1
|
Li F, Gao C, Huang Y, Qiao Y, Xu H, Liu S, Wu H. Unraveling the breast cancer tumor microenvironment: crucial factors influencing natural killer cell function and therapeutic strategies. Int J Biol Sci 2025; 21:2606-2628. [PMID: 40303301 PMCID: PMC12035885 DOI: 10.7150/ijbs.108803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/04/2025] [Indexed: 05/02/2025] Open
Abstract
Natural killer (NK) cells have emerged as a novel and effective treatment for breast cancer. Nevertheless, the breast cancer tumor microenvironment (TME) manifests multiple immunosuppressive mechanisms, impeding the proper execution of NK cell functions. This review summarizes recent research on the influence of the TME on the functionality of NK cells in breast cancer. It delves into the effects of the internal environment of the TME on NK cells and elucidates the roles of diverse stromal components, immune cells, and signaling molecules in regulating NK cell activity within the TME. It also summarizes therapeutic strategies based on small-molecule inhibitors, antibody therapies, and natural products, as well as the progress of research in preclinical and clinical trials. By enhancing our understanding of the immunosuppressive TME and formulating strategies to counteract its effects, we could fully harness the therapeutic promise of NK cells in breast cancer treatment.
Collapse
Affiliation(s)
- Feifei Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunfang Gao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Yu Qiao
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Hongxiao Xu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huangan Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| |
Collapse
|
2
|
Kim R, Kawai A, Wakisaka M, Shimoyama M, Yasuda N, Ito M, Kin T, Arihiro K. Accumulation of CD56 + CD16 - Natural Killer Cells in Response to Preoperative Chemotherapy for Breast Cancer. World J Oncol 2024; 15:682-694. [PMID: 38993254 PMCID: PMC11236370 DOI: 10.14740/wjon1885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/28/2024] [Indexed: 07/13/2024] Open
Abstract
Background The activation of the antitumor immune responses of T cells and natural killer (NK) cells is important to induce breast tumor shrinkage via preoperative chemotherapy. We evaluated how antitumor immune responses contribute to the effects of such therapy. Methods Forty-three patients with stages I - IV breast cancer who underwent surgery between August 2018 and Jun 2023 after preoperative chemotherapy were enrolled. Peripheral natural killer (pNK) cell activity was assessed by 51Cr-release assay, and the counts and percentages of CD4+, CD8+, and NK cells and their subsets in peripheral blood were measured before and after chemotherapy by two-color flow cytometry. Associations of cell population changes with chemotherapy responses were analyzed. Results On univariate analysis, relative to grade (G) ≤ 1 effects, G ≥ 2 therapeutic effects were associated significantly with human epidermal growth factor receptor 2 (HER-2)+ breast cancer (P = 0.024) and post-chemotherapy CD56+ CD16- NK cell accumulation (8.4% vs. 5.5%, P = 0.042), and tended to be associated with increased pre-chemotherapy CD56+ CD16- NK cell percentages (5.4% vs. 3.3%, P = 0.054) and pNK cell activity (42.0% vs. 34.5%, P = 0.057). The accumulation and increased percentage of CD56+ CD16- NK cells in patients with G ≥ 2 effects were not associated with changes in pNK cell activity or the disappearance of axillary lymph-node metastases. On multivariate analysis, G ≥ 2 therapeutic effects tended to be associated with higher pre-chemotherapy pNK levels (odds ratio = 0.96; 95% confidence interval: 0.921 - 1.002; P = 0.067). Conclusions The accumulation of the immunoregulatory CD56+ CD16- NK cell subset in the peripheral blood before and after chemotherapy may lead to the production of cytokines that induce an antitumor immune response. Activation of the immune response mediated by CD56+ CD16- pNK cells after chemotherapy and their high counts before chemotherapy may contribute to the improvement of therapeutic effects against breast cancer.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Ami Kawai
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Megumi Wakisaka
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Mika Shimoyama
