Open Access

Prognostic relevance of tumor‑infiltrating lymphocytes in residual tumor tissue from patients with triple‑negative breast cancer following neoadjuvant chemotherapy: A systematic review and meta‑analysis

  • Authors:
    • Bo Cao
    • Ziran Zhang
    • Chaoxian Wang
    • Xiang Lv
  • View Affiliations

  • Published online on: August 24, 2023     https://doi.org/10.3892/ol.2023.14028
  • Article Number: 441
  • Copyright: © Cao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Further adjuvant chemotherapy treatment can provide benefits to certain patients with triple‑negative breast cancer (TNBC) that fail to achieve pathological complete response (pCR) after the administration of a neoadjuvant chemotherapy (NAC) regimen. However, biomarkers suitable for identifying patients likely to experience poor prognostic outcomes after undergoing additional adjuvant chemotherapy are currently lacking. Accordingly, the present meta‑analysis was conducted to explore the relationship between tumor‑infiltrating lymphocytes (TILs) or TIL subtypes (CD4+ or CD8+) in residual tumor (RT) tissue following NAC and TNBC patient prognosis. Relevant studies published through March 2023 were identified in Pubmed, The Cochrane Library, Embase and Web of Science databases. After excluding irrelevant studies, data were extracted from the remaining reports, while study quality was analyzed with the Newcastle‑Ottawa Scale. Subsequent analyses were performed with Stata 14.0 and Review Manager 5.3. In total, seven relevant studies incorporating 1,202 patients were identified, all of which were retrospective cohort studies. Pooled analyses demonstrated that those patients exhibiting higher levels of RT TIL infiltration following NAC exhibited significantly improved recurrence‑free, metastasis‑free and event‑free survival (RFS/MFS/EFS) compared with patients with lower RT TIL infiltration levels, together with an improved distant recurrence‑free interval (DRFI) [hazard ratio (HR)=0.52; 95% confidence interval (CI)=0.39‑0.69; P<0.00001]. In addition, patients exhibiting high RT TIL infiltration exhibited improved overall survival (OS) and breast cancer‑specific survival (BCSS; HR=0.49; 95% CI=0.38‑0.65; P<0.00001). Additional subgroup analyses revealed that patients with higher TIL infiltration levels or TIL subtype (CD4+ or CD8+) infiltration exhibited improved RFS/MFS/EFS/DRFI as compared with patients with lower levels of overall TIL or TIL subtype (CD4+ or CD8+) infiltration in RT tissue (HR=0.35, 95% CI=0.20‑0.59, P<0.0001; HR=0.49, 95% CI=0.33‑0.71, P=0.0002). Consistently, the OS/BCSS of patients exhibiting high levels of overall TIL or TIL subtype (CD4+ or CD8+) infiltration was increased compared with patients with lower levels of such infiltration (HR=0.33, 95% CI=0.19‑0.59, P=0.0002; HR=0.55, 95% CI=0.41‑0.76, P=0.0002). These data thus demonstrate that levels of overall TIL infiltration or infiltration by CD4+ or CD8+ TILs in RT following NAC can be used as a biomarker to reliably predict prognostic outcomes in patients with TNBC, in addition to highlighting possible targets that may guide the further immunotherapeutic management of these patients.

Introduction

Triple-negative breast cancer (TNBC) cases account for 15–20% of total breast cancer incidence, but patients with TNBC face a worse prognosis in due to the fact that these tumor cells do not express traditional therapeutic targets, including hormone receptors and human epidermal growth factor receptor-2 (1). In addition, TNBC cases tend to be more aggressive and associated with a higher risk or recurrence, contributing to a higher overall mortality risk (2,3). As endocrine and targeted therapy strategies fail to effectively treat TNBC, patients primarily undergo chemotherapeutic and surgical treatment. No differences in survival rates have been observed between patients with TNBC who undergo adjuvant chemotherapy and those that undergo neoadjuvant chemotherapy (NAC), such that the latter has emerged as the preferred systemic treatment regimen for this cancer type (4). NAC enables the direct assessment of tumor responses to chemotherapeutic drug administration, allowing clinicians to gauge patient prognosis and to opt for further adjuvant chemotherapy as appropriate (5). Given that TNBC tumors are generally more sensitive to cytotoxic drugs, higher pathological complete response (pCR) rates tend to be observed following NAC, which is noteworthy given that those patients who achieve pCR after NAC exhibit a better prognosis compared with those who do not (6). When residual tumor (RT) tissue is evident after NAC, patients face higher recurrence rates and shorter overall survival (OS) (7). The CREATE-X trial determined that those patients with TNBC and RT who postoperatively undergo intensified adjuvant capecitabine therapy experience improved outcomes (5). However, the administration of further cytotoxic drugs can also contribute to a higher risk of toxic side effects with the potential to impact patient quality of life, including persistent peripheral neuropathy and an elevated risk of cardiac events (8). Moreover, a subset of these patients with RT fail to relapse, suggesting that some of these individuals exhibit a good prognosis (9). Thus, there is a pressing need to identify novel biomarkers capable of reliably predicting patients with RT outcomes in order to guide the identification of high-risk patients that are most likely to benefit from the administration of further adjuvant chemotherapy. Such biomarkers would provide an effective approach to the individualized treatment of patients with TNBC by minimizing the risk of unnecessary treatment-related toxicity while maximizing the odds of better therapeutic outcomes.

