3 cells stems only from the neural cell CM and not prior FBS supplementation. The control (C) and treated (T) nuclear (N) and cytoplasmic (C) cellular subfractions were annotated as follows: CN1–CN3 nuclear fractions of control cells, CC1–CC3 cytoplasmic fractions of control cells, TN1–TN3 nuclear fractions of treated cells, and TC1–TC3 cytoplasmic fractions of treated cells.

Morphological characteristics of CM-treated SKBR3 cells

Under basal conditions, the SKBR3 cells displayed a grape-like morphology with a lower intercellular adherence (Fig.  1 B), as it has been confirmed by previous studies 28 . The cells stained strongly for the ERBB2 receptor which is known to be present in high abundance on the surface of the cells (Fig.  1 C). Upon treatment with the CM, we observed the development of protruding structures resembling lamellipodia and filopodia emanating from the surface of cells, which could be also visualized by ERBB2 staining (Fig.  1 D,E). DNA content analysis by flow cytometry revealed small but consistent differences, the treated SKBR3 cells displaying a larger proportion of cells in the G1 stage of the cell cycle in comparison to the controls [83% (CV 13%) treatment vs. 74% (CV 9%) control], suggesting an inhibition of growth upon treatment with the CM. Nonetheless, the proteomic data (see further) revealed a more complex response to the CM treatment that branched out into multiple biological processes beyond cell cycle arrest such as metabolism, adhesion, and immune response.

Analysis of conditioned media using cytokine arrays

Membrane-based cytokine arrays were used to measure the relative concentration of 105 different components in the CM of serum-deprived brain and cancer cells and assess the factors relevant to intercellular communication that may affect the behavior of cells (Fig.  2 ). Detailed results of the replicate cytokine measurements including images and raw and normalized pixel intensities are provided in Supplemental file 1 . The functional impact of components that displayed noticeable pixel intensities (> ~ 2000) was evaluated. A set of ~ 50 proteins of interest emerged from the analysis: chemokines and interleukins, growth factors, and various other factors with roles in growth/development, angiogenesis, adhesion, migration, and immune response. The group of proteins with increased abundance in at least one of the brain cell secretomes included pro-inflammatory chemokines (CCL2, CCL5, CCL7/MCP3, CXCL1, CXCL5, CXCL8/IL8, CXCL10, CXCL11), homeostatic chemokines (CXCL12/SDF1), pro- and inflammatory cytokines and factors (IL6, IL15, TNFSF13B, FLT3LG, RBP4, CD14, CSF1/2), pro-inflammatory inhibitors and homeostatic cytokines (IL18BP, PTX3), growth and cell activator factors (BDNF, IGFBP2/3, PDGFA, GDF15, TNFSF13B, ANGPT1/ANGPT2, CSF1/2, CHI3L1, SPP1, DKK1), and cell adhesion and extracellular matrix (ECM) remodeling molecules (ICAM1, VCAM1, MMP9, THBS1, SERPINE1, PLAUR) (Fig.  2 A–C,F).

figure 2

Results from the cytokine microarray profiling of conditioned medium collected from serum-deprived cell cultures of ( A ) HBEC5i, ( B ) HMC3, ( C ) NHA, ( D ) SKBR3-control, and ( E ) SKBR3-stimulated cells. ( F ) Bar chart showing the average pixel intensities of the set of cytokines identified in the CM collected from the serum-starved brain and SKBR3-control cells (pixel intensities > 2000). Color code: blue-HBEC5i, green-HMC3, yellow-NHA, orange-SKBR3.

The group of proteins with increased abundance in the SKBR3 secretome (not necessarily unique to SKBR3) included cytokines (pro-inflammatory MIF, CCL20, IL17A, and anti-inflammatory TFF3) and several groups of factors and receptors with relevance to growth and development (PDGFB, FGF19, TFRC), ECM remodeling, and invasiveness (LCN2, BSG) (Fig.  2 D, F). Angiogenesis factors such as VEGFA, ANG and ENG were secreted by all cells, with ENG being more abundant in the HBEC5i and SKBR3 secretomes. A schematic representation of a subgroup of cytokines that provided visually discernable spots (pixel intensity > 7000), and of which some displayed discrepancies in abundance in the cell secretomes, is provided in a node/edge networked configuration in Fig.  3 A with edges indicating detectability in either of the four cell lines. A cutoff of ~ threefold change (FC) in normalized pixel intensities in the group of HBEC5i/HMC3/NHA brain cells and SKBR3 was used to indicate unique association with a particular cell line.

figure 3

Inflammatory cytokines

With few exceptions, all pro-inflammatory cytokines with elevated abundance in the brain cell secretomes were secreted at the basal level by the HBEC5i cells, with only a few being secreted in similar abundance by the HMC3 (CCL2, CCL5, CXCL8/IL-8, PTX3, RBP4, FLT3LG, CD14) and NHA (CCL7, CXCL8) cells. Owing to their prime location in the bloodstream, the endothelial cells are the first to come in direct contact with any infectious entity in the body. Therefore, these cells advanced mechanisms for the recognition of damage-associated molecular patterns (DAMPs) for inciting an immune response and for recruiting immune cells to the site of pathogenesis 29 . Many of these functions are facilitated by the inflammatory cytokines and chemokines that are released by the endothelial cells in their microenvironment. Elevated levels of TNFSF13B, a cytokine involved in the regulation of immune responses and stimulation of B-/T-cells, were observed only in the HBEC5i cell secretome. IL6 was also preponderantly expressed in the HBEC5i secretome. Along with its well-known inflammatory functions, this cytokine has been implicated in regulating the proliferation, angiogenesis, invasion, and metabolism of cancer cells. IL6 mediates these outcomes primarily via the STAT3 or NFKB signaling axis, and induces the expression of factors which promote enhanced malignancy (GM-CSF, CCL2, MMP, VEGF) 30 . Previous studies have confirmed a basal level expression of CCL2 and CXCL8/IL8 by the human brain endothelial cells, however, CXCL10 and CCL5 were shown to be expressed only upon pro-inflammatory cytokine stimulation 31 . The levels of CCL2 were relatively constant in all cell secretomes, but there is accumulating evidence that CCL2 induces angiogenesis via increased VEGFA expression, while CXCL5, CXCL8/IL8 and CXCL1 directly affect tumor growth and metastasis by enhancing blood vessel supply through neovascularization 32 . On the other hand, CXCL10 and CXCL11 exert angiostatic effects and initiate T-cell or NK-cell mediated immune responses 32 . Previous studies suggested that CCL7 could be involved in promoting tumor invasion and metastasis, but tumor suppressor effects of this cytokine have been also identified 33 . The detection of CCL7 in this work was, however, just above the intensity threshold setting in HBEC5i and NHA cells. IL15 was also observable in low abundance, and only in the secretome of HBEC5i cells. By stimulating the proliferation and activation of immune cells (NK-, B- and T-cells), IL15 has many protective and anti-tumor roles. It is explored as a potential therapeutic agent by itself or as a target for the development of IL15 agonists and a number of immunotherapies 34 . IL18BPa (IL18 binding protein) which is an inhibitor of the proinflammatory IL18 cytokine was upregulated in HBEC5i, while the homeostatic/tissue remodeling PTX3 (Pentraxin-3) in the HMC3 and HBEC5i secretomes. IL18BPa is believed to play a buffering role for IL18 35 , and PTX3 to exert multiple tumor suppressive and supportive roles via complex involvement in mediating immune responses, ECM remodeling and angiogenic programs 36 , 37 . CXCL12 (SDF1α) was elevated in the HBEC5i secretome relative to SKBR3, and while this is a homeostatic chemokine, it is known to exert various roles in pathogenic conditions 38 , 39 . Different splice variants have specific activities, which are also controlled by PTMs 38 , and hypoxia and growth arrest in different cell types can elevate the expression level of this cytokine 38 . Increased CXCL12 levels produced under hypoxia by ovarian cancers have been shown to promote the expansion of endothelial cells and angiogenesis 40 . Altogether, aberrant expression of CXCL12 in the tumor microenvironment was correlated with tumor growth, proliferation, inhibition of apoptosis, and driving cancer cell migration to distant sites 39 . The cytokine signals along the MAPK, PI3K/AKT, Wnt and NFKB pathways to induce the expression of VEGF, FGF, cyclooxygenase-2 and IL6, all of which are critical mediators of angiogenesis 39 . CXCL12 was also observed in the NHA medium. Additional pro-inflammatory proteins such as the Fms-Related Tyrosine Kinase 3 Ligand and growth factor (FLT3LG), retinol binding protein 4 (RBP4), and receptor CD14, albeit in lower abundance than the other cytokines, complemented the ability of HBEC5i cells to stimulate the proliferation or activity of immune cells 41 . In addition, the cell adhesion glycoproteins VCAM1 and ICAM1 with roles in mediating cell adhesion, motility and maintaining tissue architecture, were also predominantly expressed in the HBEC5i secretomes, especially VCAM1 (ICAM1 was observable in low abundance). VCAM1 is overexpressed on endothelial cells under inflammatory conditions to allow for the adhesion and rolling of immune cells on the endothelium surface 42 . It has particular relevance during cancer metastasis, as it is one of the molecules that tumor cells bind for transmigrating across the endothelial barrier 42 .

