Next Article in Journal
Engineering and Expression Strategies for Optimization of L-Asparaginase Development and Production
Next Article in Special Issue
Sequential Therapy with Ropeginterferon Alfa-2b and Anti-Programmed Cell Death 1 Antibody for Inhibiting the Recurrence of Hepatitis B-Related Hepatocellular Carcinoma: From Animal Modeling to Phase I Clinical Results
Previous Article in Journal
Cytokine Levels in Saliva Are Associated with Salivary Gland Fibrosis and Hyposalivation in Mice after Fractionated Radiotherapy of the Head and Neck
Previous Article in Special Issue
Biomarkers for Immune Checkpoint Inhibitor Response in NSCLC: Current Developments and Applicability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Impact of CD38- and IgκC-Positive Tumor-Infiltrating Plasma Cells in Triple-Negative Breast Cancer

Department of Obstetrics and Gynecology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(20), 15219; https://doi.org/10.3390/ijms242015219
Submission received: 20 September 2023 / Revised: 12 October 2023 / Accepted: 13 October 2023 / Published: 16 October 2023
(This article belongs to the Special Issue Molecular Advances in Cancer Immunotherapy)

Abstract

:
Due to a higher mutational load, triple-negative breast cancer (TNBC) is characterized by a higher immunogenicity compared to other subtypes. In this context, we analyzed the prognostic significance of tumor-infiltrating plasma cells in a cohort of 107 triple-negative breast cancer patients. Tumor-infiltrating plasma cells were analyzed via immunohistochemistry using the plasma cell markers CD38 and IgκC. The prognostic impact of the CD38 and IgκC expression was evaluated using the Kaplan–Meier plots and Cox regression analyses. A Spearman-Rho correlation coefficient was used to evaluate a possible association between plasma cell infiltration and the BRCA mutation status. The study cohort consisted of 107 patients with early-stage TNBC, who were treated between 2009 and 2016 at the Department of Gynecology and Obstetrics, University Medical Center Mainz, Germany. The median follow-up was five years. The Kaplan–Meier survival analysis showed that higher tumor infiltration with CD38-positive plasma cells was associated with significantly longer metastasis-free survival (MFS) (p = 0.039 Log Rank). In the multivariate Cox regression analysis for metastasis-free survival, in which additional clinicopathological factors (age, tumor size, nodal status, and grading) were considered, CD38 was identified as an independent prognostic factor within the analyzed cohort (HR 0.438, 95% CI 0.195–0.983; p = 0.045). In addition to the CD38 expression, the nodal status was also identified as an independent prognostic factor in multivariate Cox regression. Regarding the IgκC expression, a higher IgκC expression was shown to be associated with a better outcome, although this effect was not statistically significant. Furthermore, we were able to show a significant correlation between plasma cell infiltration and the BRCA mutation status. A favorable prognostic significance of tumor-infiltrating plasma cells could be demonstrated in triple-negative breast cancer immunohistochemically analyzed for the CD38 and IgκC expression. CD38 was identified as an independent prognostic factor via multivariate Cox regression.

1. Introduction

Triple-negative breast cancer, which is defined by a lack of expression of the estrogen, progesterone, and human epidermal growth factor receptor 2 (HER2) receptors, represents approximately 10–15% of all breast cancers and is characterized by more aggressive tumor growth, poorer differentiation, and a higher proliferation index (Ki-67), as well as the associated unfavorable prognosis [1]. The immune system plays an increasingly important role in the development of targeted therapeutic strategies in breast cancer. Using mRNA sequencing data from The Cancer Genome Atlas (TCGA), it has been shown that a high expression of T-cell and B-cell signatures has a favorable effect on the overall survival in many tumor entities, including breast cancer [2]. Due to a higher mutational load, and consequently, a higher number of immunogenic tumor-associated antigens, triple-negative breast cancer has a stronger immunogenic potential compared to the other subtypes [3]. Tumor-associated antigens (TAA) can be recognized by the immune system and elicit an immune response that, in the best case, leads to tumor suppression [4]. In this context, tumor-infiltrating lymphocytes (TILs) play an important role: numerous studies have already shown that TILs have a strong prognostic and predictive influence in triple-negative breast cancer [5,6,7,8,9,10,11], with higher numbers of tumor-infiltrating lymphocytes being associated with a higher rate of pathologic complete remission (pCR) and improved survival. In a recent retrospective analysis by de Jong et al. in which a total of 441 younger patients with early node-negative TNBC who did not receive (neo)adjuvant systemic therapy were included, the presence of stromal TILs (sTILs) was identified as an independent prognostic parameter; an increased number of stromal TILs was associated with longer MFS and OS [12]. Based on the excellent prognosis of those younger patients (age at diagnosis <40) with early node-negative TNBC, small tumor size, and a high level of sTILs, the authors postulated that de-escalating strategies should be evaluated in prospective clinical trials, demonstrating a possible clinical impact of sTILs in early triple-negative breast cancer.
Stanton and colleagues showed in a systematic review that the extent of tumor-infiltrating lymphocytes varies both within and between subtypes, with triple-negative breast cancer often having high levels of TILs [13]. TILs are composed of various immune cells (T-lymphocytes, B-lymphocytes, and macrophages) that interact with cytokines, immunoglobulins, interferons, and other immune-associated mediators to form the so-called tumor microenvironment. Following the presentation of antigens by antigen-presenting cells (APCs), various immune responses can be elicited. Depending on the predominant immune cell type, either a tumor-inhibiting effect (acute inflammation by CD8 lymphocytes (Th1) or M1 macrophages) or a Th2-driven tumor-promoting effect (chronic inflammation by M2 macrophages, regulatory T cells, or immune checkpoints such as programmed cell death protein 1 (PD-1) or its ligand (PD-L1) can result. The complex interaction between the immune system and the tumor is illustrated via the so-called “Cancer Immunity Cycle” [4]. Most studies analyzing the prognostic and/or predictive role of TILs in breast cancer have focused on the cellular immune system, particularly cytotoxic T-cells. For example, several studies have demonstrated that increased numbers of tumor-infiltrating lymphocytes or T-cell transcripts are associated with improved prognosis in rapidly proliferating breast cancers, such as triple-negative breast cancer [14,15,16]. Recently, immune checkpoint inhibitors in combination with chemotherapy have been successfully used in the treatment of early and advanced triple-negative breast cancer, which release the “brakes” on the immune system and enhance naturally occurring immune responses against the tumor [17,18,19]. In contrast, the role of B lymphocytes, especially tumor-infiltrating plasma cells, is controversial [20]. Using the gene expression analysis, we and others have shown that the expression of B-cell-associated genes (collectively referred to as B-cell metagene) is associated with a favorable prognosis in patients with early node-negative breast cancer [21,22,23]. Further studies have confirmed the prognostic significance of IgκC as a representative marker of the B-cell metagene at both the mRNA and protein levels [24,25]. Furthermore, IgκC has been identified as a predictive marker for response to anthracycline-based chemotherapy [24]. Using fluorescence microscopy, IgκC was detected in IRF4-positive tumor-infiltrating plasma cells [24]. The primary objective of the current study was to evaluate the prognostic impact of tumor-infiltrating plasma cells in a cohort of 107 patients with early triple-negative breast cancer. The expression of the plasma cell markers CD38 and IgκC, was determined via immunohistochemistry. Additionally, we analyzed whether there was a correlation between plasma cell infiltration (the expression of CD38 and IgκC) and the histological degree of differentiation and the proliferation Index Ki67, respectively. Other secondary objectives were to determine whether the expression levels of CD38 and IgκC correlated with each other and to evaluate the extent to which there was a correlation between the BRCA mutation (at the germline level) and the level of plasma cell infiltration.