Malignant tumor development is strongly influenced by the composition of the tumor microenvironment (TME), which consists of extracellular matrix components and a range of cell types. Tumor-infiltrating lymphocytes (TILs) within the TME are particularly relevant, given that they comprise the majority of this niche and are also vital to the effective control of tumorigenesis (10,11). TILs include a range of mononuclear immune cell populations including natural killer cells, dendritic cells, CD4+ T cells and CD8+ T cells (12,13). TIL infiltration has been reported in up to 75% of TNBC tumors, of which ~20% exhibit particularly high levels of such infiltration (14). In one recent meta-analysis focused on patients with TNBC undergoing NAC, a higher level of total TIL or TIL subtype (CD4+ or CD8+) infiltration prior to treatment was associated with higher pCR rates and improved post-treatment OS and recurrence-free survival (RFS) (15). Cytotoxic drug treatment can contribute to the mobilization and activation of TILs within the TME, thus reflecting TME responses to chemotherapy. Accordingly, analyses of TILs present in RT tissue can potentially offer additional prognostic information beyond that provided by analyses of pre-treatment TILs (16). The evaluation of RT TILs is also currently recommended by both the International Immuno-Tumor Biomarkers Working Group (14) and the International Immuno-Tumor Biomarkers Working Group.

There have been relatively few studies focused on the clinical relevance of TIL infiltration in RT tissue following NAC in patients with TNBC, and the prognostic value of such infiltration is still subject to controversy. As such, the present meta-analysis focused on those patients with TNBC that failed to achieve pCR after NAC in order to assess the prognostic value of TILs in RT tissue.

Materials and methods

Search strategy

Relevant studies published as of March 2023 were identified by searching the Pubmed (https://pubmed.ncbi.nlm.nih.gov/), Embase (www.embase.com/), The Cochrane Library (https://www.cochranelibrary.com/), and Web of Science (https://www.webofknowledge.com/) databases using the following search terms: ‘Triple negative breast cancer’, ‘tumor-infiltrating lymphocytes’, ‘neoadjuvant chemotherapy’ and ‘residual tumors’. Subject headings combined with free words were used for study retrieval.

Inclusion and exclusion criteria

Studies eligible for inclusion were: i) Published in English; ii) studies enrolling patients with TNBC that did not achieve pCR following NAC; iii) studies assessing the association between RT TILs and TIL subtypes (CD8+ or CD4+) in RT tissue and patient prognosis; iv) original research studies of any design; and v) studies in which prognosis-related hazard ratios (HRs) and 95% confidence intervals (CIs) were provided or could be calculated indirectly. Studies were excluded if they were: i) Case reports, systematic reviews or literature reviews; ii) articles enrolling patients that achieved pCR following NAC; iii) studies lacking outcome indicators; or iv) studies for which HRs and 95% CIs could not be established.

Data extraction and quality analyses

Study screening was independently performed by two investigators, who initially conducted title, abstract and full-text review based on the established inclusion and exclusion criteria. Discrepancies were resolved by discussion and consensus. Relevant data were extracted from eligible studies, including: First author, publication year, country, sample size, TILs subtype, high infiltration threshold, detection method, outcome indicators, HRs and 95% CIs.

Literature quality evaluation

Included study quality was assessed using the Newcastle-Ottawa Quality Assessment Scale (NOS) (17). Possible NOS scores were between 0 and 9, and high-quality studies were those with scores of ≥6.