In microglia, the presence of IL6, CXCL8/IL8, CCL5, PTX3 was consistent with the role of these cytokines in the regulation of leukocyte activation and chemotaxis that is invoked in the brain in the case of neuropathological and inflammatory conditions, and the detection of PTX3 and FLT3LG with the involvement of microglia in mediating inflammation and phagocytosis, respectively 43 . Likewise, the secretion of CCL7 by NHA cells was associated with promoting microglia-mediated inflammation after brain injury 44 .

In the SKBR3 secretome, pro-inflammatory cytokines were present, but generally in low abundance (excepting IL17A and MIF which were also present in the brain cell secretomes) or had mostly tumor-promoting functions (CCL20). Cytokines such as IL11, IL22, RETN and TNF which are associated with inflammatory responses were observable at very low intensities, but their emerging role in promoting the progression of cancer has been recognized 45 , 46 , 47 , 48 . The other cytokines have been shown to promote cell renewal, tissue regeneration, recruitment of immune cells and activation of pro-survival and mitogenic signaling (IL17A 49 ); to induce cell invasiveness, angiogenesis and cell survival pathways (macrophage migration inhibitory factor (MIF 50 ); or, to stimulate invasiveness via MMP secretion and impart chemotherapeutic resistance (chemokine CCL20 51 ).

Growth factors

We further assessed the differences in the concentration of growth and stimulating factors between the SKBR3-control and the brain cells (Fig.  2 A–D) 41 , 52 , 53 , 54 , 55 . The analysis revealed that the level of many pro-tumor factors, especially of those that are implicated in growth/proliferation, angiogenesis, migration, and metastatic progression was either similar or higher in the SKBR3 than in the brain cell secretomes (PDGFB, FGF19, GDF15, MIF, LCN2, TFF3, VEGFA, ANG, ENG, ANGPT2, TFRC, SERPINE1, PLAUR, BSG) (Figs.  2 F, 3 A). Specific biological processes sustained by these proteins included regulation of ERK1/ERK2 and MAPK signaling cascades, cell adhesion, cell migration and invasion, blood vessel development and angiogenesis, and response to stress/defense and inflammation. Selected GO biological processes represented by minimum 5 proteins from either of the cell secretomes, with a fold-enrichment of > 4, are provided in Fig.  3 B. The multifunctional platelet derived growth factor subunit B (PDGFB) with roles in cell proliferation/survival and migration, in particular, was highly abundant in SKBR3 and essentially missing from all brain cells. GDF15 was expressed by all cells, and increased levels have been shown to be associated with advanced cancer and poor patient outcomes, as this factor promotes mechanisms of immune evasion in cancer cells and enhances cell viability and metastasis 52 . Others, however, have suggested a conflicting response to GDF15, when it was observed to induce apoptosis in some cancer cells 52 . Further evidence for the secretion of cancer progression supportive factors in SKBR3 was presented by the higher expression of lipocalin-2 (LCN2) which has important ramifications in EMT-related processes, invasion, and cell migration 53 and of the anti-inflammatory trefoil factor 3 (TFF3) which supports invasion and metastasis in several carcinomas 54 .

All three brain cell types secreted some if not all of the above cancer-supportive factors. In a broader context, these proteins are involved in developmental processes, and regulate cell proliferation, mitogenesis, survival, tissue repair, and chemotaxis (Fig.  3 B). A few factors that were identifiable in SKBR3 were present in higher abundance in the brain cell secretomes (IGFBP2/3, PDGFA, SPP1, CSF1/2), some being particularly elevated in NHA (IGFBP2, ANGPT1, CHI3L1, SPP1) or HMC3 (PDGFA, BDNF) (Fig.  3 A). NHA exclusively expressed the angiopoietin (ANGPT1) and chitinase 3 like 1 (CHI3L1) proteins. Together with VEGFA, angiopoietins secreted by astrocytes are important for governing the angiogenic remodeling processes for the development of vasculature in the brain 56 . Increased levels of CHI3L1 have been associated with neurodegenerative and inflammatory brain diseases, but aberrant expression has been also observed in brain tumors and metastasis. It has been suggested that CHI3L1 expression by activated astrocytes may play a role in tumor progression, angiogenesis, immune escape, and resistance to therapeutic drugs 57 . It is also worth highlighting osteopontin (SPP1) which plays a crucial role in driving metastasis, invasion, angiogenesis, chemotaxis, and suppression of anti-tumor immune responses 58 . SPP1 is known to be secreted by both cancer cells and the cells of the TME, and to have an overall positive impact on the metastatic progression of cancer 58 . HMC3 revealed the expression of the BDNF involved in the development and survival of CNS cells. Interestingly, BDNF activates TrkB (Tropomyosin-Related Kinase B or Neurotrophic Receptor Tyrosine Kinase 2) and HER2 receptors and enhances the proliferation and survival of brain metastatic HER2 + breast cancer cells 59 . On the other hand, while the exact function of PDGFA in microglia is not fully understood, this growth factor is recognized for its important contributions to supporting cell proliferation, angiogenesis, migration, as well as chemotaxis 41 . The matrix remodeling proteins and enzymes (SERPINE1, PLAUR, PTX3, LCN2, BSG, THBS1, MMP9), together with the CAMs (ICAM, VCAM1) that were overexpressed in the HBEC5i secretome, formed a group that plays key roles in mediating cell–cell/cell–matrix interactions and ECM remodeling processes that ultimately affect, again, the adhesion, migration, and the invasion capabilities of cancer cells.

The culture medium collected from the SKBR3 cells grown in brain cell-conditioned media generated a signal for almost all factors that produced intense spots in the brain and SKBR3-control secretions (Fig.  2 E). It was therefore difficult to conclusively determine which cytokines and biological processes were upregulated in the SKBR3 secretions after treatment with conditioned media from the brain cells.

Proteomic analysis and quantification of the CM-treated versus non-treated SKBR3 cells

The impact of treating the SKBR3 cells with the brain cell-conditioned media was next evaluated quantitatively at the proteome level. The expected outcome was the identification of novel proteins that mediate an early response in SKBR3 cells to the factors that they encounter in a foreign microenvironment (i.e., the brain TME). Three biological replicates of SKBR3-treated (T) cells were compared to three biological replicates of SKBR3-control (C) cells. Two complementary approaches were used for assessing protein differential expression, i.e., based on LC peak area and PSM count measurements. The two approaches produced rather complementary results due to: (a) the actual use of two distinct methods for assessing protein abundance; (b) the use of two distinct approaches to validate the PSMs, with the area-based measurements being enabled by a workflow that included the Percolator node that uses a semi-supervised machine learning approach to distinguish the correct from the incorrect PSMs, in contrast to the PSM count measurements that relied on using the results from the Target and Decoy database searches; and (c) the possible software-related misrepresentation of area measurements because some chromatographic peaks did not have actual PSMs associated with them in all treatment and control samples. The complementarity of such results is not surprising when only few proteins change abundance or the changes in abundance are small, and it was noted in previous studies, as well 60 . Longer exposure of the SKBR3 cells (> 24 h) to the brain CM could have amplified the changes in the proteome profiles, however, the prolonged lack of serum from the culture medium would have biased the results by inducing an excessive stress response.

