trying...
383082982024020720241218
1756-05001712024Feb02BMC research notesBMC Res NotesThe relationship between high ratios of CD4/FOXP3 and CD8/CD163 and the improved survivability of metastatic triple-negative breast cancer patients: a multicenter cohort study.44444410.1186/s13104-024-06704-zTriple-negative breast cancer (TNBC) has been documented as the most aggressive subtype of breast cancer. This study aimed to analyze antitumor and protumor immune activities, and their ratios as significant prognostic biomarkers in metastatic TNBC (mTNBC).A multicenter cohort study was conducted among 103 de novo mTNBC patients. The expression of CD8 and CD163 was evaluated using immunohistochemistry staining, CD4 and FOXP3 using double-staining immunohistochemistry, and PD-L1 using immunohistochemistry and RT-PCR.Multivariate analysis revealed that high CD4/FOXP3 (HR 1.857; 95% CI 1.049-3.288; p = 0.034) and the CD8/CD163 ratio (HR 2.089; 95% CI 1.174-3.717; p = 0.012) yield significantly improved 1 year overall survival (OS). Kaplan-Meier analysis showed that high levels of CD4 (p = 0.023), CD8 (p = 0.043), CD4/FOXP3 (p = 0.016), CD8/FOXP3 (p = 0.005), CD8/CD163 (p = 0.005) ratios were significantly associated with higher rate of 1 year OS. Furthermore, 1 year OS was directly correlated with antitumor CD4 (R = 0.233; p = 0.018) and CD8 (R = 0.219; p = 0.026) and was indirectly correlated with protumor CD163 and FOXP3 through CD4/FOXP3 (R = 0.282; p = 0.006), CD4/CD163 (R = 0.239; p = 0.015), CD8/FOXP3 (R = 0.260; p = 0.008), and CD8/CD163 (R = 0.258; p = 0.009).This is the first study to demonstrate that high levels of CD4/FOXP3 and CD8/CD163 significantly improved the 1 year OS in de novo mTNBC patients. Thus, we recommend the application of these markers as prognosis determination and individual treatment decision.© 2024. The Author(s).TenggaraJeffry BetaJBDivision of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia. jeffry.tenggara@yahoo.com.Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia. jeffry.tenggara@yahoo.com.RachmanAndhikaADivision of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia.Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia.PrihartonoJoedoJDepartment of Community Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.RachmadiLisnawatiLDepartment of Anatomical Pathology, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.PanigoroSonar SoniSSDepartment of Surgical Oncology, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.HeriyantoDidik SetyoDSDepartment of Anatomical Pathology, Dr. Sardjito Hospital-Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.SutandyoNoorwatiNDivision of Hematology and Medical Oncology, Department of Internal Medicine, Dharmais National Cancer Hospital, Jakarta, Indonesia.NasutionIntan RussiannaIRDivision of Hematology and Medical Oncology, Gatot Soebroto Army Hospital Jakarta, Jakarta, Indonesia.RahadiatiFamilia BellaFBDepartment of Anatomical Pathology, Gatot Soebroto Army Hospital Jakarta, Jakarta, Indonesia.StevenRicciRDivision of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia.Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia.BetsyRachelleRDepartment of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.JuanputraSamuelSDepartment of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.SudoyoAru WisaksonoAWDivision of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia.Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia.engMulticenter StudyJournal Article20240202
EnglandBMC Res Notes1014627681756-05000B7-H1 Antigen0Forkhead Transcription Factors0FOXP3 protein, human0CD4 Antigens0CD8 Antigens0CD163 antigenIMHumansB7-H1 AntigenCD8-Positive T-LymphocytesmetabolismCohort StudiesForkhead Transcription FactorsgeneticsLymphocytes, Tumor-InfiltratingmetabolismpathologyNeoadjuvant TherapyTriple Negative Breast NeoplasmspathologyCD4 AntigensCD8 AntigensCD163CD4CD8FOXP3MetastaticSurvivalTriple-negative breast cancerThe authors declared that they had no competing interests.
202371920241242024256422024230422024222358202422epublish38308298PMC1083586410.1186/s13104-024-06704-z10.1186/s13104-024-06704-zAlmansour NM. Triple-negative breast cancer: a brief review about epidemiology, risk factors, signaling pathways, treatment and role of artificial intelligence. Front Mol Biosci. 2022 doi: 10.3389/fmolb.2022.836417.10.3389/fmolb.2022.836417PMC882442735145999Oshi M, Newman S, Tokumaru Y, Yan L, Matsuyama R, Endo I, et al. Inflammation is associated with worse outcome in the whole cohort but with better outcome in triple-negative subtype of breast cancer patients. J Immunol Res. 2020;2020:1–17. doi: 10.1155/2020/5618786.10.1155/2020/5618786PMC778787133457427Zheng H, Siddharth S, Parida S, Wu X, Sharma D. Tumor microenvironment: key players in triple negative breast cancer immunomodulation. Cancers. 2021;13:3357. doi: 10.3390/cancers13133357.10.3390/cancers13133357PMC826909034283088Anders CK, Carey LA. Biology, metastatic patterns, and treatment of patients with triple-negative breast cancer. Clin Breast Cancer. 2009;9:S73–81. doi: 10.3816/CBC.2009.s.008.10.3816/CBC.2009.s.008PMC291976119596646Bou Zerdan M, Ghorayeb T, Saliba F, Allam S, Bou Zerdan M, Yaghi M, et al. Triple negative breast cancer: updates on classification and treatment in 2021. Cancers. 2022;14:1253. doi: 10.3390/cancers14051253.10.3390/cancers14051253PMC890918735267561Kassam F, Enright K, Dent R, Dranitsaris G, Myers J, Flynn C, et al. Survival outcomes for patients with metastatic triple-negative breast cancer: implications for clinical practice and trial design. Clin Breast Cancer. 2009;9:29–33. doi: 10.3816/CBC.2009.n.005.10.3816/CBC.2009.n.00519299237Kazmi S, Chatterjee D, Raju D, Hauser R, Kaufman PA. Overall survival analysis in patients with metastatic breast cancer and liver or lung metastases treated with eribulin, gemcitabine, or capecitabine. Breast Cancer Res Treat. 2020;184:559–565. doi: 10.1007/s10549-020-05867-0.10.1007/s10549-020-05867-0PMC759918632808239Benchama O, Malamas MS, Praveen K, Ethier EC, Williams MK, Makriyannis A, et al. Inhibition of triple negative breast cancer-associated inflammation and progression by N- acylethanolamine acid amide hydrolase (NAAA) Sci Rep. 2022;12:22255. doi: 10.1038/s41598-022-26564-6.10.1038/s41598-022-26564-6PMC978904036564457Lan T, Chen L, Wei X. inflammatory cytokines in cancer: comprehensive understanding and clinical progress in gene therapy. Cells. 2021;10:100. doi: 10.3390/cells10010100.10.3390/cells10010100PMC782794733429846Salem ML, Attia ZI, Galal SM. Acute inflammation induces immunomodulatory effects on myeloid cells associated with anti-tumor responses in a tumor mouse model. J Adv Res. 2016;7:243–253. doi: 10.1016/j.jare.2015.06.001.10.1016/j.jare.2015.06.001PMC476779826966565Marusyk A, Tabassum DP, Altrock PM, Almendro V, Michor F, Polyak K. Non-cell-autonomous driving of tumour growth supports sub-clonal heterogeneity. Nature. 2014;514:54–58. doi: 10.1038/nature13556.10.1038/nature13556PMC418496125079331Lee JM, Lee M-H, Garon E, Goldman JW, Salehi-Rad R, Baratelli FE, et al. Phase I trial of intratumoral injection of CCL21 gene-modified dendritic cells in lung cancer elicits tumor-specific immune responses and CD8+ T-cell Infiltration. Clin Cancer Res. 2017;23:4556–4568. doi: 10.1158/1078-0432.CCR-16-2821.10.1158/1078-0432.CCR-16-2821PMC559926328468947Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 2010;28:2784–2795. doi: 10.1200/JCO.2009.25.6529.10.1200/JCO.2009.25.6529PMC288185520404251Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline update. J Clin Oncol. 2013;31:3997–4013. doi: 10.1200/JCO.2013.50.9984.10.1200/JCO.2013.50.998424101045Tavares MC, Sampaio CD, Lima GE, Andrade VP, Gonçalves DG, Macedo MP, et al. A high CD8 to FOXP3 ratio in the tumor stroma and expression of PTEN in tumor cells are associated with improved survival in non-metastatic triple-negative breast carcinoma. BMC Cancer. 2021;21:901. doi: 10.1186/s12885-021-08636-4.10.1186/s12885-021-08636-4PMC834397334362334Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs working group 2014. Ann Oncol. 2015;26:259–271. doi: 10.1093/annonc/mdu450.10.1093/annonc/mdu450PMC626786325214542Yip WK, Abdullah MA, Yusoff SM, Seow HF. Increase in tumour-infiltrating lymphocytes with regulatory T cell immunophenotypes and reduced ζ-chain expression in nasopharyngeal carcinoma patients. Clin Exp Immunol. 2009;155:412–422. doi: 10.1111/j.1365-2249.2008.03793.x.10.1111/j.1365-2249.2008.03793.xPMC266951719220831Bankhead P, Loughrey MB, Fernández JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: open source software for digital pathology image analysis. Sci Rep. 2017;7:16878. doi: 10.1038/s41598-017-17204-5.10.1038/s41598-017-17204-5PMC571511029203879Park Y, Koh J, Na HY, Kwak Y, Lee K-W, Ahn S-H, et al. PD-L1 testing in gastric cancer by the combined positive score of the 22C3 PharmDx and SP263 assay with clinically relevant cut-offs. Cancer Res Treat. 2020;52:661–670. doi: 10.4143/crt.2019.718.10.4143/crt.2019.718PMC737386232019283Yarso K, Bellynda M, Azmiardi A, Wasita B, Heriyanto D, Astuti I, et al. Chemotherapy negates the effect of SDF1 mRNA to distant metastasis and poor overall survival in breast cancer patients. Asian Pac J Cancer Prev. 2021;22:757–766. doi: 10.31557/APJCP.2021.22.3.757.10.31557/APJCP.2021.22.3.757PMC828665733773539Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol. 2010;185:2273–2284. doi: 10.4049/jimmunol.1000901.10.4049/jimmunol.1000901PMC292348320644162National Comprehensive Cancer Network. NCCN Guidelines. 2022. https://www.nccn.org/guidelines/category_1. Accessed 19 Nov 2022.Tay RE, Richardson EK, Toh HC. Revisiting the role of CD4+ T cells in cancer immunotherapy—new insights into old paradigms. Cancer Gene Ther. 2021;28:5–17. doi: 10.1038/s41417-020-0183-x.10.1038/s41417-020-0183-xPMC788665132457487Borst J, Ahrends T, Bąbała N, Melief CJM, Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18:635–647. doi: 10.1038/s41577-018-0044-0.10.1038/s41577-018-0044-030057419Hor JL, Whitney PG, Zaid A, Brooks AG, Heath WR, Mueller SN. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity. 2015;43:554–565. doi: 10.1016/j.immuni.2015.07.020.10.1016/j.immuni.2015.07.02026297566Smith CM, Wilson NS, Waithman J, Villadangos JA, Carbone FR, Heath WR, et al. Cognate CD4+ T cell licensing of dendritic cells in CD8+ T cell immunity. Nat Immunol. 2004;5:1143–1148. doi: 10.1038/ni1129.10.1038/ni112915475958Laidlaw BJ, Craft JE, Kaech SM. The multifaceted role of CD4+ T cells in CD8+ T cell memory. Nat Rev Immunol. 2016;16:102–111. doi: 10.1038/nri.2015.10.10.1038/nri.2015.10PMC486001426781939Kennedy R, Celis E. Multiple roles for CD4 + T cells in anti-tumor immune responses. Immunol Rev. 2008;222:129–144. doi: 10.1111/j.1600-065X.2008.00616.x.10.1111/j.1600-065X.2008.00616.x18363998Jackute J, Zemaitis M, Pranys D, Sitkauskiene B, Miliauskas S, Bajoriunas V, et al. The prognostic influence of tumor infiltrating Foxp3+CD4+, CD4+ and CD8+ T cells in resected non-small cell lung cancer. J Inflamm. 2015;12:63. doi: 10.1186/s12950-015-0108-x.10.1186/s12950-015-0108-xPMC465729626604855Mandapathil M, Szczepanski MJ, Szajnik M, Ren J, Lenzner DE, Jackson EK, et al. increased ectonucleotidase expression and activity in regulatory T cells of patients with head and neck cancer. Clin Cancer Res. 2009;15:6348–6357. doi: 10.1158/1078-0432.CCR-09-1143.10.1158/1078-0432.CCR-09-1143PMC276333519825957Liu C, Sun B, Hu X, Zhang Y, Wang Q, Yue J, et al. Stereotactic ablative radiation therapy for pulmonary recurrence-based oligometastatic non-small cell lung cancer: survival and prognostic value of regulatory T cells. Int J Radiation Oncol Biol Phys. 2019;105:1055–64. doi: 10.1016/j.ijrobp.2019.08.012.10.1016/j.ijrobp.2019.08.01231437470Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression — implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16:356–371. doi: 10.1038/s41571-019-0175-7.10.1038/s41571-019-0175-730705439Ren X, Song Y, Pang J, Chen L, Zhou L, Liang Z, et al. Prognostic value of various immune cells and Immunoscore in triple-negative breast cancer. Front Immunol. 2023 doi: 10.3389/fimmu.2023.1137561.10.3389/fimmu.2023.1137561PMC1011782837090736Farhood B, Najafi M, Mortezaee K. CD8 + cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234:8509–8521. doi: 10.1002/jcp.27782.10.1002/jcp.2778230520029Zhang L, Zhang W, Li Z, Lin S, Zheng T, Hao B, et al. Mitochondria dysfunction in CD8+ T cells as an important contributing factor for cancer development and a potential target for cancer treatment: a review. J Exp Clin Cancer Res. 2022;41:227. doi: 10.1186/s13046-022-02439-6.10.1186/s13046-022-02439-6PMC930605335864520Hudson K, Cross N, Jordan-Mahy N, Leyland R. The extrinsic and intrinsic roles of PD-L1 and Its receptor PD-1: implications for immunotherapy treatment. Front Immunol. 2020 doi: 10.3389/fimmu.2020.568931.10.3389/fimmu.2020.568931PMC760940033193345Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang T-H, et al. The immune landscape of cancer. Immunity. 2018;48:812–830.e14. doi: 10.1016/j.immuni.2018.03.023.10.1016/j.immuni.2018.03.023PMC598258429628290Zhu Y, Zhang H, Pan C, He G, Cui X, Yu X, et al. Integrated tumor genomic and immune microenvironment analysis identifies predictive biomarkers associated with the efficacy of neoadjuvant therapy for triple-negative breast cancer. Cancer Med. 2023;12:5846–5858. doi: 10.1002/cam4.5372.10.1002/cam4.5372PMC1002816736271505Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18:10. doi: 10.1186/s12943-018-0928-4.10.1186/s12943-018-0928-4PMC633284330646912Medrek C, Pontén F, Jirström K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012;12:306. doi: 10.1186/1471-2407-12-306.10.1186/1471-2407-12-306PMC341478222824040Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 2015;21:24–33. doi: 10.1016/j.molmed.2014.10.009.10.1016/j.molmed.2014.10.009PMC428282525440090Purwanto I, Heriyanto DS, Ghozali A, Widodo I, Dwiprahasto I, Aryandono T, et al. Overexpression of programmed death-ligand 1 receptor MRNA as an independent negative prognostic factor for triple negative breast cancer. World J Oncol. 2020;11:216–222. doi: 10.14740/wjon1302.10.14740/wjon1302PMC757527533117465Magaki S, Hojat SA, Wei B, So A, Yong WH. An introduction to the performance of immunohistochemistry. Berlin: Springer; 2019.PMC674999830539453Wang R, Zhu Y, Liu X, Liao X, He J, Niu L. The clinicopathological features and survival outcomes of patients with different metastatic sites in stage IV breast cancer. BMC Cancer. 2019;19:1091. doi: 10.1186/s12885-019-6311-z.10.1186/s12885-019-6311-zPMC685291331718602
trying2...
trying...
537238501MCID_676f086f293dc21275038228 39729313 39729292 39729291 39729247 39729236 breast cancer "breast neoplasms"[MeSH Terms] OR ("breast"[All Fields] AND "neoplasms"[All Fields]) OR "breast neoplasms"[All Fields] OR ("breast"[All Fields] AND "cancer"[All Fields]) OR "breast cancer"[All Fields] "breast neoplasms"[MeSH Terms] OR ("breast"[All Fields] AND "neoplasms"[All Fields]) OR "breast neoplasms"[All Fields] OR ("breast"[All Fields] AND "cancer"[All Fields]) OR "breast cancer"[All Fields] trying2...
trying...
3972931320241227