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Naomi Yasuda
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Mitsuya Ito
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Takanori Kin
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
3
|
Yi JI, Schneider J, Lim ST, Park B, Suh YJ. Interferon-Gamma Secretion Is Significantly Decreased in Stage III Breast Cancer Patients. Int J Mol Sci 2024; 25:4561. [PMID: 38674146 PMCID: PMC11050491 DOI: 10.3390/ijms25084561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Even though some studies have shown possible clinical relationship between molecular subtypes and tumor infiltrating natural killer (NK) cells around tumors, there are few studies showing the clinical relevance of peripheral NK cell activity at diagnosis in female patients with invasive breast cancer. A total of 396 female invasive breast cancer patients who received curative surgical treatment from March 2017 to July 2021 were retrospectively analyzed. NK cell activation-induced interferon-gamma (IFN-γ) secretion measured by enzyme-linked immunosorbent assay was used to measure the activity of peripheral NK cells. Statistical analyses were performed to determine clinical relationships with major clinicopathologic parameters. Quadripartite NK cell activity measured by induced interferon-gamma showed significant relevance with staging and body mass index, and some of the inflammatory serological markers, namely N/L (neutrophil/lymphocyte), P/N (platelet/neutrophil), and P/L (platelet/lymphocyte), showed significantly different NK activity in each interval by univariate analysis. A binary subgroup analysis, setting the IFN-γ secretion cut-off at 100 pg/mL, showed that stage III was significantly increased and axillary lymph node metastasis positivity was increased in the group of IFN-γ < 100 pg/mL, and IFN-γ secretion decreased with an increasing N stage, increased BMI (body mass index), and decreased production of IFN-γ. Following this, the same binary analysis, but with the IFN-γ secretion cut-off at 250 pg/mL, also showed that secretion in stage III was increased in those concentrations with <250 pg/mL, axillary lymph node positivity appeared to be correlated, and BMI ≥ 30 increased in prevalence. Additional ANOVA post hoc tests (Bonferroni) were performed on some serological markers that tended to be somewhat inconsistent. By subgroup analysis with Bonferroni adjustment between the IFN-γ secretion and TNM stage, no significant difference in IFN-γ secretion could be identified at stages I, II, and IV, but at stage III, the IFN-γ secretion < 100 pg/mL was significantly higher than 250 ≤ IFN-γ secretion < 500 pg/mL or IFN-γ secretion ≥ 500 pg/mL. According to this study, stage III was significantly associated with the lowest IFN-γ secretion. Compared to a higher level of IFN-γ secretion, a lower level of IFN-γ secretion seemed to be associated with increased body mass index. Unlike when IFN-γ secretion was analyzed in quartiles, as the IFN-γ secretion fell below 100 pg/mL, the correlation between axillary lymph node positivity and increased N stage, increased BMI, and increased N/L and P/L, which are suggested poor prognostic factors, became more pronounced. We think a peripheral IFN-γ secretion test might be convenient and useful tool for pretreatment risk assessment and selecting probable candidates for further treatment such as immunotherapy in some curable but high-risk invasive breast cancer patients, compared to other costly assaying of tissue NK cell activity at diagnosis.
Collapse
Affiliation(s)
- Jung Im Yi
- Department of Surgery, The Catholic University of Korea St. Vincent’s Hospital, Suwon 16247, Republic of Korea; (J.I.Y.); (S.T.L.); (B.P.)
| | - Jean Schneider
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Seung Taek Lim
- Department of Surgery, The Catholic University of Korea St. Vincent’s Hospital, Suwon 16247, Republic of Korea; (J.I.Y.); (S.T.L.); (B.P.)
| | - Byeongkwan Park
- Department of Surgery, The Catholic University of Korea St. Vincent’s Hospital, Suwon 16247, Republic of Korea; (J.I.Y.); (S.T.L.); (B.P.)
| | - Young Jin Suh
- Department of Surgery, The Catholic University of Korea St. Vincent’s Hospital, Suwon 16247, Republic of Korea; (J.I.Y.); (S.T.L.); (B.P.)
