Th2 infiltration is a better predictor of survival than tumor-infiltrating lymphocytes (TILs) in triple-negative breast cancer (TNBC) ===================================================================================================================================== * Susie Brousse * Florence Godey * Patrick Tas * Boris Campillo-Gimenez * Elodie Lafont * Amanda Poissonnier * Jean Levêque * Vincent Lavoué * Matthieu Le Gallo ## Abstract **Purpose** Triple-negative breast cancers (TNBC) account for 15% of all breast cancers but carry the worst prognosis. Because of their heterogenicity, these tumors are not all prone to targeted therapies. However, due to their high immune infiltration, targeting their immune microenvironment is of tremendous interest and is becoming the standard of care for high-risk early-stage TNBC. Nevertheless, the characterization of this immune infiltrate is often limited to general tumor-infiltrating lymphocytes (TILs) counting, without characterization of lymphocytes subtypes. Thus, we aimed at precisely characterizing these sub-populations and evaluating their prognostic significance. **Methods** We selected 91 TNBC tumors for which we had both the TILs count on hematoxylin and eosin (H&E) slides determined by an expert pathologist and the immune microenvironment cell subtypes characterization using flow cytometry (FC). We then compared the prognostic value of immune microenvironment subpopulations vs total TILs count. **Results** TNBCs contained a mean of 22.8±25.9% TILs in the tumor surface area, including mainly CD4+ helper T lymphocytes (14.1%), mostly Th2 (11.7%), CD8+ cytotoxic T lymphocytes (11.1%), and myeloid cells (8.4%) including antigen presenting cells (APC). The TILs count was correlated with the abundance of these cellular subpopulations (p≤0.004). TILs percentage was predictive of overall survival (OS) in univariate analysis (p=0.044), high APC infiltration was predictive of relapse-free survival (RFS) in univariate analysis (p≤0.030), and Th2 infiltration was predictive of both RFS and OS in univariate (p=0.009, 0.008 respectively) and multivariate analyses (p=0.002, 0.010 respectively). **Conclusion** The characterization of TILs composition is essential to better understand the potential antitumoral functions of these cells, and to strongly improve the associated prognostic and predictive values. We here demonstrate that Th2 subpopulation is associated with a better overall survival in TNBC and could be of use to predict response to the newly used immunotherapies. Keywords * Triple-negative breast cancers * tumor-infiltrating lymphocytes * immune response * Th2 signature * prognostic and predictive factors ## Introduction Representing 25% of newly diagnosed cancers among women, breast cancer is a worldwide health concern with 685,000 associated deaths in 2020 according to the World Health Organization, making this pathology the leading cause of female death by cancer in most countries (1,2). Triple negative breast cancers (TNBC) are defined as negative for the expression of hormonal receptors (HR) to estrogen (ER), progesterone (PR), and Human Epidermal Growth Factor 2 (Her2) receptors, being mostly constituted of basal-like molecular subtype (3,4). Whilst they represent 10 to 20% of all breast tumors (2,5), they are a major health concern due to their epidemiology, being frequently diagnosed among younger non-menopausal women, and to their quickly unfavorable evolution (6–8) associated with a high proliferation index, frequent relapse after chemotherapy (9,10), and early metastatic disease (pulmonary, hepatic, neurologic) (8,11). Besides, we have too few clinically available targeted therapy (5,9) since they represent a very heterogenous disease made of up to 7 subtypes (12–14). Even though there are some promising therapeutic options (12,13,15–18), TNBC do not share a common antigenic target, making it difficult to find broadly efficient therapies. Nevertheless, their immunogenicity could offer some therapeutic options, since an immune infiltrate is detected in up to 75% of TNBC, much more than in luminal subtypes (19), and its composition is very distinct from the other molecular subtypes (19,20). Hence, immunotherapy recently became available and recommended for immunosuppressive TNBC, thanks to the the newly FDA-approved Atezolizumab (21) and Pembrolizumab (22), sometimes depending on PD-L1 expression. However, PD1/PD-L1 expression does not select responding patients accurately enough (22,23). So, in order to better identify which patients could benefit from immunotherapies and to identify new potential targets, it is essential to better understand the role and composition of the immune infiltrate in TNBCs. The tumor-infiltrating lymphocytes (TILs) density has been shown to be predictive of response to neoadjuvant chemotherapy (NAC) and of patient outcome (24–26), especially in TNBC (24,25,27,28). CD8+ cytotoxic T cells (TC) were first proved to be markers of good outcome, especially in TNBCs (26,29,30), as well as CD4+ helper and CD4+ follicular helper (fh) TC (31,32). But TNBC can escape immune cell-mediated cytotoxicity by developing an immunotolerance thanks to immune checkpoints and regulatory TC (Treg) activation (33). Moreover, the modulation of the expression of some immunological-related genes (34), and different immune signatures such as T helper cell type 1 (35), low M2-like macrophages (36) and gene cluster Immunity 2 (37) also have prognostic value. That’s why further classification in immunocompetent or tolerant TC is necessary to evaluate their prognostic significance. Nevertheless, to assess immune infiltrate in daily practice, pathologists currently only measure TILs density (38,39), since it is predictive of pathological complete response (pCR) to NAC, and of significantly better recurrence-free (RFS) and overall survival (OS) (40). However, no therapeutic option derives from this measurement since the TILs subtypes are not searched for. Hence, we here aim to characterize the immune microenvironment of TNBC tumors by flow cytometry and thus to better specify the prognostic value of TILs subtypes. ## Methods ### Patients 123 TNBC patients were recruited at the time of surgery, before any chemotherapy and radiotherapy was administered, from January 2013 to December 2018 in Centre Eugène Marquis in Rennes, France. All participants provided informed consent and the study was approved by the local Ethics Committee Review Board and complied with local ethics guidelines. 91 patients were finally selected (Supplementary Figure 1). ### Sample collection After surgical resection, TNBC tumor fresh tissue was taken by the pathologist in the operative room and processed into two fragments. The first one, fixed in 10% neutral buffered formalin for standard histochemical analysis with Hematoxylin and Eosin (H&E), was analyzed for TILs quantification and requalified for research purposes. The second one was cut into small pieces (<2 mm), stored in cold RPMI medium (Gibco), and processed within two hours with Human tumor dissociation kit (Miltenyi Biotec GmbH) in a gentle MACS Dissociator, according to the manufacturer’s recommendations. Macroscopic pieces were eliminated using a Corning cell strainer (70 μm), dissociated cells were then washed twice in RPMI (20 mL) and counted using a hemocytometer. Living cells were immediately analyzed using multiparameter flow cytometry (FC). ### Analyses of TILs by FC Tumor cells (50,000 cells) were suspended in PBS supplemented with 2% BSA, 2% FCS, and FcR block (Miltenyi Biotec GmbH®) at 4°C for 20 minutes. Cells were then stained for 30 minutes at 4°C with fluorochrome-coupled antibodies targeting CD45-PeVio770 (clone 5B1), CD3-FITC (clone REA613), CD4-APCVio770 (clone VIT4), CD8-FITC (clone BW135/80), CD11c-APC-Vio770 (clone REA618), CD83-APC (clone HB15), CD86-FITC (cloneFM95), CXCR3-PE (clone REA232), CCR6-APC (clone REA190) (Miltenyi Biotec GmbH), CD14-APC (clone MφP9), CD24-PE (clone ML5), CD44-APC (clone M44-26) (BD Biosciences, France). Matching isotypic antibodies purchased from the same manufacturers were used as control. Cells were then washed twice in PBS supplemented with 2% BSA and 2% FCS and resuspended in PBS. To assess cell viability, cells were incubated with 7-AAD (BD Biosciences) for 