2574-38057122024Dec02JAMA network openJAMA Netw OpenChanges to the US Preventive Services Task Force Screening Guidelines and Incidence of Breast Cancer.e2452688e245268810.1001/jamanetworkopen.2024.52688The 2009 US Preventive Services Task Force breast cancer screening guideline changes led to decreases in screening mammography, raising concern about potential increases in late-stage disease and more invasive surgical treatments.To investigate the incidence of breast cancer by stage at diagnosis and surgical treatment before and after the 2009 guideline changes.This population-based, epidemiologic cohort study of women aged 40 years or older used 2004 to 2019 data from the National Cancer Institute's Surveillance, Epidemiology, and End Results Program. Age- and stage-specific breast cancer incidence rates and the proportion of breast cancers treated by partial mastectomy, total mastectomy, and total mastectomy with reconstruction were calculated. Data analyses were conducted from August 2023 to February 2024.Age group (40-49, 50-74, and ≥75 years).Annual percent changes (APCs) in stage-specific breast cancer incidence and proportions of cases treated with each surgery type.This cohort study included 2 022 250 women (354 263 [17.5%] aged 40-49 years, 1 279 542 [63.2%] aged 50-74 years, and 388 445 [19.2%] aged ≥75 age group, from a total of 2 023 541 women) diagnosed with breast cancer. Rates of in situ breast cancer decreased since 2009 (eg, APC, -0.69 [95% CI, -2.77 to -0.18] for women aged 50-74 years). Localized breast cancer rates increased steadily during 2004 to 2019 in women aged 40 to 74 years (eg, APC, 1.18 [95% CI, 1.02-1.34] for women aged 50-74 years), with no evidence of a change in trend during the study period. Regional cancer rates decreased or did not change. Distant cancer rates were flat since 2012 among women aged 40 to 74 years and increased steadily for those 75 years or older during 2004 to 2019 (APC, 1.40 [95% CI, 1.00-1.82]). The proportion of cases treated with partial mastectomy decreased during 2004 to 2012 (eg, APC, -0.77 [95% CI, -2.96 to -0.03] among women aged 50-74 years with localized cancer), whereas the proportion of cases treated with total mastectomy with reconstruction increased (eg, APC, 20.17 [95% CI, 16.50-33.16]). During 2012 to 2019, the proportion of cases treated with total mastectomy decreased (eg, APC, -2.44 [95% CI, -3.45 to -1.61] for women aged 50-74 years with localized cancer), and the proportion of cases treated with partial mastectomy increased (eg, APC, 1.70 [95% CI, 0.90-4.08] for women aged 50-74 years).In this cohort study, in situ breast cancer decreased since 2009, consistent with decreasing use of screening mammography since the 2009 guideline changes, but this decrease did not appear to have translated to more advanced breast cancer stages at diagnosis or decreases in the proportion of cases treated with partial mastectomy. Further research is needed to understand the long-standing increase in localized invasive breast cancer and the decrease in regional invasive breast cancer observed during the past 20 years in the context of decreased breast cancer screening.Zhang-PetersenCarinaCDepartment of Surgery, University of Vermont, Burlington.SowdenMichelleMDepartment of Surgery, University of Vermont, Burlington.University of Vermont Cancer Center, Burlington.ChenJenniferJLarner College of Medicine, University of Vermont, Burlington.BurnsJuliaJDepartment of Surgery, University of Vermont, Burlington.SpragueBrian LBLDepartment of Surgery, University of Vermont, Burlington.University of Vermont Cancer Center, Burlington.engJournal Article20241202
United StatesJAMA Netw Open1017292352574-3805IM
202412271220202412271220202412271133epublish3972931310.1001/jamanetworkopen.2024.526882828497
3972929220241227
1880-42332024Dec27Breast cancer (Tokyo, Japan)Breast CancerClinicopathological significance of androgen receptor expression and tumor infiltrating lymphocytes in triple-negative breast cancer: a retrospective cohort study.10.1007/s12282-024-01662-7Triple-negative breast cancer (TNBC) is a serious disease with limited treatment options. We explored the significance of androgen receptor (AR) expression and tumor-infiltrating lymphocytes (TILs) in predicting neoadjuvant chemotherapy (NAC) resistance in TNBC, hypothesizing that AR/TIL classification using pretreatment biopsies can identify NAC-resistant subgroups and improve the understanding of apocrine differentiation.This retrospective study included 156 consecutive patients with TNBC treated with NAC. AR immunostaining was defined positive if ≥ 1% of the tumor cell nuclei were stained. Stromal TIL levels were assessed, with high levels defined as ≥ 50%. Apocrine differentiation was detected using an anti-15-PGDH antibody. The pathological response to NAC was evaluated.Overall, 36% (n = 56) of the patients achieved a pathological complete response (pCR). AR+/TILlow tumors had a high non-pCR rate (76%, 42/55) and were resistant to NAC. Kaplan-Meier plots showed significant differences in overall survival (OS) and distant metastasis-free survival (DMFS) among the four AR/TIL subgroups (OS: p = 0.013; DMFS: p = 0.0016). All 11 cases with some degree of apocrine differentiation were AR+/TILlow, 15-PGDH-positive, and NAC-resistant. AR+/TILlow status was significantly associated with a high likelihood of non-pCR (OR = 0.26, p = 0.009). Multivariate analysis confirmed pCR as an independent predictor of better prognosis (OS, HR = 0.13, p = 0.006; DMFS, HR = 0.15, p = 0.002), whereas AR+/TILlow status was not significantly associated with OS or DMFS.AR/TIL classification using pretreatment biopsies identified TNBC subgroups with distinct NAC responses and prognoses. AR+/TILlow TNBC, including apocrine differentiation cases, were NAC-resistant, highlighting the need for alternative therapies.© 2024. The Author(s), under exclusive licence to The Japanese Breast Cancer Society.UshigusaTakeshiT0000-0002-5946-3742Department of Pathology, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo-ku, Tokyo, 1048560, Japan. taushig@luke.ac.jp.HirakawaNamiNDepartment of Pathology, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo-ku, Tokyo, 1048560, Japan.KajiuraYukaYDepartment of Breast Surgery, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo-ku, Tokyo, 1048560, Japan.YoshidaAtsushiA0000-0001-5536-132XDepartment of Breast Surgery, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo-ku, Tokyo, 1048560, Japan.YamauchiHidekoHUniversity of Hawai'i Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.KanomataNaokiN0000-0002-0172-4524Department of Pathology, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo-ku, Tokyo, 1048560, Japan.eng22H04343Japan Society for the Promotion of ScienceZK2022-04St. Luke's Health Science Research FundJournal Article20241227
JapanBreast Cancer1008882011340-6868IM15-prostaglandin dehydrogenaseAndrogen receptorApocrineTriple-negative breast cancerTumor-infiltrating lymphocytesDeclarations. Conflict of interest: The authors declare that they have no conflicts of interest related to this article. Ethical approval: The study was approved by the Institutional Review Board (22R010). All procedures performed in studies involving human participants adhered to the ethical standards of the institutional research committee, the 1964 Helsinki Declaration, and its later amendments or comparable ethical standards. Patient consent: Informed consent was obtained from all participants in the study.
202412271220202412271220202461720241220202412271116aheadofprint3972929210.1007/s12282-024-01662-710.1007/s12282-024-01662-7Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:164–72. https://doi.org/10.1016/s0140-6736(13)62422-8 .10.1016/s0140-6736(13)62422-824529560Liedtke C, Mazouni C, Hess KR, André F, Tordai A, Mejia JA, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 2023;41:1809–15. https://doi.org/10.1200/jco.22.02572 .10.1200/jco.22.0257236989609Gerratana L, Basile D, Buono G, De Placido S, Giuliano M, Minichillo S, et al. Androgen receptor in triple negative breast cancer: a potential target for the targetless subtype. Cancer Treat Rev. 2018;68:102–10. https://doi.org/10.1016/j.ctrv.2018.06.005 .10.1016/j.ctrv.2018.06.00529940524Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121:2750–67. https://doi.org/10.1172/JCI45014 .10.1172/JCI45014216331663127435Thompson KJ, Leon-Ferre RA, Sinnwell JP, Zahrieh DM, Suman VJ, Metzger FO, et al. Luminal androgen receptor breast cancer subtype and investigation of the microenvironment and neoadjuvant chemotherapy response. NAR Cancer. 2022. https://doi.org/10.1093/narcan/zcac018 .10.1093/narcan/zcac018357343919204893Masuda H, Baggerly KA, Wang Y, Zhang Y, Gonzalez-Angulo AM, Meric-Bernstam F, et al. Differential response to neoadjuvant chemotherapy among 7 triple-negative breast cancer molecular subtypes. Clin Cancer Res. 2013;19:5533–40. https://doi.org/10.1158/1078-0432.Ccr-13-0799 .10.1158/1078-0432.Ccr-13-079923948975Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol. 2015;26:259–71. https://doi.org/10.1093/annonc/mdu450 .10.1093/annonc/mdu45025214542Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018;19:40–50. https://doi.org/10.1016/s1470-2045(17)30904-x .10.1016/s1470-2045(17)30904-x29233559El Bairi K, Haynes HR, Blackley E, Fineberg S, Shear J, Turner S, et al. The tale of TILs in breast cancer: a report from the international immuno-oncology biomarker working group. NPJ Breast Cancer. 2021;7:150. https://doi.org/10.1038/s41523-021-00346-1 .10.1038/s41523-021-00346-1348533558636568Gatalica Z, Kuzmova N, Rose I, Ulamec M, Peric-Balja M, Skenderi F, et al. The assessment of tumor-infiltrating lymphocytes in invasive apocrine carcinoma of the breast in relation to the HER2 status. Biomol Biomed. 2024;24:256–61. https://doi.org/10.17305/bb.2023.9868 .10.17305/bb.2023.98683778256210950344Sun X, Zuo K, Yao Q, Zhou S, Shui R, Xu X, et al. Invasive apocrine carcinoma of the breast: clinicopathologic features and comprehensive genomic profiling of 18 pure triple-negative apocrine carcinomas. Mod Pathol. 2020;33:2473–82. https://doi.org/10.1038/s41379-020-0589-x .10.1038/s41379-020-0589-x32504034Board WCoTE. Breast tumours. World Health Organization classification of tumours. 5th ed. Lyon: International Agency for Research on Cancer; 2019.Schwartz CJ, Ruiz J, Bean GR, Sirohi D, Joseph NM, Hosfield EM, et al. Triple-negative apocrine carcinomas: toward a unified group with shared molecular features and clinical behavior. Mod Pathol. 2023;36:100125. https://doi.org/10.1016/j.modpat.2023.100125 .10.1016/j.modpat.2023.10012536870308Wolff AC, Somerfield MR, Dowsett M, Hammond MEH, Hayes DF, McShane LM, et al. Human epidermal growth factor receptor 2 testing in breast cancer: ASCO–College of American pathologists guideline update. J Clin Oncol. 2023;41:3867–72. https://doi.org/10.1200/jco.22.02864 .10.1200/jco.22.0286437284804Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American society of clinical oncology/College of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 2010;28:2784–95. https://doi.org/10.1200/jco.2009.25.6529 .10.1200/jco.2009.25.6529204042512881855(UICC) UfICC. TNM classification of malignant tumours. 8th ed. Oxford: Wiley-Blackwell; 2017.Celis JE, Cabezón T, Moreira JM, Gromov P, Gromova I, Timmermans-Wielenga V, et al. Molecular characterization of apocrine carcinoma of the breast: validation of an apocrine protein signature in a well-defined cohort. Mol Oncol. 2009;3:220–37. https://doi.org/10.1016/j.molonc.2009.01.005 .10.1016/j.molonc.2009.01.005193935835527852Hanamura T, Kitano S, Kagamu H, Yamashita M, Terao M, Okamura T, et al. Expression of hormone receptors is associated with specific immunological profiles of the breast cancer microenvironment. Breast Cancer Res. 2023;25:13. https://doi.org/10.1186/s13058-023-01606-7 .10.1186/s13058-023-01606-7367212189887885Leon-Ferre RA, Jonas SF, Salgado R, Loi S, De Jong V, Carter JM, et al. Tumor-infiltrating lymphocytes in triple-negative breast cancer. JAMA. 2024;331:1135–44. https://doi.org/10.1001/jama.2024.3056 .10.1001/jama.2024.30563856383410988354Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA, et al. Phase II trial of bicalutamide in patients with androgen receptor–positive, estrogen receptor–negative metastatic breast cancer. Clin Cancer Res. 2013;19:5505–12. https://doi.org/10.1158/1078-0432.Ccr-12-3327 .10.1158/1078-0432.Ccr-12-3327239659014086643Traina TA, Miller K, Yardley DA, Eakle J, Schwartzberg LS, O’Shaughnessy J, et al. Enzalutamide for the treatment of androgen receptor–expressing triple-negative breast cancer. J Clin Oncol. 2018;36:884–90. https://doi.org/10.1200/jco.2016.71.3495 .10.1200/jco.2016.71.3495293730715858523Bonnefoi H, Grellety T, Tredan O, Saghatchian M, Dalenc F, Mailliez A, et al. A phase II trial of abiraterone acetate plus prednisone in patients with triple-negative androgen receptor positive locally advanced or metastatic breast cancer (UCBG 12–1). Ann Oncol. 2016;27:812–8. https://doi.org/10.1093/annonc/mdw067 .10.1093/annonc/mdw06727052658Jiang Y-Z, Liu Y, Xiao Y, Hu X, Jiang L, Zuo W-J, et al. Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: the FUTURE trial. Cell Res. 2021;31:178–86. https://doi.org/10.1038/s41422-020-0375-9 .10.1038/s41422-020-0375-932719455Hu T, Liu Y, Wu J, Hu XL, Zhao G, Liang B, et al. Triple-negative apocrine breast carcinoma has better prognosis despite poor response to neoadjuvant chemotherapy. J Clin Med. 2022;11:1607. https://doi.org/10.3390/jcm11061607 .10.3390/jcm11061607353299348949126Ogiya A, Horii R, Osako T, Ito Y, Iwase T, Eishi Y, et al. Apocrine metaplasia of breast cancer: clinicopathological features and predicting response. Breast Cancer. 2010;17:290–7. https://doi.org/10.1007/s12282-009-0178-9 .10.1007/s12282-009-0178-919789945Celis JE, Gromov P, Cabezón T, Moreira JM, Friis E, Jirström K, et al. 15-prostaglandin dehydrogenase expression alone or in combination with ACSM1 defines a subgroup of the apocrine molecular subtype of breast carcinoma. Mol Cell Proteomics. 2008;7:1795–809. https://doi.org/10.1074/mcp.R800011-MCP200 .10.1074/mcp.R800011-MCP20018632593Tai H-H. Prostaglandin catabolic enzymes as tumor suppressors. Cancer Metastasis Rev. 2011;30:409–17. https://doi.org/10.1007/s10555-011-9314-z .10.1007/s10555-011-9314-z22020925Asano Y, Kashiwagi S, Onoda N, Kurata K, Morisaki T, Noda S, et al. Clinical verification of sensitivity to preoperative chemotherapy in cases of androgen receptor-expressing positive breast cancer. Br J Cancer. 2016;114:14–20. https://doi.org/10.1038/bjc.2015.434 .10.1038/bjc.2015.434267574224716546Zuo T, Wilson P, Cicek AF, Harigopal M. Androgen receptor expression is a favorable prognostic factor in triple-negative breast cancers. Hum Pathol. 2018;80:239–45. https://doi.org/10.1016/j.humpath.2018.06.003 .10.1016/j.humpath.2018.06.00329902579Astvatsaturyan K, Yue Y, Walts AE, Bose S. Androgen receptor positive triple negative breast cancer: clinicopathologic, prognostic, and predictive features. PLoS ONE. 2018;13:e0197827. https://doi.org/10.1371/journal.pone.0197827 .10.1371/journal.pone.0197827298834875993259Sridhar N, Glisch C, Jawa Z, Chaudhary LN, Kamaraju S, Burfeind J, et al. Androgen receptor expression in patients with triple negative breast cancer treated with neoadjuvant chemotherapy: a single institution study. J Cancer. 2022;13:2472–6. https://doi.org/10.7150/jca.67536 .10.7150/jca.67536357118339174868Choi JE, Kang SH, Lee SJ, Bae YK. Androgen receptor expression predicts decreased survival in early stage triple-negative breast cancer. Ann Surg Oncol. 2015;22:82–9. https://doi.org/10.1245/s10434-014-3984-z .10.1245/s10434-014-3984-z25145503Thike AA, Chong LY-Z, Cheok PY, Li HH, Wai-Cheong Yip G, Huat Bay B, et al. Loss of androgen receptor expression predicts early recurrence in triple-negative and basal-like breast cancer. Mod Pathol. 2014;27:352–60. https://doi.org/10.1038/modpathol.2013.145 .10.1038/modpathol.2013.14523929266Mangia A, Saponaro C, Vagheggini A, Opinto G, Centonze M, Vicenti C, et al. Should tumor infiltrating lymphocytes, androgen receptor, and FOXA1 expression predict the clinical outcome in triple negative breast cancer patients? Cancers. 2019;11:1393. https://doi.org/10.3390/cancers11091393 .10.3390/cancers11091393315404866769726Asano Y, Kashiwagi S, Goto W, Tanaka S, Morisaki T, Takashima T, et al. Expression and clinical significance of androgen receptor in triple-negative breast cancer. Cancers. 2017;9:4. https://doi.org/10.3390/cancers9010004 .10.3390/cancers9010004280678095295775