| |
Collapse
|
4
|
Dou T, Li J, Zhang Y, Pei W, Zhang B, Wang B, Wang Y, Jia H. The cellular composition of the tumor microenvironment is an important marker for predicting therapeutic efficacy in breast cancer. Front Immunol 2024; 15:1368687. [PMID: 38487526 PMCID: PMC10937353 DOI: 10.3389/fimmu.2024.1368687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
At present, the incidence rate of breast cancer ranks first among new-onset malignant tumors in women. The tumor microenvironment is a hot topic in tumor research. There are abundant cells in the tumor microenvironment that play a protumor or antitumor role in breast cancer. During the treatment of breast cancer, different cells have different influences on the therapeutic response. And after treatment, the cellular composition in the tumor microenvironment will change too. In this review, we summarize the interactions between different cell compositions (such as immune cells, fibroblasts, endothelial cells, and adipocytes) in the tumor microenvironment and the treatment mechanism of breast cancer. We believe that detecting the cellular composition of the tumor microenvironment is able to predict the therapeutic efficacy of treatments for breast cancer and benefit to combination administration of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Binyue Zhang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
5
|
Quail DF, Park M, Welm AL, Ekiz HA. Breast Cancer Immunity: It is TIME for the Next Chapter. Cold Spring Harb Perspect Med 2024; 14:a041324. [PMID: 37188526 PMCID: PMC10835621 DOI: 10.1101/cshperspect.a041324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Our ability to interrogate the tumor immune microenvironment (TIME) at an ever-increasing granularity has uncovered critical determinants of disease progression. Not only do we now have a better understanding of the immune response in breast cancer, but it is becoming possible to leverage key mechanisms to effectively combat this disease. Almost every component of the immune system plays a role in enabling or inhibiting breast tumor growth. Building on early seminal work showing the involvement of T cells and macrophages in controlling breast cancer progression and metastasis, single-cell genomics and spatial proteomics approaches have recently expanded our view of the TIME. In this article, we provide a detailed description of the immune response against breast cancer and examine its heterogeneity in disease subtypes. We discuss preclinical models that enable dissecting the mechanisms responsible for tumor clearance or immune evasion and draw parallels and distinctions between human disease and murine counterparts. Last, as the cancer immunology field is moving toward the analysis of the TIME at the cellular and spatial levels, we highlight key studies that revealed previously unappreciated complexity in breast cancer using these technologies. Taken together, this article summarizes what is known in breast cancer immunology through the lens of translational research and identifies future directions to improve clinical outcomes.
Collapse
Affiliation(s)
- Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec H3A 1A3, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec H3A 1A3, Canada
- Departments of Biochemistry, Oncology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce, 35430 Urla, Izmir, Turkey
| |
Collapse
|
6
|
Zhang J, Yang Y, Wei Y, Li L, Wang X, Ye Z. Hsa-miR-301a-3p inhibited the killing effect of natural killer cells on non-small cell lung cancer cells by regulating RUNX3. Cancer Biomark 2023:CBM220469. [PMID: 37302028 DOI: 10.3233/cbm-220469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most commonly diagnosed solid tumor. Natural killer (NK) cell-based immunotherapy is a promising anti-tumor strategy in various cancers including NSCLC. OBJECTIVE We aimed to investigate the specific mechanisms that regulate the killing effect of NK cells to NSCLC cells. METHODS Reverse transcription-quantitative PCR (RT-qPCR) assay was applied to measure the levels of hsa-microRNA (miR)-301a-3p and Runt-related transcription factor 3 (RUNX3). Enzyme-linked immunosorbent assay (ELISA) was used to measure the levels of IFN-γ and TNF-α. Lactate dehydrogenase assay was applied to detect the killing effect of NK cells. Dualluciferase reporter assay and RNA immunoprecipitation (RIP) assay were carried out to confirm the regulatory relationship between hsa-miR-301a-3p and RUNX3. RESULTS A low expression of hsa-miR-301a-3p was observed in NK cells stimulated by IL-2. The levels of IFN-γ and TNF-α were increased in NK cells of the IL-2 group. Overexpression of hsa-miR-301a-3p reduced the levels of IFN-γ and TNF-α as well as the killing effect of NK cells. Furthermore, RUNX3 was identified to be a target of hsamiR-301a-3p. hsa-miR-301a-3p suppressed the cytotoxicity of NK cells to NSCLC cells by inhibiting the expression of RUNX3. We found hsa-miR-301a-3p promoted tumor growth by suppressing the killing effect of NK cells against NSCLC cells in vivo. CONCLUSIONS Hsa-miR-301a-3p suppressed the killing effect of NK cells on NSCLC cells by targeting RUNX3, which may provide promising strategies for NK cell-based antitumor therapies.