On the average, ~ 3200/4700 proteins per cell fraction were identified in the SKBR3 cells when using the Percolator and Target/Decoy validator nodes, respectively, with over 70% of proteins being identified with two or more unique peptides (Supplemental file 2 ). The reproducibility of protein identifications in biological replicates for the workflow that used the Percolator node is illustrated in Fig.  4 (# protein IDs, overlaps, PSM correlations), and the results of the quantitative comparisons in Fig.  5 A–E,G [box and whisker plots of raw and normalized Log10(Protein Abundance) and Volcano plots] and Supplemental file 3 . There were no major overlaps between the proteins that displayed differences in counts in the nuclear and cytoplasmic fractions, demonstrating the complementarity of the data (Fig.  5 F and H). Taken altogether, the area and PSM-based upregulated datasets comprised 96/91 proteins, respectively, and the downregulated ones 256/46 proteins, combined from the analysis of both nuclear and cytoplasmic samples. A few representative proteins are illustrated in Fig.  6 A. Selected proteins were validated using PRM-MS analysis and a few others by Western blotting (Supplemental file 4 ).

figure 4

Evaluation of reproducibility in protein identifications. ( A ) Stacked bar chart showing the percentage of proteins identified by 2 or more unique peptides in a cell state and fraction (~ 70%, bars shaded in gray). The white boxes in the center display the total number of identified proteins in each cell state and fraction. ( B – E ) Venn diagrams displaying the overlap between the proteins identified with high/medium FDR and ≥ 2 unique peptides in three biological replicates of various SKBR3 cell fractions: ( B ) CN, ( C ) CC, ( D ) TN, and ( E ) TC. ( F – I ) Scatterplots displaying the degree of correlation between the PSMs of two biological replicates for ( F ) CN, ( G ) CC, ( H ) TN, and ( I ) TC datasets. Heatmaps on the right show the Pearson correlation coefficients for every comparison.

figure 5

Overview of the proteomic label-free differential expression analysis approach. ( A – D ) Boxplots of area-based protein abundances for three biological and three technical replicates of SKBR3 cell fractions: ( A ) before and ( B ) after normalization for the quantitative analysis of TC vs CC fractions; ( C ) before and ( D ) after normalization for the quantitative analysis of TN vs CN fractions. ( E , G ) Volcano plots illustrating the results of differential expression analysis for proteins that displayed minimum 2-FC up- (red) or downregulation (blue) with a p-value ≤ 0.05 in TN vs CN and TC vs CC: ( E ) area-based measurements, and ( G ) PSM count-based measurements. ( F, H ) Venn diagrams showing the overlap between the proteins in the N and C fractions that were either up- or down-regulated in the area-based and PSM count-based measurements.

figure 6

GO Biological processes represented by the proteins that changed abundance in the proteomic profiling of SKBR3 cells treated with CM medium from brain cells. ( A ) Box-whisker plots displaying the relative spectral counts for the selected proteins identified in the control and treated proteomic datasets. ( B ) Enriched biological processes represented by the combined nuclear/cytoplasmic proteins detected by the area and PSM-count measurements with increased or decreased abundance, respectively, in CM-treated vs non-treated SKBR3 cells. ( C ) Protein counts that emerged from the area and PSM measurements in the CM-treated vs non-treated SKBR3 cells mapped to GO biological processes of relevance to cancer (note: the proteins were mapped to biological processes by using GO controlled vocabulary terms).

Quantification was performed separately for the nuclear and cytoplasmic fractions, however, the results were combined for a thorough understanding of the cellular biology. The short lists of differentially expressed proteins yielded only very few enriched biological functional categories. When combined, however, the nuclear and cytoplasmic results, as assessed either via area or PSM measurements, revealed the main processes that were affected by treating the cells with brain cell conditioned medium. The processes that were sustained by proteins with increased counts were dominated by altered gene expression, chromosome organization, trafficking, protein localization, and interferon signaling. The processes that were supported by proteins with decreased counts (represented mostly by the cytoplasmic cluster generated by area measurements) were broadly associated with various metabolic and transport processes, and included an over-representation of cytoplasmic translation and electron transport/mitochondrial respiratory processes (Fig.  6 B and Supplemental file 5 ). Ribosome biogenesis was represented by proteins with both increased and decreased counts. Many of the above processes are mediated via posttranslational activation/deactivation or protein shuttling between various cellular organelles. Therefore, increased or decreased protein abundance measurements may not be reflective of actual protein up- or down-regulation, but rather of changes in PTMs or cellular location. Altogether, however, the results suggest that the treatment of cells with conditioned medium altered the cellular transcriptional/translational machinery with a net outcome of slowed metabolic processes. Ribosome biogenesis, for example, is an energy-intensive process, and a disruption in the availability of nutrients or energy in the cell that can affect any step of the biogenesis process leading to either up- or down-regulated ribosome components, has been shown to result in altered cell cycle progression or even cell death 61 .

It is important to reiterate that upon treatment with the brain cell-conditioned media the SKBR3 cells were exposed to many inflammatory molecules that were either absent or present in low abundance in the secretome of the SKBR3-control cells. In the same time, the SKBR3-control cells secreted larger amounts of several important growth factors that may or may not have been at optimum levels in the brain cell-conditioned media to elicit a signaling response in the SKBR3 cells. Therefore, to derive a better understanding of the processes that were dysregulated in the SKBR3 treated cells, the proteins were further mapped directly to specific GO terms reflective of cancer-supportive processes and metastatic progression (Fig.  6 C). The complex interplay between these proteins in affecting the cell fate is exemplified in a Sankey diagram that presents a selected list of SKBR3 proteins with changed abundance, matched by at least three unique peptides, mapped to such processes (Fig.  7 and Supplemental file 6 ).

figure 7

Sankey diagram illustrating the mapping of dysregulated nuclear/cytoplasmic proteins with increased abundance to key biological processes of relevance to metastatic progression of cancer (note: only proteins identified by at least three unique peptides are shown).

Based on our previous work on proteomic profiling of SKBR3 cells that has identified a broad range of receptors in the cell membrane (EGFR, FGFR, CSFR, IL, CD44/Cd47, IL, PDGFRA, TNFR, VEGFA receptor FLT1, ephrin, integrin, neuropilin NRP1, activin, chemokine ACKR2, adiponectin ADIPOR1, sortilin SORT1, low density lipoprotein LRP, syndecan SDC4) 62 , a depiction of the possible interactions between the SKBR3 breast cancer and the brain cells is provided in Fig.  8 . Relevant SKBR3 receptors, along with the most abundant cytokines and growth factors secreted by the brain cells, are listed in the figure. The observed changes in the SKBR3 proteome were further interpreted based on the consideration that the SKBR3 cells that were exposed to the CM from brain cells experienced an environment deprived of optimal levels of growth factors needed for proliferation, but more abundant in inflammatory molecules. Proteins with similar trends in abundance changes, as emerged from the two quantification approaches, are discussed below.

figure 8

Illustration of receptors and secreted factors that can enable interactions between the SKBR3 breast cancer and brain cells. Relevant SKBR3 receptors were detected by proteomic profiling 58 . Abundant cytokines and growth factors secreted by brain cells, but missing or detected in low abundance in the SKBR3 secretome, are depicted near each cell line. Abundant cytokines and growth factors secreted in common by all brain and/or cancer cells are shown inside the sidebars.

Effect of CM-treatment on cell cycle and proliferation

An interesting behavior of metastatic cancer cells is dormancy, a mechanism through which the metastatic cells temporarily exit the cell cycle to enter a state of quiescence and growth arrest, which allows them to survive the adverse conditions of a foreign niche 63 . Dormancy is activated in cancer cells to cope with environmental stress triggered by growth factor deprivation, oxidative stress, lack of ECM attachment and stromal cell interactions 63 , 64 . Inhibited ERK, autocrine Wnt signaling, and activation of various components of MKK4/p38/JNK, MTOR and hypoxic pathways are reflective of this state. The quiescent cells significantly reduce their metabolic activity including biosynthesis of macromolecules, carbohydrate and lipid metabolism, and mitochondrial respiration 65 . A recent study demonstrated an enrichment of stress response and cell cycle related processes in the transcriptomics data derived from the dormant cancer cells of mouse and human origin 64 . In this work, a number of transcription factors and regulators were affected by the SKBR3 stimulation process with both initiator/activator (GTF2E2, USF1, CDK5RAP3, PML, CEBPB) and repressor roles (GATAD2B, PML, ZBTB7A). In particular, ZBTB7A (zinc finger and BTB domain containing 7A protein) which is a transcriptional repressor of genes implicated in cell cycle progression and known to exert multiple roles in cancer cells that affect NFKB and Notch signaling, glycolysis, OXPHOS, transcription, chromatin organization, and DNA damage 66 , warrants further investigation to provide clarity about its contribution to tumorigenesis. In contrast, CEBPB, a transcription factor involved in the regulation of a number of immune and inflammatory responses (see also later in the text), has been recognized for both its proliferative and anti-proliferative activities 41 . Clarifying the mechanisms of its functionality, however, is challenging because the function of CEBPB in cancer cells is context specific with diverse outcomes in different types of cancers, and because its activity is regulated by a number of different PTMs 67 . While on one hand CEBPB inactivates p53 expression to improve rates of survival and reduce apoptosis in cancer cells, it has also been shown to induce growth arrest in several types of cancers 67 . In this work, CEBPB was measured with increased PSM counts, result that was validated by targeted PRM-MS analysis (Supplemental file 4 ). Several proteins from the dataset also pointed to the alteration of signaling pathways that modulate in concert cell cycle, proliferation, stress and inflammation, such as EGFR (ERBIN—Fig.  6 A), p38/MAPK (TAOK3), PI3K (PI3KR2), DNA damage and repair (RPA2; ASCC—Fig.  6 A; 14-3-3 sigma or stratifin/SFN—Supplemental file 4 ), TGFB (ZBTB7A; SDCBP), NFKB-mediated transcription and host-virus interactions (CDK5RAP3/LZAP), and anti-inflammatory processes (GNG5). The Ser/Thr protein kinase TAOK3 is a regulator and activator of the p38/MAPK14 stress-activated MAPK cascade, with implications in many cellular stress-responses including apoptosis, metabolism, cell cycle, proliferation, cytoskeleton rearrangement, and inflammation 41 , 68 . Alterations in its abundance/activity were confirmed by PRM/MS (Supplemental file 4 ). Recently, it was found to induce drug resistance in breast cancer cells via NFKB activation and pathways that can have diverse outcomes, one of which is blockade of cell cycle progression and development of quiescence or dormancy 69 , 70 . Notable were also the upregulated replication protein A2 (RPA2) that responds to cellular stress by repairing damaged DNA at cell cycle checkpoints 41 , and CDK5RAP3 and PML, due to their involvement in numerous cellular processes such as transcription regulation, DNA repair, growth suppression, apoptosis, and viral defense mechanisms. The last two proteins have been additionally recognized for their potential role in tumor suppression.