3972929120241227
1880-42332024Dec27Breast cancer (Tokyo, Japan)Breast CancerIdentifying subgroups of ypN1 breast cancer patients who may exempt from axillary lymph node dissection after neoadjuvant chemotherapy: insights from a large cohort study.10.1007/s12282-024-01663-6In patients with breast cancer staged ypN1 after neoadjuvant chemotherapy (NAC), there is limited evidence-based guidance regarding exemption from axillary lymph node dissection (ALND).This study analyzed ypN1 breast cancer patients post-NAC from the Surveillance, Epidemiology, and End Results databases. Patients were categorized into the breast-conserving surgery (BCS) group and the total mastectomy (TM) group, and further divided by the number of positive lymph nodes (LNs). The effects of three axillary management strategies, ALND, sentinel lymph node biopsy combined with radiotherapy (SLNB + RT), and ALND + RT were compared. The overall survival (OS) and breast cancer-specific survival (BCSS) of all subgroups and their independent risk factors were analyzed. Independent prognostic factors selected from multivariate Cox analysis were utilized to create nomograms for predicting OS and BCSS.A total of 3641 patients were involved, with 1331 in the BCS group and 2310 in the TM group. In the TM group, patients with 3 residual positive LNs exhibited significant improvements in OS and BCSS when treated with ALND + RT. For patients with 1 or 2 residual positive LNs in the TM group and all BCS patients, no significant survival differences in survival outcomes were observed among the three axillary management methods. The accuracy of the nomograms was validated via calibration curves, receiver operating characteristic curves, and decision curve analysis curves.For TM group patients with 3 residual positive LNs after NAC, ALND + RT is recommended. For other subgroups of ypN1 patients, SLNB + RT can be considered an alternative to ALND. The nomogram developed to predict OS and BCSS in ypN1 breast cancer patients demonstrated excellent predictive ability.© 2024. The Author(s), under exclusive licence to The Japanese Breast Cancer Society.LiuPeinanPThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.LiuDandanDThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.ZhaoChangyingCDepartment of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.WeiYumengYThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.LiuXingyuXThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.CuiHanxiaoHThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.ZhaoXuyanXThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.ChangLidanLThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.LinShuaiSThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.WuHaoHSchool of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.MaXiaobinXThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.KangHuafengHThe Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China. kanghuafeng1973@126.com.WangMengM0000-0002-7417-9192The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China. wmeng0308@126.com.eng2020YJ (ZYTS)625Xi'an Jiaotong UniversityJournal Article20241227
JapanBreast Cancer1008882011340-6868IMAxillary lymph node dissectionBreast cancerNeoadjuvant chemotherapySentinel lymph node biopsyDeclarations. Conflict of interest: All authors declare that they have no conflicts of interest to disclose. Ethical approval and informed consent. As the SEER database used in this study does not contain personally identifiable information, patient informed consent is not needed. The study received an exemption from the Ethics Committee of the Second Affiliated Hospital of Xi'an Jiaotong University. Consent for publication: All the authors declare that this manuscript is original work and has not been published previously, nor is it under consideration for publication elsewhere. All sources of funding and potential conflicts of interest have been disclosed.
202412271220202412271220202410920241220202412271116aheadofprint3972929110.1007/s12282-024-01663-610.1007/s12282-024-01663-6Bray F, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca-a Cancer J Clin. 2024;74:229–63. https://doi.org/10.3322/caac.21834 .10.3322/caac.21834Veronesi U, et al. A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med. 2003;349:546–53. https://doi.org/10.1056/NEJMoa012782 .10.1056/NEJMoa01278212904519Caudle AS, et al. Improved axillary evaluation following neoadjuvant therapy for patients with node-positive breast cancer using selective evaluation of clipped nodes: implementation of targeted axillary dissection. J Clin Oncol. 2016;34:1072–8. https://doi.org/10.1200/JCO.2015.64.0094 .10.1200/JCO.2015.64.0094268115284933133Fleissig A, et al. Post-operative arm morbidity and quality of life. Results of the ALMANAC randomised trial comparing sentinel node biopsy with standard axillary treatment in the management of patients with early breast cancer. Breast Cancer Res Treatment. 2006. https://doi.org/10.1007/s10549-005-9025-7 .10.1007/s10549-005-9025-7Mansel RE, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: The ALMANAC trial. J Natl Cancer Inst. 2006;98:599–609. https://doi.org/10.1093/jnci/djj158 .10.1093/jnci/djj15816670385Land SR, et al. Patient-reported outcomes in sentinel node-negative adjuvant breast cancer patients receiving sentinel-node biopsy or axillary dissection: National Surgical Adjuvant Breast and Bowel Project phase III protocol B-32. J Clin Oncol. 2010;28:3929–36. https://doi.org/10.1200/JCO.2010.28.2491 .10.1200/JCO.2010.28.2491206796002940391Giuliano AE, et al. Locoregional recurrence after sentinel lymph node dissection with or without axillary dissection in patients with sentinel lymph node metastases: long-term follow-up from the american college of surgeons oncology group (alliance) acosog z0011 randomized trial. Ann Surg. 2016;264:413–20. https://doi.org/10.1097/SLA.0000000000001863 .10.1097/SLA.000000000000186327513155Giuliano AE, et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis the ACOSOG Z0011 (Alliance) randomized clinical trial. Jama-J the Am Med Assoc. 2017;318:918–26. https://doi.org/10.1001/jama.2017.11470 .10.1001/jama.2017.11470Donker M, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer (EORTC 10981–22023 AMAROS): a randomised, multicentre, open-label, phase 3 non-inferiority trial. Lancet Oncol. 2014;15:1303–10. https://doi.org/10.1016/s1470-2045(14)70460-7 .10.1016/s1470-2045(14)70460-7254396884291166Tinterri C, et al. Preservation of axillary lymph nodes compared with complete dissection in t1–2 breast cancer patients presenting one or two metastatic sentinel lymph nodes: the sinodar-one multicenter randomized clinical trial. Ann Surg Oncol. 2022;29:5732–44. https://doi.org/10.1245/s10434-022-11866-w .10.1245/s10434-022-11866-w35552930Frei E. Clinical cancer-research - an embattled species. Cancer. 1982;50:1979–92.10.1002/1097-0142(19821115)50:10<1979::AID-CNCR2820501002>3.0.CO;2-D7127245Shien T, Iwata H. Adjuvant and neoadjuvant therapy for breast cancer. Jpn J Clin Oncol. 2020;50:225–9. https://doi.org/10.1093/jjco/hyz213 .10.1093/jjco/hyz21332147701Rastogi P, et al. Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J Clin Oncol. 2008;26:778–85. https://doi.org/10.1200/jco.2007.15.0235 .10.1200/jco.2007.15.023518258986van der Hage JA, et al. Preoperative chemotherapy in primary operable breast cancer: results from the european organization for research and treatment of cancer trial 10902. J Clin Oncol. 2001;19:4224–37. https://doi.org/10.1200/jco.2001.19.22.4224 .10.1200/jco.2001.19.22.422411709566Boughey JC, et al. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast cancer the ACOSOG Z1071 (Alliance) clinical trial. Jama-J Am Med Assoc. 2013;310:1455–61. https://doi.org/10.1001/jama.2013.278932 .10.1001/jama.2013.278932Heidinger M, Knauer M, Tausch C, Weber WP. Tailored axillary surgery - A novel concept for clinically node positive breast cancer. Breast. 2023;69:281–9. https://doi.org/10.1016/j.breast.2023.03.005 .10.1016/j.breast.2023.03.0053692230510034500Kunkler IH, Canney P, van Tienhoven G, Russell NS. Elucidating the role of chest wall irradiation in “intermediate-risk” breast cancer: the MRC/EORTC SUPREMO trial. Clin Oncol (R Coll Radiol). 2008;20:31–4.10.1016/j.clon.2007.10.00418345543Pilewskie M, Morrow M. Axillary nodal management following neoadjuvant chemotherapy: a review. JAMA Oncol. 2017;3:549–55. https://doi.org/10.1001/jamaoncol.2016.4163 .10.1001/jamaoncol.2016.4163279187535580251Giuliano AE, et al. Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: a randomized clinical trial. Arch Surg. 2011;146:980–980.EBCTCG. Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 2018. https://doi.org/10.1016/S1470-2045(17)30777-5 .10.1016/S1470-2045(17)30777-5Barrio AV, et al. Nodal recurrence in patients with node-positive breast cancer treated with sentinel node biopsy alone after neoadjuvant chemotherapy-a rare event. JAMA Oncol. 2021;7:1851–5. https://doi.org/10.1001/jamaoncol.2021.4394 .10.1001/jamaoncol.2021.439434617979Kuehn T, et al. Sentinel-lymph-node biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA): a prospective, multicentre cohort study. Lancet Oncology. 2013;14:609–18. https://doi.org/10.1016/s1470-2045(13)70166-9 .10.1016/s1470-2045(13)70166-923683750Boileau J-F, et al. Sentinel node biopsy after neoadjuvant chemotherapy in biopsy-proven node-positive breast cancer: the SN FNAC study. J Clin Oncol. 2015;33:258-U150. https://doi.org/10.1200/jco.2014.55.7827 .10.1200/jco.2014.55.782725452445Classe J-M, et al. Sentinel lymph node biopsy without axillary lymphadenectomy after neoadjuvant chemotherapy is accurate and safe for selected patients: the GANEA 2 study. Breast Cancer Res Treat. 2019;173:343–52. https://doi.org/10.1007/s10549-018-5004-7 .10.1007/s10549-018-5004-730343457Montagna G, et al. Omission of axillary dissection following nodal downstaging with neoadjuvant chemotherapy. JAMA Oncol. 2024;10:831–2. https://doi.org/10.1001/jamaoncol.2024.0578 .10.1001/jamaoncol.2024.0578Magnoni F, et al. Axillary surgery in breast cancer: An updated historical perspective. Semin Oncol. 2020;47:341–52. https://doi.org/10.1053/j.seminoncol.2020.09.001 .10.1053/j.seminoncol.2020.09.00133131896Beck AC, Morrow M. Axillary lymph node dissection: Dead or still alive? Breast. 2023;69:469–75. https://doi.org/10.1016/j.breast.2023.01.009 .10.1016/j.breast.2023.01.0093670267210300611Gerber B, et al. Axillary lymph node dissection in early-stage invasive breast cancer: is it still standard today? Breast Cancer Res Treat. 2011;128:613–24. https://doi.org/10.1007/s10549-011-1532-0 .10.1007/s10549-011-1532-021523451Harlow SP, et al. Prerandomization surgical training for the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-32 trial - A randomized phase III clinical trial to compare sentinel node resection to conventional axillary dissection in clinically node-negative breast cancer. Ann Surg. 2005;241:48–54. https://doi.org/10.1097/01.sla.0000149429.39656.94 .10.1097/01.sla.0000149429.39656.94156219901356845Galimberti V, et al. Axillary dissection versus no axillary dissection in patients with sentinel-node micrometastases (IBCSG 23–01): a phase 3 randomised controlled trial. Lancet Oncology. 2013;14:297–305. https://doi.org/10.1016/s1470-2045(13)70035-4 .10.1016/s1470-2045(13)70035-423491275Tinterri C, et al. Sentinel lymph node biopsy versus axillary lymph node dissection in breast cancer patients undergoing mastectomy with one to two metastatic sentinel lymph nodes: sub-analysis of the SINODAR-ONE multicentre randomized clinical trial and reopening of enrolment. Br J Surg. 2023;110:1143–52. https://doi.org/10.1093/bjs/znad215 .10.1093/bjs/znad2153747157410492188Cao S, et al. Feasibility and reliability of sentinel lymph node biopsy after neoadjuvant chemotherapy in breast cancer patients with positive axillary nodes at initial diagnosis: An up-to-date meta-analysis of 3,578 patients. Breast (Edinburgh, Scotland). 2021;59:256–69. https://doi.org/10.1016/j.breast.2021.07.015 .10.1016/j.breast.2021.07.01534325383Swarnkar PK, Tayeh S, Michell MJ, Mokbel K. The Evolving role of marked lymph node biopsy (MLNB) and targeted axillary dissection (TAD) after neoadjuvant chemotherapy (NACT) for node-positive breast cancer: systematic review and pooled analysis. Cancers (Basel). 2021. https://doi.org/10.3390/cancers13071539 .10.3390/cancers1307153933810544Gasparri ML, et al. Axillary surgery after neoadjuvant therapy in initially node-positive breast cancer: international EUBREAST survey. Br J Surg. 2022;109:857–63. https://doi.org/10.1093/bjs/znac217 .10.1093/bjs/znac21735766257Choy N, et al. Initial results with preoperative tattooing of biopsied axillary lymph nodes and correlation to sentinel lymph nodes in breast cancer patients. Ann Surg Oncol. 2015;22:377–82. https://doi.org/10.1245/s10434-014-4034-6 .10.1245/s10434-014-4034-625164040Caudle AS, et al. Improved axillary evaluation following neoadjuvant therapy for patients with node-positive breast cancer using selective evaluation of clipped nodes: implementation of targeted axillary dissection. J Clin Oncol. 2016. https://doi.org/10.1200/jco.2015.64.0094 .10.1200/jco.2015.64.0094268115284933133Diego EJ, et al. Axillary staging after neoadjuvant chemotherapy for breast cancer: a pilot study combining sentinel lymph node biopsy with radioactive seed localization of pre-treatment positive axillary lymph nodes. Ann Surg Oncol. 2016;23:1549–53. https://doi.org/10.1245/s10434-015-5052-8 .10.1245/s10434-015-5052-826727919Ling DC, Iarrobino NA, Champ CE, Soran A, Beriwal S. Regional recurrence rates with or without complete axillary dissection for breast cancer patients with node-positive disease on sentinel lymph node biopsy after neoadjuvant chemotherapy. Adv Radiat Oncol. 2020;5:163–70. https://doi.org/10.1016/j.adro.2019.09.006 .10.1016/j.adro.2019.09.00632280815Almahariq MF, et al. Omission of axillary lymph node dissection is associated with inferior survival in breast cancer patients with residual n1 nodal disease following neoadjuvant chemotherapy. Ann Surg Oncol. 2021;28:930–40. https://doi.org/10.1245/s10434-020-08928-2 .10.1245/s10434-020-08928-232712895Zhu T, et al. Multifactor artificial intelligence model assists axillary lymph node surgery in breast cancer after neoadjuvant chemotherapy: multicenter retrospective cohort study. Int J Surg. 2023;109:3383–94. https://doi.org/10.1097/js9.0000000000000621 .10.1097/js9.00000000000006213783094310651262
3972924720241227