Collapse
|
7
|
Zhang W, Xu K, Li Z, Wang L, Chen H. Tumor immune microenvironment components and the other markers can predict the efficacy of neoadjuvant chemotherapy for breast cancer. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1579-1593. [PMID: 36652115 DOI: 10.1007/s12094-023-03075-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023]
Abstract
Breast cancer is an epithelial malignant tumor that occurs in the terminal ducts of the breast. Neoadjuvant chemotherapy (NACT) is an important part of breast cancer treatment. Its purpose is to use systemic treatment for some locally advanced breast cancer patients, to decrease the tumor size and clinical stage so that non-operable breast cancer patients can have a chance to access surgical treatment, or patients who are not suitable for breast-conserving surgery can get the opportunity of breast-conserving. However, some patients who do not respond to NACT will lead deterioration in their condition. Therefore, prediction of NACT efficacy in breast cancer is vital for precision therapy. The tumor microenvironment (TME) has a crucial role in the carcinogenesis and therapeutic response of breast cancer. In this review, we summarized the immune cells, immune checkpoints, and other biomarkers in the TME that can evaluate the efficacy of NACT in treating breast cancer. We believe that the detection and evaluation of the TME components in breast cancer are helpful to predict the efficacy of NACT, and the prediction methods are in the prospect. In addition, we also summarized other predictive factors of NACT, such as imaging examination, biochemical markers, and multigene/multiprotein profiling.
Collapse
Affiliation(s)
- Weiqian Zhang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Ke Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Zhengfa Li
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Linwei Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Honglei Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China. .,Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
8
|
lncRNA MANCR Inhibits NK Cell Killing Effect on Lung Adenocarcinoma by Targeting miRNA-30d-5p. Cell Microbiol 2022. [DOI: 10.1155/2022/4928635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background. NK cells are imperative in spontaneous antitumor response of various cancers. Currently, lncRNAs are considered important modulators of the tumor microenvironment. This study investigated the molecular mechanism by which mitotically associated long noncoding RNA (MANCR) controls killing effect of NK cells on lung adenocarcinoma (LUAD) in the tumor microenvironment. Methods. The interplay between MANCR and miRNA-30d-5p was analyzed by bioinformatics. Expression of MANCR mRNA and miRNA-30d-5p was examined using qRT-PCR. Dual-luciferase reporter and RIP assays were utilized to verify the targeted relationship between MANCR and miRNA-30d-5p. To investigate regulation of MANCR/miRNA-30d-5p axis in NK cell killing effect on LUAD cells, western blot tested the protein level of perforin and granzyme B. ELISA determined the level of IFN-γ. CytoTox 96 Non-Radioactive Cytotoxicity Assay kit was applied for cytotoxicity detection of NK cells. Perforin and granzyme B fluorescence intensity was measured via immunofluorescence, and cell apoptosis levels were also revealed via flow cytometry. Results. MANCR was found to be upregulated, while miRNA-30d-5p expression was downregulated in LUAD tissues. Overexpression of MANCR in LUAD cells significantly reduced NK cell IFN-γ secretion, expression of granzyme B and perforin, and NK cell killing effect. In addition, MANCR could target and downregulate miRNA-30d-5p expression, and miRNA-30d-5p overexpression reversed the inhibition of NK cell killing effect caused by MANCR overexpression. Conclusion. MANCR inhibited the killing effect of NK cells on LUAD via targeting and downregulating miRNA-30d-5p and provided new ideas for antitumor therapy based on tumor microenvironment.