Proliferation markers such as KI-67 or PCNA did not display differential expression by MS analysis. This, however, was not unexpected as both cell states were deprived of serum components for 48 h. Under proliferative conditions, in the presence of FBS, the PSMs of KI-67 typically increase ~ 5–tenfold. The mass spectrometry results for a negative regulator of cell cycle, 14-3-3 sigma, indicated changes in the protein abundance, but the changes were small and did not pass all data filtering criteria. Nonetheless, Western blot analysis confirmed an increase in 14-3-3 expression, corroborating the more advanced state of arrest of CM-treated SKBR3 cells (Supplemental file 4 ). Altogether, the findings suggest that the cells demonstrated a more complex response, rather than merely exhibiting a more pronounced cell cycle arrest. The cells possibly initiated adaptive mechanisms to survive the nutrient-limited stress conditions and respond to additional insults inflicted by the brain cell-secreted factors. Cancer cell quiescence has been also linked to increased resistance to apoptosis 71 . The results indicate, however, that although the cells activated biological mechanisms in response to nutritional and inflammatory stress, the cells were not apoptotic.

Effect of CM-treatment on cancer cell metabolism and response to stress

Scarcity of nutrients and unfavorable growth conditions drive cancer cells towards quiescence, which results in reduced metabolic activity, biosynthetic rates and respiration 65 . Recent studies have shown that dormant/quiescent cells switch their carbohydrate metabolism from mitochondrial OXPHOS to fatty acid oxidation, in order to secure a minimal supply of energy and prevent oxidative damage caused by the reactive oxygen species released during mitochondrial respiration 72 , 73 . Several metabolism and oxidative stress associated proteins were downregulated according to the area-based measurements, including the ribosomal subunit proteins involved in translation (RPLs, RPSs), the large neutral amino acid transporter LAT1 (SLC7A5), the mitochondrial OXPHOS/electron transport and ATP production subunit proteins, and various antioxidant proteins (PRDX1). PRM/MS (LAT1, RPL8, PRDX1) and Western blot (LAT1) analysis validated the MS results (Supplemental file 4 ). Ribosome biogenesis is an energy consuming process, necessary for rapid cell growth and division, as a result of which ribosomes have a central role in a number of oncogenic signaling networks 61 , 74 . Under conditions of nutritional stress, cells can curtail the biogenesis of ribosomes to conserve energy, and, consequently, limit the G1/S progression of cells 75 . Reduced levels of solute carriers such as of the nucleotide-sugar transmembrane transporter (SLC35A4), that are implicated in cellular response to stress by regulating translation processes, corroborated the results. Lower expression of LAT1 was also suggestive of impaired proliferation in the CM-treated cells. The LAT1 transporter allows for an increased uptake of essential amino acids from the external microenvironment, in exchange for glutamine, which is pumped out of the cells 76 . LAT1 overexpression was found associated with highly proliferating cancer cells growing under low nutrient conditions and hypoxia, and is being evaluated for its therapeutic target potential 76 . The cluster also included proteins that mediate transport across the mitochondrial membrane, such as the glutamate carrier SLC25A22 and amino acid transporter SFXN1, all of which pointed toward restricted mitochondrial metabolism. These results correlated with the upregulation of the transcriptional repressor of genes involved in OXPHOS and glycolysis (ZBTB7A). Overall, the results indicate that the nutritional stress conditions induced by serum-deprivation and CM-stimulation impeded ribosomal protein synthesis pathways and reduced mitochondrial respiration. Furthermore, the inflammatory conditions mediated by the cytokines and chemokines triggered an overexpression of proteins that respond to these stimuli by regulating changes in cancer cell proliferation, growth and survival.

Effect of CM-treatment on cancer cell adhesion/migration and EMT

Epithelial cells are characterized as having an apical-basal polarity, with junction proteins binding the cells together in a monolayer, attached to the basement membrane 77 . During cancer progression, tumor cells undergo EMT (epithelial to mesenchymal transition) which is marked by the loss of certain adhesion and junction proteins and acquisition of mesenchymal features, such as cytoskeletal rearrangements, that induce invasive and migratory capacities in the cancer cells 77 . The CM treatment of SKBR3 resulted in changes in the expression level of several proteins that have known implications in adhesion, migration and EMT-related processes.

Evidence for EMT type behavior and development of cell motility in the CM-treated SKBR3 cells was provided by the overexpression of syntenin-1 and annexin proteins. Syntenin-1 (SDCBP) activates cancer cell invasion and migration through actin remodeling processes during the late stages of cancer metastasis 78 (Fig.  6 A and Supplemental file 4 ). It colocalizes with growth factor receptors, the focal adhesion kinase complex, and other integrin-associated signal transducers to activate p38MAPK/JNK downstream signaling 78 , 79 . This, ultimately triggers cytoskeleton rearrangement processes, which regulate adhesion and migration in cancer cells 79 . A critical outcome of this pathway is the activation of mesenchymal proteins such as SLUG that induce EMT 79 , 80 . The cytoplasmic fractions of the treated cells displayed an upregulation of annexin A1 (ANXA1), which has the ability to bind the actin cytoskeleton, which in turn is crucial for the initiation of EMT, migration, and invasive properties in cancer cells 81 . The multifunctional ZBTB7A, as noted above, has been shown to also mediate EMT via the NFKB signaling pathway in a number of breast cancer cell lines 82 . As noted earlier, treatment with the CM induced the formation of protruding lamellipodia and filopodia on the surface of SKBR3 cells (Figs.  1 D and 1E). In transmigration assays that exposed the SKBR3 cells to HMC3 secreted factors similar changes in morphology were observed, however, cell migration within 24 h of exposure was very slow, and a Western blot of MIEN1 (Migration and Invasion Enhancer 1) that induces the formation of filopodia 41 did not show a change in the abundance of this protein (Supplemental file 4 ).

A few additional proteins of potential interest included the downregulated YBX1 and LAMP1, and upregulated ICAM1 and S100 Ca-binding proteins. PRM/MS conducted for a few of these proteins, confirmed their change in abundance (Supplemental file 4 ). Notably, the DNA/RNA binding protein YBX1 participates in a range of cellular transcription/translation processes and augments metastatic processes such as invasion and cell migration 83 , development of drug resistance 84 , and elevated glycolysis 85 . Its activity, however, depends on phosphorylation, and further investigation is required to provide clarity on its activation status in the CM-treated SKBR3 cells. YBX1 and the previously discussed TAOK3 are both included in the DRMref database, which provides a comprehensive map of drug resistance mechanisms in human cancer 86 . The lysosome associated membrane glycoprotein LAMP1 provides glycan ligands to selectins and promotes cell–cell adhesion in metastatic cancer cells 87 . ICAM1, an intercellular cell adhesion molecule, is involved in cell–cell and cell-ECM adhesion and was found to be expressed by metastasized cancer cells as they interact with the endothelial cells to undergo trans-endothelial migration. S100 calcium-binding proteins bind to a variety of cell surface receptors on other cells to induce metastatic progression 88 . These proteins carry out, however, a broader range of functions that present relevance to metastatic processes, both in the intra- and extracellular environments. The intracellular functions of S100 proteins include involvement in various aspects of cell cycle regulation, promotion of cell survival and invasion via the NFKB signaling pathway, metabolism, and trafficking. The extracellular functions include involvement in pro-inflammatory, anti-microbial and death-inducing activities 41 . Binding to receptors on macrophages/microglia or endothelial cells facilitates their recruitment and angiogenesis at the site of tumorigenesis 88 . In this study, S100A7 and S100A9 were found upregulated in the CM-treated cells (Fig.  6 A and Supplemental file 4 ), while other isoforms (A4, A6, A13, and A16) appeared to be rather downregulated, pinpointing again to the need for careful interpretation of results and further studies.