1432-133515112024Dec27Journal of cancer research and clinical oncologyJ Cancer Res Clin OncolClinicopathological characteristics and long-term prognosis of triple-negative breast cancer patients with HER2-Low expression: a retrospective propensity score-matched cohort study.242410.1007/s00432-024-06069-7The objective of the current research was to assess the clinicopathological characteristics and long-term prognosis of triple-negative breast cancer (TNBC) patients with human epidermal growth factor receptor 2 (HER2)-low status following breast surgery.A total of 202 TNBC patients treated at Qingdao Central Hospital from January 2010 to December 2019 were included, comprising 71 HER2-low and 131 HER2-zero patients. Propensity score matching (PSM) was applied to minimize differences between the cohorts.HER2-low TNBC patients had lower histological grade, lower Ki-67 expression levels, and a higher prevalence of hypertension compared to HER2-zero TNBC patients. Before and after PSM, the HER2-low group consistently exhibited a lower recurrence rate and longer RFS compared to HER2-zero TNBC patients. HER2-low status was validated as an independent low-risk factor for RFS both pre-PSM (HR 0.354, 95% CI 0.178-0.706, p = 0.003) and post-PSM (HR 0.405, 95% CI 0.185-0.886, p = 0.024). No statistically significant differences in mortality rate and OS were observed, both before and after PSM.HER2-low and HER2-zero TNBC patients show significant clinicopathological differences. Compared to HER2-zero, HER2-low status is linked to better long-term prognosis and serves as an independent low-risk factor for RFS in TNBC patients.© 2024. The Author(s).LiuXinXQingdao Medical College, Qingdao University, Qingdao, 266071, Shandong, China.Department of Breast Surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, 266042, Shandong, China.ZhaoKaihuaKDepartment of Breast Surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, 266042, Shandong, China.ZhangZiyanZDepartment of Breast Surgery, Women and Children's Hospital, Qingdao University, Qingdao, 266034, Shandong, China.LiuMeiyanMQingdao Medical College, Qingdao University, Qingdao, 266071, Shandong, China.ChuHongwuHGuangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China. chuhw5@mail2.sysu.edu.cn.ZouXiaoXDepartment of Breast Surgery, Xiangdong Hospital Affiliated to Hunan Normal University, Liling, 412200, Hunan, China. 18660229101@qq.com.engJournal Article20241227
GermanyJ Cancer Res Clin Oncol79020600171-5216IMClinicopathological characteristicsHER2-LowLong-term prognosisTriple-negative breast cancerDeclarations. Ethical approval: The study was conducted in accordance with the Declaration of Helsinki and was approved by the Ethics Committee of Qingdao Central Hospital. Written informed consent requirement was waived by the Ethics Committee. Consent to publish: Not applicable. Competing interests: The authors declare no competing interests.
2024122712202024122712202024111320241217202412271114epublish3972924710.1007/s00432-024-06069-710.1007/s00432-024-06069-7Allison KH, Hammond MEH, Dowsett M, McKernin SE, Carey LA, Fitzgibbons PL et al (2020) Estrogen and progesterone receptor testing in breast Cancer: ASCO/CAP Guideline Update. J Clin Oncol 38(12):1346–1366. https://doi.org/10.1200/jco.19.0230910.1200/jco.19.0230931928404Almstedt K, Heimes AS, Kappenberg F, Battista MJ, Lehr HA, Krajnak S et al (2022) Long-term prognostic significance of HER2-low and HER2-zero in node-negative breast cancer. Eur J Cancer 173:10–19. https://doi.org/10.1016/j.ejca.2022.06.01210.1016/j.ejca.2022.06.01235839597Berrino E, Annaratone L, Bellomo SE, Ferrero G, Gagliardi A, Bragoni A et al (2022) Integrative genomic and transcriptomic analyses illuminate the ontology of HER2-low breast carcinomas. Genome Med 14(1):98. https://doi.org/10.1186/s13073-022-01104-z10.1186/s13073-022-01104-z360388849426037Bianchini G, De Angelis C, Licata L, Gianni L (2022) Treatment landscape of triple-negative breast cancer - expanded options, evolving needs. Nat Rev Clin Oncol 19(2):91–113. https://doi.org/10.1038/s41571-021-00565-210.1038/s41571-021-00565-234754128Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I et al (2024) Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 74(3):229–263. https://doi.org/10.3322/caac.2183410.3322/caac.2183438572751Chen L, Liu CC, Zhu SY, Ge JY, Chen YF, Ma D et al (2023) Multiomics of HER2-low triple-negative breast cancer identifies a receptor tyrosine kinase-relevant subgroup with therapeutic prospects. JCI Insight 8(22). https://doi.org/10.1172/jci.insight.172366Corti C, Giugliano F, Nicolò E, Tarantino P, Criscitiello C, Curigliano G (2023) HER2-Low breast Cancer: a New Subtype? Curr Treat Options Oncol 24(5):468–478. https://doi.org/10.1007/s11864-023-01068-110.1007/s11864-023-01068-136971965Dai LJ, Ma D, Xu YZ, Li M, Li YW, Xiao Y et al (2023) Molecular features and clinical implications of the heterogeneity in Chinese patients with HER2-low breast cancer. Nat Commun 14(1):5112. https://doi.org/10.1038/s41467-023-40715-x10.1038/s41467-023-40715-x3760791610444861Denkert C, Seither F, Schneeweiss A, Link T, Blohmer JU, Just M et al (2021) Clinical and molecular characteristics of HER2-low-positive breast cancer: pooled analysis of individual patient data from four prospective, neoadjuvant clinical trials. Lancet Oncol 22(8):1151–1161. https://doi.org/10.1016/s1470-2045(21)00301-610.1016/s1470-2045(21)00301-634252375Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA et al (2007) Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13(15 Pt 1):4429–4434. https://doi.org/10.1158/1078-0432.Ccr-06-304510.1158/1078-0432.Ccr-06-304517671126Gampenrieder SP, Dezentjé V, Lambertini M, de Nonneville A, Marhold M, Le Du F et al (2023) Influence of HER2 expression on prognosis in metastatic triple-negative breast cancer-results from an international, multicenter analysis coordinated by the AGMT Study Group. ESMO Open 8(1):100747. https://doi.org/10.1016/j.esmoop.2022.10074710.1016/j.esmoop.2022.10074736563519Giuliano AE, Edge SB, Hortobagyi GN (2018) Eighth Edition of the AJCC Cancer staging Manual: breast Cancer. Ann Surg Oncol 25(7):1–3. https://doi.org/10.3389/fonc.2023.115778910.3389/fonc.2023.1157789Han BY, Chen C, Luo H, Lin CJ, Han XC, Nasir J et al (2024) Clinical sequencing defines the somatic and germline mutation landscapes of Chinese HER2-Low breast Cancer. Cancer Lett 588:216763. https://doi.org/10.1016/j.canlet.2024.21676310.1016/j.canlet.2024.21676338403109Horisawa N, Adachi Y, Takatsuka D, Nozawa K, Endo Y, Ozaki Y et al (2022) The frequency of low HER2 expression in breast cancer and a comparison of prognosis between patients with HER2-low and HER2-negative breast cancer by HR status. Breast Cancer 29(2):234–241. https://doi.org/10.1007/s12282-021-01303-310.1007/s12282-021-01303-334622383Hu XE, Yang P, Chen S, Wei G, Yuan L, Yang Z et al (2023) Clinical and biological heterogeneities in triple-negative breast cancer reveals a non-negligible role of HER2-low. Breast Cancer Res 25(1):34. https://doi.org/10.1186/s13058-023-01639-y10.1186/s13058-023-01639-y3699801410061837Jin J, Li B, Cao J, Li T, Zhang J, Cao J et al (2023) Analysis of clinical features, genomic landscapes and survival outcomes in HER2-low breast cancer. J Transl Med 21(1):360. https://doi.org/10.1186/s12967-023-04076-910.1186/s12967-023-04076-93726441710236705Li Y, Sun Y, Kulke M, Hechler T, Van der Jeught K, Dong T et al (2021) Targeted immunotherapy for HER2-low breast cancer with 17p loss. Sci Transl Med 13(580). https://doi.org/10.1126/scitranslmed.abc6894Li Y, Zhang H, Merkher Y, Chen L, Liu N, Leonov S et al (2022) Recent advances in therapeutic strategies for triple-negative breast cancer. J Hematol Oncol 15(1):121. https://doi.org/10.1186/s13045-022-01341-010.1186/s13045-022-01341-0360389139422136Li Y, Tsang JY, Tam F, Loong T, Tse GM (2023) Comprehensive characterization of HER2-low breast cancers: implications in prognosis and treatment. EBioMedicine 91:104571. https://doi.org/10.1016/j.ebiom.2023.10457110.1016/j.ebiom.2023.1045713706834910130469Li H, Plichta JK, Li K, Jin Y, Thomas SM, Ma F et al (2024) Impact of HER2-low status for patients with early-stage breast cancer and non-pCR after neoadjuvant chemotherapy: a National Cancer Database Analysis. Breast Cancer Res Treat 204(1):89–105. https://doi.org/10.1007/s10549-023-07171-z10.1007/s10549-023-07171-z38066250Liu Y, Hu Y, Xue J, Li J, Yi J, Bu J et al (2023) Advances in immunotherapy for triple-negative breast cancer. Mol Cancer 22(1):145. https://doi.org/10.1186/s12943-023-01850-710.1186/s12943-023-01850-73766003910474743Löb S, Linsmeier E, Herbert SL, Schlaiß T, Kiesel M, Wischhusen J et al (2023) Prognostic effect of HER2 evolution from primary breast cancer to breast cancer metastases. J Cancer Res Clin Oncol 149(8):5417–5428. https://doi.org/10.1007/s00432-022-04486-010.1007/s00432-022-04486-036451043Miglietta F, Griguolo G, Bottosso M, Giarratano T, Lo Mele M, Fassan M et al (2021) Evolution of HER2-low expression from primary to recurrent breast cancer. NPJ Breast Cancer 7(1):137. https://doi.org/10.1038/s41523-021-00343-410.1038/s41523-021-00343-4346423488511010Modi S, Jacot W, Yamashita T, Sohn J, Vidal M, Tokunaga E et al (2022) Trastuzumab Deruxtecan in previously treated HER2-Low advanced breast Cancer. N Engl J Med 387(1):9–20. https://doi.org/10.1056/NEJMoa220369010.1056/NEJMoa22036903566578210561652Peiffer DS, Zhao F, Chen N, Hahn OM, Nanda R, Olopade OI et al (2023) Clinicopathologic characteristics and prognosis of ERBB2-Low breast Cancer among patients in the National Cancer Database. JAMA Oncol 9(4):500–510. https://doi.org/10.1001/jamaoncol.2022.747610.1001/jamaoncol.2022.7476368211259951099Sanomachi T, Okuma HS, Kitadai R, Kawachi A, Yazaki S, Tokura M et al (2023) Low HER2 expression is a predictor of poor prognosis in stage I triple-negative breast cancer. Front Oncol 13:1157789. https://doi.org/10.3389/fonc.2023.115778910.3389/fonc.2023.11577893705154510083471Schettini F, Chic N, Brasó-Maristany F, Paré L, Pascual T, Conte B et al (2021) Clinical, pathological, and PAM50 gene expression features of HER2-low breast cancer. NPJ Breast Cancer 7(1):1. https://doi.org/10.1038/s41523-020-00208-210.1038/s41523-020-00208-2333979687782714Schettini F, Blondeaux E, Molinelli C, Bas R, Kim HJ, Di Meglio A et al (2024) Characterization of HER2-low breast cancer in young women with germline BRCA1/2 pathogenetic variants: results of a large international retrospective cohort study. Cancer 130(16):2746–2762. https://doi.org/10.1002/cncr.3532310.1002/cncr.3532338752572Shao Y, Guan H, Luo Z, Yu Y, He Y, Chen Q et al (2024) Clinicopathological characteristics and value of HER2-low expression evolution in breast cancer receiving neoadjuvant chemotherapy. Breast 73:103666. https://doi.org/10.1016/j.breast.2023.10366610.1016/j.breast.2023.10366638159433Shi Z, Liu Y, Fang X, Liu X, Meng J, Zhang J (2024) Efficacy and prognosis of HER2-Low and HER2-Zero in triple-negative breast cancer after neoadjuvant chemotherapy. Sci Rep 14(1):16899. https://doi.org/10.1038/s41598-024-67795-z10.1038/s41598-024-67795-z3904375611266405So JY, Ohm J, Lipkowitz S, Yang L (2022) Triple negative breast cancer (TNBC): non-genetic tumor heterogeneity and immune microenvironment: emerging treatment options. Pharmacol Ther 237:108253. https://doi.org/10.1016/j.pharmthera.2022.10825310.1016/j.pharmthera.2022.108253358723329378710Tan R, Ong WS, Lee KH, Lim AH, Park S, Park YH et al (2022) HER2 expression, copy number variation and survival outcomes in HER2-low non-metastatic breast cancer: an international multicentre cohort study and TCGA-METABRIC analysis. BMC Med 20(1):105. https://doi.org/10.1186/s12916-022-02284-610.1186/s12916-022-02284-6352963008928638Tang Y, Shen G, Xin Y, Li Z, Zheng Y, Wang M et al (2023) The association between HER2-low expression and prognosis of breast cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 15:17588359231156669. https://doi.org/10.1177/1758835923115666910.1177/17588359231156669368729489983100Tarantino P, Hamilton E, Tolaney SM, Cortes J, Morganti S, Ferraro E et al (2020) HER2-Low breast Cancer: pathological and clinical Landscape. J Clin Oncol 38(17):1951–1962. https://doi.org/10.1200/jco.19.0248810.1200/jco.19.0248832330069Tarantino P, Gandini S, Nicolò E, Trillo P, Giugliano F, Zagami P et al (2022) Evolution of low HER2 expression between early and advanced-stage breast cancer. Eur J Cancer 163:35–43. https://doi.org/10.1016/j.ejca.2021.12.02210.1016/j.ejca.2021.12.02235032815Tarantino P, Viale G, Press MF, Hu X, Penault-Llorca F, Bardia A et al (2023) ESMO expert consensus statements (ECS) on the definition, diagnosis, and management of HER2-low breast cancer. Ann Oncol 34(8):645–659. https://doi.org/10.1016/j.annonc.2023.05.00810.1016/j.annonc.2023.05.00837269905Tuluhong D, Li X, Gao H, Zhu Y, Li Q, Wang S (2023) Molecular characteristics and prognosis of breast cancer patients with different level of HER2 positivity after adjuvant and neoadjuvant chemotherapy. Eur J Cancer Prev 32(4):377–387. https://doi.org/10.1097/cej.000000000000081310.1097/cej.000000000000081337302017Weng L, Zhou J, Guo S, Xu N, Ma R (2024) The molecular subtyping and precision medicine in triple-negative breast cancer—based on Fudan TNBC classification. Cancer Cell Int 24(1):120. https://doi.org/10.1186/s12935-024-03261-010.1186/s12935-024-03261-03855542910981301Wolff AC, Somerfield MR, Dowsett M, Hammond MEH, Hayes DF, McShane LM et al (2023) Human epidermal growth factor receptor 2 testing in breast Cancer: ASCO-College of American Pathologists Guideline Update. J Clin Oncol 41(22):3867–3872. https://doi.org/10.1200/jco.22.0286410.1200/jco.22.0286437284804Won HS, Ahn J, Kim Y, Kim JS, Song JY, Kim HK et al (2022) Clinical significance of HER2-low expression in early breast cancer: a nationwide study from the Korean breast Cancer Society. Breast Cancer Res 24(1):22. https://doi.org/10.1186/s13058-022-01519-x10.1186/s13058-022-01519-x353070148935777Zhang H, Katerji H, Turner BM, Audeh W, Hicks DG (2022a) HER2-low breast cancers: incidence, HER2 staining patterns, clinicopathologic features, MammaPrint and BluePrint genomic profiles. Mod Pathol 35(8):1075–1082. https://doi.org/10.1038/s41379-022-01019-510.1038/s41379-022-01019-535184150Zhang G, Ren C, Li C, Wang Y, Chen B, Wen L et al (2022b) Distinct clinical and somatic mutational features of breast tumors with high-, low-, or non-expressing human epidermal growth factor receptor 2 status. BMC Med 20(1):142. https://doi.org/10.1186/s12916-022-02346-910.1186/s12916-022-02346-9354845939052533Zheng L, Zhang Y, Wang Z, Wang H, Hao C, Li C et al (2023) Comparisons of clinical characteristics, prognosis, epidemiological factors, and genetic susceptibility between HER2-low and HER2-zero breast cancer among Chinese females. Cancer Med 12(14):14937–14948. https://doi.org/10.1002/cam4.612910.1002/cam4.61293738746910417066Zhou S, Liu T, Kuang X, Zhen T, Shi H, Lin Y et al (2023) Comparison of clinicopathological characteristics and response to neoadjuvant chemotherapy between HER2-low and HER2-zero breast cancer. Breast 67:1–7. https://doi.org/10.1016/j.breast.2022.12.00610.1016/j.breast.2022.12.00636535072
3972923620241227