2. Results

To evaluate the impact of plasma cell infiltration in the below-mentioned cohort of triple-negative breast cancer patients, the plasma cell markers CD38 and IgκC were analyzed via immunohistochemistry using a semi-quantitative scoring method as previously described in [25,26]. To dichotomize the patients, tumor slides with IgκC/CD38 score 0 and 1+ were considered as low plasma cell infiltration, whereas cases with 2+ and 3+ were high plasma cell infiltration, respectively.
The immunohistochemical examination of tumor slides showed 55.1% of tumor slides as weakly positive or negative for the IgκC expression (Score 0, 1+), with 44.9% being strongly IgκC positive (Score 2+, 3+). Concerning the CD38 expression, 56.1% of the tumor slides were weakly positive or negative, whereas 43.9% showed a strong CD38 expression (Score 2+, 3+).

2.1. Prognostic Impact of IgκC Expression

The Kaplan–Meier analysis demonstrated that a higher IgκC expression had a weak trend to be associated with a better outcome (longer MFS), although this effect did not reach the statistical significance level (p = 0.35 Log Rank) (Figure 1).
The univariate and multivariate Cox Regression analysis, adjusted for the clinical–pathological parameters such as age, tumor size, lymph node status, and histological grade of differentiation, failed to show a statistically significant impact of the IgκC expression on metastasis-free survival in the observed cohort (Table 1 and Table 2). However, the lymph node status was shown to be a significant prognostic factor in both the univariate and multivariate Cox regression analysis.

2.2. Prognostic Impact of CD38 Expression

Using the Kaplan–Meier survival analysis, it was demonstrated that a higher number of CD38-positive tumor-infiltrating plasma cells was associated with a significantly longer metastasis-free survival within the observed collective (p = 0.039 Log Rank) (Figure 2). In the multivariate Cox regression analysis adjusted for other clinicopathological factors (age, tumor size, nodal status, and grading), CD38 was identified as an independent prognostic factor within the analyzed collective (HR 0.438, 95% CI 0.195–0.983; p = 0.045). In addition to the CD38 expression, the nodal status was also shown to be an independent prognostic factor in multivariate Cox regression (Table 3).

2.3. Validation of the Prognostic Impact of IgκC and CD38 Expression Using Gene Expression Data from an Independent, Publicly Available TNBC Cohort

To validate our immunohistochemical results on the prognostic impact of the plasma cell markers IgκC and CD38 in a larger, independent cohort of TNBC, we used the publicly available gene expression data of IgκC and CD38 with their associated tumor characteristics and follow-up data [27] in a cohort of 424 TNBC samples. Using two different probe sets for the IgκC mRNA expression, the Kaplan–Meier plots show that a higher IgκC mRNA expression was associated with a significantly longer MFS (probe set 214669_x_at: p = 0.049 Log Rank; probe set 216576_x_at: p = 0.018 Log Rank, Figure 3). These validation results on the mRNA gene expression data of IgκC are basically in line with our immunohistochemical results showing a trend that a higher IgκC expression at the protein level correlates with a longer MFS. With regard to CD38, the mRNA expression data do not show a significant effect on MFS (Figure 4).

2.4. Correlation between the Strength of Plasma Cell Infiltration (CD38 and IgκC Expression) and the Tumor Grade as Well as the Proliferation Index Ki67

In addition, we investigated whether there was a correlation between the IgκC and CD38 expression on the one hand and the histological degree of differentiation and the proliferation index Ki67 on the other hand. Using ordinal regression, a significant correlation between the CD38 expression and the proliferation index Ki67 was found: a higher CD38 expression was associated with a higher proliferation index (p = 0.024).
In contrast, there was no significant correlation between IgκC and the proliferation index Ki67 or the histological grade of differentiation, nor between the CD38 expression and the histological grade of differentiation.

2.5. Correlation of the Level of Tumor-Infiltrating Plasma Cells and BRCA Mutation Status

Using the Spearman-Rho correlation, we could show a strong, statistically significant correlation between the level of the CD38 expression and the level of the IgκC expression (r = 0.588, p < 0.001). In a subsequent step, we were able to demonstrate a significant correlation between the presence of a BRCA mutation (at the germline level) and the extent of plasma cell infiltration (determined as the mean of the IgκC and CD38 expression) using the Spearman-Rho correlation coefficient: the presence of a BRCA mutation was associated with a stronger plasma cell infiltration (r = 0.211; p = 0.029) (Figure 5).