Effect of CM-treatment on the mechanisms of immune evasion

Upregulated ISG15 (interferon-induced ubiquitin-like protein) and MX1 (interferon-inducible protein p78 or myxoma resistance protein 1) were two proteins that were potentially reflective of the SKBR3 cell response to exposure to the cytokines from the conditioned medium. ISG15 plays multiple roles in innate immune responses that include antiviral activities, cell–cell signaling, and chemotactic activity 41 . It can act either in free form or by binding to target proteins, including MX1, a protein that impedes viral replication. Previous studies have demonstrated that ISG15 enhances EGFR recycling on the surface of breast cancer cells, as a result of which the cells display an increased membrane localization of EGFR and aberrant tumorigenic signaling 89 . A recent study identified MX1 as a potent inducer of migration, EMT, and invasion in cancer cells due to its association with EGFR signaling 90 . According to the cytokine arrays, a small increase in the abundance of interferon γ in the secretome of CM-treated SKBR3 cells was observable, but further studies would be needed to clarify the complex mechanisms that are responsible for the upregulation of these proteins. In the same time, the lysosomal thiol reductase IFI30 which is involved in MHC/HLA class I/II antigen presentation and which is known to be induced by interferon-γ in non-professional antigen presenting cells, was downregulated, suggesting a loss of HLA presentation. HLA proteins were practically undetectable in the SKBR3 cells, so comparisons between CM-treated and non-treated cells could not be made. The inability to detect HLA class I molecules was, however, not surprising, as cancer cells downregulate the HLA class I presentation machinery to evade detection and destruction by immune cells (T/CD8 cells) 91 . Nonetheless, HLA-E and HLA-C expression was detected in both cell states. HLA-E is a ligand for the CD94/NKG2A inhibitory receptor expressed by NK- and T/CD8-cells, and HLA-C for the NK-cell KIR receptors 92 , 93 . Both proteins were detected with low counts, and changes in their expression were not observed. Complementary cell-membrane protein enrichment protocols could provide additional insights into how the cancer cells evade cytotoxic immune surveillance at a metastatic site.

Conclusions

The findings of this study provide preliminary, yet novel, insights into the early response of breast cancer cells to in vitro conditions that mimic the neural niche. The results led to the hypothesis that unfavorable environmental conditions that support cell cycle arrest, further mediated by inflammatory molecules or suboptimal concentrations of growth factors, drive the cancer cells into quiescence, and possibly dormancy, in the foreign perivascular metastatic niche. The pro-inflammatory chemokines and cytokines secreted by the brain cells have pleiotropic roles with anti-tumor activities and simultaneous functions in facilitating the recruitment of immune cells and promoting neovascularization to enhance blood vessel supply in a hypoxic TME. The pro-tumorigenic growth factors, on the other hand, can counteract the detrimental effects of the pro-inflammatory mediators and promote viability of cancer cells. Altogether, however, the SKBR3 cells secreted cancer-promoting growth factors in higher abundance than the brain cells, and displayed signs of impaired growth when subjected to the brain cell-conditioned medium. Such pilot studies can provide crucial insights into the initial response of cancer cells in a foreign niche, especially as the concept of dormancy is difficult to replicate experimentally under both in vivo and in vitro conditions. Furthermore, such exploratory results can pave the way for future research into critical questions such as how cancer cells adapt and/or mutate to nutritionally deprived conditions, and how the combination of factors in the neural niche protects and later supports the revival of dormant cells. Moreover, the detection of early metastatic markers not just from cancer but the TME will be critical to running accurate diagnostic tests, devising adequate treatment regimens with improved monitoring and better survival outcomes, and minimizing the negative impact on brain function. Finally, the extrapolation of this study to primary cells, fluids or tumors collected from brain metastatic patients can provide a stepping stone in the discovery efforts of novel drug targets and therapeutic strategies that target not just the cancer cells but also the tumor microenvironment.

Data availability

The mass spectrometry raw files were deposited to the ProteomeXchange Consortium via the PRIDE 94 partner repository with the following dataset identifier: PXD046330.

Ostrom, Q. T., Wright, C. H. & Barnholtz-Sloan, J. S. Brain metastases: Epidemiology. Handb. Clin. Neurol. 149 , 27–42 (2018).

Article   PubMed   Google Scholar  

Kuksis, M. et al. The incidence of brain metastases among patients with metastatic breast cancer: A systematic review and meta-analysis. Neuro-oncology 23 (6), 894–904 (2021).

Carbonell, W. S., Ansorge, O., Sibson, N. & Muschel, R. The vascular basement membrane as “soil” in brain metastasis. PloS One 4 (6), e5857 (2009).

Article   ADS   PubMed   PubMed Central   Google Scholar  

Neophytou, C. M., Kyriakou, T.-C. & Papageorgis, P. Mechanisms of metastatic tumor dormancy and implications for cancer therapy. Int. J. Mol. Sci. 20 (24), 6158 (2019).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Lau, L. W., Cua, R., Keough, M. B., Haylock-Jacobs, S. & Yong, V. W. Pathophysiology of the brain extracellular matrix: A new target for remyelination. Nat. Rev. Neurosci. 14 (10), 722–729 (2013).

Article   CAS   PubMed   Google Scholar  

Chen, Q. et al. (201Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533 (7604), 493–498 (2016).

Article   ADS   CAS   PubMed   PubMed Central   Google Scholar  

Srinivasan, E. S. et al. Salting the soil: Targeting the microenvironment of brain metastases. Mol. Cancer Ther. 20 (3), 455–466 (2021).

Cacho-Díaz, B. et al. Tumor microenvironment differences between primary tumor and brain metastases. J. Transl. Med. 18 , 1–12 (2020).

Article   PubMed   PubMed Central   Google Scholar  

Sinha, A. et al. Proteomic profiling of secreted proteins, exosomes, and microvesicles in cell culture conditioned media. In Methods Mol. Biol: The Surfaceome Vol. 1722 (eds Boheler, K. R. & Gundry, R. L.) 91–102 (Humana Press, 2018).

Google Scholar  

Proia, P. et al. Astrocytes shed extracellular vesicles that contain fibroblast growth factor-2 and vascular endothelial growth factor. Int. J. Mol. Med. 21 (1), 63–67 (2008).

CAS   PubMed   Google Scholar  

Lorger, M. & Felding-Habermann, B. Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis. Am. J. Pathol 176 (6), 2958–2971 (2010).

Zhang, L. et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 527 (7576), 100–104 (2015).

Lorger, M. Tumor microenvironment in the brain. Cancers 4 (1), 218–243 (2012).

Hambardzumyan, D., Gutmann, D. H. & Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 19 (1), 20–27 (2016).

Pistono, C., Bister, N., Stanová, I. & Malm, T. Glia-derived extracellular vesicles: Role in central nervous system communication in health and disease. Front. Cell Dev. Biol. 8 , 623771 (2021).

Igarashi, Y. et al. Glial cell line-derived neurotrophic factor induces barrier function of endothelial cells forming the blood–brain barrier. Biochem. Biophys. Res. Commun. 261 (1), 108–112 (1999).

Linnerbauer, M. & Rothhammer, V. Protective functions of reactive astrocytes following central nervous system insult. Front. Immunol. 11 , 573256 (2020).

Araki, T., Ikegaya, Y. & Koyama, R. The effects of microglia-and astrocyte-derived factors on neurogenesis in health and disease. Eur. J. Neurosci. 54 (5), 5880–5901 (2021).

Choy, C. et al. Cooperation of neurotrophin receptor TrkB and Her2 in breast cancer cells facilitates brain metastases. Breast Cancer Res. 19 (1), 1–11 (2017).

Article   Google Scholar  

Parida, P. K. et al. Metabolic diversity within breast cancer brain-tropic cells determines metastatic fitness. Cell Metab. 34 (1), 90–105 (2022).

Memorial Sloan Kettering Institute/SK-BR-3 Human Breast Cancer Cell Line. https://www.mskcc.org/research-advantage/support/technology/tangible-material/human-breast-cell-line-sk-br-3 .

Tenga, M. J. & Lazar, I. M. Proteomic snapshot of breast cancer cell cycle: G1/S transition point. Proteomics 13 (1), 48–60 (2013).