2730-60111512024Dec27Discover oncologyDiscov OncolAdvancements in research and clinical management of interstitial lung injury associated with ADC drugs administration in breast cancer.84384310.1007/s12672-024-01705-7Antibody-drug conjugates (ADCs) represent a novel class of targeted anti-tumor medications that utilize the covalent linkage between monoclonal antibodies and cytotoxic agents. This unique mechanism combines the cytotoxic potency of drugs with the targeting specificity conferred by antigen recognition. However, it is essential to recognize that many ADCs still face challenges related to off-target toxicity akin to cytotoxic payloads, as well as targeted toxicity and other potential life-threatening adverse effects, such as treatment-induced interstitial lung injury. Currently, of the four approved ADC drugs for breast cancer, several reports have documented post-treatment lung injury-related fatalities. As a result, treatment-induced interstitial lung injury due to ADC drugs has become a clinical concern. In this review article, we delve into the factors associated with ADC-induced interstitial lung injury in patients with advanced-stage breast cancer and highlight strategies expected to decrease the incidence of ADC-related interstitial lung injury in the years ahead. These efforts are directed at enhancing treatment outcomes in both advanced and early-stage cancer patients while also providing insights into the development and innovation of ADC drugs and bolstering clinicians' understanding of the diagnosis and management of ADC-associated interstitial lung injury.© 2024. The Author(s).ZhuJia-YuJYDepartment of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.JiangRui-YuanRYDepartment of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.ZhangHuan-PingHPDepartment of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.Department of Graduate Student, Wenzhou Medical University, No. 270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China.FangZi-RuZRDepartment of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.ZhouHuan-HuanHHZhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.WeiQingQZhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China. Weiqingmd@163.com.WangXiaojiaXZhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China. wxiaojia0803@163.com.eng2012C13019-1Major Science and Technology Projects of Zhejiang Province2021RC043Science and Technology Program offered by the Health Bureau of Zhejiang ProvinceY-pierrefabre202101-0126Beijing Xisike Clinical Oncology Research FoundationJournal ArticleReview20241227
United StatesDiscov Oncol1017751422730-6011Adverse reactionAntibody–drug conjugateBreast cancerDrug-induced interstitial lung injuryTargeted therapyDeclarations. Ethics approval and consent to participate: Not applicable. Consent for publication: All authors consent to the publication of this work in Discover Oncology. Competing interests: The authors declare no competing interests.
202412271221202412271220202482620241213202412271114epublish3972923610.1007/s12672-024-01705-710.1007/s12672-024-01705-7Bray F, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229–63. https://doi.org/10.3322/caac.21834 .10.3322/caac.2183438572751Huppert LA, Gumusay O, Idossa D, Rugo HS. Systemic therapy for hormone receptor-positive/human epidermal growth factor receptor 2-negative early stage and metastatic breast cancer. CA Cancer J Clin. 2023. https://doi.org/10.3322/caac.21777 .10.3322/caac.2177736939293Waks AG, Winer EP. Breast cancer treatment: a review. JAMA. 2019;321:288–300. https://doi.org/10.1001/jama.2018.19323 .10.1001/jama.2018.1932330667505Breast cancer. Nat Rev Dis Primers 2019;5:67. https://doi.org/10.1038/s41572-019-0122-z .Yin L, Duan J-J, Bian X-W, Yu S-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020;22:61. https://doi.org/10.1186/s13058-020-01296-5 .10.1186/s13058-020-01296-5325177357285581Haque R, et al. Impact of breast cancer subtypes and treatment on survival: an analysis spanning two decades. Cancer Epidemiol Biomarkers Prev. 2012;21:1848–55. https://doi.org/10.1158/1055-9965.EPI-12-0474 .10.1158/1055-9965.EPI-12-0474229894613467337Waks AG, Winer EP. Breast cancer treatment. JAMA. 2019;321:316–316. https://doi.org/10.1001/jama.2018.20751 .10.1001/jama.2018.2075130667503Lau KH, Tan AM, Shi Y. New and emerging targeted therapies for advanced breast cancer. Int J Mol Sci. 2022;23(4):2288.10.3390/ijms23042288352164058874375Kunte S, Abraham J, Montero AJ. Novel HER2-targeted therapies for HER2-positive metastatic breast cancer. Cancer. 2020;126:4278–88. https://doi.org/10.1002/cncr.33102 .10.1002/cncr.3310232721042Swain SM, Shastry M, Hamilton ET. Targeting HER2-positive breast cancer: advances and future directions. Nat Rev Drug Discov. 2023;22:101–26. https://doi.org/10.1038/s41573-022-00579-0 .10.1038/s41573-022-00579-036344672Slamon DJ, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–12. https://doi.org/10.1126/science.2470152 .10.1126/science.24701522470152Oh D-Y, Bang Y-J. HER2-targeted therapies — a role beyond breast cancer. Nat Rev Clin Oncol. 2020;17:33–48. https://doi.org/10.1038/s41571-019-0268-3 .10.1038/s41571-019-0268-331548601Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the “biological missile” for targeted cancer therapy. Signal Transduct Target Ther. 2022;7:93. https://doi.org/10.1038/s41392-022-00947-7 .10.1038/s41392-022-00947-7353183098941077Ye F, et al. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Mol Cancer. 2023;22:105. https://doi.org/10.1186/s12943-023-01805-y .10.1186/s12943-023-01805-y3741516410324146Smolarz B, Nowak AZ, Romanowicz H. Breast cancer-epidemiology, classification, pathogenesis and treatment (review of literature). Cancers. 2022;14(10):2569.10.3390/cancers14102569356261739139759Planes-Laine G, et al. PD-1/PD-L1 targeting in breast cancer: the first clinical evidences are emerging—a literature review. Cancers. 2019;11(7):1033.10.3390/cancers11071033313366856679223Keam SJ. Trastuzumab deruxtecan: first approval. Drugs. 2020;80:501–8. https://doi.org/10.1007/s40265-020-01281-4 .10.1007/s40265-020-01281-432144719Indini A, Rijavec E, Grossi F. Trastuzumab deruxtecan: changing the destiny of HER2 expressing solid tumors. Int J Mol Sci. 2021;22(9):4774. https://doi.org/10.3390/ijms22094774 .10.3390/ijms22094774339463108125530American Association for Cancer Research. SG improves OS in HR+/HER2− breast cancer. Cancer Discov. 2022;12:2714–5. https://doi.org/10.1158/2159-8290.Cd-nb2022-0061 .10.1158/2159-8290.Cd-nb2022-0061Yang H, Ganguly A, Cabral F. Inhibition of cell migration and cell division correlates with distinct effects of microtubule inhibiting drugs. J Biol Chem. 2010;285:32242–50. https://doi.org/10.1074/jbc.M110.160820 .10.1074/jbc.M110.160820206967572952225Birrer MJ, Moore KN, Betella I, Bates RC. Antibody-drug conjugate-based therapeutics: state of the science. J Natl Cancer Inst. 2019;111:538–49. https://doi.org/10.1093/jnci/djz035 .10.1093/jnci/djz03530859213Jin Y, Schladetsch MA, Huang X, Balunas MJ, Wiemer AJ. Stepping forward in antibody-drug conjugate development. Pharmacol Ther. 2022;229:107917. https://doi.org/10.1016/j.pharmthera.2021.107917 .10.1016/j.pharmthera.2021.10791734171334Khongorzul P, Ling CJ, Khan FU, Ihsan AU, Zhang J. Antibody-drug conjugates: a comprehensive review. Mol Cancer Res. 2020;18:3–19. https://doi.org/10.1158/1541-7786.MCR-19-0582 .10.1158/1541-7786.MCR-19-058231659006Yaghoubi S, et al. Potential drugs used in the antibody-drug conjugate (ADC) architecture for cancer therapy. J Cell Physiol. 2020;235:31–64. https://doi.org/10.1002/jcp.28967 .10.1002/jcp.2896731215038Staudacher AH, Brown MP. Antibody drug conjugates and bystander killing: is antigen-dependent internalisation required? Br J Cancer. 2017;117:1736–42. https://doi.org/10.1038/bjc.2017.367 .10.1038/bjc.2017.367290651105729478Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol. 2009;9:353–63. https://doi.org/10.1038/nri2545 .10.1038/nri2545193654082818721Kepp O, Tesniere A, Zitvogel L, Kroemer G. The immunogenicity of tumor cell death. Curr Opin Oncol. 2009;21:71–6. https://doi.org/10.1097/CCO.0b013e32831bc375 .10.1097/CCO.0b013e32831bc37519125021Nagata S, Tanaka MP. Programmed cell death and the immune system. Nat Rev Immunol. 2017;17:333–40. https://doi.org/10.1038/nri.2016.153 .10.1038/nri.2016.15328163302Obeid M, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. https://doi.org/10.1038/nm1523 .10.1038/nm152317187072Clarke C, Smyth MJ. Calreticulin exposure increases cancer immunogenicity. Nat Biotechnol. 2007;25:192–3. https://doi.org/10.1038/nbt0207-192 .10.1038/nbt0207-19217287754Garg AD, et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012;31:1062–79. https://doi.org/10.1038/emboj.2011.497 .10.1038/emboj.2011.497222521283298003Rios-Doria J, et al. Antibody-drug conjugates bearing pyrrolobenzodiazepine or tubulysin payloads are immunomodulatory and synergize with multiple immunotherapies. Cancer Res. 2017;77:2686–98. https://doi.org/10.1158/0008-5472.Can-16-2854 .10.1158/0008-5472.Can-16-285428283653Iwata TN, et al. A HER2-targeting antibody-drug conjugate, trastuzumab deruxtecan (DS-8201a), enhances antitumor immunity in a mouse model. Mol Cancer Ther. 2018;17:1494–503. https://doi.org/10.1158/1535-7163.Mct-17-0749 .10.1158/1535-7163.Mct-17-074929703841Zammarchi F, et al. CD25-targeted antibody-drug conjugate depletes regulatory T cells and eliminates established syngeneic tumors via antitumor immunity. J Immunother Cancer. 2020. https://doi.org/10.1136/jitc-2020-000860 .10.1136/jitc-2020-000860329129227482493Vivier E, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–9. https://doi.org/10.1126/science.1198687 .10.1126/science.1198687212123483089969Nucera S, Conti C, Martorana F, Wilson B, Genta S. Antibody-drug conjugates to promote immune surveillance: lessons learned from breast cancer. Biomedicines. 2024;12:1491.10.3390/biomedicines120714913906206511274676D’Amico L, et al. A novel anti-HER2 anthracycline-based antibody-drug conjugate induces adaptive anti-tumor immunity and potentiates PD-1 blockade in breast cancer. J Immunother Cancer. 2019;7:16. https://doi.org/10.1186/s40425-018-0464-1 .10.1186/s40425-018-0464-1306654636341578Natsume A, Niwa R, Satoh M. Improving effector functions of antibodies for cancer treatment: enhancing ADCC and CDC. Drug Design Dev Ther. 2009;3:7–16. https://doi.org/10.2147/DDDT.S4378 .10.2147/DDDT.S4378Radocha J, van de Donk N, Weisel K. Monoclonal antibodies and antibody drug conjugates in multiple myeloma. Cancers. 2021. https://doi.org/10.3390/cancers13071571 .10.3390/cancers13071571338054818037134Tai YT, et al. Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood. 2014;123:3128–38. https://doi.org/10.1182/blood-2013-10-535088 .10.1182/blood-2013-10-535088245692624023420Mahalingaiah PK, et al. Potential mechanisms of target-independent uptake and toxicity of antibody-drug conjugates. Pharmacol Ther. 2019;200:110–25. https://doi.org/10.1016/j.pharmthera.2019.04.008 .10.1016/j.pharmthera.2019.04.00831028836Hafeez U, Parakh S, Gan HK, Scott AM. Antibody-drug conjugates for cancer therapy. Molecules. 2020. https://doi.org/10.3390/molecules25204764 .10.3390/molecules25204764330813837587605Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs. 2016;8:659–71. https://doi.org/10.1080/19420862.2016.1156829 .10.1080/19420862.2016.1156829270458004966843Li L, et al. Antibody-drug conjugates in HER2-positive breast cancer. Chin Med J. 2021;135:261–7. https://doi.org/10.1097/cm9.0000000000001932 .10.1097/cm9.0000000000001932349356888812658Wolska-Washer A, Robak T. Safety and tolerability of antibody-drug conjugates in cancer. Drug Saf. 2019;42:295–314. https://doi.org/10.1007/s40264-018-0775-7 .10.1007/s40264-018-0775-7306497476399172Lu J, Jiang F, Lu A, Zhang G. Linkers having a crucial role in antibody–drug conjugates. Int J Mol Sci. 2016;17(4):561.10.3390/ijms17040561270893294849017Tsuchikama K, An Z. Antibody-drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell. 2018;9:33–46. https://doi.org/10.1007/s13238-016-0323-0 .10.1007/s13238-016-0323-027743348Masters JC, Nickens DJ, Xuan D, Shazer RL, Amantea M. Clinical toxicity of antibody drug conjugates: a meta-analysis of payloads. Invest New Drugs. 2018;36:121–35. https://doi.org/10.1007/s10637-017-0520-6 .10.1007/s10637-017-0520-629027591Hinrichs MJ, Dixit R. Antibody drug conjugates: nonclinical safety considerations. AAPS J. 2015;17:1055–64. https://doi.org/10.1208/s12248-015-9790-0 .10.1208/s12248-015-9790-0260246564540738Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107:1039–46. https://doi.org/10.1111/cas.12966 .10.1111/cas.12966271669744946713Menderes G, et al. SYD985, a novel duocarmycin-based HER2-targeting antibody-drug conjugate, shows antitumor activity in uterine and ovarian carcinosarcoma with HER2/Neu expression. Clin Cancer Res. 2017;23:5836–45. https://doi.org/10.1158/1078-0432.CCR-16-2862 .10.1158/1078-0432.CCR-16-2862286797745626613Ogitani Y, et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22:5097–108. https://doi.org/10.1158/1078-0432.Ccr-15-2822 .10.1158/1078-0432.Ccr-15-282227026201Saber H, Leighton JK. An FDA oncology analysis of antibody-drug conjugates. Regulat Toxicol Pharmacol. 2015;71:444–52. https://doi.org/10.1016/j.yrtph.2015.01.014 .10.1016/j.yrtph.2015.01.014Nessler I, Menezes B, Thurber GM. Key metrics to expanding the pipeline of successful antibody-drug conjugates. Trends Pharmacol Sci. 2021;42:803–12. https://doi.org/10.1016/j.tips.2021.07.005 .10.1016/j.tips.2021.07.005344560948519343Adams GP, et al. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Can Res. 2001;61:4750–5.Cilliers C, Menezes B, Nessler I, Linderman J, Thurber GM. Improved tumor penetration and single-cell targeting of antibody-drug conjugates increases anticancer efficacy and host survival. Can Res. 2018;78:758–68. https://doi.org/10.1158/0008-5472.Can-17-1638 .10.1158/0008-5472.Can-17-1638Modi S, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 2020;382:610–21. https://doi.org/10.1056/NEJMoa1914510 .10.1056/NEJMoa191451031825192Banerji U, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019;20:1124–35. https://doi.org/10.1016/S1470-2045(19)30328-6 .10.1016/S1470-2045(19)30328-631257177Zhang J, et al. Phase I trial of a novel anti-HER2 antibody-drug conjugate, ARX788, for the treatment of HER2-positive metastatic breast cancer. Clin Cancer Res. 2022;28:4212–21. https://doi.org/10.1158/1078-0432.CCR-22-0456 .10.1158/1078-0432.CCR-22-0456Hamilton E, et al. Abstract PD3-07: Trastuzumab deruxtecan (T-DXd; DS-8201) with nivolumab in patients with HER2-expressing, advanced breast cancer: A 2-part, phase 1b, multicenter, open-label study. Cancer Res. 2021. https://doi.org/10.1158/1538-7445.SABCS20-PD3-07 .10.1158/1538-7445.SABCS20-PD3-07Wei Q, et al. The promise and challenges of combination therapies with antibody-drug conjugates in solid tumors. J Hematol Oncol. 2024;17(1):1. https://doi.org/10.1186/s13045-023-01509-2 .10.1186/s13045-023-01509-23817820010768262Martin M, et al. Trastuzumab emtansine (T-DM1) plus docetaxel with or without pertuzumab in patients with HER2-positive locally advanced or metastatic breast cancer: results from a phase Ib/IIa study. Ann Oncol. 2016;27:1249–56. https://doi.org/10.1093/annonc/mdw157 .10.1093/annonc/mdw15727052654Levy B, et al. MA13.07 TROPION-Lung 02: initial results for datopotamab deruxtecan plus pembrolizumab and platinum chemotherapy in advanced NSCLC. J Thorac Oncol. 2022;17:S91. https://doi.org/10.1016/j.jtho.2022.07.152 .10.1016/j.jtho.2022.07.152Janjigian YY, et al. Dose-escalation and dose-expansion study of trastuzumab deruxtecan (T-DXd) monotherapy and combinations in patients (pts) with advanced/metastatic HER2+ gastric cancer (GC)/gastroesophageal junction adenocarcinoma (GEJA): DESTINY-Gastric03. J Clin Oncol. 2022;40:295–295. https://doi.org/10.1200/JCO.2022.40.4_suppl.295 .10.1200/JCO.2022.40.4_suppl.295Mamounas EP, et al. Adjuvant T-DM1 versus trastuzumab in patients with residual invasive disease after neoadjuvant therapy for HER2-positive breast cancer: subgroup analyses from KATHERINE. Ann Oncol. 