3. Discussion

The present retrospective study demonstrated a favorable prognostic impact of tumor-infiltrating plasma cells within the described collective of 107 triple-negative breast cancer patients, which were analyzed immunohistochemically with regard to the expression of the plasma cell markers CD38 and IgκC. Furthermore, CD38 was identified as an independent prognostic factor via multivariate Cox regression. Additionally, we could show a significant correlation between the CD38 expression and the proliferation index Ki67: a higher number of CD38-positive, tumor-infiltrating plasma cells was associated with a higher percentage of Ki67-positive tumor cells.
These results confirmed the prognostic significance of IgκC as a representative marker of the B-cell metagene and underlined that the humoral immune response plays an important role in antitumor immunity. These findings are in line with previous studies of our group and others. Refs. [21,22,23,24,25,28]: using the gene expression analysis, IgκC has already been shown to be a prognostic marker in a large cohort consisting of 965 node-negative breast cancer patients who did not receive any systemic therapy in the adjuvant setting [24], where a higher IgκC mRNA expression was associated with a better prognosis [24]. This effect was particularly pronounced in patients with estrogen receptor (ER)-negative, highly proliferating breast cancer. Additionally, IgκC was identified as a predictive marker for the response to neoadjuvant anthracycline-based chemotherapy in a cohort of 845 breast cancer patients, where the higher IgκC expression was correlated with a higher pCR rate. Further immunohistochemical studies confirmed that IgκC is mainly produced by activated tumor-infiltrating plasma cells. In a retrospective immunohistochemical study of Chen et al., the prognostic impact of the IgκC expression was evaluated in a cohort of 335 node-negative breast cancer patients who did not receive any adjuvant therapy after operation and irradiation [25]. Of these, 160 patients had infiltration with IgκC-positive, tumor-infiltrating plasma cells, which showed a statistically significant longer survival time. In estrogen receptor-negative tumors and in luminal B breast carcinomas, the significance was particularly emphasized. In the present work, adjuvant chemotherapy could be a possible bias for the evaluation of the prognosis. In contrast, regarding the above-mentioned study from Schmidt et al. [24] and the immunohistochemical study of Chen et al. [25], only patients who did not receive any adjuvant systemic treatment entered their studies, allowing for the validation of IgκC as a possible prognostic factor. Furthermore, the above-cited studies included all breast cancer subtypes. In contrast, the present study focused on triple-negative breast cancer samples. In our retrospective immunohistochemical study, there was a trend for longer metastasis-free survival in tumors with a higher IgκC expression. In contrast to the above-mentioned studies, statistical significance was not reached. This could be due to the small number of cases or the restriction to triple-negative breast cancer or the chemotherapy administered. Contrasting these findings, another retrospective study analyzed the prognostic significance of IgκC, whose expression was evaluated at the protein level using immunohistochemistry [26]. Thereby, a significant prognostic influence of the IgκC expression could be confirmed even in patients treated with adjuvant chemotherapy. A higher IgκC expression was associated with longer metastasis-free survival. These retrospective results regarding the prognostic significance of the IgκC expression in adjuvant chemotherapy-treated patients with early breast cancer were confirmed using the retrospective–prospective data. The importance of the IgκC gene expression as a positive prognostic marker was demonstrated within a large cohort of patients with early breast cancer treated in the prospective randomized FinHer trial [29]. Thus, it was shown that a higher IgκC expression was associated with longer metastasis-free survival, particularly in patients with triple-negative breast cancer.
These results are basically in line with the findings of our present retrospective immunohistochemical study which included 107 patients with early triple-negative breast cancer, from whom 79 received an adjuvant chemotherapy, showing a trend for a higher IgκC expression being associated with a better clinical outcome. These immunohistochemical results are supported by the validation of the IgκC expression using publicly available mRNA data from a larger, independent TNBC cohort: here, two probe sets were able to demonstrate that a higher IgκC expression was associated with significantly longer MFS (Figure 3).
Furthermore, in the present study, we demonstrated a prognostic effect of CD38-positive, tumor-infiltrating plasma cells, where a higher expression of CD38 was associated with a longer MFS. Additionally, CD38 could be identified as a positive independent prognostic marker (using the multivariate Cox Regression analyses). These results could be confirmed for relapse-free survival (RFS) and disease-free survival (DFS) by other studies, i.e., the plasma cell marker CD38, among other markers of tumor-infiltrating lymphocytes, was used in a Japanese retrospective study including 114 patients with triple-negative breast cancer. The authors postulated a significant prognostically favorable impact of CD38-positive, tumor-infiltrating plasma cells for relapse-free survival [28]. These findings are in line with the results of a retrospective study published in 2018 by Yeong et al. [30]. Using immunohistochemistry, the authors evaluated the prognostic impact of plasma cell-associated markers such as CD38 in a cohort of 269 TNBC samples. They revealed a statistically significant longer disease-free survival (DFS) in patients with a higher density of CD38-positive plasma cells (p = 0.004). Furthermore, they could show a significant correlation between the amount of CD38-positive plasma cells and the expression of B-cell-associated genes such as IgκC, IGHM (Immunoglobulin Heavy Constant Mu), and IGHG (Immunoglobin Heavy Constant Gamma) [30].
Antitumor immunity represents an important mechanism in tumor biology in general, not only in breast cancer but also in other tumor entities. Schmidt et al. were able to show that the humoral immune response has a favorable prognostic effect in non-small cell lung cancer (NSCLC) [24]. In a cohort of 196 NSCLC patients, the gene expression analyses revealed that the upregulation of the B-cell metagene and a higher IgκC expression were associated with longer survival. Within the subgroup analysis, this effect was significant only in adenocarcinomas, but not in squamous cell carcinomas. In addition, a retrospective Japanese study could show that the presence of TILs also has a favorable prognostic effect in endometrial carcinoma [31]. Based on the immunohistochemical analyses, it could be demonstrated that the B-cell infiltration (measured via the immunohistochemical expression of CD20 and CD38) correlated positively with the number of TILs and was associated with a better outcome.
The key secondary objective of our study was to examine whether a BRCA mutation was associated with a higher rate of tumor-infiltrating plasma cells. We showed a positive correlation of tumor-infiltrating plasma cells and mutations in BRCA. This finding may be explained via genetic instability and the resulting higher number of immunogenic antigens in the tumor, especially in the presence of a BRCA mutation [32]. The use of PARPi is known to increase the genomic instability already present in the tumor, so treatment with PARPi (poly ADP-ribose polymerase inhibitor) may increase the sensitivity to immunotherapy in BRCAm tumors. In a retrospective study, Grandal et al. showed that in patients with luminal breast cancer, the BRCA mutation status was associated with higher pCR rates and higher numbers of TILs after neoadjuvant chemotherapy [33], but they did not find a correlation between immune infiltration at the baseline and the BRCA mutation status. Another study by Solinas et al. demonstrated that there was a significantly higher rate of TIL-positive tumors in the BRCA-mutated (BRCAm) TNBC group compared to the BRCA-wildtype (wt) group (p = 0.037) [34]. However, the study cited above did not find a significant difference in the composition of TILs in the BRCAm TNBC group compared to the BRCAwt group. A further study by Telli et al. [35] evaluated the correlation between TILs and the BRCA mutation status in a cohort of 161 TNBC patients pooled from five phase II clinical trials of platinum-based neoadjuvant therapy. They did not find a significant association between TILs and the BRCA mutation status in the observed cohort of TNBC patients. Using TCGA data, Kraya et al. were able to show that BRCA-mutated breast cancer tumors dispose of a higher immunogenicity compared with those without mutation. In contrast, they found a negative correlation between HRD (Homologous recombination deficiency) scores and the gene expression-based immune markers [36]. The above-mentioned studies show that, in conclusion, the association between immune infiltration and BRCA mutation in triple-negative breast cancer is not yet clearly established and requires further studies with larger sample sizes.
The strength of our retrospective study is that we showed a positive prognostic impact of CD38- and IgκC-positive tumor-infiltrating plasma cells in TNBC patients. The differences in prognostic significance between the protein and the gene expression could be explained via post-translational changes and the different sample sizes analyzed for immunohistochemistry and the gene expression, respectively. Furthermore, a correlation between tumor-infiltrating plasma cells and BRCA mutations could be demonstrated. Potential weaknesses of the present study include the small sample size of 107 tumors, differences in systemic treatment (chemotherapy vs. no systemic therapy), differences in the analysis methods and sample size between the finding and validation cohorts (immunohistochemistry vs. gene expression), and the retrospective study design.
A further potential weakness of the retrospective study presented here is that the prognostic significance of stromal TILs was not analyzed, assuming the fact that the significance of stromal TILs in early triple-negative breast cancer is sufficiently well known [5,6,7,8,9,10,11,12]. However, it is not possible to conclude from this study whether the amount of plasma cell infiltration provides additional prognostic information compared to stromal TILs.