Statistical analysis

Stata 14.0 (version 12.0; StataCorp LP) and Rev Man 5.3 (RevMan 5.3; Cochrane) were used to conduct this meta-analysis as per the Cochrane International Collaboration Network Systematic Review Guidelines. Pooled effect sizes for survival data were calculated using HRs and 95% CIs, and P<0.05 was selected as the cut-off for statistical significance. The I2 statistic and Cochrane's Q test were used to assess the heterogeneity of included studies, with P<0.10 or I2>50% being considered indicative of significant heterogeneity. Random and fixed effect models were used when significant heterogeneity was and was not detected, respectively (18). Additional analyses of heterogeneity were performed by specifically analyzing results pertaining to TILs and TILs subtypes (CD8+ or CD4+). Egger's test and funnel plots were used to gauge publication bias, with P<0.05 as the cut-off for publication bias detection. P<0.05 was considered to indicate a statistically significant difference.

Results

Study selection and characteristics

The initial study search yielded 208 potentially relevant studies, of which seven enrolling 1,202 patients with TNBC were ultimately included in this analysis (Fig. 1).

All studies eligible for inclusion in the present meta-analysis were retrospective cohort studies. TILs were classified into intratumoral TILs (iTILs) and stromal TILs (sTILs) based on their distributions. CD4+ and CD8+ T cells were the primary TIL subtypes, as distinguished through immunohistochemical staining, and the thresholds used to define high TIL or TIL subtype (CD4+ or CD8+) infiltration varied across studies (Table I).

Table I.

Baseline characteristics of included studies.

Table I.

Baseline characteristics of included studies.

First authorYearCountrySample sizeType of TILsThresholdTILs evaluation methodOutcome(Refs.)
Dieci et al2014France278iTILs and/or sTILs60%H&EMFS/OS(27)
Miyashita et al2015Japan101 CD8+Not specified IHC  RFS/BCSS(36)
Luen et al2019Australia375sTILs20%H&ERFS/OS(19)
Pinard et al2019France109 CD8+0.164 IHC  DRFI(32)
2019France109 CD4+0.066 IHC  DRFI(32)
Wang et al2021China109sTILs30%H&ERFS/OS(29)
Da Silva et al2022Brazilian134 CD8+Not specified IHC  EFS(31)
2022Brazilian134 CD4+Not specified IHC  EFS/OS(31)
Lejeune et al2023Spain96 CD4+Not specified IHC  RFS/OS(28)

[i] TILs, tumor-infiltrating lymphocytes; iTILs, intratumoral tumor-infiltrating lymphocytes; sTILs, stromal tumor-infiltrating lymphocytes; H&E, hematoxylin-eosin staining; IHC, immunohistochemistry; MFS, metastasis-free survival; RFS, recurrence-free survival; DRFI, distant recurrence-free interval; EFS, event-free survival; OS, overall survival; BCSS, breast cancer-specific survival.

Analyses of study quality

All seven of the included studies exhibited NOS scores >7, and were thus classified as high-quality studies (Fig. 2).

Association between RT TIL infiltration and RFS/MFS/EFS/DRFI

In total, the seven included studies enrolled 1,202 patients. A high degree of heterogeneity was detected among these studies (I2=74.2%; P=0.0001), and results were thus analyzed with a random effects model. In sensitivity analyses, the iterative omission of individual studies from pooled analyses did not impact the overall results, indicating that this selected random effects model yielded stable and reliable results. Pooled analyses revealed that those patients with high levels of total TIL or TIL subtype (CD4+ or CD8+) infiltration in RT tissue following NAC exhibited significantly improved recurrence-free survival (RFS)/metastasis-free survival (MFS)/event-free survival (EFS)/distant recurrence-free interval (DRFI) as compared with patients with lower infiltration levels (HR=0.52; 95% CI=0.39–0.69; P<0.00001) (Fig. 3A).

When subgroup analyses for this endpoint were conducted for studies analyzing total TILs and TIL subtypes (CD4+ or CD8+), a total of three studies were included that provided results for overall TIL infiltration. Pooled analyses of these studies were conducted with a random effects model owing to a high degree of heterogeneity (I2=83%; P=0.002), and the exclusion of the study by Luen et al (2019) (19) eliminated this heterogeneity (I2=0%; P=0.39). When results were analyzed with a fixed effects model for this subgroup, patients with high levels of TIL infiltration exhibited improved RFS/MFS/EFS/DFRI compared with patients with low TIL infiltration (HR=0.35; 95% CI=0.20–0.59; P<0.0001) (Fig. 3B). In addition, four studies reported results for the TIL subtype (CD4+ or CD8+) subgroup, and a random effects model was used owing to moderate heterogeneity (I2=58%; P=0.04). Pooled analyses for this subgroup similarly confirmed that patients with high levels of TIL infiltration exhibited improved RFS/MFS/EFS/DRFI compared with patients with low levels of TIL infiltration (HR=0.49; 95% CI=0.33–0.71; P=0.0002) (Fig. 3B). Sensitivity analyses revealed no changes in the combined effect size when alternating between fixed and random effects models, confirming that these results are robust.