2021;32:1005–14. https://doi.org/10.1016/j.annonc.2021.04.011 .10.1016/j.annonc.2021.04.01133932503Hurvitz SA, et al. TRIO-US B-12 TALENT: Phase II neoadjuvant trial evaluating trastuzumab deruxtecan with or without anastrozole for HER2-low, HR+ early-stage breast cancer. J Clin Oncol. 2022. https://doi.org/10.1200/JCO.2022.40.16_suppl.TPS623 .10.1200/JCO.2022.40.16_suppl.TPS623Schmid P, et al. 166MO Datopotamab deruxtecan (Dato-DXd) + durvalumab (D) as first-line (1L) treatment for unresectable locally advanced/metastatic triple-negative breast cancer (a/mTNBC): initial results from BEGONIA, a phase Ib/II study. Ann Oncol. 2022;33:199. https://doi.org/10.1016/j.annonc.2022.03.185 .10.1016/j.annonc.2022.03.185Patel JD, et al. Sacituzumab govitecan (SG) + pembrolizumab (pembro) in first-line (1L) metastatic non-small cell lung cancer (mNSCLC) with PD-L1 ≥ 50%: cohort A of EVOKE-02. J Clin Oncol. 2024;42:8592–8592. https://doi.org/10.1200/JCO.2024.42.16_suppl.8592 .10.1200/JCO.2024.42.16_suppl.8592Zhu Z, et al. Incidence of antibody-drug conjugates-related pneumonitis in patients with solid tumors: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2023;184:103960. https://doi.org/10.1016/j.critrevonc.2023.103960 .10.1016/j.critrevonc.2023.10396036907365André F, et al. Trastuzumab deruxtecan versus treatment of physician’s choice in patients with HER2-positive metastatic breast cancer (DESTINY-Breast02): a randomised, open-label, multicentre, phase 3 trial. Lancet. 2023;401:1773–85. https://doi.org/10.1016/s0140-6736(23)00725-0 .10.1016/s0140-6736(23)00725-037086745Tarantino P, et al. Interstitial lung disease induced by anti-ERBB2 antibody-drug conjugates: a review. JAMA Oncol. 2021;7:1873–81. https://doi.org/10.1001/jamaoncol.2021.3595 .10.1001/jamaoncol.2021.359534647966Mathur R, Weiner GJ. Picking the optimal target for antibody-drug conjugates. Am Soc Clin Oncol Educ Book. 2013. https://doi.org/10.14694/EdBook_AM.2013.33.e103 .10.14694/EdBook_AM.2013.33.e10323714470Ritchie M, Tchistiakova L, Scott N. Implications of receptor-mediated endocytosis and intracellular trafficking dynamics in the development of antibody drug conjugates. MAbs. 2013;5(1):13–21. https://doi.org/10.4161/mabs.22854 .10.4161/mabs.22854232214643564878Saber H, Leighton JK. An FDA oncology analysis of antibody-drug conjugates. Regul Toxicol Pharmacol. 2015;71:444–52. https://doi.org/10.1016/j.yrtph.2015.01.014 .10.1016/j.yrtph.2015.01.01425661711Beck A, Goetsch L, Dumontet C, Corvaïa NS. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 2017;16:315–37. https://doi.org/10.1038/nrd.2016.268 .10.1038/nrd.2016.26828303026Kumagai K, et al. Interstitial pneumonitis related to trastuzumab deruxtecan, a human epidermal growth factor receptor 2-targeting Ab-drug conjugate, in monkeys. Cancer Sci. 2020;111:4636–45. https://doi.org/10.1111/cas.14686 .10.1111/cas.14686330519387734153Zhao H, et al. Inhibition of megakaryocyte differentiation by antibody-drug conjugates (ADCs) is mediated by macropinocytosis: implications for ADC-induced thrombocytopenia. Mol Cancer Ther. 2017;16:1877–86. https://doi.org/10.1158/1535-7163.Mct-16-0710 .10.1158/1535-7163.Mct-16-071028655784Aoyama M, Tada M, Yokoo H, Demizu Y, Ishii-Watabe A. Fcγ receptor-dependent internalization and off-target cytotoxicity of antibody-drug conjugate aggregates. Pharm Res. 2022;39:89–103. https://doi.org/10.1007/s11095-021-03158-x .10.1007/s11095-021-03158-x34961908Ahmadi M, et al. Small amounts of sub-visible aggregates enhance the immunogenic potential of monoclonal antibody therapeutics. Pharm Res. 2015;32:1383–94. https://doi.org/10.1007/s11095-014-1541-x .10.1007/s11095-014-1541-x25319104Tada M, Aoyama M, Ishii-Watabe A. Fcγ receptor activation by human monoclonal antibody aggregates. J Pharm Sci. 2020;109:576–83. https://doi.org/10.1016/j.xphs.2019.10.046 .10.1016/j.xphs.2019.10.04631676270Nguyen TD, Bordeau BM, Balthasar JP. Mechanisms of ADC toxicity and strategies to increase ADC tolerability. Cancers. 2023;15(3):713.10.3390/cancers15030713367656689913659Bruggeman CW, et al. Tissue-specific expression of IgG receptors by human macrophages ex vivo. PLoS ONE. 2019;14:e0223264. https://doi.org/10.1371/journal.pone.0223264 .10.1371/journal.pone.0223264316138766793881Aegerter H, Lambrecht BN, Jakubzick CV. Biology of lung macrophages in health and disease. Immunity. 2022;55:1564–80. https://doi.org/10.1016/j.immuni.2022.08.010 .10.1016/j.immuni.2022.08.010361038539533769Silver RF, et al. Diversity of human and macaque airway immune cells at baseline and during tuberculosis infection. Am J Respir Cell Mol Biol. 2016;55:899–908. https://doi.org/10.1165/rcmb.2016-0122OC .10.1165/rcmb.2016-0122OC275094885248955Barletta KE, et al. Leukocyte compartments in the mouse lung: distinguishing between marginated, interstitial, and alveolar cells in response to injury. J Immunol Methods. 2012;375:100–10. https://doi.org/10.1016/j.jim.2011.09.013 .10.1016/j.jim.2011.09.01321996427Schneider C, et al. Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat Immunol. 2014;15:1026–37. https://doi.org/10.1038/ni.3005 .10.1038/ni.300525263125Roberts AW, et al. Tissue-resident macrophages are locally programmed for silent clearance of apoptotic cells. Immunity. 2017;47:913-927.e916. https://doi.org/10.1016/j.immuni.2017.10.006 .10.1016/j.immuni.2017.10.006291502395728676Zhou X, Liu X, Huang L. Macrophage-mediated tumor cell phagocytosis: opportunity for nanomedicine intervention. Adv Funct Mater. 2021;31:2006220. https://doi.org/10.1002/adfm.202006220 .10.1002/adfm.20200622033692665Conner SD, Schmid SL. Regulated portals of entry into the cell. Nature. 2003;422:37–44. https://doi.org/10.1038/nature01451 .10.1038/nature0145112621426Lin JH. Pharmacokinetics of biotech drugs: peptides, proteins and monoclonal antibodies. Curr Drug Metab. 2009;10:661–91. https://doi.org/10.2174/138920009789895499 .10.2174/13892000978989549919702530Muro S, Koval M, Muzykantov V. Endothelial endocytic pathways: gates for vascular drug delivery. Curr Vasc Pharmacol. 2004;2:281–99. https://doi.org/10.2174/1570161043385736 .10.2174/157016104338573615320826Doherty GJ, McMahon HT. Mechanisms of endocytosis. Annu Rev Biochem. 2009;78:857–902. https://doi.org/10.1146/annurev.biochem.78.081307.110540 .10.1146/annurev.biochem.78.081307.11054019317650Koganemaru S, et al. Potential mechanisms of interstitial lung disease induced by antibody-drug conjugates based on quantitative analysis of drug distribution. Mol Cancer Ther. 2024. https://doi.org/10.1158/1535-7163.Mct-24-0267 .10.1158/1535-7163.Mct-24-026739450538Freites-Martinez A, Santana N, Arias-Santiago S, Viera A. Using the common terminology criteria for adverse events (CTCAE - Version 5.0) to evaluate the severity of adverse events of anticancer therapies. Actas dermo-sifiliograficas. 2021;112:90–2. https://doi.org/10.1016/j.ad.2019.05.009 .10.1016/j.ad.2019.05.00932891586Skeoch S, et al. Drug-induced interstitial lung disease: a systematic review. J Clin Med. 2018. https://doi.org/10.3390/jcm7100356 .10.3390/jcm7100356303266126209877Abuhelwa Z, Alloghbi A, Alqahtani A, Nagasaka M. Trastuzumab deruxtecan-induced interstitial lung disease/pneumonitis in ERBB2-positive advanced solid malignancies: a systematic review. Drugs. 2022;82:979–87. https://doi.org/10.1007/s40265-022-01736-w .10.1007/s40265-022-01736-w357591219276583Jian Z, et al. criteria for the management of targeted drug-induced interstitial lung disease in solid tumors. China Oncol. 2021;31:241–9. https://doi.org/10.19401/j.cnki.1007-3639.2021.04.001 .10.19401/j.cnki.1007-3639.2021.04.001Bardia A, et al. Clinical practices and institutional protocols on prophylaxis, monitoring, and management of selected adverse events associated with trastuzumab deruxtecan. Oncologist. 2022;27:637–45. https://doi.org/10.1093/oncolo/oyac107 .10.1093/oncolo/oyac107356429079355822中国医师协会肿瘤医师分会乳腺癌学组 & 中国抗癌协会国际医疗交流分会. 中国乳腺癌抗体药物偶联物安全性管理专家共识. 中华肿瘤杂志 2022;44:913–27.Johkoh T, et al. Chest CT diagnosis and clinical management of drug-related pneumonitis in patients receiving molecular targeting agents and immune checkpoint inhibitors: a position paper from the Fleischner Society. Radiology. 2021;298:550–66. https://doi.org/10.1148/radiol.2021203427 .10.1148/radiol.202120342733434111Verheijden G, et al. Toward clinical development of SYD985, a novel HER2-targeting antibody-drug conjugate (ADC). J Clin Oncol. 2014;32:626–626. https://doi.org/10.1200/jco.2014.32.15_suppl.626 .10.1200/jco.2014.32.15_suppl.626Manich CS, et al. LBA15 Primary outcome of the phase III SYD985.002/TULIP trial comparing [vic-]trastuzumab duocarmazine to physician’s choice treatment in patients with pre-treated HER2-positive locally advanced or metastatic breast cancer. Ann Oncol. 2021;32(5):1288–9.10.1016/j.annonc.2021.08.2088Kuhlman JE. The role of chest computed tomography in the diagnosis of drug-related reactions. J Thorac Imaging. 1991;6:52–61. https://doi.org/10.1097/00005382-199101000-00008 .10.1097/00005382-199101000-000081703580Nishino M, et al. Trastuzumab deruxtecan-related interstitial lung disease/pneumonitis: computed tomography imaging patterns to guide diagnosis and management. JCO Precis Oncol. 2023;7:e2300391. https://doi.org/10.1200/PO.23.00391 .10.1200/PO.23.0039138061008Chugh K, Jatwani S. Transbronchial biopsy vs bronchoalveolar lavage in interstitial lung disease. Curr Opin Pulmonary Med. 2022;28:3–8. https://doi.org/10.1097/mcp.0000000000000847 .10.1097/mcp.0000000000000847Costabel U, Uzaslan E, Guzman J. Bronchoalveolar lavage in drug-induced lung disease. Clin Chest Med. 2004;25:25–35. https://doi.org/10.1016/S0272-5231(03)00143-6 .10.1016/S0272-5231(03)00143-615062594Hutchinson JP, Fogarty AW, McKeever TM, Hubbard RB. In-hospital mortality after surgical lung biopsy for interstitial lung disease in the United States 2000 to 2011. Am J Respir Crit Care Med. 2016;193:1161–7. https://doi.org/10.1164/rccm.201508-1632OC .10.1164/rccm.201508-1632OC26646481Yamakawa H, et al. Anti-inflammatory and/or anti-fibrotic treatment of MPO-ANCA-positive interstitial lung disease: a short review. J Clin Med. 2022;11(13):3835. https://doi.org/10.3390/jcm11133835 .10.3390/jcm11133835358071209267459Yang S, et al. Serum oncomarkers in patients with MPO-ANCA-positive vasculitis: diagnostic and prognostic predictive values for interstitial lung disease. Lung. 2022;200:331–8. https://doi.org/10.1007/s00408-022-00532-3 .10.1007/s00408-022-00532-335426513Zhang T, Shen P, Duan C, Gao L. KL-6 as an immunological biomarker predicts the severity, progression, acute exacerbation, and poor outcomes of interstitial lung disease: a systematic review and meta-analysis. Front Immunol. 2021;12:745233. https://doi.org/10.3389/fimmu.2021.745233 .10.3389/fimmu.2021.745233349561798699527Kobayashi J, Kitamura S. KL-6: a serum marker for interstitial pneumonia. Chest. 1995;108:311–5. https://doi.org/10.1378/chest.108.2.311 .10.1378/chest.108.2.3117634858Ohnishi H, et al. Circulating KL-6 levels in patients with drug induced pneumonitis. Thorax. 2003;58:872–5. https://doi.org/10.1136/thorax.58.10.872 .10.1136/thorax.58.10.872145149421746480Hackshaw MD, et al. Incidence of pneumonitis/interstitial lung disease induced by HER2-targeting therapy for HER2-positive metastatic breast cancer. Breast Cancer Res Treat. 2020;183:23–39. https://doi.org/10.1007/s10549-020-05754-8 .10.1007/s10549-020-05754-8325919877376509Swain SM, et al. Multidisciplinary clinical guidance on trastuzumab deruxtecan (T-DXd)-related interstitial lung disease/pneumonitis-Focus on proactive monitoring, diagnosis, and management. Cancer Treatment Rev. 2022;106:102378. https://doi.org/10.1016/j.ctrv.2022.102378 .10.1016/j.ctrv.2022.102378Yong WP, et al. Clinical best practices in optimal monitoring, early diagnosis, and effective management of antibody–drug conjugate-induced interstitial lung disease or pneumonitis: a multidisciplinary team approach in Singapore. Expert Opin Drug Metab Toxicol. 2022;18:805–15. https://doi.org/10.1080/17425255.2022.2162383 .10.1080/17425255.2022.216238336636012Xu C, et al. Clinical best practices in interdisciplinary management of human epidermal growth factor receptor 2 antibody-drug conjugates-induced interstitial lung disease/pneumonitis: an expert consensus in China. Cancer. 2024;130:3054–66. https://doi.org/10.1002/cncr.35475 .10.1002/cncr.3547539092590von Minckwitz G, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019;380:617–28. https://doi.org/10.1056/NEJMoa1814017 .10.1056/NEJMoa1814017Krop IE, et al. Trastuzumab emtansine versus treatment of physician’s choice in patients with previously treated HER2-positive metastatic breast cancer (TH3RESA): final overall survival results from a randomised open-label phase 3 trial. Lancet Oncol. 2017;18:743–54. https://doi.org/10.1016/s1470-2045(17)30313-3 .10.1016/s1470-2045(17)30313-328526538Wuerstlein R, et al. Final results of the global and Asia cohorts of KAMILLA, a phase IIIB safety trial of trastuzumab emtansine in patients with HER2-positive advanced breast cancer. ESMO Open. 2022;7:100561. https://doi.org/10.1016/j.esmoop.2022.100561 .10.1016/j.esmoop.2022.100561360843959588895Beeram M, et al. A phase 1 study of weekly dosing of trastuzumab emtansine (T-DM1) in patients with advanced human epidermal growth factor 2-positive breast cancer. Cancer. 2012;118:5733–40. https://doi.org/10.1002/cncr.27622 .10.1002/cncr.2762222648179Hurvitz SA, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. Lancet. 2023;401:105–17. https://doi.org/10.1016/s0140-6736(22)02420-5 .10.1016/s0140-6736(22)02420-536495879Modi S, et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N Engl J Med. 2022;387:9–20. https://doi.org/10.1056/NEJMoa2203690 .10.1056/NEJMoa22036903566578210561652Tsurutani J, et al. Targeting HER2 with trastuzumab deruxtecan: a dose-expansion, phase I study in multiple advanced solid tumors. Cancer Discov. 2020;10:688–701. https://doi.org/10.1158/2159-8290.Cd-19-1014 .10.1158/2159-8290.Cd-19-1014322135408292921Meric-Bernstam F, et al. Efficacy and safety of trastuzumab deruxtecan (T-DXd) in patients (pts) with HER2-expressing solid tumors: DESTINY-PanTumor02 (DP-02) interim results. J Clin Oncol. 2023;41:LBA3000. https://doi.org/10.1200/JCO.2023.41.17_suppl.LBA3000 .10.1200/JCO.2023.41.17_suppl.LBA3000Xu Y, et al. Phase I study of the recombinant humanized anti-HER2 monoclonal antibody-MMAE conjugate RC48-ADC in patients with HER2-positive advanced solid tumors. Gastric Cancer. 2021;24:913–25. https://doi.org/10.1007/s10120-021-01168-7 .10.1007/s10120-021-01168-7339450498205919Zhang J, et al. Phase I trial of a novel anti-HER2 antibody-drug conjugate, ARX788, for the treatment of HER2-positive metastatic breast cancer. Clin Cancer Res. 2022. https://doi.org/10.1158/1078-0432.ccr-22-0456 .10.1158/1078-0432.ccr-22-0456362605249890931Saura Manich C, et al. LBA15 Primary outcome of the phase III SYD985002/TULIP trial comparing [vic-]trastuzumab duocarmazine to physician’s choice treatment in patients with pre-treated HER2-positive locally advanced or metastatic breast cancer. Ann Oncol. 2021;32:S1288. https://doi.org/10.1016/j.annonc.2021.08.2088 .10.1016/j.annonc.2021.08.2088Banerji UT, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lanct Oncol. 2019;20:1124–35. https://doi.org/10.1016/s1470-2045(19)30328-6 .10.1016/s1470-2045(19)30328-6
trying2...
The relationship between high ratios of CD4/FOXP3 and CD8/CD163 and the improved survivability of metastatic triple-negative breast cancer patients: a multicenter cohort study. | LitMetric