4. Material and Methods

4.1. Study Patients

The patient collective consisted of 107 patients who had been treated for triple-negative breast cancer between 2009 and 2016 at University Hospital Mainz and received breast-conserving surgery or mastectomy. Following surgical treatment, 79 patients received chemotherapy and 28 of the 107 patients were without chemotherapy, although there is a general recommendation for (neo)adjuvant chemotherapy in triple-negative tumors upon a tumor size pT1b and larger. According to the results of the follow up questionnaire and our database, the most common reasons for this were general refusal or the discontinuation of chemotherapy in the case of intolerance due to side effects. At the time of the diagnosis, the median age was 55 years. The proportion of BRCA mutation-associated triple-negative breast cancers was 15.9%, including 14.02% with BRCA-1 mutation and 1.88% with BRCA-2 mutation. The median follow up was five years.
At the time of the last follow-up, 88.8% of the patients were without metastases, whereas 11.2% developed distant metastases. The recurrence of their diseases was reported by 3.7% of the patients; thus, 96.3% were without recurrence.
To perform the immunohistochemistry of CD 38 and IgκC, tissue samples were available from the whole study cohort of 107 patients. Tumor slides were provided by the tissue bank of the University Medical Center Mainz in accordance with the regulations of the tissue biobank and the approval of the ethics committee of University Medical Center Mainz.
Patients’ characteristics are given in Table 4.

4.2. Immunostaining

For immunostaining, formalin-fixed and paraffin-embedded tumor slides were stained with commercially available monoclonal antibodies binding IgκC (Clone KP-53; Santa Cruz Biotechnology Company, Santa Cruz, CA, USA) and CD38 (Sigma-Aldrich Chemie GmbH, Schnelldorf, Germany), according to the standard procedures as previously described [26]. All slides were analyzed using a Leica light microscope (Leica Microsystem Vertrieb Company, Wetzlar, Germany) by three of the authors (N.R., A.-S.H., and M.S.) trained in histological and immunohistochemical diagnostics, unaware of the clinical outcome.

4.3. Evaluation of Immunostaining

For the immunohistochemical evaluation of IgκC- and/or CD38-positive tumor-infiltrating plasma cells, a semiquantitative scoring method was used as previously described [25]: 0, no IgκC/CD38-positive infiltrate; 1+, weak IgκC/CD38-positive infiltrate; 2+, moderate IgκC/CD38-positive infiltrate; and 3+, strong IgκC/CD38-positive infiltrate. To dichotomize the patients, tumor slides with a IgκC/CD38 score 0 and 1+ were considered as low plasma cell infiltration, whereas cases with 2+ and 3+ were high plasma cell infiltration, respectively.