Ahuja, S. & Lazar, I. M. Systems-level proteomics evaluation of microglia response to tumor-supportive anti-inflammatory cytokines. Front. Immunol. 12 , 646043 (2021).

MacLean, B. et al. Skyline: An open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26 (7), 966–968 (2010).

Winkler, F. The brain metastatic niche. J. Mol. Med. (Berl) 93 (11), 1213–1220 (2015).

Montagner, M. & Sahai, E. In vitro models of breast cancer metastatic dormancy. Front. Cell. Dev. Biol. 8 , 37 (2020).

Zheng, X. et al. Proteomic analysis for the assessment of different lots of fetal bovine serum as a raw material for cell culture. Part IV. Application of proteomics to the manufacture of biological drugs. Biotechnol. Prog. 22 (5), 1294–1300 (2006).

Kenny, P. A. et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol. Oncol. 1 (1), 84–96 (2007).

Mai, J., Virtue, A., Shen, J., Wang, H. & Yang, X. F. An evolving new paradigm: Endothelial cells-conditional innate immune cells. J. Hematol. Oncol. 6 (1), 1–13 (2013).

Kumari, N., Dwarakanath, B. S., Das, A. & Bhatt, A. N. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumor Biol. 37 (9), 11553–11572 (2016).

Article   CAS   Google Scholar  

Subileau, E. A. et al. Expression of chemokines and their receptors by human brain endothelium: Implications for multiple sclerosis. J Neuropathol. Exp. Neurol. 68 (3), 227–240 (2009).

Keeley, E. C., Mehrad, B. & Strieter, R. M. Chemokines as mediators of neovascularization. Arterioscler. Thromb. Vasc. Biol. 28 (11), 1928–1936 (2008).

Chang, T.-T., Chen, C. & Chen, J.-W. CCL7 as a novel inflammatory mediator in cardiovascular disease, diabetes mellitus, and kidney disease. Cardiovasc. Diabetol. 21 , 185 (2022).

Cai, M. et al. Research progress of interleukin-15 in cancer immunotherapy. Front. Pharmacol. 14 , 1184703 (2023).

Harel, M., Fauteux-Daniel, S., Girard-Guyonvarc’h, C. & Gabay, C. Balance between interleukin-18 and interleukin-18 binding protein in auto-inflammatory diseases. Cytokine 150 , 155781 (2022).

Doni, A. et al. The long pentraxin PTX3 as a link between innate immunity, tissue remodeling, and cancer. Front. Immunol. 10 , 712 (2019).

Garlanda, C., Bottazzi, B., Magrini, E., Inforzato, A. & Mantovani, A. PTX3, a humoral pattern recognition molecule, in innate immunity, tissue repair, and cancer. Physiol. Rev. 98 (2), 623–639 (2018).

Janssens, R., Struyf, S. & Proost, P. The unique structural and functional features of CXCL12. Cell. Mol. Immunol. 15 (4), 299–311 (2018).

Guo, F. et al. CXCL12/CXCR4: A symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene 35 (7), 816–826 (2016).

Kryczek, I. et al. CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Res. 65 (2), 465–472 (2005).

Stelzer, G. et al. The GeneCards suite: From gene data mining to disease genome sequence analyses. Curr. Protoc. Bioinform. 54 (1), 1–30 (2016).

Article   MathSciNet   Google Scholar  

Schlesinger, M. & Bendas, G. Vascular cell adhesion molecule-1 (VCAM-1)—An increasing insight into its role in tumorigenicity and metastasis. Int. J. Cancer 136 (11), 2504–2514 (2015).

Jeon, H., Lee, S., Lee, W.-H. & Suk, K. Analysis of glial secretome: The long pentraxin PTX3 modulates phagocytic activity of microglia. J. Neuroimmunol. 229 (1–2), 63–72 (2010).

Xue, J. et al. Astrocyte-derived CCL7 promotes microglia-mediated inflammation following traumatic brain injury. Int. Immunopharmacol. 99 , 107975 (2021).

Fung, K. Y. et al. Emerging roles for IL-11 in inflammatory diseases. Cytokine 149 , 155750 (2022).

Markota, A., Endres, S. & Kobold, S. Targeting interleukin-22 for cancer therapy. Hum. Vaccin. Immunother. 14 (8), 2012–2015 (2018).

Sudan, S. K. et al. Resistin: An inflammatory cytokine with multi-faceted roles in cancer. Biochim. Biophys. Acta (BBA) Rev Cancer 1874 (2), 188419 (2020).

Wang, X. & Lin, Y. Tumor necrosis factor and cancer, buddies or foes?. Acta Pharmacol. Sin. 29 (11), 1275–1288 (2008).

Zhao, J., Chen, X., Herjan, T. & Li, X. The role of interleukin-17 in tumor development and progression. J. Exp. Med. 217 (1), e20190297 (2020).

Verjans, E. et al. Dual role of macrophage migration inhibitory factor (MIF) in human breast cancer. BMC Cancer 9 (1), 1–18 (2009).

Kadomoto, S., Izumi, K. & Mizokami, A. The CCL20-CCR6 axis in cancer progression. Int. J. Mol. Sci. 21 (15), 5186 (2020).

Wischhusen, J., Melero, I. & Fridman, W. H. Growth/differentiation factor-15 (GDF-15): From biomarker to novel targetable immune checkpoint. Front. Immunol. 11 , 951 (2020).

Hu, C. et al. Lipocalin 2: A potential therapeutic target for breast cancer metastasis. Onco Targets Ther. 11 , 8099–8106 (2018).

Pandey, V. et al. Trefoil factor 3 promotes metastatic seeding and predicts poor survival outcome of patients with mammary carcinoma. Breast Cancer Res. 16 (5), 1–20 (2014).

Breznik, B., Mitrović, A., Lah, T. T. & Kos, J. Cystatins in cancer progression: More than just cathepsin inhibitors. Biochimie 166 , 233–250 (2019).

Acker, T., Beck, H. & Plate, K. H. Cell type specific expression of vascular endothelial growth factor and angiopoietin-1 and-2 suggests an important role of astrocytes in cerebellar vascularization. Mech. Dev. 108 (1–2), 45–57 (2001).

Li, F., Liu, A., Zhao, M. & Luo, L. Astrocytic chitinase-3-like protein 1 in neurological diseases: Potential roles and future perspectives. J. Neurochem. 165 (6), 772–790 (2023).

Butti, R. et al. Osteopontin signaling in shaping tumor microenvironment conducive to malignant progression. Adv. Exp. Med. Biol. 1329 , 419–441 (2021).

Choy, C. et al. Cooperation of neurotrophin receptor TrkB and Her2 in breast cancer cells facilitates brain metastases. Breast Cancer Res. 19 , 51 (2017).

Old, W. M. et al. Comparison of label-free methods for quantifying human proteins by shotgun proteomics. Mol. Cell. Proteomics 4 (10), 1487–1502 (2005).

Turi, Z., Lacey, M., Mistrik, M. & Moudry, P. Impaired ribosome biogenesis: Mechanisms and relevance to cancer and aging. Aging (Albany NY) 11 (8), 2512–2540 (2019).

Karcini, A. & Lazar, I. M. The SKBR3 Cell-membrane proteome reveals telltales of aberrant cancer cell proliferation and targets for precision medicine applications. Sci. Rep. 12 , 10847 (2022).

Vera-Ramirez, L. & Hunter, K. W. Tumor cell dormancy as an adaptive cell stress response mechanism. F1000Res. 6 , 2134 (2017).

Uzuner, D. et al. Transcriptional landscape of cellular networks reveals interactions driving the dormancy mechanisms in cancer. Sci. Rep. 11 (1), 1–10 (2021).

Marescal, O. & Cheeseman, I. M. Cellular mechanisms and regulation of quiescence. Dev. Cell 55 (3), 259–271 (2020).

Constantinou, C. et al. The multi-faceted functioning portrait of LRF/ZBTB7A. Hum. Genomics 13 (1), 1–14 (2019).

Zahnow, C. A. CCAAT/enhancer-binding protein β: Its role in breast cancer and associations with receptor tyrosine kinases. Expert Rev. Mol. Med. 11 , e12 (2009).

Fang, C. Y., Lai, T. C., Hsiao, M. & Chang, Y. C. The diverse roles of TAO kinases in health and diseases. Int. J. Mol. Sci. 21 (20), 7463 (2020).

Lai, T. C. et al. Kinase shRNA screening reveals that TAOK3 enhances microtubule-targeted drug resistance of breast cancer cells via the NF-κB signaling pathway. Cell Commun. Signal. 18 (1), 1–14 (2020).