Background: Triple-negative breast cancer (TNBC) has been documented as the most aggressive subtype of breast cancer. This study aimed to analyze antitumor and protumor immune activities, and their ratios as significant prognostic biomarkers in metastatic TNBC (mTNBC).

Methods: A multicenter cohort study was conducted among 103 de novo mTNBC patients. The expression of CD8 and CD163 was evaluated using immunohistochemistry staining, CD4 and FOXP3 using double-staining immunohistochemistry, and PD-L1 using immunohistochemistry and RT-PCR.

Results: Multivariate analysis revealed that high CD4/FOXP3 (HR 1.857; 95% CI 1.049-3.288; p = 0.034) and the CD8/CD163 ratio (HR 2.089; 95% CI 1.174-3.717; p = 0.012) yield significantly improved 1 year overall survival (OS). Kaplan-Meier analysis showed that high levels of CD4 (p = 0.023), CD8 (p = 0.043), CD4/FOXP3 (p = 0.016), CD8/FOXP3 (p = 0.005), CD8/CD163 (p = 0.005) ratios were significantly associated with higher rate of 1 year OS. Furthermore, 1 year OS was directly correlated with antitumor CD4 (R = 0.233; p = 0.018) and CD8 (R = 0.219; p = 0.026) and was indirectly correlated with protumor CD163 and FOXP3 through CD4/FOXP3 (R = 0.282; p = 0.006), CD4/CD163 (R = 0.239; p = 0.015), CD8/FOXP3 (R = 0.260; p = 0.008), and CD8/CD163 (R = 0.258; p = 0.009).