4.4. Statistical Analysis

Statistical analyses were performed using the SPSS statistical software program, version 28.0 (IBM SPSS Statistics for Windows, Version 28.0., Released 2021 NY, USA, IBM Corp.
Patients’ characteristics were given in absolute and relative numbers. The prognostic significance of the immunohistochemically determined the IgκC and CD38 expression for MFS was examined via the Kaplan–Meier survival analysis as well as the univariate and multivariate Cox Regression analysis adjusted for age (≤50 vs. >50 years), pT-stage (≤2 vs. >2 cm), histological grade (GI + GII vs. GIII), and lymph node status (negative vs. positive). The significance of Kaplan–Meier survival analysis was assessed using the p value of the Log-Rank test. A possible correlation between the strength of plasma cell infiltration on the one hand and the histological degree of differentiation or the proliferation index Ki67 on the other hand was calculated using ordinal regression. The Spearman-Rho correlation coefficient was used to assess whether there was a correlation effect between the IgκC and C38 expression as well as a possible correlation between the intensity of plasma cell infiltration and the presence of a BRCA mutation (at the germline level).

4.5. Validation of Immunohistochemical Results Using mRNA Expression Data of IgκC and CD38 of an Independent TNBC Cohort

To validate our immunohistochemical results concerning the prognostic impact of the plasma cell markers IgκC and CD38 in a larger, independent cohort of TNBC, we used the publicly available gene expression data of IgκC and CD38 with their associated tumor characteristics, as well as the follow-up data [29] in a cohort of 424 TNBC samples.

5. Conclusions

In the present immunohistochemical retrospective study, we were able to show the prognostic significance of the humoral immune response in the observed collective of 107 patients with early triple-negative breast cancer. A higher number of CD38-positive tumor-infiltrating plasma cells was associated with a significantly longer MFS and BRCA mutations. Furthermore, CD38 was identified as an independent prognostic factor via the multivariate Cox regression analysis.

Author Contributions

Conceptualization, A.-S.H. and M.S.; Data curation, N.R. and M.S.; Formal analysis, A.-S.H. and N.R.; Investigation, M.S.; Methodology, A.-S.H., N.R. and M.S.; Project administration, A.-S.H.; Resources, W.B., A.H. and M.S.; Supervision, M.S.; Validation, A.-S.H., N.R. and M.S.; Visualization, A.-S.H.; Writing—original draft, A.-S.H. and M.S.; Writing—review and editing, K.A., S.K., R.S., K.S., A.L., M.J.B., W.B., A.H. and M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was approved by the Ethics Committee of Rhineland-Palatinate, Germany [no. 837.139.05 (4797)]. Written informed consent was obtained from all patients, and all clinical investigations were conducted ethically in accordance with ethical and legal standards and in consideration of the Declarations of Helsinki.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge Martina Seehase and Susanne Gebhard for excellent administrative and technical support. We would also like to acknowledge the tissue biobank of the University Medical Center Mainz for providing tumor slides (in accordance with the regulations of the tissue biobank and the approval of the ethics committee of University Medical Center Mainz).

Conflicts of Interest

The authors declare that this study was not funded by external sponsors. Listed below are the personal conflicts of interest of the (co-)authors. The funders listed were not involved in the study design, collection, analysis, interpretation of data, the writing of this article, or the decision to submit it for publication. A.-S. Heimes received honoraria from Pfizer Pharma GmbH, Roche Pharma AG, Daiichy Sankyo GmbH, Medupdate GmbH and Streamedup!GmbH K. Almstedt received speaker honoraria from Roche Pharma AG, Pfizer Pharma GmbH and AstraZeneca. K. Stewen received honoraria from StreamedUp!GmbH and Sanofi-Aventis GmbH R. Schwab received honoraria from Roche Pharma AG, AstraZeneca and Streamedup!GmbH M. J. Battista received honoraria and expenses from Astra Zeneca, Clovis Oncology, GSK, MSD, Pharma Mar, Roche and Tesaro Bio Germany GmbH. He is consultant to Eisai, GSK, MSD, Pharma Mar, Roche Pharma AG and Tesaro Bio Germany GmbH. He received funded research from Astra Zeneca, Clovis Oncology, MSD and Novartis. S. Krajnak received speaker honoraria from Roche Pharma AG and Novartis Pharma GmbH Germany, research funding from Novartis Pharma GmbH Germany and travel reimbursement from PharmaMar and Novartis Pharma GmbH Germany. A. Hasenburg received honoraria from AstraZeneca, Celgen, GSK, LEO Pharma, MedConcept GmbH, Med update GmbH, Medicultus, Pfizer, Promedicis GmbH, Softconsult, Roche Pharma AG, Streamedup!GmbH, Tesaro Bio Germany GmbH. She is a member of the advisory board of AstraZeneca, GSK, LEO Pharma, PharmaMar, Promedicis GmbH, Roche Pharma AG, Tesaro Bio Germany GmbH, MSD Sharp&Dohme GmbH M. Schmidt reports personal fees from AstraZeneca, BioNTech, Daiichi Sankyo, Eisai, Lilly, MSD, Novartis, Pantarhei Bioscience, Pfizer, Roche, and SeaGen outside the submitted work. Institutional research funding from AstraZeneca, BioNTech, Eisai, Genentech, German Breast Group, Novartis, Palleos, Pantarhei Bioscience, Pierre Fabre, and SeaGen. In addition, Marcus Schmidt has a patent for EP 2390370 B1 issued and a patent for EP 2951317 B1 issued. All other authors declare that they have no conflicts of interest.