10 min prior to cytometry analysis. Data were acquired using a FACS Canto II (BD Biosciences) or a Novocyte cytometer (ACEA Biosciences) and analyzed using Novoexpress software. Tumor and immune cells were defined as described in Table 1 and Supplementary Figure 2. The different TILs populations were thus measured, and their abundance, overall and relative over total CD45+ immune cells, was quantified for each tumor (Figure 1). ![Figure 1-](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/06/06/2023.06.02.23289891/F1.medium.gif) [Figure 1-](http://medrxiv.org/content/early/2023/06/06/2023.06.02.23289891/F1) Figure 1- Tumor-Infiltrating Lymphocytes (TILs) and their characterization by Flow Cytometry (FC). Corresponding H&E pathological slides (A) and FC analysis (B) of a highly infiltrated group C tumor, BT127, showing 70% of TILs infiltration (A) and 20% of Th2/CD45 +cells (B), and a poorly infiltrated group A one, BT82, showing 5% of TILs stromal infiltration (A), and 6% of Th2/CD45+ cells (B). TILs count consists of mononuclear cells count in stromal fibrous spans (pink). Th2 cells are defined as living T helper lymphocytes (CD4+) expressing neither CXCR3 (contrary to Th1 and Th17 lymphocytes) nor CCR6 (contrary to Th17 lymphocytes). C: Complete characterization of the immune microenvironment of all tumors by FC showing the relative abundance of the different subpopulations (Percentage values in Supplementary Table 3). TC: T lymphocyte cells; APC: Antigen-presenting cells. View this table: [Table 1-](http://medrxiv.org/content/early/2023/06/06/2023.06.02.23289891/T1) Table 1- TILs characterization by Flow Cytometry (FC) according to the expression of their surface markers. BCSC: Breast Cancer Stem Cells; DBCC: Differentiated Breast Cancer Cells; APC: Antigen-Presenting Cells. ### TILs quantification H&E FFPE slides were obtained from Ouest pathology after pathological report was done. Retrospective TILs count, defined as the percentage of tumor stroma surface occupied by TILs, was done based on the 2014 TILs working group guidelines updated in 2018 (38,39). Group A, B, and C were defined as TILs infiltration <20%, ≥20% and <50%, and ≥ 50% respectively (Figure 1). ### Statistical analyses Patients and tumor characteristics were compared between TILs or Th2 groups using Chi-squared tests or Fisher’s exact tests for qualitative variables, and either Student’s t tests, Mann Whitney U tests, or ANOVA and Kruskall-Wallis tests (when more than 2 groups were compared) for quantitative ones. Pearson or Spearman correlations were used to study the link between TILs percentage and other immune subpopulations abundance. Relapse-free survival (RFS) was defined as the time from surgery to relapse or death and overall survival (OS) as the time from diagnosis to death. Patients lost to follow-up were censored at the date of last news. Kaplan-Meier curves were estimated for RFS and OS, and log-rank tests were performed to compare survival distributions. Univariate and multivariate Cox regression analyses were performed to calculate unadjusted and adjusted Hazard Ratios (HR) and were presented with 95% Confidence Intervals (CI95%). Bayesian Information Criterion (BIC) was used to select independent parameters according to a forward step-by-step selection procedure. Whole statistical analysis was performed with R and RStudio® softwares. All reported p values were two-sided and considered statistically significant when <0.05. ## Results ### Population description 91 patients got a complete immune characterization (TILs count by pathologists and subpopulations characterization by FC) (Supplementary Figure 1). Their characteristics are precisely described in Supplementary Table 1 and 2. Briefly, our population consisted of females with a mean age of 58 (±14.5) years. Of these women, 15.7% and 11.8% carried a BRCA1 or BRCA2 mutation respectively. Their tumors were mostly high grade (93.4%) and highly proliferative (mean Ki67: 64.8±23.8%) invasive ductal carcinomas (85.7%) of 22.9±11.6mm with 1.5±3.6 metastatic lymph nodes. Therapy mostly consisted of lumpectomy or breast conserving surgery (75.8%), with axillary treatment (94.4%), followed by chemotherapy (85.7%), and radiotherapy (86.5%). The median follow-up was 28.0 months (Supplementary Table 2). ### TILs count, characterization using FC, and their correlation Most TNBC (58.2%) were poorly infiltrated Group A tumors, versus 23.1% Group B and 18.7% Group C, for a TILs count of 22.8±25.9% (median 10.0% [0.0-90.0]). TILs consisted of 3.1% macrophages, 8.4% myeloid cells, 11.1% CD8+ cytotoxic TC, and 14.1% CD4+ helper TC (1.0% Th1, 0.8% Th17, and 11.7% Th2 subsets) (Figure 1 and Supplementary Table 3). The correlation between TILs groups and immune cells measured by FC was linear and significant for total leukocytes (p≤0.001), helper TC (p≤0.001), cytotoxic TC (p≤0.001), Th17 and Th2 responses (p≤0.01), and antigen presenting cells including dendritic ones (p≤0.001 & p≤0.01 respectively) (Figure 2). Pearson and Spearman correlations showed the same results when considering TILs percentages and total leukocytes (p=0.002), myeloid cells (p<0.001), cytotoxic TC (p=0.002), helper TC (p<0.001), and Th17 (p=0.022) or Th2 expressing TCs (p=0.002). We noted that the presence of Th1 TCs was not significantly correlated with TILs group (p=0.14) or percentage (p=0.218). ![Figure 2-](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/06/06/2023.06.02.23289891/F2.medium.gif) [Figure 2-](http://medrxiv.org/content/early/2023/06/06/2023.06.02.23289891/F2) Figure 2- Correlation of TILs groups with immune subpopulations relative abundance. Correlation of TILs group as defined by the pathologist with total leukocytes, including helper CD4+ and cytotoxic CD8+ T cells, antigen-presenting cells (APC) or dendritic cells, and tumor cells characterized by flow cytometry (FC). Helper T cells could either trigger a Th1, Th17, or Th2 response. Only populations with significant correlation are shown on the figure. p values were derived from a Holm and a Kruskal-Wallis tests (for the comparison of respectively 2 and 3 groups). ns: non-significant; * p≤ 0.05; ** p≤ 0.005; \***| p≤ 0.001. ### Correlation of demographical and tumoral characteristics with immune cells TILs groups and percentages were not significantly correlated to patients’ clinical characteristics, except for BMI (p=0.038 and 0.007 respectively) and the presence of BRCA mutations (p=0.009 and 0.016 respectively). Regarding tumor characteristics, only DCIS (p=0.037) and grade (p=1.8.10−9) were correlated with TILs percentages. Th2 response was only significantly related to intravascular tumor emboli (p=0.047) and there was a trend of an association with both lymph node involvement and grade (p=0.123 and 0.180 respectively) (Supplementary Table 4). ### RFS and OS analyses 16 patients (17.6%) relapsed, mostly by distant metastasis (15.4%), and 10 (11%) died of cancer-related cause resulting in a 5-year RFS and OS of 77% and 83% respectively (Supplementary Table 2 and Supplementary Figure 3). Only BMI was significantly predictive of relapse and OS (p=0.033 and 0.034 respectively) (Table 2). Number of foci and pathological size were significantly predictive of OS (p=0.009 and 0.010 respectively). Pathological size and nodal involvement were also strongly predictive of both RFS and OS in multivariate analyses (p<0.05)(Table 2 and Figure 3). All the non-significative variables are shown in Supplementary table 5. ![Figure 3-](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/06/06/2023.06.02.23289891/F3.medium.gif) [Figure 3-](http://medrxiv.org/content/early/2023/06/06/2023.06.02.23289891/F3) Figure 3- Relapse-free (RFS) and overall survival (OS) analyses. RFS and OS according to tumor pathological size (median=20mm), pathologically confirmed lymph node metastasis, dendritic cells percentage by FC (median=0.07%), tumor-infiltrating lymphocytes (TILs) percentage (median =10.0%), and Th2 lymphocytes percentage by FC (median=7.98%). Kaplan Meier curves were estimated, log rank tests (for p values) were performed to compare survival distributions. p values in bold font are significant ones. View this table: [Table 2-](http://medrxiv.org/content/early/2023/06/06/2023.06.02.23289891/T2) Table 2- Correlation of patient, tumor, immunity, and