Wagner, E. F. & Nebreda, Á. R. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat. Rev. Cancer 9 (8), 537–549 (2009).

Park, S. Y. & Nam, J. S. The force awakens: Metastatic dormant cancer cells. Exp. Mol. Med. 52 (4), 569–581 (2020).

Dias, I. B., Bouma, H. R. & Henning, R. H. Unraveling the big sleep: Molecular aspects of stem cell dormancy and hibernation. Front. Physiol. 12 , 624950 (2021).

Movahed, Z. G., Rastegari-Pouyani, M., Hossein Mohammadi, M. & Mansouri, K. Cancer cells change their glucose metabolism to overcome increased ROS: One step from cancer cell to cancer stem cell?. Biomed. Pharmacother. 112 , 108690 (2019).

El Khoury, W. & Nasr, Z. Deregulation of ribosomal proteins in human cancers. Biosci. Rep. 41 (12), BSR20211577 (2021).

Boulon, S., Westman, B. J., Hutten, S., Boisvert, F. M. & Lamond, A. I. The nucleolus under stress. Mol. Cell 40 (2), 216–227 (2010).

Häfliger, P. & Charles, R. P. The L-type amino acid transporter LAT1-An emerging target in cancer. Int. J. Mol. Sci. 20 (10), 2428 (2019).

Ribatti, D., Tamma, R. & Annese, T. Epithelial-mesenchymal transition in cancer: A historical overview. Transl. Oncol. 13 (6), 100773 (2020).

Das, S. K. et al. MDA-9/Syntenin (SDCBP): Novel gene and therapeutic target for cancer metastasis. Pharmacol. Res. 155 , 104695 (2020).

Pradhan, A. K. et al. MDA-9/Syntenin/SDCBP: New insights into a unique multifunctional scaffold protein. Cancer Metastasis Rev. 39 (3), 769–781 (2020).

Menezes, M. E. et al. MDA-9/Syntenin (SDCBP) modulates small GTPases RhoA and Cdc42 via transforming growth factor β1 to enhance epithelial–mesenchymal transition in breast cancer. Oncotarget 7 (49), 80175–80189 (2016).

Araújo, T. G. et al. Annexin A1 as a regulator of immune response in cancer. Cells 10 (9), 2245 (2021).

Mao, A. et al. ZBTB7A promotes migration, invasion and metastasis of human breast cancer cells through NF-κB-induced epithelial–mesenchymal transition in vitro and in vivo. J. Biochem. 166 (6), 485–493 (2019).

Lim, J. P. et al. YBX1 gene silencing inhibits migratory and invasive potential via CORO1C in breast cancer in vitro. BMC Cancer 17 (1), 1–15 (2017).

Kuwano, M., Shibata, T., Watari, K. & Ono, M. Oncogenic Y-box binding protein-1 as an effective therapeutic target in drug-resistant cancer. Cancer Sci. 110 (5), 1536–1543 (2019).

Xu, L., Li, H., Wu, L. & Huang, S. YBX1 promotes tumor growth by elevating glycolysis in human bladder cancer. Oncotarget 8 (39), 65946–65956 (2017).

Liu, X. et al. DRMref: comprehensive reference map of drug resistance mechanisms in human cancer. Nucleic Acids Res. 52 (D1), D1253–D1264 (2024).

Sarafian, V. et al. Expression of Lamp-1 and Lamp-2 and their interactions with galectin-3 in human tumor cells. Int. J. Cancer 75 (1), 105–111 (1998).

Bresnick, A. R., Weber, D. J. & Zimmer, D. B. S100 proteins in cancer. Nat. Rev. Cancer 15 (2), 96–109 (2015).

Bolado-Carrancio, A. et al. ISGylation drives basal breast tumour progression by promoting EGFR recycling and Akt signalling. Oncogene 40 (44), 6235–6247 (2021).

Aljohani, A. I. et al. Myxovirus resistance 1 (MX1) is an independent predictor of poor outcome in invasive breast cancer. Breast Cancer Res. Treat. 181 (3), 541–551 (2020).

Dhatchinamoorthy, K., Colbert, J. D. & Rock, K. L. Cancer immune evasion through loss of MHC class I antigen presentation. Front. Immunol. 12 , 636568 (2021).

Borst, L., van der Burg, S. H. & van Hall, T. The NKG2A-HLA-E axis as a novel checkpoint in the tumor microenvironment. Clin. Cancer Res. 26 (21), 5549–5556 (2020).

Blais, M. E., Dong, T. & Rowland-Jones, S. HLA-C as a mediator of natural killer and T-cell activation: Spectator or key player?. Immunology 133 (1), 1–7 (2011).

Perez-Riverol, Y. et al. The PRIDE database resources in 2022: A Hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50 (D1), D543–D552 (2022).

Download references

Acknowledgements

This work was supported by an award from the National Institute of General Medical Sciences (Grant No. 1R01GM121920) to IML.

Author information

Authors and affiliations.

Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA

Shreya Ahuja & Iulia M. Lazar

Fralin Life Sciences Institute, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA

Iulia M. Lazar

Carilion School of Medicine, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA

Division of Systems Biology/AIS, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA

You can also search for this author in PubMed   Google Scholar

Contributions

SA performed the experiments; SA and IML analyzed the data and wrote the manuscript; IML coordinated the work. All authors reviewed and approved the final version of the manuscript.

Corresponding author

Correspondence to Iulia M. Lazar .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Supplementary information 1., supplementary information 2., supplementary information 3., supplementary information 4., supplementary information 5., supplementary information 6., rights and permissions.

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/ .

Reprints and permissions

About this article

Cite this article.

Ahuja, S., Lazar, I.M. Proteomic insights into breast cancer response to brain cell-secreted factors. Sci Rep 14 , 19351 (2024). https://doi.org/10.1038/s41598-024-70386-7

Download citation

Received : 22 February 2024

Accepted : 16 August 2024

Published : 21 August 2024

DOI : https://doi.org/10.1038/s41598-024-70386-7

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

By submitting a comment you agree to abide by our Terms and Community Guidelines . If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

breast cancer research and treatment impact factor 2023

IMAGES

  1. Understanding The Impact Factor Of Breast Cancer Research And Treatment

    breast cancer research and treatment impact factor 2023

  2. (Latest) Cancers Impact Factor 2023

    breast cancer research and treatment impact factor 2023

  3. Understanding The Impact Factor Of Breast Cancer Research And Treatment

    breast cancer research and treatment impact factor 2023

  4. Breast Cancer Research and Treatment impact factor and...

    breast cancer research and treatment impact factor 2023

  5. Breast Cancer Research impact factor and citations:...

    breast cancer research and treatment impact factor 2023

  6. Breast Cancer Treatment Market Share, Forecast 2023

    breast cancer research and treatment impact factor 2023

COMMENTS

  1. Home

    Breast Cancer Research and Treatment is a comprehensive forum dedicated to all aspects of breast cancer research. ... Journal Impact Factor 3.0 (2023) 5-year Journal Impact Factor 3.9 (2023) Submission to first decision (median) 14 days. Downloads 1,529,254 (2023) Latest issue

  2. Breast Cancer Research and Treatment

    The Impact IF 2022 of Breast Cancer Research and Treatment is 3.49, which is computed in 2023 as per its definition. Breast Cancer Research and Treatment IF is decreased by a factor of 0.57 and approximate percentage change is -14.04% when compared to preceding year 2021, which shows a falling trend. The impact IF, also denoted as Journal impact score (JIS), of an academic journal is a measure ...

  3. Breast cancer highlights from 2023: Knowledge to guide practice and

    The guidelines and consensus publication (Table 1) represent collaborative efforts aimed at improving the standard of diagnosis and treatment for breast cancer patients both in early and metastatic breast cancer.These could be integrated in the routine clinical practice of breast units and teams and can help to guide difficult decisions, in the case of expert consensus recommendations, mainly ...

  4. Breast Cancer Research and Treatment Impact Factor IF 2024|2023|2022

    Breast Cancer Research and Treatment Impact Factor, IF, number of article, detailed information and journal factor. ISSN: 0167-6806. ... Breast Cancer Research and Treatment provides the surgeon, radiotherapist, medical oncologist, endocrinologist, epidemiologist, immunologist or cell biologist investigating problems in breast cancer a single ...

  5. Volumes and issues

    Search all Breast Cancer Research and Treatment articles. Volume 207 August - September 2024. Issue 2 September 2024. Issue 1 August 2024. Volume 206 July - August 2024. Issue 3 August 2024. Issue 2 July 2024. Issue 1 July 2024. Volume 205 May - June 2024.