References

  1. Metzger-Filho, O.; Tutt, A.; de Azambuja, E.; Saini, K.S.; Viale, G.; Loi, S.; Bradbury, I.; Bliss, J.M.; Azim, H.A., Jr.; Ellis, P.; et al. Dissecting the heterogeneity of triple-negative breast cancer. J. Clin. Oncol. 2012, 30, 1879–1887. [Google Scholar] [CrossRef]
  2. Iglesia, M.D.; Parker, J.S.; Hoadley, K.A.; Serody, J.S.; Perou, C.M.; Vincent, B.G. Genomic Analysis of Immune Cell Infiltrates Across 11 Tumor Types. J. Natl. Cancer Inst. 2016, 108, djw144. [Google Scholar] [CrossRef]
  3. Narang, P.; Chen, M.; Sharma, A.A.; Anderson, K.S.; Wilson, M.A. The neoepitope landscape of breast cancer: Implications for immunotherapy. BMC Cancer 2019, 19, 200. [Google Scholar] [CrossRef]
  4. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef]
  5. Denkert, C.; Loibl, S.; Noske, A.; Roller, M.; Muller, B.M.; Komor, M.; Budczies, J.; Darb-Esfahani, S.; Kronenwett, R.; Hanusch, C.; et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J. Clin. Oncol. 2010, 28, 105–113. [Google Scholar] [CrossRef]
  6. Denkert, C.; von Minckwitz, G.; Brase, J.C.; Sinn, B.V.; Gade, S.; Kronenwett, R.; Pfitzner, B.M.; Salat, C.; Loi, S.; Schmitt, W.D.; 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. 2015, 33, 983–991. [Google Scholar] [CrossRef]
  7. Denkert, C.; von Minckwitz, G.; Darb-Esfahani, S.; Lederer, B.; Heppner, B.I.; Weber, K.E.; 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. 2018, 19, 40–50. [Google Scholar] [CrossRef]
  8. Loi, S.; Drubay, D.; Adams, S.; Pruneri, G.; Francis, P.A.; Lacroix-Triki, M.; Joensuu, H.; Dieci, M.V.; Badve, S.; Demaria, S.; et al. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J. Clin. Oncol. 2019, 37, 559–569. [Google Scholar] [CrossRef]
  9. Hida, A.I.; Watanabe, T.; Sagara, Y.; Kashiwaba, M.; Sagara, Y.; Aogi, K.; Ohi, Y.; Tanimoto, A. Diffuse distribution of tumor-infiltrating lymphocytes is a marker for better prognosis and chemotherapeutic effect in triple-negative breast cancer. Breast Cancer Res. Treat. 2019, 178, 283–294. [Google Scholar] [CrossRef]
  10. Adams, S.; Gray, R.J.; Demaria, S.; Goldstein, L.; Perez, E.A.; Shulman, L.N.; Martino, S.; Wang, M.; Jones, V.E.; Saphner, T.J.; et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J. Clin. Oncol. 2014, 32, 2959–2966. [Google Scholar] [CrossRef]
  11. Ibrahim, E.M.; Al-Foheidi, M.E.; Al-Mansour, M.M.; Kazkaz, G.A. The prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancer: A meta-analysis. Breast Cancer Res. Treat. 2014, 148, 467–476. [Google Scholar] [CrossRef]
  12. de Jong, V.M.T.; Wang, Y.; Ter Hoeve, N.D.; Opdam, M.; Stathonikos, N.; Jozwiak, K.; Hauptmann, M.; Cornelissen, S.; Vreuls, W.; Rosenberg, E.H.; et al. Prognostic Value of Stromal Tumor-Infiltrating Lymphocytes in Young, Node-Negative, Triple-Negative Breast Cancer Patients Who Did Not Receive (neo)Adjuvant Systemic Therapy. J. Clin. Oncol. 2022, 40, 2361–2374. [Google Scholar] [CrossRef]
  13. Stanton, S.E.; Adams, S.; Disis, M.L. Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review. JAMA Oncol. 2016, 2, 1354–1360. [Google Scholar] [CrossRef]
  14. Burugu, S.; Asleh-Aburaya, K.; Nielsen, T.O. Immune infiltrates in the breast cancer microenvironment: Detection, characterization and clinical implication. Breast Cancer 2017, 24, 3–15. [Google Scholar] [CrossRef]
  15. Peng, G.L.; Li, L.; Guo, Y.W.; Yu, P.; Yin, X.J.; Wang, S.; Liu, C.P. CD8+ cytotoxic and FoxP3+ regulatory T lymphocytes serve as prognostic factors in breast cancer. Am. J. Transl. Res. 2019, 11, 5039–5053. [Google Scholar]
  16. Mahmoud, S.M.; Paish, E.C.; Powe, D.G.; Macmillan, R.D.; Grainge, M.J.; Lee, A.H.; Ellis, I.O.; Green, A.R. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J. Clin. Oncol. 2011, 29, 1949–1955. [Google Scholar] [CrossRef]
  17. Schmid, P.; Cortes, J.; Pusztai, L.; McArthur, H.; Kummel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N. Engl. J. Med. 2020, 382, 810–821. [Google Scholar] [CrossRef]
  18. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.-A.; Wright, C.S.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2019, 380, 987–988. [Google Scholar] [CrossRef]
  19. Cortes, J.; Cescon, D.W.; Rugo, H.S.; Nowecki, Z.; Im, S.A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; 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 2020, 396, 1817–1828. [Google Scholar] [CrossRef]
  20. Fremd, C.; Schuetz, F.; Sohn, C.; Beckhove, P.; Domschke, C. B cell-regulated immune responses in tumor models and cancer patients. Oncoimmunology 2013, 2, e25443. [Google Scholar] [CrossRef]
  21. Schmidt, M.; Bohm, D.; von Torne, C.; Steiner, E.; Puhl, A.; Pilch, H.; Lehr, H.A.; Hengstler, J.G.; Kolbl, H.; Gehrmann, M. The humoral immune system has a key prognostic impact in node-negative breast cancer. Cancer Res. 2008, 68, 5405–5413. [Google Scholar] [CrossRef]