Conclusion: This is the first study to demonstrate that high levels of CD4/FOXP3 and CD8/CD163 significantly improved the 1 year OS in de novo mTNBC patients. Thus, we recommend the application of these markers as prognosis determination and individual treatment decision.

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC10835864PMC
http://dx.doi.org/10.1186/s13104-024-06704-zDOI Listing

Publication Analysis

Top Keywords

breast cancer
12
cd4/foxp3 cd8/cd163
8
cd8/cd163 improved
8
triple-negative breast
8
novo mtnbc
8
mtnbc patients
8
improved 1 year
8
high levels
8
cd4/foxp3
5
cd8/cd163
5

Similar Publications

Importance: The 2009 US Preventive Services Task Force breast cancer screening guideline changes led to decreases in screening mammography, raising concern about potential increases in late-stage disease and more invasive surgical treatments.

Objective: To investigate the incidence of breast cancer by stage at diagnosis and surgical treatment before and after the 2009 guideline changes.

Design, Setting, And Participants: This population-based, epidemiologic cohort study of women aged 40 years or older used 2004 to 2019 data from the National Cancer Institute's Surveillance, Epidemiology, and End Results Program.

View Article and Find Full Text PDF

Background: Triple-negative breast cancer (TNBC) is a serious disease with limited treatment options. We explored the significance of androgen receptor (AR) expression and tumor-infiltrating lymphocytes (TILs) in predicting neoadjuvant chemotherapy (NAC) resistance in TNBC, hypothesizing that AR/TIL classification using pretreatment biopsies can identify NAC-resistant subgroups and improve the understanding of apocrine differentiation.

Methods: This retrospective study included 156 consecutive patients with TNBC treated with NAC.

View Article and Find Full Text PDF

Background: In patients with breast cancer staged ypN1 after neoadjuvant chemotherapy (NAC), there is limited evidence-based guidance regarding exemption from axillary lymph node dissection (ALND).

Methods: This study analyzed ypN1 breast cancer patients post-NAC from the Surveillance, Epidemiology, and End Results databases. Patients were categorized into the breast-conserving surgery (BCS) group and the total mastectomy (TM) group, and further divided by the number of positive lymph nodes (LNs).

View Article and Find Full Text PDF

Purpose: The objective of the current research was to assess the clinicopathological characteristics and long-term prognosis of triple-negative breast cancer (TNBC) patients with human epidermal growth factor receptor 2 (HER2)-low status following breast surgery.

Methods: A total of 202 TNBC patients treated at Qingdao Central Hospital from January 2010 to December 2019 were included, comprising 71 HER2-low and 131 HER2-zero patients. Propensity score matching (PSM) was applied to minimize differences between the cohorts.

View Article and Find Full Text PDF

Antibody-drug conjugates (ADCs) represent a novel class of targeted anti-tumor medications that utilize the covalent linkage between monoclonal antibodies and cytotoxic agents. This unique mechanism combines the cytotoxic potency of drugs with the targeting specificity conferred by antigen recognition. However, it is essential to recognize that many ADCs still face challenges related to off-target toxicity akin to cytotoxic payloads, as well as targeted toxicity and other potential life-threatening adverse effects, such as treatment-induced interstitial lung injury.

View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!