  6. BREAST CANCER RESEARCH AND TREATMENT

    Journal Impact Factor (JIF): 3 5-year Impact Factor: 3.9 Best ranking: ... are now ranked by JIF as the same with journals of SCIE and SSCI in the release of JCR 2023 (in 2024). ... » BREAST CANCER RESEARCH AND TREATMENT. Abbreviation: BREAST CANCER RES TR ISSN: 0167-6806 eISSN: 1573-7217

  7. Volume 197, Issue 1

    Comments on: The clinical relevance of various methods of classifying ipsilateral breast tumor recurrence as either true local recurrence or new primary. Fei-Lin Qu. Jun-Jie Li. Zhi-Ming Shao. Letter to the Editor 30 October 2022 Pages: 243 - 244. Volume 197, issue 1 articles listing for Breast Cancer Research and Treatment.

  8. Breast Cancer Research and Treatment

    Scope. Breast Cancer Research and Treatment provides the surgeon, radiotherapist, medical oncologist, endocrinologist, epidemiologist, immunologist or cell biologist investigating problems in breast cancer a single forum for communication. The journal creates a "market place" for breast cancer topics which cuts across all the usual lines of ...

  9. Breast Cancer Research and Treatment

    Breast Cancer Research and Treatment is a scientific journal focused on the treatment of and investigations in breast cancer. ... the journal has a 2020 impact factor of 4.872. References This page was last edited on 29 April 2023, at 12:55 (UTC). Text is available under the ... This page was last edited on 29 April 2023, at 12:55 ...

  10. Breast Cancer Research and Treatment Latest Journal's Impact IF 2023

    The ISSN of Breast Cancer Research and Treatment is 0167-6806 . An ISSN is an 8-digit code used to identify newspapers, journals, magazines and periodicals of all kinds and on all media-print and electronic. Breast Cancer Research and Treatment Key Factor Analysis

  11. Home page

    Breast Cancer Research is the highest ranking breast cancer-specific title in the top quartile of oncology journals worldwide. ... Critical research gaps and translational priorities for the successful prevention and treatment of breast cancer. Authors: Suzanne A Eccles, Eric O Aboagye ... Citation Impact 2023 Journal Impact Factor: 6.1 5-year ...

  12. Top Breast Cancer Research Advances of 2023

    Advertisement. Regular screening mammograms reduce the risk of dying from breast cancer by more than 65%. Advertisement. Women in their 40s who had screening mammograms were less likely to die from breast cancer if they were diagnosed with the disease. — Last updated on February 7, 2024, 10:27 AM.

  13. Breast Cancer Research and Treatment

    Top authors and change over time. The top authors publishing in Breast Cancer Research and Treatment (based on the number of publications) are: Ian O. Ellis (103 papers) published 2 papers at the last edition, 1 less than at the previous edition,; Steven A. Narod (101 papers) published 2 papers at the last edition, 1 more than at the previous edition, ...

  14. npj Breast Cancer

    npj Breast Cancer has a 2-year impact factor of 5.9 (2022), article downloads of 742,276 (2022) and 7 days from submission to first editorial decision (2022). Call for papers: AI in precision oncology

  15. Breast cancer highlights from 2023: Knowledge to guide practice and

    Abstract. This narrative work highlights a selection of published work from 2023 with potential implications for breast cancer practice. We feature publications that have provided new knowledge immediately relevant to patient care or for future research. We also highlight guidelines that have reported evidence-based or consensus recommendations ...

  16. Articles

    Breast Cancer Research is the highest ranking breast cancer-specific title in the top quartile of oncology journals worldwide. ... Development of a humanized anti-FABP4 monoclonal antibody for potential treatment of breast cancer. ... Citation Impact 2023 Journal Impact Factor: 6.1 5-year Journal Impact Factor: 7.1 Source Normalized Impact per ...

  17. Cancer Research and Treatment

    Cancer Research and Treatment: Search: ... Editor's Note on Recent Journal Impact Factor of Cancer Research and Treatment. Yong Chan Ahn. Cancer Res Treat. 2024;56(3):699-699. Published online July 9, 2024 DOI: ... Breast cancer: 795 Trastuzumab Biosimilar (HLX02), Pertuzumab Plus Chemotherapy in Patients with HER2-Positive Metastatic Breast ...

  18. Submission guidelines

    Citation Impact 2023 Journal Impact Factor: 6.1 5-year Journal Impact Factor: 7.1 Source Normalized Impact per Paper (SNIP): 1.865 SCImago Journal Rank (SJR): 2.578 Speed 2023 Submission to first editorial decision (median days): 14 Submission to acceptance (median days): 129 Usage 2023 Downloads: 2,432,781 Altmetric mentions: 1,561

  19. Volume 198, Issue 1

    A rare but persistent cause of decreased function secondary to adjuvant radiation therapy in women with breast cancer. Susan R. Harris. Letter to the Editor 05 January 2023 Pages: 185 - 186. Volume 198, issue 1 articles listing for Breast Cancer Research and Treatment.

  20. Cancer Research and Treatment Impact Factor IF 2024|2023|2022

    Cancer Research and Treatment. Journal Abbreviation: CANCER RES TREAT. Journal ISSN: 1598-2998. Year. Impact Factor (IF) Total Articles. Total Cites. 2023 (2024 update) 4.1.

  21. Breast Cancer Research Latest Journal's Impact IF 2023-2024

    About. Breast Cancer Research is an international, peer-reviewed online journal, publishing original research, reviews, editorials and reports. Open access research articles of exceptional interest are published in all areas of biology and medicine relevant to breast cancer, including normal mammary gland biology, with special emphasis on the genetic, biochemical, and cellular basis of breast ...

  22. North America Cancer Biopsy Market By Type (Liquid Biopsy, Tissue

    North America Cancer Biopsy Market By Type (Liquid Biopsy, Tissue Biopsy), By Product Type (Instruments, Kits & Consumables, Others), By Application (Breast Cancer, Lung Cancer, Others), By Country, Competition, Forecast and Opportunities, 2019-2029F - North America Cancer biopsy Market was valued at USD 10.71 Billion in 2023 and is anticipated to project impressive growth in the forecast ...

  23. Evaluating the Breast Cancer Therapeutics Market

    A recent market report published by Precedence Research, which analyzed from the base year 2022 to the forecast period of 2023-2032, revealed that in 2022 the breast cancer market size was estimated to be $28.8 billion (USD). Research projections anticipate the market will be valued at $73.68 billion (USD) by 2032 with a compound annual ...

  24. The Effects of Radiotherapy on the Sequence and Eligibility of Breast

    Immediate breast reconstruction (IBR) following a mastectomy, combined with radiotherapy, presents a multifaceted approach to breast cancer treatment, balancing oncological safety and aesthetic outcomes. IBR, typically involving the use of implants or autologous tissue, aims to restore breast morphology directly after a mastectomy, minimizing the psychological and physical impacts.

  25. Breast cancer survival improved in Gaza between 2017-2020, finds study

    A recent study shines light on the treatment and survival outcomes for women with breast cancer in Gaza before the current conflict. While over half of reported diagnoses were made at advanced ...

  26. Volume 201, Issue 1

    It is the impact of racism, not race, that causes breast cancer inequities. Francois G. Rollin. Shub S. Agrawal. Mackenzie L. W. Garcia. Letter to the Editor 19 June 2023 Pages: 149 - 150. Part of 1 collection: Cancer Disparities. Volume 201, issue 1 articles listing for Breast Cancer Research and Treatment.

  27. Cost-Effectiveness of Olaparib for Patients With Breast Cancer and

    Key Points. Question Is adjuvant olaparib cost-effective in patients with early-stage breast cancer with high-risk disease and germline BRCA1/2 mutations?. Findings In an economic evaluation using a Markov model, adjuvant olaparib therapy was associated with a 1.25-year increase in life expectancy and a 1.20-quality-adjusted life-year (QALY) increase at an incremental cost of $133 133 compared ...

  28. Qualitative exploration of patients' experiences with Intrabeam

    Objective To gather a deep qualitative understanding of the perceived benefits and impacts of External-Beam RadioTherapy (EBRT) and TARGeted Intraoperative radioTherapy (TARGIT-IORT) using Intrabeam to assess how the treatments affected patient/care partner experiences during their cancer treatment and beyond. Design and participants A patient-led working group was established to guide study ...

  29. Proteomic insights into breast cancer response to brain cell-secreted

    The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2 + and TN breast cancers frequently metastasize to the brain and stay potentially ...

  30. Breast cancer classification

    Breast cancers can be classified by different schemata. Each of these aspects influences treatment response and prognosis. Description of a breast cancer would optimally include all of these classification aspects, as well as other findings, such as signs found on physical exam.A full classification includes histopathological type, grade, stage, receptor status, and the presence or absence of ...