Association between RT TIL infiltration and OS/BCSS

In total, six of the included studies incorporating 1,093 patients reported on OS/BCSS outcomes. As a high degree of heterogeneity was detected (I2=77%; P=0.0007), a random effects model was selected. Sensitivity analyses indicated that the study conducted by Luen et al (19) had the greatest effect on this heterogeneity, which was reduced following the exclusion of this study (I2=36%; P=0.18). A fixed effects model was thus selected for pooled analyses based on this reduction in heterogeneity. This approach revealed that patients with high levels of total TIL or TIL subtype (CD4+ or CD8+) infiltration exhibited significantly improved OS/BCSS relative to patients with lower levels of such infiltration (HR=0.49; 95% CI=0.38–0.65; P<0.00001) (Fig. 4A).

Subgroup analyses of studies assessing TILs and TIL subtypes (CD4+ or CD8+) were additionally conducted for this endpoint. Just two studies were included in the TILs subgroup, and pooled analyses of these studies were performed with a fixed effects model owing to an absence of heterogeneity (I2=16%; P=0.27). The present analysis demonstrated that patients with higher levels of RT TIL infiltration following NAC exhibited better OS/BCSS as compared with patients with low levels of infiltration (HR=0.33; 95% CI=0.19–0.59; P=0.0002) (Fig. 4B). In addition, three studies reported results for TIL subtypes (CD4+ or CD8+), and these pooled analyses were also performed with a fixed effects model owing to the absence of significant heterogeneity (I2=26%; P=0.26). As aforementioned, those patients with higher levels of TIL (CD4+ or CD8+) infiltration exhibited significantly better OS/BCSS as compared with patients with lower levels of such infiltration (HR=0.55; 95% CI=0.41–0.76; P=0.0002) (Fig. 4B). Sensitivity analyses revealed no changes in the combined effect size when alternating between fixed and random effects models, confirming that these results are robust.

Publication bias

Generated funnel plots were asymmetrical, and Egger's test yielded evidence of significant publication bias (P<0.0001 and P=0.002) (Fig. 5). This suggests that these results are subject to some degree of publication bias. This may be attributable to the choice to only include English language studies, contributing to language bias. In addition, negative research results often go unpublished, contributing to further bias in the literature. These findings suggest that the present meta-analysis results are not stable, underscoring the need for further research focused on this experimental topic.

Discussion

NAC has emerged as the standard approach to TNBC management, with most patients receiving a combination of anthracyclines, alkylates and taxanes while undergoing NAC treatment (20). The administration of these cytotoxic drugs can modulate lymphocyte infiltration of the TME in a manner that induces a robust antitumor immune response (21,22). Adaptive immunity can also be triggered in response to tumor-specific antigens, promoting the infiltration of lymphocytes engaged in immune surveillance whereupon they can destroy target tumor cells (23). TIL infiltration can impact the effects of cytotoxic drugs on tumor cells, while also offering value as a biomarker associated with treatment outcomes and the overall prognosis of patients with cancer, providing guidance for treatment planning efforts. A number of studies to date have confirmed that TIL infiltration is significantly correlated with TNBC patient prognostic outcomes and treatment responses (2426). While numerous studies have explored the association between TIL infiltration and pCR following NAC, there is also some evidence that a subset of patients who fail to achieve pCR can also achieve better outcomes (19,27).

Several recent studies have documented an association between TILs present in RT tissue following the prognosis of patients with NAC and TNBC (28,29). While this suggests the existence of a relationship between TILs and patient survival, systematic high-quality studies focused on this topic are lacking at present. As such, a meta-analysis was conducted in the present study as a means of systematically reviewing published studies to explore the prognostic implications of RT TIL infiltration levels. Owing to limitations with respect to the outcome indicators used in different studies, RFS/MFS/EFS/DRFI were utilized as short-term endpoints. Pooled analyses indicated that patients exhibiting higher levels of TIL infiltration experienced better postoperative RFS/MFS/EFS/DRFI compared with patients with lower levels of such infiltration (HR=0.52; 95% CI=0.39–0.69). OS/BCSS were additionally analyzed as long-term outcomes, revealing that those patients with TNBC with higher RT TIL infiltration levels following NAC exhibited a significantly lower risk of death compared with patients with lower levels of TIL infiltration (HR=0.49; 95% CI=0.38–0.65).