  22. Bianchini, G.; Qi, Y.; Alvarez, R.H.; Iwamoto, T.; Coutant, C.; Ibrahim, N.K.; Valero, V.; Cristofanilli, M.; Green, M.C.; Radvanyi, L.; et al. Molecular anatomy of breast cancer stroma and its prognostic value in estrogen receptor-positive and -negative cancers. J. Clin. Oncol. 2010, 28, 4316–4323. [Google Scholar] [CrossRef]
  23. Gentles, A.J.; Newman, A.M.; Liu, C.L.; Bratman, S.V.; Feng, W.; Kim, D.; Nair, V.S.; Xu, Y.; Khuong, A.; Hoang, C.D.; et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 2015, 21, 938–945. [Google Scholar] [CrossRef]
  24. Schmidt, M.; Hellwig, B.; Hammad, S.; Othman, A.; Lohr, M.; Chen, Z.; Boehm, D.; Gebhard, S.; Petry, I.; Lebrecht, A.; et al. A comprehensive analysis of human gene expression profiles identifies stromal immunoglobulin kappa C as a compatible prognostic marker in human solid tumors. Clin. Cancer Res. 2012, 18, 2695–2703. [Google Scholar] [CrossRef]
  25. Chen, Z.; Gerhold-Ay, A.; Gebhard, S.; Boehm, D.; Solbach, C.; Lebrecht, A.; Battista, M.; Sicking, I.; Cotarelo, C.; Cadenas, C.; et al. Immunoglobulin kappa C predicts overall survival in node-negative breast cancer. PLoS ONE 2012, 7, e44741. [Google Scholar] [CrossRef]
  26. Heimes, A.S.; Kramer, H.; Hartner, F.; Almstedt, K.; Krajnak, S.; Battista, M.J.; Brenner, W.; Hasenburg, A.; Schmidt, M. Prognostic Impact of Immunoglobulin Kappa C in Breast Cancer Patients Treated with Adjuvant Chemotherapy. Breast Care 2021, 16, 444–451. [Google Scholar] [CrossRef]
  27. Gyorffy, B. Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer. Comput. Struct. Biotechnol. J. 2021, 19, 4101–4109. [Google Scholar] [CrossRef]
  28. Kuroda, H.; Jamiyan, T.; Yamaguchi, R.; Kakumoto, A.; Abe, A.; Harada, O.; Enkhbat, B.; Masunaga, A. Prognostic value of tumor-infiltrating B lymphocytes and plasma cells in triple-negative breast cancer. Breast Cancer 2021, 28, 904–914. [Google Scholar] [CrossRef]
  29. Schmidt, M.; Edlund, K.; Hengstler, J.G.; Heimes, A.S.; Almstedt, K.; Lebrecht, A.; Krajnak, S.; Battista, M.J.; Brenner, W.; Hasenburg, A.; et al. Prognostic Impact of Immunoglobulin Kappa C (IGKC) in Early Breast Cancer. Cancers 2021, 13, 3626. [Google Scholar] [CrossRef]
  30. Yeong, J.; Lim, J.C.T.; Lee, B.; Li, H.; Chia, N.; Ong, C.C.H.; Lye, W.K.; Putti, T.C.; Dent, R.; Lim, E.; et al. High Densities of Tumor-Associated Plasma Cells Predict Improved Prognosis in Triple Negative Breast Cancer. Front. Immunol. 2018, 9, 1209. [Google Scholar] [CrossRef]
  31. Qin, M.; Hamanishi, J.; Ukita, M.; Yamanoi, K.; Takamatsu, S.; Abiko, K.; Murakami, R.; Miyamoto, T.; Suzuki, H.; Ueda, A.; et al. Tertiary lymphoid structures are associated with favorable survival outcomes in patients with endometrial cancer. Cancer Immunol. Immunother. 2022, 71, 1431–1442. [Google Scholar] [CrossRef] [PubMed]
  32. Hunia, J.; Gawalski, K.; Szredzka, A.; Suskiewicz, M.J.; Nowis, D. The potential of PARP inhibitors in targeted cancer therapy and immunotherapy. Front. Mol. Biosci. 2022, 9, 1073797. [Google Scholar] [CrossRef] [PubMed]
  33. Grandal, B.; Evrevin, C.; Laas, E.; Jardin, I.; Rozette, S.; Laot, L.; Dumas, E.; Coussy, F.; Pierga, J.Y.; Brain, E.; et al. Impact of BRCA Mutation Status on Tumor Infiltrating Lymphocytes (TILs), Response to Treatment, and Prognosis in Breast Cancer Patients Treated with Neoadjuvant Chemotherapy. Cancers 2020, 12, 3681. [Google Scholar] [CrossRef] [PubMed]
  34. Solinas, C.; Marcoux, D.; Garaud, S.; Vitoria, J.R.; Van den Eynden, G.; de Wind, A.; De Silva, P.; Boisson, A.; Craciun, L.; Larsimont, D.; et al. BRCA gene mutations do not shape the extent and organization of tumor infiltrating lymphocytes in triple negative breast cancer. Cancer Lett. 2019, 450, 88–97. [Google Scholar] [CrossRef] [PubMed]
  35. Telli, M.L.; Chu, C.; Badve, S.S.; Vinayak, S.; Silver, D.P.; Isakoff, S.J.; Kaklamani, V.; Gradishar, W.; Stearns, V.; Connolly, R.M.; et al. Association of Tumor-Infiltrating Lymphocytes with Homologous Recombination Deficiency and BRCA1/2 Status in Patients with Early Triple-Negative Breast Cancer: A Pooled Analysis. Clin. Cancer Res. 2020, 26, 2704–2710. [Google Scholar] [CrossRef]
  36. Kraya, A.A.; Maxwell, K.N.; Wubbenhorst, B.; Wenz, B.M.; Pluta, J.; Rech, A.J.; Dorfman, L.M.; Lunceford, N.; Barrett, A.; Mitra, N.; et al. Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer. Clin. Cancer Res. 2019, 25, 4363–4374. [Google Scholar] [CrossRef]
Figure 1. Kaplan–Meier Diagram of IgκC expression (p = 0.35, Log Rank) in terms of MFS (metastasis-free survival) (months).
Figure 1. Kaplan–Meier Diagram of IgκC expression (p = 0.35, Log Rank) in terms of MFS (metastasis-free survival) (months).
Ijms 24 15219 g001
Figure 2. Kaplan–Meier Diagram of CD38 expression (p = 0.039 Log Rank) in terms of MFS (metastasis-free survival) (months).