treatment characteristics to relapse-free survival (RFS) and overall survival (OS). p values for survival status are obtained using Wilcoxon test for quantitative variables and Chi2 or Fisher exact test for qualitative ones, depending if the group do or do not contain at least 5 values respectively. p values for relapse free and survival regression are obtained using log rank test for univariate (univar) analyses and Cox model for multivariate ones (multivar), using BIC condition for prognostic usefulness. p values <0.05 appear in bold font in the text. *p values/ HR [CI95%]. BMI : Body mass index; DCIS : Ductal carcinoma in-situ; IV : Intra-vascular; TILs : Tumor-infiltrating lymphocytes; FC : Flow cytometry; BCSC : Breast Cancer Stem Cells; DBCC : Differentiated Breast Cancer Cells;DC : Dendritic Cells; pTNM : pathological staging of tumor size(T), lymph node involvement(N), and metastatic status(M) of breast cancer according to international guidelines. The TILs percentage was predictive of OS (p=0.044) (Figure 3 and Table 2) but only tended to be of RFS (p=0.071). The percentages of CD45+ leukocytes, CD4+ helper TC, and macrophages were not significantly predictive of RFS or OS, and we observed a non-significant trend with CD8+ cytotoxic TCs (p=0.072 and 0.188 respectively). However, we discovered that a high Th2 infiltration was strongly predictive of both RFS and OS (Figure 3) in univariate (p=0.009 and 0.008 respectively) and multivariate (p=0.002 and 0.010 respectively) analyses (Table 2), while Th1 and Th17 were not. Myeloid cells including dendritic ones were also significantly predictive of relapse (p=0.012 and 0.030 respectively) (Table 2). When considering the different Th populations, only Th2 was a better predictive factor of survival, especially OS, than total leukocytes (Figure 4). In accord, almost all patients having relapsed or died were Th2 low (below the median), even though some of them where highly infiltrated by lymphocytes. Th1 and Th17 did not add the same prognostic value, and Th17 was particularly poorly correlated to RFS and OS (Figure 4). ![Figure 4-](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/06/06/2023.06.02.23289891/F4.medium.gif) [Figure 4-](http://medrxiv.org/content/early/2023/06/06/2023.06.02.23289891/F4) Figure 4- Relapse and overall survival status of patients depending on Th subpopulation versus total CD45+ leukocytes infiltration measured by flow cytometry (FC). Repartition of free from relapse (no relapse in blue) patients versus having-relapsed ones (relapse in red) (A,C,E) and alive (red) versus deceased (blue) ones (B,D,F) ones according to Th2(A,B), Th1 (C,D), Th17 (E,F) infiltrations vs all leukocytes. Dashed lines represent medians of different subpopulations infiltrations. ## Discussion The emergence of immunotherapy represents a new hope for TNBC patients since these tumors are frequently highly infiltrated by immune cells and may thus respond to immune checkpoint inhibitors. However, markers than can robustly predict responsiveness to immune checkpoint therapy are lacking in TNBC. Furthermore, the nature of this immune infiltrate remains largely undocumented. Hence, it is essential to better characterize it to identify patients that would benefit the most from these new therapies. ### TILs content, methods of investigation, limits, and interpretation TILs are needed to recognize disseminated tumor cells or death-associated molecular patterns (DAMPs) to trigger further immune recruitment, infiltration, and activation of immunogenic cancer cell death (41). Regardless of their localization, the characterization of TILs subtypes is critical, since they can have either a pro-or an anti-tumorigenic function (42). Memory, helper, or CD4+ fh TC can modulate the immune response, while cytotoxic CD8+ TC play a main role in cancer killing. FOXP3+ Treg or the activation of immune checkpoints (PD1/PD-L1) and CTLA4 are crucial in immune tolerance (33). Immune escape (drop in CD8+ and CD4+ TC) and tolerance (by PD-L1, PD-L2, and HLA-1 overexpression (43) or Treg activation (44)) are associated with poor prognosis and with the development of metastatic disease. But results are still controversial (29,45–48), also depending on tumoral TILs localization (49). Zhang et al. even showed that Treg and PD-1/PD-L1 expression was correlated with better OS in TNBC That is why immunomodulatory subtypes such as Treg and Th2 CD4+ TC need to be considered and better understood in TNBC. TILs percentage is currently the only assessed parameter in the clinic but has many limits. First, there is a need for standardized TILs measurements by pathologists (38), since variability can affect the correlation of TILs with pCR (51). Moreover, the TILs working group only recommends the measurement of stromal TILs that are easier to measure than intratumoral ones (39,52). Nevertheless, results are still controversial (31), since Ruan et al. showed that both intratumoral and stromal TILs were independent predictors for pCR (28). In our study, and contrary to the data previously mentioned (24–26), stromal TILs were not significantly associated with RFS and OS, perhaps because of the limited amount of relapses and death events in the studied cohort. However, other studies also failed to show a prognostic utility of TILs count alone (53). CD8+ and CD4+ TC were not significantly predictive of RFS or OS in our cohort either, while they frequently are in TNBC (24,26,29–32,48,54), maybe for the same reasons. ### Need for reproducible immune characterization In our study, we counteracted these limitations by immunophenotyping the immune infiltrate on fresh tumor, without consideration of localization using a highly standardized and trustworthy FC method. Other studies use whole exome sequencing to assess T cell infiltration and correlate these data to prognosis and response to immunotherapy (55), or even gene-expression deconvolution to characterize T cell subtypes (53,56–58). However there is currently no consensual transcriptomic signature for Th2 cells. ### Th2 prognostic value We observed that Th2 infiltration was the most frequent one, as previously reported (20). The prognostic value of Th2 in various cancers is often unconclusive or assumed to favor pro-tumorigenic immune-tolerance because of its link to aggressive features (59–65). On the contrary, high Th2 infiltration was a strong factor of good prognosis in our cohort, as already suspected in previous studies through thymic stromal lymphopoietin (TSLP) (66,67), or collaboration with eosinophils (68,69), sometimes being as powerfully antitumoral as Th1 (70,71). Th2 levels even favorably correlated to pCR in ER+ breast cancers (72). AllergoOncology group showed that immune escape can be reversed by IgE and Th2 response (69,73,74). Th2 anti-tumoral action could be boosted by anti-TGF-β (75) and Th2/Th1 response could be recapacitated by xanthohumol (76), saikosaponin (77), anti-IL4/IL4R (78), anti-IL13 (79), anti-CCR4 (80), among others (70,81). Other interesting perspectives consist of better understanding Th2 interaction with other immune subsets, like fibroblasts (82), macrophages (83,84), dendritic cells (79,85), or eosinophils and IgE (68,86), to better control Th2 response. Finally, immunotherapies modulating Th2 response could also be used around the time of surgery that has been proved to induce immunomodulation (87). ### DC prognostic value We also showed that dendritic cells, potentially through their interaction with Th2 CD4+ TC, were correlated with better survival. On the contrary, DC have also been reported as immunosuppressive (79), especially through Th17 response (85). To face this complexity, it will be required to consider the DC subset involved (88). In addition, studying a wider signature including B-cells and myeloid-derived antigen-presenting cells (89), such as the ones used to predict response to treatment (90,91) will likely be informative. Of note, immune metagenes overexpression and innate and adoptive immune cooperation are also associated with low-risk of metastasis in TNBC (91,92). ### Clinical features and immune microenvironment Only BMI correlated with TILs infiltration in our cohort, emphasizing the complex immune relation between obesity and cancer (93), especially through Th2 processes (94). Few of the tumoral characteristics were correlated to TILs infiltration, as previously reported (20). Nevertheless, Th2 percentages was significantly correlated with intravascular tumor emboli, a variable often linked to S.B.R. grade and lymph node involvement. Th2 levels were already proved to be higher in tumor microenvironment and lymph nodes in case of aggressive, therapy-resistant, metastatic, and large or locally advanced breast cancers (30,72,95–99), confirming their abundance without proving their pejorative prognostic value. ### Strength and weakness of our study and added value Potential limitations of the applicability of our results could be the good RFS and OS of our patients compared to general TNBC populations (8), and the inclusion of patients who underwent surgery first whereas NAC is nowadays the standard of care for numerous TNBC. However, identifying a Th2 response in supposedly favorable-prognosis BC could be of tremendous interest to prevent unexpected but still early and deadly relapses. Moreover, even if TILs assessment post-NAC has some specificities (39,100), it is transposable to the ones performed pre-NAC (35,101), including for Th response in tumor (99) and in lymph nodes (95). TILs assessment post-NAC is also an independent predictor of survival (40,102), even better when used alongside Residual Cancer Burden score (RCB) (103). Besides, TILs count can also be done on pre-NAC biopsies and still carry the same prognostic value (104). Finally, modification of immune subpopulations percentages and ratios has also been proved to be of importance after NAC (47,105), and could thus be studied at both timepoints. A better understanding of the TILs in TNBC, especially Th2 (75), could eventually enable to target them more specifically (106–108), or even to use them for Adoptive cell transfer (109), CAR-TC design, or even vaccines (21,110). However, we still need to identify which tumors are immunogenic enough to benefit from it (111,112). Following the CREATE-X trial, adjuvant chemotherapy (Capecitabine) is currently given to non-pCR TNBC patients regardless of the immune infiltrate (113), while only some of them will benefit from it. Hence, immune characterization could predict chemotherapy efficacy or immune exhaustion making relevant the addition of immunotherapy to Capecitabine for some selected non-pCR TNBCs, similarly to those benefiting from Atezolizumab in metastatic settings (21). Finally, necessary future therapeutic development include further understanding of the role of other immune cells such as macrophages (114), natural killer TC (NK cells) (115), and their interactions with TILs and Th2. ## Supporting information Supplementary Tables and Figures [[supplements/289891_file03.pdf]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors ## Conflict of interest The authors declare no conflict of interest. ## Author contribution SB wrote and edited the manuscript, raised funding, designed and performed experiments, analyzed the data and generated figures, FG provided patient samples and edited the manuscript, PT analyzed IHC BCG performed the statistical analyses, EL contributed to the writing and editing of the manuscript, AP performed some of the Flow cytometry experiments, JL and VL provided patient samples. MLG supervised the work, raised funding, wrote and edited the manuscript, designed and performed experiments, analyzed the data and generated figures. ## Supplementary materials ***Supplementary Table 1- Patient and tumor characteristics.*** *For quantitative variables we give the statistical dispersion defined by the average, the standard deviation (SD), the median, the minimum and maximum (Min-Max), whereas we give percentage (%) and actual number of events (n) over total population (n total) for qualitative ones. ADD: Anxiety-depressive disorder; AHT: Arterial hypertension; BMI: Body mass index; BC: Breast carcinoma; IDC: Invasive ductal carcinoma; ILC: Invasive lobular carcinoma; DCIS: Ductal carcinoma in-situ; IV: Intra-vascular; SLN: Sentinel lymph node; LN: Lymph node; pTNM: pathological staging of tumor size(T). lymph node involvement(N). and metastatic status(M) of breast cancer according to international guidelines*. ***Supplementary Table 2- Treatment, and follow-up characteristics.*** *For quantitative variables we give the statistical dispersion defined by the average, the standard deviation (SD), the median, the minimum and maximum (Min-Max), whereas we give percentage (%) and actual number of events (n) over total population (n total) for qualitative ones. SLN: Sentinel lymph node dissection; LND: Lymph node dissection; LN: Lymph nodes. Chemotherapy regimen were mainly FEC (5-flurouracile, Epirubicine, Cyclophosphamide) and Taxanes for all but eight patients who got either Taxanes for half of them (alone or with targeted therapy), or Cyclophosphamide for the other half (+ Doxorubicine for two or Taxanes for the other two). Three of these patients also got targeted therapy by Cobimetinib for two and Olaparib for one of them*. ***Supplementary Table 3- Tumor infiltrating lymphocytes (TILs) count and immune infiltrate characterization by flow cytometry (FC).*** *For quantitative variables we give the statistical dispersion defined by the average, the standard deviation (SD), the median, the minimum and maximum (Min-Max), whereas we give percentage (%) and actual number of events (n) over total population (n total) for qualitative ones. TILs: Tumor-infiltrating lymphocytes; FC: Flow cytometry; BCSC: Breast Cancer Stem Cells; DBCC: Differentiated Breast Cancer Cells*. ***Supplementary Table 4- TILs and Th2 correlation with patient, tumor, and treatment characteristics.*** *p values are obtained thanks to a chi2 or a Fisher’s exact test (when there are less than 5 patients per group) for TILs group comparison of qualitative values and an ANOVA one for quantitative values. For TILs percentages and Th2 response comparisons, p values are obtained from a Student or an ANOVA test (when there are more than 2 groups) for qualitative values and a Pearson correlation one for quantitative values. p values in bold font are significant ones. HRT: Hormone Replacement Therapy; BMI: Body mass index; BC: Breast carcinoma; S.B.R.: Scarff-Bloom and Richardson; DCIS: Ductal carcinoma in-situ; IV: Intra-vascular; LN: Lymph Node; pTNM: pathological staging of tumor size(T), lymph node involvement (N), and metastatic status(M) of breast cancer according to international guidelines*. ***Supplementary Table 5- Correlation of patient, tumor, immunity, and treatment characteristics to relapse-free survival (RFS) or overall survival (OS).*** *p values for relapse status are obtained using Wilcoxon test for quantitative variables and Chi2 or Fisher exact test for qualitative ones (if the group does or does not contain at least 5 values respectively). p values for survival regression are obtained using log rank test for univariate (univar) analyses and Cox model for multivariate ones (multivar), using BIC condition for prognostic usefulness. p values <0.05 appear in bold font in the text. *p values/ HR [CI95%]. BMI: Body mass index; BC: Breast cancer; DCIS: Ductal carcinoma in-situ; IV: Intra-vascular; LN: Lymph Nodes; pTNM: pathological staging of tumor size (T), lymph node involvement (N), and metastatic status (M) of breast cancer according to international guidelines; TILs: Tumor-infiltrating lymphocytes; FC: Flow cytometry; BCSC: Breast Cancer Stem Cells; DBCC: Differentiated Breast Cancer Cells*. ***Supplementary Figure 1- Flow chart for patient inclusion.*** *HR+ +/− Her2+: hormonal receptors and/or Her2 positive; BC: breast cancers; TNBC: Triple-negative breast cancers; TNBC only*: exclusion of patients having developed another histological type of cancer or a recurrence as an HR+ BC; TILs: Tumor-infiltrating lymphocytes; w/o: without; w/: with; FC: Flow Cytometry. Exclusion criteria are depicted in grey*. ***Supplementary Figure 2 – Flow Cytometry (FC) characterization of tumor cells and tumor-infiltrating immune cells according to their cluster of differentiation (CD).*** *BC: Breast cancer; SC: Stem cells; DBCC: Differentiated breast cancer cells; CD4+: helper T lymphocytes; CD8+: cytotoxic T lymphocytes; BT; lab number*. ***Supplementary Figure 3- Relapse-free survival (A) and overall survival (B) for the entire cohort.*** *Kaplan Meier curves were estimated, and percentages of survival rates derived from a log-rank test*. ## Acknowledgements SB This work was funded by grants from Rennes University Hospital (SB). MLG got fundings from La Vannetaise ## Abbreviations BC : Breast Cancer BCSC : Breast Cancer Stem Cell CD : Cluster of Differentiation DBCC : Differentiated Breast Cancer Cell DC : Dendritic cells DCIS : Ductal Carcinoma In Situ FC : Flow Cytometry FFPE : Formalin-Fixed Paraffin-Embedded H&E : Hematoxylin and Eosin LN : Lymph Node NAC : Neoadjuvant chemotherapy OS : Overall Survival pCR : pathological Complete Response pTNM : pathological classification of Tumor size (T), Node involvement (N), and Metastasis (M) according to international guidelines RFS : Relapse-Free Survival TC : T Cells (Lymphocytes) Th response : T helper response TILs : Tumor-Infiltrating Lymphocytes TNBC : Triple-Negative Breast Cancer * Received June 2, 2023. * Revision received June 2, 2023. * Accepted June 6, 2023. * © 2023, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## Bibliography 1. 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. nov 2018;68(6):394–424. 2. 2.Street W. Breast Cancer Facts & Figures 2017-2018. :44. 3. 3.Badowska-Kozakiewicz AM, Budzik MP. Immunohistochemical characteristics of basal-like breast cancer. Contemp Oncol. 2016;20(6):436–43. 4. 4.Sizemore ST, Sizemore GM, Booth CN, Thompson CL, Silverman P, Bebek G, et al. Hypomethylation of the MMP7 promoter and increased expression of MMP7 distinguishes the basal-like breast cancer subtype from other triple-negative tumors. Breast Cancer Res Treat. juill 2014;146(1):25–40. 5. 5.Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. nov 2016;13(11):674–90. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrclinonc.2016.66&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27184417&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 6. 6.Kutomi G, Ohmura T, Suzuki Y, Kameshima H, Shima H, Takamaru T, et al. Clinicopathological Characteristics of Basal Type Breast Cancer in Triple-Negative Breast Cancer. J Cancer Ther. 2012;03(05):836–40. 7. 7.Li X, Yang J, Peng L, Sahin AA, Huo L, Ward KC, et al. Triple-negative breast cancer has worse overall survival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res Treat. 2017;161(2):279–87. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10549-016-4059-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27888421&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 8. 8.DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Goding Sauer A, et al. Breast cancer statistics, 2019. CA Cancer J Clin. nov 2019;69(6):438–51. 9. 9.Gluz O, Liedtke C, Gottschalk N, Pusztai L, Nitz U, Harbeck N. Triple-negative breast cancer--current status and future directions. Ann Oncol Off J Eur Soc Med Oncol. déc 2009;20(12):1913–27. 10. 10.Lin NU, Vanderplas A, Hughes ME, Theriault RL, Edge SB, Wong YN, et al. Clinicopathological Features, Patterns of Recurrence, and Survival Among Women With Triple-Negative Breast Cancer in the National Comprehensive Cancer Network. Cancer. 15 nov 2012;118(22):5463–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/cncr.27581&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22544643&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000310483800002&link_type=ISI) 11. 11.Malorni L, Shetty PB, De Angelis C, Hilsenbeck S, Rimawi MF, Elledge R, et al. Clinical and biologic features of triple-negative breast cancers in a large cohort of patients with long-term follow-up. Breast Cancer Res Treat. déc 2012;136(3):795–804. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10549-012-2315-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23124476&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 12. 12.Lehmann 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. 1 juill 2011;121(7):2750–67. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI45014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21633166&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000292413000024&link_type=ISI) 13. 13.Mayer IA, Abramson VG, Lehmann BD, Pietenpol JA. NEW STRATEGIES FOR TRIPLE NEGATIVE BREAST CANCER – DECIPHERING THE HETEROGENEITY. Clin Cancer Res Off J Am Assoc Cancer Res. 15 févr 2014;20(4):782–90. 14. 14.Jiang YZ, Ma D, Suo C, Shi J, Xue M, Hu X, et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell. 18 mars 2019;35(3):428–440.e5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ccell.2019.02.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 15. 15.Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, et al. Comprehensive Genomic Analysis Identifies Novel Subtypes and Targets of Triple-negative Breast Cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 1 avr 2015;21(7):1688–98. 16. 16.Caparica R, Lambertini M, Pondé N, Fumagalli D, de Azambuja E, Piccart M. Post-neoadjuvant treatment and the management of residual disease in breast cancer: state of the art and perspectives. Ther Adv Med Oncol. 2019;11:1758835919827714. 17. 17.Bardia A, Hurvitz SA, Tolaney SM, Loirat D, Punie K, Oliveira M, et al. Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer. N Engl J Med. 22 avr 2021;384(16):1529–41. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2028485&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 18. 18.Nagayama A, Vidula N, Bardia A. Novel Therapies for Metastatic Triple-Negative Breast Cancer: Spotlight on Immunotherapy and Antibody-Drug Conjugates. Oncol Williston Park N. 13 mai 2021;35(5):249–54. 19. 19.Stanton SE, Adams S, Disis ML. Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review. JAMA Oncol. 1 oct 2016;2(10):1354–60. 20. 20.Glajcar A, Szpor J, Hodorowicz-Zaniewska D, Tyrak KE, Okoń K. The composition of T cell infiltrates varies in primary invasive breast cancer of different molecular subtypes as well as according to tumor size and nodal status. Virchows Arch Int J Pathol. juill 2019;475(1):13–23. 21. 21.Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N Engl J Med. 29 2018;379(22):2108–21. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/nejmoa1809615&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30345906&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 22. 22.Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N Engl J Med. 27 févr 2020;382(9):810–21. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1910549&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 23. 23.Schmid P, Cortes J, Dent R, Pusztai L, McArthur H, Kümmel S, et al. Event-free Survival with Pembrolizumab in Early Triple-Negative Breast Cancer. N Engl J Med. 10 févr 2022;386(6):556–67. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2112651&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 24. 24.Ali HR, Chlon L, Pharoah PDP, Markowetz F, Caldas C. Patterns of Immune Infiltration in Breast Cancer and Their Clinical Implications: A Gene-Expression-Based Retrospective Study. PLoS Med. déc 2016;13(12):e1002194. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 25. 25.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(1):40–50. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 26. 26.Criscitiello C, Esposito A, Trapani D, Curigliano G. Prognostic and predictive value of tumor infiltrating lymphocytes in early breast cancer. Cancer Treat Rev. 24 févr 2018;50:205–7. 27. 27.Kurozumi S, Fujii T, Matsumoto H, Inoue K, Kurosumi M, Horiguchi J, et al. Significance of evaluating tumor-infiltrating lymphocytes (TILs) and programmed cell death-ligand 1 (PD-L1) expression in breast cancer. Med Mol Morphol. déc 2017;50(4):185–94. 28. 28.Ruan M, Tian T, Rao J, Xu X, Yu B, Yang W, et al. Predictive value of tumor-infiltrating lymphocytes to pathological complete response in neoadjuvant treated triple-negative breast cancers. Diagn Pathol. 31 août 2018;13(1):66. 29. 29.Tsiatas M, Kalogeras KT, Manousou K, Wirtz RM, Gogas H, Veltrup E, et al. Evaluation of the prognostic value of CD3, CD8, and FOXP3 mRNA expression in early-stage breast cancer patients treated with anthracycline-based adjuvant chemotherapy. Cancer Med. oct 2018;7(10):5066–82. 30. 30.Kaewkangsadan V, Verma C, Eremin JM, Cowley G, Ilyas M, Eremin O. Crucial Contributions by T Lymphocytes (Effector, Regulatory, and Checkpoint Inhibitor) and Cytokines (TH1, TH2, and TH17) to a Pathological Complete Response Induced by Neoadjuvant Chemotherapy in Women with Breast Cancer. J Immunol Res. 2016;2016:4757405. 31. 31.Matsumoto H, Thike AA, Li H, Yeong J, Koo SL, Dent RA, et al. Increased CD4 and CD8-positive T cell infiltrate signifies good prognosis in a subset of triple-negative breast cancer. Breast Cancer Res Treat. avr 2016;156(2):237–47. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10549-016-3743-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 32. 32.Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 1 juill 2013;123(7):2873–92. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI67428&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23778140&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000321316700017&link_type=ISI) 33. 33.Quigley DA, Kristensen V. Predicting prognosis and therapeutic response from interactions between lymphocytes and tumor cells. Mol Oncol. 1 déc 2015;9(10):2054–62. 34. 34.Martínez-Canales S, Cifuentes F, López De Rodas Gregorio M, Serrano-Oviedo L, Galán-Moya EM, Amir E, et al. Transcriptomic immunologic signature associated with favorable clinical outcome in basal-like breast tumors. PLOS ONE. 4 mai 2017;12(5):e0175128. 35. 35.Lee HJ, Lee JJ, Song IH, Park IA, Kang J, Yu JH, et al. Prognostic and predictive value of NanoString-based immune-related gene signatures in a neoadjuvant setting of triple-negative breast cancer: relationship to tumor-infiltrating lymphocytes. Breast Cancer Res Treat. juin 2015;151(3):619–27. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 36. 36.Jézéquel P, Loussouarn D, Guérin-Charbonnel C, Campion L, Vanier A, Gouraud W, et al. Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response. Breast Cancer Res BCR. 20 mars 2015;17:43. 37. 37.Bonsang-Kitzis H, Sadacca B, Hamy-Petit A, Moarii M, Pinheiro A, Laurent C, et al. Biological network-driven gene selection identifies a stromal immune module as a key determinant of triple-negative breast carcinoma prognosis. OncoImmunology. 2 janv 2016;5(1):e1061176. 38. 38.Salgado 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 Off J Eur Soc Med Oncol. févr 2015;26(2):259–71. 39. 39.Dieci MV, Radosevic-Robin N, Fineberg S, van den Eynden G, Ternes N, Penault-Llorca F, et al. 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. 1 oct 2018;52:16–25. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.semcancer.2017.10.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 40. 40.Loi S, Drubay D, Adams S, Pruneri G, Francis PA, Lacroix-Triki M, et al. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J Clin Oncol. 16 janv 2019;37(7):559–69. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1200/jco.18.01010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 41. 41.König L, Mairinger FD, Hoffmann O, Bittner AK, Schmid KW, Kimmig R, et al. Dissimilar patterns of tumor-infiltrating immune cells at the invasive tumor front and tumor center are associated with response to neoadjuvant chemotherapy in primary breast cancer. BMC Cancer. 4 févr 2019;19(1):120. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 42. 42.Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 15 2012;12(4):298–306. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrc3245&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22419253&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000302134900014&link_type=ISI) 43. 43.Ogiya R, Niikura N, Kumaki N, Bianchini G, Kitano S, Iwamoto T, et al. Comparison of tumor-infiltrating lymphocytes between primary and metastatic tumors in breast cancer patients. Cancer Sci. 1 déc 2016;107(12):1730–5. 44. 44.Shou J, Zhang Z, Lai Y, Chen Z, Huang J. Worse outcome in breast cancer with higher tumor-infiltrating FOXP3+ Tregs : a systematic review and meta-analysis. BMC Cancer. 26 2016;16:687. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 45. 45.West NR, Kost SE, Martin SD, Milne K, Deleeuw RJ, Nelson BH, et al. Tumour-infiltrating FOXP3(+) lymphocytes are associated with cytotoxic immune responses and good clinical outcome in oestrogen receptor-negative breast cancer. Br J Cancer. 15 janv 2013;108(1):155–62. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/bjc.2012.524&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23169287&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000314800600021&link_type=ISI) 46. 46.Papaioannou E, Sakellakis M, Melachrinou M, Tzoracoleftherakis E, Kalofonos H, Kourea E. A Standardized Evaluation Method for FOXP3+ Tregs and CD8+ T-cells in Breast Carcinoma: Association With Breast Carcinoma Subtypes, Stage and Prognosis. Anticancer Res. mars 2019;39(3):1217–32. 47. 47.Miyashita M, Sasano H, Tamaki K, Hirakawa H, Takahashi Y, Nakagawa S, et al. Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: a retrospective multicenter study. Breast Cancer Res BCR. 4 sept 2015;17:124. 48. 48.Sudarsa IW, Subawa DG, Adiputra PAT, Manuaba IBTW. Correlation of CD8+ Expression, Foxp3+ Expression, and CD8+/Foxp3+ Ratio with Triple Negative Breast Cancer Stage in Sanglah General Hospital. Open Access Maced J Med Sci. 31 mai 2019;7(10):1593–6. 49. 49.Liu F, Lang R, Zhao J, Zhang X, Pringle GA, Fan Y, et al. CD8+ cytotoxic T cell and FOXP3+ regulatory T cell infiltration in relation to breast cancer survival and molecular subtypes. Breast Cancer Res Treat. nov 2011;130(2):645–55. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10549-011-1647-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21717105&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000295675700029&link_type=ISI) 50. 50.Zhang L, Wang XI, Ding J, Sun Q, Zhang S. The predictive and prognostic value of Foxp3+/CD25+ regulatory T cells and PD-L1 expression in triple negative breast cancer. Ann Diagn Pathol. juin 2019;40:143–51. 51. 51.Van Bockstal MR, François A, Altinay S, Arnould L, Balkenhol M, Broeckx G, et al. Interobserver variability in the assessment of stromal tumor-infiltrating lymphocytes (sTILs) in triple-negative invasive breast carcinoma influences the association with pathological complete response: the IVITA study. Mod Pathol Off J U S Can Acad Pathol Inc. 3 juill 2021; 52. 52.Denkert C, Wienert S, Poterie A, Loibl S, Budczies J, Badve S, 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 Off J U S Can Acad Pathol Inc. 2016;29(10):1155–64. 53. 53.Craven KE, Gökmen-Polar Y, Badve SS. CIBERSORT analysis of TCGA and METABRIC identifies subgroups with better outcomes in triple negative breast cancer. Sci Rep. 25 févr 2021;11(1):4691. 54. 54.Ibrahim EM, Al-Foheidi ME, Al-Mansour MM, Kazkaz GA. The prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancer: a meta-analysis. Breast Cancer Res Treat. déc 2014;148(3):467–76. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10549-014-3185-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25361613&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 55. 55.Bentham R, Litchfield K, Watkins TBK, Lim EL, Rosenthal R, Martínez-Ruiz C, et al. Using DNA sequencing data to quantify T cell fraction and therapy response. Nature. 8 sept 2021;1–6. 56. 56.Hao Y, Yan M, Heath BR, Lei YL, Xie Y. Fast and robust deconvolution of tumor infiltrating lymphocyte from expression profiles using least trimmed squares. PLOS Comput Biol. 6 mai 2019;15(5):e1006976. 57. 57.Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling Tumor Infiltrating Immune Cells with CIBERSORT. Methods Mol Biol Clifton NJ. 2018;1711:243–59. 58. 58.Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 15 nov 2017;18(1):220. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10/gckmjs&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31601707&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 59. 59.Ruffell B, DeNardo DG, Affara NI, Coussens LM. Lymphocytes in cancer development: polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. févr 2010;21(1):3–10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cytogfr.2009.11.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20005150&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000276592700002&link_type=ISI) 60. 60.Johansson M, DeNardo DG, Coussens LM. Polarized immune responses differentially regulate cancer development. Immunol Rev. avr 2008;222:145–54. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1600-065X.2008.00600.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18363999&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000254274900009&link_type=ISI) 61. 61.Faucheux L, Grandclaudon M, Perrot-Dockès M, Sirven P, Berger F, Hamy AS, et al. A multivariate Th17 metagene for prognostic stratification in T cell non-inflamed triple negative breast cancer. Oncoimmunology. 2019;8(9):e1624130. 62. 62.Kristensen VN, Vaske CJ, Ursini-Siegel J, Van Loo P, Nordgard SH, Sachidanandam R, et al. Integrated molecular profiles of invasive breast tumors and ductal carcinoma in situ (DCIS) reveal differential vascular and interleukin signaling. Proc Natl Acad Sci U S A. 21 févr 2012;109(8):2802–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTA5LzgvMjgwMiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA2LzA2LzIwMjMuMDYuMDIuMjMyODk4OTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 63. 63.Zhang Q, Qin J, Zhong L, Gong L, Zhang B, Zhang Y, et al. CCL5-Mediated Th2 Immune Polarization Promotes Metastasis in Luminal Breast Cancer. Cancer Res. 15 oct 2015;75(20):4312–21. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3NS8yMC80MzEyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDYvMDYvMjAyMy4wNi4wMi4yMzI4OTg5MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 64. 64.Espinoza JA, Jabeen S, Batra R, Papaleo E, Haakensen V, Timmermans Wielenga V, et al. Cytokine profiling of tumor interstitial fluid of the breast and its relationship with lymphocyte infiltration and clinicopathological characteristics. Oncoimmunology. 2016;5(12):e1248015. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/2162402X.2016.1248015&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28123884&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 65. 65.Chen X, Wang J, Wang R, Su Q, Luan J, Huang H, et al. Th1-, Th2-, and Th17-associated cytokine expression in hypopharyngeal carcinoma and clinical significance. Eur Arch Oto-Rhino-Laryngol Off J Eur Fed Oto-Rhino-Laryngol Soc EUFOS Affil Ger Soc Oto-Rhino-Laryngol - Head Neck Surg. févr 2016;273(2):431–8. 66. 66.Demehri S, Cunningham TJ, Manivasagam S, Ngo KH, Moradi Tuchayi S, Reddy R, et al. Thymic stromal lymphopoietin blocks early stages of breast carcinogenesis. J Clin Invest. 1 avr 2016;126(4):1458–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI83724&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 67. 67.Boieri M, Malishkevich A, Guennoun R, Marchese E, Kroon S, Trerice KE, et al. CD4+ T helper 2 cells suppress breast cancer by inducing terminal differentiation. J Exp Med. 4 juill 2022;219(7):e20201963. 68. 68.Ellyard JI, Simson L, Parish CR. Th2-mediated anti-tumour immunity: friend or foe? Tissue Antigens. juill 2007;70(1):1–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1399-0039.2007.00869.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17559575&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000247173300001&link_type=ISI) 69. 69.Jensen-Jarolim E, Bax HJ, Bianchini R, Capron M, Corrigan C, Castells M, et al. AllergoOncology – the impact of allergy in oncology: EAACI position paper. Allergy. juin 2017;72(6):866–87. 70. 70.Mattes J, Hulett M, Xie W, Hogan S, Rothenberg ME, Foster P, et al. Immunotherapy of cytotoxic T cell-resistant tumors by T helper 2 cells: an eotaxin and STAT6-dependent process. J Exp Med. 3 févr 2003;197(3):387–93. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjk6IjE5Ny8zLzM4NyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA2LzA2LzIwMjMuMDYuMDIuMjMyODk4OTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 71. 71.Nishimura T, Iwakabe K, Sekimoto M, Ohmi Y, Yahata T, Nakui M, et al. Distinct role of antigen-specific T helper type 1 (Th1) and Th2 cells in tumor eradication in vivo. J Exp Med. 6 sept 1999;190(5):617–27. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjk6IjE5MC81LzYxNyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA2LzA2LzIwMjMuMDYuMDIuMjMyODk4OTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 72. 72.Le L, Tokumaru Y, Oshi M, Asaoka M, Yan L, Endo I, et al. Th2 cell infiltrations predict neoadjuvant chemotherapy response of estrogen receptor-positive breast cancer. Gland Surg. janv 2021;10(1):154–65. 73. 73.Jensen-Jarolim E, Bax HJ, Bianchini R, Crescioli S, Daniels-Wells TR, Dombrowicz D, et al. AllergoOncology: Opposite outcomes of immune tolerance in allergy and cancer. Allergy. 2018;73(2):328–40. 74. 74.Wulaningsih W, Holmberg L, Garmo H, Karagiannis SN, Ahlstedt S, Malmstrom H, et al. Investigating the association between allergen-specific immunoglobulin E, cancer risk and survival. Oncoimmunology. juin 2016;5(6):e1154250. 75. 75.Li S, Liu M, Do MH, Chou C, Stamatiades EG, Nixon BG, et al. Cancer immunotherapy via targeted TGF-β signalling blockade in TH cells. Nature. nov 2020;587(7832):121–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-020-2850-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 76. 76.Zhang W, Pan Y, Gou P, Zhou C, Ma L, Liu Q, et al. Effect of xanthohumol on Th1/Th2 balance in a breast cancer mouse model. Oncol Rep. janv 2018;39(1):280–8. 77. 77.Zhao X, Liu J, Ge S, Chen C, Li S, Wu X, et al. Saikosaponin A Inhibits Breast Cancer by Regulating Th1/Th2 Balance. Front Pharmacol. 2019;10:624. 78. 78.Bankaitis KV, Fingleton B. Targeting IL4/IL4R for the treatment of epithelial cancer metastasis. Clin Exp Metastasis. déc 2015;32(8):847–56. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10585-015-9747-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26385103&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 79. 79.Aspord C, Pedroza-Gonzalez A, Gallegos M, Tindle S, Burton EC, Su D, et al. Breast cancer instructs dendritic cells to prime interleukin 13-secreting CD4+ T cells that facilitate tumor development. J Exp Med. 14 mai 2007;204(5):1037–47. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjEwOiIyMDQvNS8xMDM3IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDYvMDYvMjAyMy4wNi4wMi4yMzI4OTg5MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 80. 80.Berlato C, Khan MN, Schioppa T, Thompson R, Maniati E, Montfort A, et al. A CCR4 antagonist reverses the tumor-promoting microenvironment of renal cancer. J Clin Invest. 1 mars 2017;127(3):801–13. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI82976&link_type=DOI) 81. 81.Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev J Clin Ther. août 2003;8(3):223–46. 82. 82.Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer. Cancer Cell. 12 2018;33(3):463–479.e10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ccell.2018.01.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 83. 83.Mandal PK, Biswas S, Mandal G, Purohit S, Gupta A, Majumdar Giri A, et al. CCL2 conditionally determines CCL22-dependent Th2-accumulation during TGF-β-induced breast cancer progression. Immunobiology. 2018;223(2):151–61. 84. 84.Sun L, Chen B, Jiang R, Li J, Wang B. Resveratrol inhibits lung cancer growth by suppressing M2-like polarization of tumor associated macrophages. Cell Immunol. 2017;311:86–93. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cellimm.2016.11.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27825563&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 85. 85.Grandclaudon M, Perrot-Dockès M, Trichot C, Karpf L, Abouzid O, Chauvin C, et al. A Quantitative Multivariate Model of Human Dendritic Cell-T Helper Cell Communication. Cell. 3 oct 2019;179(2):432–447.e21. 86. 86.Jensen-Jarolim E, Turner MC, Karagiannis SN. AllergoOncology: IgE- and IgG4-mediated immune mechanisms linking allergy with cancer and their translational implications. J Allergy Clin Immunol. 2017;140(4):982–4. 87. 87.Boomsma MF, Garssen B, Slot E, Berbee M, Berkhof J, Meezenbroek E de J, et al. Breast cancer surgery-induced immunomodulation. J Surg Oncol. 1 nov 2010;102(6):640–8. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20677220&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 88. 88.Michea P, Noël F, Zakine E, Czerwinska U, Sirven P, Abouzid O, et al. Adjustment of dendritic cells to the breast-cancer microenvironment is subset specific. Nat Immunol. 2018;19(8):885–97. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41590-018-0145-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 89. 89.Alistar A, Chou JW, Nagalla S, Black MA, D’Agostino R, Miller LD. Dual roles for immune metagenes in breast cancer prognosis and therapy prediction. Genome Med. 2014;6(10):80. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13073-014-0080-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25419236&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 90. 90.Bertucci F, Ueno NT, Finetti P, Vermeulen P, Lucci A, Robertson FM, et al. Gene expression profiles of inflammatory breast cancer: correlation with response to neoadjuvant chemotherapy and metastasis-free survival. Ann Oncol Off J Eur Soc Med Oncol. févr 2014;25(2):358–65. 91. 91.Nagalla S, Chou JW, Willingham MC, Ruiz J, Vaughn JP, Dubey P, et al. Interactions between immunity, proliferation and molecular subtype in breast cancer prognosis. Genome Biol. 29 avr 2013;14(4):R34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/gb-2013-14-4-r34&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23618380&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 92. 92.Ascierto ML, Idowu MO, Zhao Y, Khalak H, Payne KK, Wang XY, et al. Molecular signatures mostly associated with NK cells are predictive of relapse free survival in breast cancer patients. J Transl Med. 2013;11:145–145. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1479-5876-11-145&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23758773&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 93. 93.Conroy MJ, Dunne MR, Donohoe CL, Reynolds JV. Obesity-associated cancer: an immunological perspective. Proc Nutr Soc. mai 2016;75(2):125–38. 94. 94.Schwartz C, O’Grady K, Lavelle EC, Fallon PG. Interleukin 33: an innate alarm for adaptive responses beyond Th2 immunity-emerging roles in obesity, intestinal inflammation, and cancer. Eur J Immunol. 2016;46(5):1091–100. 95. 95.Kaewkangsadan V, Verma C, Eremin JM, Cowley G, Ilyas M, Eremin O. Tumour-draining axillary lymph nodes in patients with large and locally advanced breast cancers undergoing neoadjuvant chemotherapy (NAC): the crucial contribution of immune cells (effector, regulatory) and cytokines (Th1, Th2) to immune-mediated tumour cell death induced by NAC. BMC Cancer. 2 févr 2018;18(1):123. 96. 96.Matsuura K, Yamaguchi Y, Osaki A, Ohara M, Okita R, Emi A, et al. FOXP3 expression of micrometastasis-positive sentinel nodes in breast cancer patients. Oncol Rep. nov 2009;22(5):1181–7. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19787238&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 97. 97.Lefrançois P, Xie P, Gunn S, Gantchev J, Villarreal AM, Sasseville D, et al. In silico analyses of the tumor microenvironment highlight tumoral inflammation, a Th2 cytokine shift and a mesenchymal stem cell-like phenotype in advanced in basal cell carcinomas. J Cell Commun Signal. juin 2020;14(2):245–54. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12079-020-00563-6&link_type=DOI) 98. 98.Jabeen S, Espinoza JA, Torland LA, Zucknick M, Kumar S, Haakensen VD, et al. Noninvasive profiling of serum cytokines in breast cancer patients and clinicopathological characteristics. Oncoimmunology. 2019;8(2):e1537691. 99. 99.Verma C, Eremin JM, Robins A, Bennett AJ, Cowley GP, El-Sheemy MA, et al. Abnormal T regulatory cells (Tregs: FOXP3+, CTLA-4+), myeloid-derived suppressor cells (MDSCs: monocytic, granulocytic) and polarised T helper cell profiles (Th1, Th2, Th17) in women with large and locally advanced breast cancers undergoing neoadjuvant chemotherapy (NAC) and surgery: failure of abolition of abnormal treg profile with treatment and correlation of treg levels with pathological response to NAC. J Transl Med. 15 janv 2013;11:16. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1479-5876-11-16&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23320561&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 100.100.Provenzano E, Bossuyt V, Viale G, Cameron D, Badve S, Denkert C, et al. Standardization of pathologic evaluation and reporting of postneoadjuvant specimens in clinical trials of breast cancer: recommendations from an international working group. Mod Pathol Off J U S Can Acad Pathol Inc. sept 2015;28(9):1185–201. 101.101.Denkert C, von Minckwitz G, Brase JC, Sinn BV, Gade S, Kronenwett R, et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol Off J Am Soc Clin Oncol. 20 mars 2015;33(9):983–91. 102.102.Dieci MV, Criscitiello C, Goubar A, Viale G, Conte P, Guarneri V, et al. Prognostic value of tumor-infiltrating lymphocytes on residual disease after primary chemotherapy for triple-negative breast cancer: a retrospective multicenter study. Ann Oncol Off J Eur Soc Med Oncol. mars 2014;25(3):611–8. 103.103.Asano Y, Kashiwagi S, Goto W, Takada K, Takahashi K, Hatano T, et al. Prediction of survival after neoadjuvant chemotherapy for breast cancer by evaluation of tumor-infiltrating lymphocytes and residual cancer burden. BMC Cancer. 28 2017;17(1):888. 104.104.Denkert C, Minckwitz G von, Darb-Esfahani S, Heppner BI, Klauschen F, Furlanetto J, et al. Abstract S1-09: Evaluation of tumor-infiltrating lymphocytes (TILs) as predictive and prognostic biomarker in different subtypes of breast cancer treated with neoadjuvant therapy - A metaanalysis of 3771 patients. Cancer Res. 15 févr 2017;77(4 Supplement):S1-09–S1-09. 105.105.García-Martínez E, Gil GL, Benito AC, González-Billalabeitia E, Conesa MAV, García García T, et al. Tumor-infiltrating immune cell profiles and their change after neoadjuvant chemotherapy predict response and prognosis of breast cancer. Breast Cancer Res BCR. 29 nov 2014;16(6):488. 106.106. Moreno Ayala MA, Gottardo MF, Imsen M, Asad AS, Bal de Kier Joffé E, Casares N, et al. Therapeutic blockade of Foxp3 in experimental breast cancer models. Breast Cancer Res Treat. nov 2017;166(2):393–405. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10549-017-4414-2&link_type=DOI) 107.107.Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer. 2016;4:59. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiaml0YyI7czo1OiJyZXNpZCI7czo2OiI0LzEvNTkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNi8wNi8yMDIzLjA2LjAyLjIzMjg5ODkxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 108.108.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. avr 2016;13(4):228–41. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrclinonc.2015.215&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26667975&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 109.109.Lee HJ, Kim YA, Sim CK, Heo SH, Song IH, Park HS, et al. Expansion of tumor-infiltrating lymphocytes and their potential for application as adoptive cell transfer therapy in human breast cancer. Oncotarget. 26 déc 2017;8(69):113345–59. 110.110.Monnot GC, Romero P. Rationale for immunological approaches to breast cancer therapy. The Breast. 24 févr 2018;37:187–95. 111.111.Andre F, Dieci MV, Dubsky P, Sotiriou C, Curigliano G, Denkert C, et al. Molecular pathways: involvement of immune pathways in the therapeutic response and outcome in breast cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 1 janv 2013;19(1):28–33. 112.112.Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell. 14 déc 2015;28(6):690–714. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ccell.2015.10.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26678337&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 113.113.Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 01 2017;376(22):2147–59. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1612645&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom) 114.114.CRUK IMAXT Grand Challenge Team, Ali HR, Jackson HW, Zanotelli VRT, Danenberg E, Fischer JR, et al. Imaging mass cytometry and multiplatform genomics define the phenogenomic landscape of breast cancer. Nat Cancer [Internet]. 17 févr 2020 [cité 19 févr 2020]; Disponible sur: [http://www.nature.com/articles/s43018-020-0026-6](http://www.nature.com/articles/s43018-020-0026-6) 115.115.Roberti MP, Rocca YS, Amat M, Pampena MB, Loza J, Coló F, et al. IL-2- or IL-15-activated NK cells enhance Cetuximab-mediated activity against triple-negative breast cancer in xenografts and in breast cancer patients. Breast Cancer Res Treat. déc 2012;136(3):659–71. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23065032&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F06%2F06%2F2023.06.02.23289891.atom)