Different TIL subtypes play specific roles in the context of antitumor immunity, engaging in cross-regulatory interactions that can shape immune response induction (30). The presence of specific TIL subtypes in RT tissue can contribute to a better or worse patient prognosis, and high levels of infiltration for certain TILs may thus provide a robust means of detecting patients in particular prognostic subgroups (31,32). CD4+ and CD8+ T cells are the primary TIL subtypes (31). CD4+ T cells primarily act as helper T cells that are reliant on antigen-presenting cells to support the activation of other immune cell types following the recognition of soluble tumor-derived antigens, ultimately exerting antitumor roles (33). Through this activity, CD4+ T cells have the potential to increase the numbers of other TILs in the TME or to enhance their functionality (33). Cytolytic CD8+ T cells can directly destroy target tumor cells, making them particularly important mediators of antitumor immunity (34). Greater levels of CD4+ and CD8+ T cell infiltration in the TME have been linked to higher pCR rates and better prognostic outcomes in patients with TNBC (33,35,36). Accordingly, subgroup analyses were conducted in the present study based on the TIL subtypes included in the analyzed studies. With respect to short-term outcomes, high levels of TIL or TIL subtype (CD4+ or CD8+) infiltration were associated with a better prognosis (HR=0.35, 95% CI=0.20–0.59; HR=0.49, 95% CI=0.33–0.71), suggesting a higher risk of metastasis and disease recurrence in individuals with low levels of TIL infiltration. Similar findings were also observed with respect to long-term outcome indicators (HR=0.33, 95% CI=0.19–0.59; HR=0.55, 95% CI, 0.41–0.76).

These meta-analysis results highlight a clear relationship between RT TIL infiltration following prognosis of patients with NAC and TNBC. Residual cancer burden (RCB) has also been used to predict the outcomes of patients with TNBC based on RT levels following NAC (37), suggesting that combining analyses of RCB and RT TIL infiltration profiles may improve prognostic assessment efforts. Indeed, Luen et al (19) observed a significant interactive effect of TIL infiltration and RCB on prognosis, while Asano et al (38) determined that the combined assessment of TILs and RCB can predict post-NAC breast cancer recurrence more sensitively compared with TILs alone. Given that TNBC tumors exhibit unique immunological characteristics that can better sensitize these tumors to immunotherapeutic treatment, there is growing interest in the post-NAC immunotherapy-based management of this cancer type (39). As a result, TIL profiles represent increasingly important targets that can be used to stratify patients in post-NAC immunotherapy clinical trials. For example, results from the KEYNOTE-086 study highlighted the fact that sTILs can predict pembrolizumab efficacy when used as a first-line treatment for metastatic TNBC (40). The KEYNOTE-173 study revealed that there was a significant association between high levels of TILs and improved pathological complete response or objective response rate in patients with TNBC who underwent treatment with pembrolizumab (41). These findings were similar to the results of the KEYNOTE-086 study. TIL immune checkpoint receptor expression has been linked to better immunotherapy responses (42,43). Low levels of TIL infiltration may be indicative of the need for chemotherapeutic treatment using cytotoxic drugs and of the potential for the addition of PD-1/PD-L1 therapy. Higher TIL infiltration and lower disease burden, by contrast, may suggest that single-agent immunotherapy represents a viable treatment strategy (44).

The present study is subject to certain limitations. For one, the included studies employed different methods when detecting TILs/TIL subtypes and when selecting the threshold values used to define high levels of TIL infiltration. This, coupled with the inconsistent outcome indicators across studies, is likely to have impacted the overall accuracy of these results. In addition, the pre-NAC TIL infiltration profiles in the patients included in these studies were not evaluated, and the prognostic relevance of dynamic changes in TIL infiltration was thus not assessed, highlighting a promising avenue for future research. Lastly, owing to the limited number of relevant studies published to date, the overall sample size in this meta-analysis was somewhat small, potentially contributing to some degree of bias that may have impacted study results. Additional large-scale studies will be essential to explore the correlation between post-NAC TILs in RT tissue and TNBC patient prognostic outcomes in order to provide a comprehensive and evidence-based approach that can better guide clinical decision-making.