Figure 2. Kaplan–Meier Diagram of CD38 expression (p = 0.039 Log Rank) in terms of MFS (metastasis-free survival) (months).
Ijms 24 15219 g002
Figure 3. Kaplan–Meier plots of IgκC mRNA expression (probe set 214669_x_at and 216576_x_at) of an independent TNBC cohort showing IgκC as a positive prognostic marker in terms of MFS.
Figure 3. Kaplan–Meier plots of IgκC mRNA expression (probe set 214669_x_at and 216576_x_at) of an independent TNBC cohort showing IgκC as a positive prognostic marker in terms of MFS.
Ijms 24 15219 g003
Figure 4. Kaplan–Meier plot of CD38 mRNA expression of an independent TNBC cohort: CD38 mRNA expression has no significant impact on MFS.
Figure 4. Kaplan–Meier plot of CD38 mRNA expression of an independent TNBC cohort: CD38 mRNA expression has no significant impact on MFS.
Ijms 24 15219 g004
Figure 5. Box Plot Diagram showing the correlation between BRCA mutation (BRCAm) and the extent of plasma cell infiltration (r = 0.211, p = 0.029).
Figure 5. Box Plot Diagram showing the correlation between BRCA mutation (BRCAm) and the extent of plasma cell infiltration (r = 0.211, p = 0.029).
Ijms 24 15219 g005
Table 1. Univariate Cox Regression Analysis in regard of MFS (metastasis-free survival).
Table 1. Univariate Cox Regression Analysis in regard of MFS (metastasis-free survival).
Multivariate Cox Regression MFS
VariableHazard Ratio95% CIp-Value
IgκC
Higher expression vs. lower expression
0.9100.406–2.0350.818
Age
>50/=50 vs. <50
0.5370.160–1.8020.314
Tumor Size
pT2–4 vs. pT1
2.2190.537–9.1680.271
Lymph node status
pN1–3 vs. pN0
4.7561.216–18.6010.025
Grading
G 1–2 vs. G 3
0.4940.143–1.7120.266
Table 2. Multivariate Cox Regressions analysis of IgκC in terms of MFS (metastasis-free survival) adjusted for age, tumor size, lymph node status, and tumor grade of differentiation.
Table 2. Multivariate Cox Regressions analysis of IgκC in terms of MFS (metastasis-free survival) adjusted for age, tumor size, lymph node status, and tumor grade of differentiation.
Univariate Cox Regression MFS
VariableHazard Ratio95% CIp-Value
IgκC
Higher expression vs. lower expression
0.8520.415–1.7510.664
CD38
Higher expression vs. lower expression
0.5080.252–1.0270.059
age
>50/= 50 vs. <50
0.6770.218–2.0990.499
Tumor size
pT2–4 vs. pT1
2.6410.715–9.7580.145
Lymph node status
pN1–3 vs. pN0
5.5721.508–20.5910.010
Grading
G 1–2 vs. G 3
0.4970.158–1.5660.233
Table 3. Multivariate Cox Regressions analysis of CD38 in terms of MFS (metastasis-free survival) adjusted for age, tumor size, lymph node status, and tumor grade of differentiation.
Table 3. Multivariate Cox Regressions analysis of CD38 in terms of MFS (metastasis-free survival) adjusted for age, tumor size, lymph node status, and tumor grade of differentiation.
Multivariate Cox Regression MFS
VariableHazard Ratio95% CIp-Value
CD38
Higher expression vs. lower expression
0.4380.195–0.9830.045
age
>50/= 50 vs. <50
0.4960.152–1.6190.245
Tumor size
pT2–4 vs. pT1
1.8690.459–7.6030.382
Lymph node status
pN1–3 vs. pN0
6.6621.639–27.0730.008
Grading
G 1–2 vs. G 3
0.8750.228–3.3660.846
Table 4. Patients’ characteristics (in absolute and relative numbers).
Table 4. Patients’ characteristics (in absolute and relative numbers).
NumberPercentage
Age at diagnosis
<504441%
>506359%
Tumor size
pT14946%
pT2–45854%
Lymph node status
pN06864%
pN1–33936%
Tumor grade
G 1–22826%
G 37974%
Ki67%
≤20%76.5%
>20%5450.5%
Missing value4643%
IgκC expression
weak (Score 0/1)5955.1%
strong (Score 2/3)4844.9%
CD38 expression
weak (Score 0/1)6056.1%
strong (Score 2/3)4743.9%
Chemotherapy
Yes7973.8%
No2826.2%
BRCA Mutation
BRCAwt (wildtype)9084.1%
BRCAm (mutation)1715.9%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Heimes, A.-S.; Riedel, N.; Almstedt, K.; Krajnak, S.; Schwab, R.; Stewen, K.; Lebrecht, A.; Battista, M.J.; Brenner, W.; Hasenburg, A.; et al. Prognostic Impact of CD38- and IgκC-Positive Tumor-Infiltrating Plasma Cells in Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2023, 24, 15219. https://doi.org/10.3390/ijms242015219

AMA Style

Heimes A-S, Riedel N, Almstedt K, Krajnak S, Schwab R, Stewen K, Lebrecht A, Battista MJ, Brenner W, Hasenburg A, et al. Prognostic Impact of CD38- and IgκC-Positive Tumor-Infiltrating Plasma Cells in Triple-Negative Breast Cancer. International Journal of Molecular Sciences. 2023; 24(20):15219. https://doi.org/10.3390/ijms242015219

Chicago/Turabian Style

Heimes, Anne-Sophie, Natali Riedel, Katrin Almstedt, Slavomir Krajnak, Roxana Schwab, Kathrin Stewen, Antje Lebrecht, Marco Johannes Battista, Walburgis Brenner, Annette Hasenburg, and et al. 2023. "Prognostic Impact of CD38- and IgκC-Positive Tumor-Infiltrating Plasma Cells in Triple-Negative Breast Cancer" International Journal of Molecular Sciences 24, no. 20: 15219. https://doi.org/10.3390/ijms242015219

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop