Impact of stromal tumor-infiltrating lymphocytes (sTILs) on response to neoadjuvant chemotherapy in triple-negative early breast cancer in the WSG-ADAPT TN trial

Cornelia Kolberg-Liedtke, Friedrich Feuerhake, Madlen Garke, Matthias Christgen, Ronald Kates, Eva Maria Grischke, Helmut Forstbauer, Michael Braun, Mathias Warm, John Hackmann, Christoph Uleer, Bahriye Aktas, Claudia Schumacher, Sherko Kuemmel, Rachel Wuerstlein, Monika Graeser, Ulrike Nitz, Hans Kreipe, Oleg Gluz, Nadia Harbeck, Cornelia Kolberg-Liedtke, Friedrich Feuerhake, Madlen Garke, Matthias Christgen, Ronald Kates, Eva Maria Grischke, Helmut Forstbauer, Michael Braun, Mathias Warm, John Hackmann, Christoph Uleer, Bahriye Aktas, Claudia Schumacher, Sherko Kuemmel, Rachel Wuerstlein, Monika Graeser, Ulrike Nitz, Hans Kreipe, Oleg Gluz, Nadia Harbeck

Abstract

Background: Higher density of stromal tumor-infiltrating lymphocytes (sTILs) at baseline has been associated with increased rates of pathological complete response (pCR) after neoadjuvant chemotherapy (NACT) in triple-negative breast cancer (TNBC). While evidence supports favorable association of pCR with survival in TNBC, an independent impact of sTILs (after adjustment for pCR) on survival is not yet established. Moreover, the impact of sTIL dynamics during NACT on pCR and survival in TNBC is unknown.

Methods: The randomized WSG-ADAPT TN phase II trial compared efficacy of 12-week nab-paclitaxel with gemcitabine versus carboplatin. This preplanned translational analysis assessed impacts of sTIL measurements at baseline (sTIL-0) and after 3 weeks of chemotherapy (sTIL-3) on pCR and invasive disease-free survival (iDFS). Predictive performance of sTIL-0 and sTIL-3 for pCR was quantified by ROC analysis and logistic regression; Kaplan-Meier estimation and Cox regression (with mediation analysis) were used to determine their impact on iDFS.

Results: For prediction of pCR, the AUC statistics for sTIL-0 and sTIL-3 were 0.60 and 0.63, respectively, in all patients; AUC for sTIL-3 was higher in NP/G. The positive predictive value (PPV) of "lymphocyte-predominant" status (sTIL-0 ≥ 60%) at baseline was 59.3%, though only 13.0% of patients had this status. To predict non-pCR, the cut point sTIL-0 ≤ 10% yielded PPV = 69.5% while addressing 33.8% of patients. Higher sTIL levels (particularly at 3 weeks) were independently and favorably associated with better iDFS, even after adjusting for pCR. For example, the adjusted hazard ratio for 3-week sTILs ≥ 60% (vs. < 60%) was 0.48 [0.23-0.99]. Low cellularity in 3-week biopsies was the strongest individual predictor for pCR (in both therapy arms), but not for iDFS.

Conclusion: The independent impact of sTILs on iDFS suggests that favorable immune response can influence key tumor biological processes for long-term survival. The results suggest that the reliability of pCR following neoadjuvant therapy as a surrogate for survival could vary among subgroups in TNBC defined by immune response or other factors. Dynamic measurements of sTILs under NACT could support immune response-guided patient selection for individualized therapy approaches for both very low levels (more effective therapies) and very high levels (de-escalation concepts).

Trial registration: Clinical trials No: NCT01815242, retrospectively registered January 25, 2013.

Keywords: 3-Week biopsy; Neoadjuvant chemotherapy; Pathologic complete response; Triple-negative breast cancer; sTils.

Conflict of interest statement

CKL reports stock by Theraklion and Phaon Scientific (self and family), honoraria by Roche, AstraZeneca, Celgene, Novartis, Pfizer, Lilly, Hexal, Amgen, SonoScape (self) and Genomic Health, Amgen, AstraZeneca, Riemser, Carl Zeiss MediTec, TEVA Pharmaceuticals Industries, Theraklion, Janssen-Cilag, GlaxoSmithKline, LIV Pharma (family), Consulting to Roche, Novartis, Pfizer, Celgene, Phaon Scientific (self) and Pfizer, Novartis, SurgVision, CarlZeissMeditec, Amgen, Onkowissen (family); research funding by Roche, Novartis, Pfizer (self) as well as Travel and Accomodation by Roche, Daiichi Sankyo, Novartis (self) and Carl Zeiss Meditec, LIV Pharma, Novartis, Amgen, Pfizer, Daiichi Sankyo (family). FF reports research funding by IOMX Therapeutics AG (Research Collaboration). MG none. MC none. RK reports honoraria by Amgen, Celgene, Genomic Health, NanoString Technologies, Novartis pharma, Pfizer Pharmaceuticals and Roche (family); consulting/advisory role by Agendia, AstraZeneca, Celgene, Daiichi Sankyo, Lilly, Merck Sharp & Dohme, Novartis pharma, Odonate Therapeutics, Pfizer Pharmaceuticals, Roche/Genentech, Sandoz (family); Research funding by the West German Study Group (self) and Genentech, Novartis, Boehringer Ingelheim and Pfizer (self). EG none. HF none. MB reports honoraria by AstraZeneca, Exact Sciences, Novartis, Pfizer, Roche, Teva; Consulting/Advisory Role by AstraZeneca, Exact Sciences, Novartis, Puma, Roche and Travel/Accomodation by AstraZeneca, Celgene, Medac, Novartis, Roche. MW none. JH none. CU reports Consulting/Advisory Role by Tesaro, Novartis, Roche, Research Funding by West German Study Group, German Breast Group, Novartis, AstraZeneca, Tesaro, Palleos Healthcare Services Gmbh, Pierre Fabre, AGO-Studiengrouppe; Travel/Accomodation by German Breast Group, West German Study Group, AGO-Studiengruppe; Speakers’ Bureau by Pfizer, Novartis, Roche, PharmaMar, AstraZeneca. BA reports Consulting/Advisory Role by Pfizer, Roche Pharma, Merck Sharp & Dohme, onkowissen.de, Novartis Pharma, AstraZeneca, PharmaMar, Lilly, promedicis; Travel /Accomodation by astraZeneca, Roche, Novartis, Pfizer. CS none. SK reports Consulting/Advisory Role by Roche, Genomic Health, Novartis, AstraZeneca, Amgen, Celgene, SOMATEX Medical Technologies, Daiichi Sankyo, PFM medical, Pfizer, MSD, Puma Biotechnology; Travel/accommodation by Roche, Daiichi Sankyo; other relationship as Co-Director West German Study Group. RW report Served as advisor, consultant, speaker and travel grants by Agendia, Amgen, Aristo, Astra Zeneca, Boeringer Ingelheim, Carl Zeiss, Celgene, Clinsol, Daiichi Sankyo, Eisai, Exact Sciences, Genomic Health, Gilead, Glaxo Smith Kline, Hexal, Lilly, Medstrom Medical, MSD, Mundipharma, Nanostring, Novartis, Odonate, Paxman, Palleos, Pfizer, Pierre Fabre, PumaBiotechnolgogy, Riemser, Roche, Sandoz/Hexal, Seattle Genetics /Seagen, Tesaro Bio, Teva, Veracyte, Viatris. MG none. UN reports honoraria by Agendia, Amgen, Celgene, Genomic Health, NanoString Technologies, Novartis pharma, Pfizer Pharmaceuticals, Roche/Genentech, Teva; Consulting/Advisory Role by Genomic Health, Roche; Research Funding by Agendia, Amgen, Celgene, Genomic Health, NanoString Technologies, Roche, Sanofi; Expert Testimony by Genomic Health; Travel/Accommodation by Genomic Health, Pfizer Pharmaceuticals, Roche; other relationship as Co-Director West German Study Group. HK none. OG reports honoraria by Genomic Health/Exact Sciences, Roche, Celgene, Pfizer, Novartis, NanoString Technologies, AstraZeneca; Consulting/Advisory Rola by Celgene, Genomic Health/Exact Sciences, Lilly, MSD, Novartis, Pfizer, Roche; Travel/Accomodation by Roche; other relationship as Co-Director West German Study Group. NH reports honoraria by Amgen, AstraZeneca, Genomic Health, Novartis, Pfizer, Pierre Fabre, Roche, Zodiac Pharma; Consulting/Advisory Role by Agendia, AstraZeneca, Celgene, Daiichi Sankyo, Lilly, Merck Sharp & Dohme, Novartis, Odonate Therapeutics, Pfizer, Pierre Fabre, Roche/Genentech, Sandoz, Seattle Genetics, West German Study Group (family); Research Funding by Lilly, Merck Sharp & Dohme, Novartis, Pfizer, Roche/Genentech; other relationship as Co-Director West German Study Group.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Sensitivity (long-dashed curves), specificity (short-dashed curves), PPV (dotted curves) and % addressed (solid curve) as a function of cut point in ROC analysis for A sTIL-0 and B sTIL-3 (lower panel). Vertical lines indicate particular cut points discussed in the text. The rightmost cut point corresponds to “lymphocyte-predominant” status
Fig. 2
Fig. 2
iDFS in Kaplan–Meier analysis for TIL+ (baseline lymphocyte-predominant status) versus TIL− in A all patients, B patients with non-pCR and C patients with pCR
Fig. 3
Fig. 3
iDFS in Kaplan–Meier analysis for 3-week measurements, i.e., 3wTIL+ versus 3wTIL− versus 3wLC (low cellularity), in A all patients, B patients with non-pCR and C patients with pCR and D iDFS in Kaplan–Meier analysis for subgroups defined (see text) by sTIL transitions from baseline to 3 weeks

References

    1. Gluz O, Liedtke C, Gottschalk N, et al. Triple-negative breast cancer–current status and future directions. Ann Oncol. 2009;20:1913–1927. doi: 10.1093/annonc/mdp492.
    1. Liedtke C, Hess KR, Karn T, et al. The prognostic impact of age in patients with triple-negative breast cancer. Breast Cancer Res Treat. 2013;138(2):591–599. doi: 10.1007/s10549-013-2461-x.
    1. Carey LA, Dees EC, Sawyer L, et al. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res. 2007;13(8):2329–2334. doi: 10.1158/1078-0432.CCR-06-1109.
    1. Liedtke C, Mazouni C, Hess KR, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 2008;26(8):1275–1281. doi: 10.1200/JCO.2007.14.4147.
    1. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–172. doi: 10.1016/S0140-6736(13)62422-8.
    1. Funt SA, Chapman PB. The role of neoadjuvant trials in drug development for solid tumors. Clin Cancer Res. 2016;22(10):2323–2328. doi: 10.1158/1078-0432.CCR-15-1961.
    1. Arnedos M, Roulleaux Dugage M, Perez-Garcia J, Cortes J. Window of opportunity trials for biomarker discovery in breast cancer. Curr Opin Oncol. 2019;31(6):486–492. doi: 10.1097/CCO.0000000000000583.
    1. Gluz O, Nitz U, Liedtke C, Christgen M, Grischke EM, Forstbauer H, Braun M, Warm M, Hackmann J, Uleer C, Aktas B, Schumacher C, Bangemann N, Lindner C, Kuemmel S, Clemens M, Potenberg J, Staib P, Kohls A, von Schumann R, Kates R, Kates R, Schumacher J, Wuerstlein R, Kreipe HH, Harbeck N. Comparison of neoadjuvant nab-paclitaxel+carboplatin vs nab-paclitaxel+gemcitabine in triple-negative breast cancer: randomized WSG-ADAPT-TN trial results. J Natl Cancer Inst. 2018;110(6):628–637. doi: 10.1093/jnci/djx258.
    1. Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ, et al. Clinical relevance of host immunity in breast cancer: From TILs to the clinic. Nat Rev Clin Oncol. 2016;13:228–241. doi: 10.1038/nrclinonc.2015.215.
    1. Denkert C, Loibl S, Noske A, Roller M, Müller BM, Komor M, Budczies J, Darb-Esfahani S, Kronenwett R, Hanusch C, von Törne C, Weichert W, Engels K, Solbach C, Schrader I, Dietel M, von Minckwitz G. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28(1):105–13. 10.1200/JCO.2009.23.7370. Epub 2009 Nov 16. Erratum in: J Clin Oncol. 2010;28(4):708.
    1. Loi S, Drubay D, Adams S, et al. Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. J Clin Oncol. 2019;37(7):559–569. doi: 10.1200/JCO.18.01010.
    1. Kolberg-Liedtke C, Gluz O, Heinisch F, et al. Association of TILs with clinical parameters, Recurrence Score® results, and prognosis in patients with early HER2-negative breast cancer (BC)—a translational analysis of the prospective WSG PlanB trial. Breast Cancer Res. 2020;22(1):47. doi: 10.1186/s13058-020-01283-w.
    1. Luen SJ, Salgado R, Dieci MV, Vingiani A, Curigliano G, Gould RE, Castaneda C, D'Alfonso T, Sanchez J, Cheng E, Andreopoulou E, Castillo M, Adams S, Demaria S, Symmans WF, Michiels S, Loi S. Prognostic implications of residual disease tumor-infiltrating lymphocytes and residual cancer burden in triple-negative breast cancer patients after neoadjuvant chemotherapy. Ann Oncol. 2019;30(2):236–242. doi: 10.1093/annonc/mdy547.
    1. Nuciforo P, Pascual T, Cortés J, et al. A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade. Ann Oncol. 2018;29(1):170–177. doi: 10.1093/annonc/mdx647.
    1. Mazouni C, Peintinger F, Wan-Kau S, et al. Residual ductal carcinoma in situ in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy does not adversely affect patient outcome. J Clin Oncol. 2007;25(19):2650–2655. doi: 10.1200/JCO.2006.08.2271.
    1. Brambilla E, et al. Prognostic effect of tumor lymphocytic infiltration in resectable non-small-cell lung cancer. J Clin Oncol. 2016;34:1223–1230. doi: 10.1200/JCO.2015.63.0970.
    1. Baron RM, Kenny DA. The moderator-mediator variable distinction in social psychological research—conceptual, strategic, and statistical considerations. J Pers Soc Psychol. 1986;51(6):1173–1182. doi: 10.1037/0022-3514.51.6.1173.
    1. Denkert 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.
    1. Bai Y-G, Gao G-X, Zhang H, Zhang S, Liu Y-H, Duan X-N, Ling Xu. Prognostic value of tumor-infiltrating lymphocyte subtypes in residual tumors of patients with triple-negative breast cancer after neoadjuvant chemotherapy. Chin Med J (Engl) 2020;133(5):552–560. doi: 10.1097/CM9.0000000000000656.
    1. Loi S, Sirtaine N, Piette F, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02–98. J Clin Oncol. 2013;31(7):860–867. doi: 10.1200/JCO.2011.41.0902.
    1. Telli ML, Chu C, Badve SS, et al. Association of tumor-infiltrating lymphocytes with homologous recombination deficiency and BRCA1/2 status in patients with early triple-negative breast cancer: A pooled analysis. Clin Cancer Res. 2020;26(11):2704–2710. doi: 10.1158/1078-0432.CCR-19-0664.
    1. Skriver SK, Jensen MB, Knoop AS, Ejlertsen B, Laenkholm AV. Tumour-infiltrating lymphocytes and response to neoadjuvant letrozole in patients with early oestrogen receptor-positive breast cancer: analysis from a nationwide phase II DBCG trial. Breast Cancer Res. 2020;22(1):46. doi: 10.1186/s13058-020-01285-8.
    1. Lee H, Lee M, Seo J-H, Gong G, Lee HJ. Changes in tumor-infiltrating lymphocytes after neoadjuvant chemotherapy and clinical significance in triple negative breast cancer. Anticancer Res. 2020;40(4):1883–1890. doi: 10.21873/anticanres.14142.
    1. Denkert C, et al. Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: results of the ring studies of the international immuno-oncology biomarker working group. Mod Pathol. 2016;29:1155–1164. doi: 10.1038/modpathol.2016.109.
    1. Wein L, et al. Clinical validity and utility of tumor-infiltrating lymphocytes in routine clinical practice for breast cancer patients: current and future directions. Front Oncol. 2017;7:156. doi: 10.3389/fonc.2017.00156.
    1. Dieci MV, Radosevic-Robin N, Fineberg S, van den Eynden G, Ternes N, Penault-Llorca F, Pruneri G, D'Alfonso TM, Demaria S, Castaneda C, Sanchez J, Badve S, Michiels S, Bossuyt V, Rojo F, Singh B, Nielsen T, Viale G, Kim SR, Hewitt S, Wienert S, Loibl S, Rimm D, Symmans F, Denkert C, Adams S, Loi S, Salgado R; International Immuno-Oncology Biomarker Working Group on Breast Cancer. Update on tumor-infiltrating lymphocytes (TILs) in breast cancer, including recommendations to assess TILs in residual disease after neoadjuvant therapy and in carcinoma in situ: A report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer. Semin Cancer Biol. 2018;52(Pt 2):16–25. 10.1016/j.semcancer.2017.10.003.
    1. Amgad M, Stovgaard ES, Balslev E, et al. Report on computational assessment of Tumor Infiltrating Lymphocytes from the International Immuno-Oncology Biomarker Working Group. NPJ Breast Cancer. 2020;6:16. doi: 10.1038/s41523-020-0154-2.
    1. Kos Z, Roblin E, Kim RS, Michiels S, Gallas BD, Chen W, van de Vijver KK, Goel S, Adams S, Demaria S, Viale G, Nielsen TO, Badve SS, Symmans WF, Sotiriou C, Rimm DL, Hewitt S, Denkert C, Loibl S, Luen SJ, Bartlett JMS, Savas P, Pruneri G, Dillon DA, Cheang MCU, Tutt A, Hall JA, Kok M, Horlings HM, Madabhushi A, van der Laak J, Ciompi F, Laenkholm AV, Bellolio E, Gruosso T, Fox SB, Araya JC, Floris G, Hudeček J, Voorwerk L, Beck AH, Kerner J, Larsimont D, Declercq S, Van den Eynden G, Pusztai L, Ehinger A, Yang W, AbdulJabbar K, Yuan Y, Singh R, Hiley C, Bakir MA, Lazar AJ, Naber S, Wienert S, Castillo M, Curigliano G, Dieci MV, André F, Swanton C, Reis-Filho J, Sparano J, Balslev E, Chen IC, Stovgaard EIS, Pogue-Geile K, Blenman KRM, Penault-Llorca F, Schnitt S, Lakhani SR, Vincent-Salomon A, Rojo F, Braybrooke JP, Hanna MG, Soler-Monsó MT, Bethmann D, Castaneda CA, Willard-Gallo K, Sharma A, Lien HC, Fineberg S, Thagaard J, Comerma L, Gonzalez-Ericsson P, Brogi E, Loi S, Saltz J, Klaushen F, Cooper L, Amgad M, Moore DA, Salgado R, International Immuno-Oncology Biomarker Working Group Pitfalls in assessing stromal tumor infiltrating lymphocytes (sTILs) in breast cancer. NPJ Breast Cancer. 2020;6:17. doi: 10.1038/s41523-020-0156-0.
    1. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363(20):1938–1948. doi: 10.1056/NEJMra1001389.
    1. Gluz O, Kolberg-Liedtke C, Prat A, Christgen M, Gebauer D, Kates R, Paré L, Grischke EM, Forstbauer H, Braun M, Warm M, Hackmann J, Uleer C, Aktas B, Schumacher C, Kuemmel S, Wuerstlein R, Pelz E, Nitz U, Kreipe HH, Harbeck N. Efficacy of deescalated chemotherapy according to PAM50 subtypes, immune and proliferation genes in triple-negative early breast cancer: primary translational analysis of the WSG-ADAPT-TN trial. Int J Cancer. 2020;146(1):262–271. doi: 10.1002/ijc.32488.
    1. De Jong VMT, Wang Y, Opdam M, ter Hoeve N, Jóźwiak K, Hauptmann M, et al. Prognostic value of tumour infiltrating lymphocytes in young triple negative breast cancer patients who did not receive adjuvant systemic treatment; by the PARADIGM study group. Ann Oncol. 2020;31(suppl_4):S303–S339. doi: 10.1016/j.annonc.2020.08.281.

Source: PubMed

3
Subscribe