The present meta-analysis results suggest that TIL infiltration in RT tissue following NAC is a valuable prognostic indicator associated with TNBC patient outcomes. Specifically, higher levels of TIL infiltration were found to be associated with a lower risk of disease recurrence, metastasis and death such that patients exhibiting higher levels of RT TIL infiltration tended to exhibit a better prognosis compared with patients with lower levels of RT TIL infiltration. Analyzing levels of total TIL or TIL subtype (CD4+ or CD8+) infiltration in RT tissue following NAC can thus improve the ability of clinicians to predict the efficacy of further adjuvant chemotherapy administration and the prognosis of patients with TNBC.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BC and ZZ contributed to the conception and design of the study. CW and XL prepared the materials, collected the data and performed the analysis. ZZ drafted the manuscript. BC and ZZ confirm the authenticity of all the raw data. All authors revised the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Authors' information

ORCID: Dr Ziran Zhang, orcid.org/0000-0002-7835-8788.

Competing interests

The authors declare that they have no competing interests.

References

1 

Garrido-Castro AC, Lin NU and Polyak K: Insights into molecular classifications of triple-negative breast cancer: Improving patient selection for treatment. Cancer Discov. 9:176–198. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Diana A, Carlino F, Franzese E, Oikonomidou O, Criscitiello C, De Vita F, Ciardiello F and Orditura M: Early triple negative breast cancer: Conventional treatment and emerging therapeutic landscapes. Cancers (Basel). 12:8192020. View Article : Google Scholar : PubMed/NCBI

3 

Won KA and Spruck C: Triple-negative breast cancer therapy: Current and future perspectives (Review). Int J Oncol. 57:1245–1261. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Gupta GK, Collier AL, Lee D, Hoefer RA, Zheleva V, Siewertsz van Reesema LL, Tang-Tan AM, Guye ML, Chang DZ, Winston JS, et al: Perspectives on triple-negative breast cancer: Current treatment strategies, unmet needs, and potential targets for future therapies. Cancers (Basel). 12:23922020. View Article : Google Scholar : PubMed/NCBI

5 

Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, Kuroi K, Im SA, Park BW, Kim SB, et al: Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med. 376:2147–2159. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, et al: Pathological complete response and long-term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet. 384:164–172. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Liedtke C, Mazouni C, Hess KR, André F, Tordai A, Mejia JA, Symmans WF, Gonzalez-Angulo AM, Hennessy B, Green M, et al: Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 26:1275–1281. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Isakoff SJ: Triple-negative breast cancer: Role of specific chemotherapy agents. Cancer J. 16:53–61. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Symmans WF, Wei C, Gould R, Yu X, Zhang Y, Liu M, Walls A, Bousamra A, Ramineni M, Sinn B, et al: Long-Term prognostic risk after neoadjuvant chemotherapy associated with residual cancer burden and breast cancer subtype. J Clin Oncol. 35:1049–1060. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, Mariette C, Chaput N, Mira JP, Delaloge S, et al: The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev. 220:47–59. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Zitvogel L and Kroemer G: The immune response against dying tumor cells: Avoid disaster, achieve cure. Cell Death Differ. 15:1–2. 2008. View Article : Google Scholar : PubMed/NCBI

12 

García-Teijido P, Cabal ML, Fernández IP and Pérez YF: Tumor-Infiltrating lymphocytes in triple negative breast cancer: The future of immune targeting. Clin Med Insights Oncol. 10 (Suppl 1):S31–S39. 2016.PubMed/NCBI

13 

Ravelli A, Roviello G, Cretella D, Cavazzoni A, Biondi A, Cappelletti MR, Zanotti L, Ferrero G, Ungari M, Zanconati F, et al: Tumor-infiltrating lymphocytes and breast cancer: Beyond the prognostic and predictive utility. Tumour Biol. 39:10104283176950232017. View Article : Google Scholar : PubMed/NCBI

14 

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, 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. 52((Pt 2)): 16–25. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Gao G, Wang Z, Qu X and Zhang Z: Prognostic value of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer: A systematic review and meta-analysis. BMC Cancer. 20:1792020. View Article : Google Scholar : PubMed/NCBI

16 

Demaria S, Volm MD, Shapiro RL, Yee HT, Oratz R, Formenti SC, Muggia F and Symmans WF: Development of tumor-infiltrating lymphocytes in breast cancer after neoadjuvant paclitaxel chemotherapy. Clin Cancer Res. 7:3025–3030. 2001.PubMed/NCBI

17 

Wells GA, Shea B, O'Connell D, Peterson J, Welch V, Losos M and Tugwell P: The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Non-Randomised Studies in Meta-Analyses. Symposium on Systematic Reviews: Beyond the Basics. 2014.

18 

DerSimonian R and Laird N: Meta-analysis in clinical trials revisited. Contemp Clin Trials. 45((Pt A)): 139–145. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Luen SJ, Salgado R, Dieci MV, Vingiani A, Curigliano G, Gould RE, Castaneda C, D'Alfonso T, Sanchez J, Cheng E, et al: Prognostic implications of residual disease tumor-infiltrating lymphocytes and residual cancer burden in triple-negative breast cancer patients after neoadjuvant chemotherapy. Ann Oncol. 30:236–242. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Park JH, Ahn JH and Kim SB: How shall we treat early triple-negative breast cancer (TNBC): From the current standard to upcoming immuno-molecular strategies. ESMO Open. 3 (Suppl 1):e0003572018. View Article : Google Scholar : PubMed/NCBI

21 

Andre F, Dieci MV, Dubsky P, Sotiriou C, Curigliano G, Denkert C and Loi S: Molecular pathways: Involvement of immune pathways in the therapeutic response and outcome in breast cancer. Clin Cancer Res. 19:28–33. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Ladoire S, Mignot G, Dabakuyo S, Arnould L, Apetoh L, Rébé C, Coudert B, Martin F, Bizollon MH, Vanoli A, et al: In situ immune response after neoadjuvant chemotherapy for breast cancer predicts survival. J Pathol. 224:389–400. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Lee H, Lee M, Seo JH, Gong G and Lee HJ: Changes in tumor-infiltrating lymphocytes after neoadjuvant chemotherapy and clinical significance in triple negative breast cancer. Anticancer Res. 40:1883–1890. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, Budczies J, Huober J, Klauschen F, Furlanetto J, 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. 19:40–50. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Pruneri G, Gray KP, Vingiani A, Viale G, Curigliano G, Criscitiello C, Láng I, Ruhstaller T, Gianni L, Goldhirsch A, et al: Tumor-infiltrating lymphocytes (TILs) are a powerful prognostic marker in patients with triple-negative breast cancer enrolled in the IBCSG phase III randomized clinical trial 22–00. Breast Cancer Res Treat. 158:323–331. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Pruneri G, Vingiani A, Bagnardi V, Rotmensz N, De Rose A, Palazzo A, Colleoni AM, Goldhirsch A and Viale G: Clinical validity of tumor-infiltrating lymphocytes analysis in patients with triple-negative breast cancer. Ann Oncol. 27:249–256. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Dieci MV, Criscitiello C, Goubar A, Viale G, Conte P, Guarneri V, Ficarra G, Mathieu MC, Delaloge S, Curigliano G and Andre F: Prognostic value of tumor-infiltrating lymphocytes on residual disease after primary chemotherapy for triple-negative breast cancer: A retrospective multicenter study. Ann Oncol. 25:611–618. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Lejeune M, Reverté L, Sauras E, Gallardo N, Bosch R, Roso A, Petit A, Peg V, Riu F, García-Fontgivell J, et al: Prognostic implications of the residual tumor microenvironment after neoadjuvant chemotherapy in triple-negative breast cancer patients without pathological complete response. Cancers (Basel). 15:5972023. View Article : Google Scholar : PubMed/NCBI

29 

Wang Y, Zong B, Yu Y, Wang Y, Tang Z, Chen R, Huang M and Liu S: Ki67 index changes and tumor-infiltrating lymphocyte levels impact the prognosis of triple-negative breast cancer patients with residual disease after neoadjuvant chemotherapy. Front Oncol. 11:6686102021. View Article : Google Scholar : PubMed/NCBI

30 

Ghajar CM: On leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol. 4:a0132762012. View Article : Google Scholar : PubMed/NCBI

31 

da Silva JL, de Albuquerque LZ, Rodrigues FR, de Mesquita GG, Fernandes PV, Thuler LCS and de Melo AC: Prognostic influence of residual tumor-infiltrating lymphocyte subtype after neoadjuvant chemotherapy in triple-negative breast cancer. Front Oncol. 11:6367162021. View Article : Google Scholar : PubMed/NCBI

32 

Pinard C, Debled M, Ben Rejeb H, Velasco V, Tunon de Lara C, Hoppe S, Richard E, Brouste V, Bonnefoi H and MacGrogan G: Residual cancer burden index and tumor-infiltrating lymphocyte subtypes in triple-negative breast cancer after neoadjuvant chemotherapy. Breast Cancer Res Treat. 179:11–23. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, Ravoet M, Le Buanec H, Sibille C, Manfouo-Foutsop G, et al: CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 123:2873–2892. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Lin KR, Pang DM, Jin YB, Hu Q, Pan YM, Cui JH, Chen XP, Lin YX, Mao XF, Duan HB and Luo W: Circulating CD8+ T-cell repertoires reveal the biological characteristics of tumors and clinical responses to chemotherapy in breast cancer patients. Cancer Immunol Immunother. 67:1743–1752. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Matsumoto H, Thike AA, Li H, Yeong J, Koo SL, Dent RA, Tan PH and Iqbal J: Increased CD4 and CD8-positive T cell infiltrate signifies good prognosis in a subset of triple-negative breast cancer. Breast Cancer Res Treat. 156:237–247. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Miyashita M, Sasano H, Tamaki K, Hirakawa H, Takahashi Y, Nakagawa S, Watanabe G, Tada H, Suzuki A, Ohuchi N and Ishida T: 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. 17:1242015. View Article : Google Scholar : PubMed/NCBI

37 

Sheri A, Smith IE, Johnston SR, A'Hern R, Nerurkar A, Jones RL, Hills M, Detre S, Pinder SE, Symmans WF and Dowsett M: Residual proliferative cancer burden to predict long-term outcome following neoadjuvant chemotherapy. Ann Oncol. 26:75–80. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Asano Y, Kashiwagi S, Goto W, Takada K, Takahashi K, Hatano T, Noda S, Takashima T, Onoda N, Tomita S, et al: Prediction of survival after neoadjuvant chemotherapy for breast cancer by evaluation of tumor-infiltrating lymphocytes and residual cancer burden. BMC Cancer. 17:8882017. View Article : Google Scholar : PubMed/NCBI

39 

Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, Gallardo C, Lipatov O, Barrios CH, Holgado E, et al: Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 396:1817–1828. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Loi S, Adams S, Schmid P, Cortés J, Cescon DW, Winer EP, Toppmeyer DL, Rugo HS, De Laurentiis M, Nanda R, et al: Relationship between tumor infiltrating lymphocyte (TIL) levels and response to pembrolizumab (pembro) in metastatic triple-negative breast cancer (mTNBC): Results from KEYNOTE-086. Ann Oncol. 28:V6082017. View Article : Google Scholar

41 

Schmid P, Salgado R, Park YH, Muñoz-Couselo E, Kim SB, Sohn J, Im SA, Foukakis T, Kuemmel S, Dent R, et al: Pembrolizumab plus chemotherapy as neoadjuvant treatment of high-risk, early-stage triple-negative breast cancer: Results from the phase 1b open-label, multicohort KEYNOTE-173 study. Ann Oncol. 31:569–581. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Ellis PM, Vella ET and Ung YC: Immune checkpoint inhibitors for patients with advanced non-small-cell lung cancer: A systematic review. Clin Lung Cancer. 18:444–459.e1. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Kluger HM, Zito CR, Turcu G, Baine MK, Zhang H, Adeniran A, Sznol M, Rimm DL, Kluger Y, Chen L, et al: PD-L1 studies across tumor types, its differential expression and predictive value in patients treated with immune checkpoint inhibitors. Clin Cancer Res. 23:4270–4279. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Blackley EF and Loi S: Targeting immune pathways in breast cancer: Review of the prognostic utility of TILs in early stage triple negative breast cancer (TNBC). Breast. 48 (Suppl 1):S44–S48. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 26 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cao B, Zhang Z, Wang C and Lv X: Prognostic relevance of tumor‑infiltrating lymphocytes in residual tumor tissue from patients with triple‑negative breast cancer following neoadjuvant chemotherapy: A systematic review and meta‑analysis. Oncol Lett 26: 441, 2023
APA
Cao, B., Zhang, Z., Wang, C., & Lv, X. (2023). Prognostic relevance of tumor‑infiltrating lymphocytes in residual tumor tissue from patients with triple‑negative breast cancer following neoadjuvant chemotherapy: A systematic review and meta‑analysis. Oncology Letters, 26, 441. https://doi.org/10.3892/ol.2023.14028
MLA
Cao, B., Zhang, Z., Wang, C., Lv, X."Prognostic relevance of tumor‑infiltrating lymphocytes in residual tumor tissue from patients with triple‑negative breast cancer following neoadjuvant chemotherapy: A systematic review and meta‑analysis". Oncology Letters 26.4 (2023): 441.
Chicago
Cao, B., Zhang, Z., Wang, C., Lv, X."Prognostic relevance of tumor‑infiltrating lymphocytes in residual tumor tissue from patients with triple‑negative breast cancer following neoadjuvant chemotherapy: A systematic review and meta‑analysis". Oncology Letters 26, no. 4 (2023): 441. https://doi.org/10.3892/ol.2023.14028