Collapse
|
9
|
Razeghian E, Kameh MC, Shafiee S, Khalafi F, Jafari F, Asghari M, Kazemi K, Ilkhani S, Shariatzadeh S, Haj-Mirzaian A. The role of the natural killer (NK) cell modulation in breast cancer incidence and progress. Mol Biol Rep 2022; 49:10935-10948. [PMID: 36008609 DOI: 10.1007/s11033-022-07865-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/11/2022] [Indexed: 01/11/2023]
Abstract
The importance of the immune system on tumor surveillance has been investigated for many years, and its impact on controlling tumor progression has been verified. An important subgroup of the innate immune system is natural killer (NK) cells, whose essential function in modulating tumor behavior and suppressing metastasis and tumor growth has been demonstrated. The first idea of NK cells' crucial biological processes was demonstrated through their potent ability to conduct direct cellular cytotoxicity, even without former sensitization. These properties of NK cells allow them to recognize transformed cells that have attenuated self-ligand and express stress-induced ligands. Furthermore, secretion of various cytokines and chemokines after their activation leads to tumor elimination via either direct cytotoxic effect on malignant cells or activation of the adaptive immune system. In addition, novel immunotherapeutic approaches tend to take advantage of NK cells' ability, leading to antibody-based approaches, the formation of engineered CAR-NK cells, and adoptive cell transfer. However, the restricted functionality of NK cells and the inability to infiltrate tumors are its blind spots in breast cancer patients. In this review, we gathered newly acquired data on the biology and functions of NK cells in breast cancer and proposed ways to employ this knowledge for novel therapeutic approaches in cancers, particularly breast cancer.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mahdis Chahar Kameh
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Shafiee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farima Khalafi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fehimeh Jafari
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadali Asghari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiarash Kazemi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Ilkhani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arvin Haj-Mirzaian
- Department of Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Depression in breast cancer patients: Immunopathogenesis and immunotherapy. Cancer Lett 2022; 536:215648. [DOI: 10.1016/j.canlet.2022.215648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 01/10/2023]
|
11
|
Zhang J, Pan S, Jian C, Hao L, Dong J, Sun Q, Jin H, Han X. Immunostimulatory Properties of Chemotherapy in Breast Cancer: From Immunogenic Modulation Mechanisms to Clinical Practice. Front Immunol 2022; 12:819405. [PMID: 35069604 PMCID: PMC8766762 DOI: 10.3389/fimmu.2021.819405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) is the most common malignancy among females. Chemotherapy drugs remain the cornerstone of treatment of BC and undergo significant shifts over the past 100 years. The advent of immunotherapy presents promising opportunities and constitutes a significant complementary to existing therapeutic strategies for BC. Chemotherapy as a cytotoxic treatment that targets proliferation malignant cells has recently been shown as an effective immune-stimulus in multiple ways. Chemotherapeutic drugs can cause the release of damage-associated molecular patterns (DAMPs) from dying tumor cells, which result in long-lasting antitumor immunity by the key process of immunogenic cell death (ICD). Furthermore, Off-target effects of chemotherapy on immune cell subsets mainly involve activation of immune effector cells including natural killer (NK) cells, dendritic cells (DCs), and cytotoxic T cells, and depletion of immunosuppressive cells including Treg cells, M2 macrophages and myeloid-derived suppressor cells (MDSCs). Current mini-review summarized recent large clinical trials regarding the combination of chemotherapy and immunotherapy in BC and addressed the molecular mechanisms of immunostimulatory properties of chemotherapy in BC. The purpose of our work was to explore the immune-stimulating effects of chemotherapy at the molecular level based on the evidence from clinical trials, which might be a rationale for combinations of chemotherapy and immunotherapy in BC.
Collapse
Affiliation(s)
- Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen Jian
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Li Hao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jie Dong
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Qingqing Sun
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
12
|
Kos K, Aslam MA, van de Ven R, Wellenstein MD, Pieters W, van Weverwijk A, Duits DEM, van Pul K, Hau CS, Vrijland K, Kaldenbach D, Raeven EAM, Quezada SA, Beyaert R, Jacobs H, de Gruijl TD, de Visser KE. Tumor-educated T regs drive organ-specific metastasis in breast cancer by impairing NK cells in the lymph node niche. Cell Rep 2022; 38:110447. [PMID: 35235800 DOI: 10.1016/j.celrep.2022.110447] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 11/01/2021] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is accompanied by systemic immunosuppression, which facilitates metastasis formation, but how this shapes organotropism of metastasis is poorly understood. Here, we investigate the impact of mammary tumorigenesis on regulatory T cells (Tregs) in distant organs and how this affects multi-organ metastatic disease. Using a preclinical mouse mammary tumor model that recapitulates human metastatic breast cancer, we observe systemic accumulation of activated, highly immunosuppressive Tregs during primary tumor growth. Tumor-educated Tregs show tissue-specific transcriptional rewiring in response to mammary tumorigenesis. This has functional consequences for organotropism of metastasis, as Treg depletion reduces metastasis to tumor-draining lymph nodes, but not to lungs. Mechanistically, we find that Tregs control natural killer (NK) cell activation in lymph nodes, thereby facilitating lymph node metastasis. In line, an increased Treg/NK cell ratio is observed in sentinel lymph nodes of breast cancer patients compared with healthy controls. This study highlights that immune regulation of metastatic disease is highly organ dependent.
Collapse
Affiliation(s)
- Kevin Kos
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Muhammad A Aslam
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Rieneke van de Ven
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam and Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, the Netherlands
| | - Max D Wellenstein
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Wietske Pieters
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Antoinette van Weverwijk
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Danique E M Duits
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Kim van Pul
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam and Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, the Netherlands
| | - Cheei-Sing Hau
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Kim Vrijland
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Daphne Kaldenbach
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Elisabeth A M Raeven
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Sergio A Quezada
- Cancer Immunology Unit, University College London Cancer Institute, WC1E 6DD London, UK
| | - Rudi Beyaert
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Heinz Jacobs
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam and Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, the Netherlands
| | - Karin E de Visser
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
13
|
Molecular Mechanisms, Biomarkers and Emerging Therapies for Chemotherapy Resistant TNBC. Int J Mol Sci 2022; 23:ijms23031665. [PMID: 35163586 PMCID: PMC8836182 DOI: 10.3390/ijms23031665] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with high recurrence rates, high incidence of distant metastases, and poor overall survival (OS). Taxane and anthracycline-containing chemotherapy (CT) is currently the main systemic treatment option for TNBC, while platinum-based chemotherapy showed promising results in the neoadjuvant and metastatic settings. An early arising of intrinsic or acquired CT resistance is common and represents the main hurdle for successful TNBC treatment. Numerous mechanisms were uncovered that can lead to the development of chemoresistance. These include cancer stem cells (CSCs) induction after neoadjuvant chemotherapy (NACT), ATP-binding cassette (ABC) transporters, hypoxia and avoidance of apoptosis, single factors such as tyrosine kinase receptors (EGFR, IGFR1), a disintegrin and metalloproteinase 10 (ADAM10), and a few pathological molecular pathways. Some biomarkers capable of predicting resistance to specific chemotherapeutic agents were identified and are expected to be validated in future studies for a more accurate selection of drugs to be employed and for a more tailored approach, both in neoadjuvant and advanced settings. Recently, based on specific biomarkers, some therapies were tailored to TNBC subsets and became available in clinical practice: olaparib and talazoparib for BRCA1/2 germline mutation carriers larotrectinib and entrectinib for neurotrophic tropomyosin receptor kinase (NTRK) gene fusion carriers, and anti-trophoblast cell surface antigen 2 (Trop2) antibody drug conjugate therapy for heavily pretreated metastatic TNBC (mTNBC). Further therapies targeting some pathologic molecular pathways, apoptosis, miRNAS, epidermal growth factor receptor (EGFR), insulin growth factor 1 receptor (IGF-1R), and androgen receptor (AR) are under investigation. Among them, phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and EGFR inhibitors as well as antiandrogens showed promising results and are under evaluation in Phase II/III clinical trials. Emerging therapies allow to select specific antiblastics that alone or by integrating the conventional therapeutic approach may overcome/hinder chemoresistance.
Collapse
|
14
|
Toffoli EC, Sweegers MG, Bontkes HJ, Altenburg TM, Verheul HM, van der Vliet HJ, de Gruijl TD, Buffart LM. Effects of physical exercise on natural killer cell activity during (neo)adjuvant chemotherapy: A randomized pilot study. Physiol Rep 2021; 9:e14919. [PMID: 34110712 PMCID: PMC8191403 DOI: 10.14814/phy2.14919] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/14/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Natural killer (NK) cells are a population of innate immune cells known to play a pivotal role against tumor spread. In multiple murine models, it was shown that physical exercise had the potential to increase NK cell antitumor activity through their mobilization and tissue redistribution in an interleukin (IL)-6 and epinephrine-dependent manner. The translation of this finding to patients is unclear. In this randomized pilot trial, we analyzed blood samples of patients with resectable breast or colon cancer who were randomized into an evidence-based moderate-high intensity resistance and aerobic exercise intervention (n = 8) or a control group (n = 6) during the first 9-12 weeks of (neo)adjuvant chemotherapy. In this pilot, we did not solely focus on statistical significance, but also explored whether average between-group differences reached 10%. NK cell degranulation was preserved in the exercise group whereas it decreased in the control group resulting in a between-group difference of 11.4% CD107a+ degranulated NK cells (95%CI = 0.57;22.3, p = 0.04) in the presence and 13.8% (95%CI = -2.5;30.0, p = 0.09) in the absence of an anti-epidermal growth factor receptor monoclonal antibody (EGFR-mAb). In line, the between-group difference of tumor cell lysis was 7.4% (95%CI = -9.1;23.9, p = 0.34), and 13.7% (95%CI = -10.1;37.5, p = 0.23) in favor of the exercise group in the presence or absence of EGFR mAb, respectively. Current explorative analyses showed that exercise during (neo)adjuvant chemotherapy may benefit NK cell activity. Future studies with a larger sample size are needed to confirm this finding and to establish its clinical potential. Trial registration: Dutch trial register number NTR4105.
Collapse
Affiliation(s)
- Elisa C. Toffoli
- Department of Medical OncologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Maike G. Sweegers
- Department of Epidemiology and BiostatisticsAmsterdam UMCVrije Universiteit AmsterdamAmsterdam Public HealthAmsterdamThe Netherlands
| | - Hetty J. Bontkes
- Department of Clinical ChemistryAmsterdam UMCVrije Universiteit AmsterdamAmsterdamNetherlands
| | - Teatske M. Altenburg
- Department of Public and Occupational HealthAmsterdam Public Health Research InstituteAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Henk M.W. Verheul
- Department of Medical OncologyRadboud University Medical CenterRadboud Institute for Health SciencesNijmegenThe Netherlands
| | - Hans J. van der Vliet
- Department of Medical OncologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Lava TherapeuticsUtrechtNetherlands
| | - Tanja D. de Gruijl
- Department of Medical OncologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Laurien M. Buffart
- Department of PhysiologyRadboud University Medical CenterRadboud Institute for Health SciencesNijmegenThe Netherlands
| |
Collapse
|
15
|
Batalha S, Ferreira S, Brito C. The Peripheral Immune Landscape of Breast Cancer: Clinical Findings and In Vitro Models for Biomarker Discovery. Cancers (Basel) 2021; 13:1305. [PMID: 33804027 PMCID: PMC8001103 DOI: 10.3390/cancers13061305] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the deadliest female malignancy worldwide and, while much is known about phenotype and function of infiltrating immune cells, the same attention has not been paid to the peripheral immune compartment of breast cancer patients. To obtain faster, cheaper, and more precise monitoring of patients' status, it is crucial to define and analyze circulating immune profiles. This review compiles and summarizes the disperse knowledge on the peripheral immune profile of breast cancer patients, how it departs from healthy individuals and how it changes with disease progression. We propose this data to be used as a starting point for validation of clinically relevant biomarkers of disease progression and therapy response, which warrants more thorough investigation in patient cohorts of specific breast cancer subtypes. Relevant clinical findings may also be explored experimentally using advanced 3D cellular models of human cancer-immune system interactions, which are under intensive development. We review the latest findings and discuss the strengths and limitations of such models, as well as the future perspectives. Together, the scientific advancement of peripheral biomarker discovery and cancer-immune crosstalk in breast cancer will be instrumental to uncover molecular mechanisms and putative biomarkers and drug targets in an all-human setting.
Collapse
Affiliation(s)
- Sofia Batalha
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal;
- Instituto de Tecnologia Química e Biológica António Xavier, University Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia Ferreira
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Rua Prof Lima Basto, 1099-023 Lisboa, Portugal;
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal;
- Instituto de Tecnologia Química e Biológica António Xavier, University Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
16
|
Kim R, Kin T. Clinical Perspectives in Addressing Unsolved Issues in (Neo)Adjuvant Therapy for Primary Breast Cancer. Cancers (Basel) 2021; 13:926. [PMID: 33672204 PMCID: PMC7927115 DOI: 10.3390/cancers13040926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 01/13/2023] Open
Abstract
The treatment of primary breast cancer has evolved over the past 50 years based on the concept that breast cancer is a systemic disease, with the escalation of adjuvant and neoadjuvant therapies and de-escalation of breast cancer surgery. Despite the development of these therapies, recurrence with distant metastasis during the 10 years after surgical treatment is observed, albeit infrequently. Recent advances in genomic analysis based on circulating tumor cells and circulating tumor DNA have enabled the development of targeted therapies based on genetic mutations in residual tumor cells. A paradigm shift involving the application of neoadjuvant chemotherapy (NAC) has enabled the prediction of treatment response and long-term prognoses; additional adjuvant chemotherapy targeting remaining tumor cells after NAC improves survival. The activation of antitumor immunity by anticancer agents may be involved in the eradication of residual tumor cells. Elucidation of the manner in which antitumor immunity is induced by anticancer agents and unknown factors, and the overcoming of drug resistance via the targeted eradication of residual tumor cells based on genomic profiles, will inevitably lead to the achievement of 0% distant recurrence and a complete cure for primary breast cancer.
Collapse
Affiliation(s)
- Ryungsa Kim
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome, Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
| | - Takanori Kin
- Department of Breast Surgery, Hiroshima City Hospital, 7-33, Moto-machi, Naka-ku, Hiroshima 730-8518, Japan;
| |
Collapse
|
17
|
Makowska A, Meier S, Shen L, Busson P, Baloche V, Kontny U. Anti-PD-1 antibody increases NK cell cytotoxicity towards nasopharyngeal carcinoma cells in the context of chemotherapy-induced upregulation of PD-1 and PD-L1. Cancer Immunol Immunother 2021; 70:323-336. [PMID: 32737537 PMCID: PMC7889576 DOI: 10.1007/s00262-020-02681-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a highly malignant epithelial cancer linked to Epstein-Barr virus (EBV) infection. Tumors are characterized by a lymphomononuclear infiltrate and the number of natural killer (NK) cells in tumors appears to be of prognostic significance. Standard treatment for NPC in adolescents and young adults consists of induction chemotherapy followed by radiochemotherapy. Though survival rates are above 80%, the majority of patients suffer from long-term side-effects, mainly related to radiotherapy. The addition of immunotherapy to induction chemotherapy could improve tumor response. METHODS We have investigated the killing of NPC cells by NK cells in the context of chemotherapy, using a panel of three nasopharyngeal carcinoma cell lines and a patient-derived xenograft. Cytotoxicity was measured using the calcein-release assay, while the contribution of different checkpoints and signaling pathways to killing was studied by siRNA-mediated gene silencing and chemical inhibitors. RESULTS Chemotherapeutics cisplatin, 5-fluorouracil and gemcitabine sensitized NPC cells to killing by NK cells. Chemotherapeutics led to upregulation of PD-1 in NK cells and PD-L1 in NPC cells via NF-κB. Inhibition of the PD-L1/PD-1 checkpoint by an anti-PD-1 antibody or siRNA increased NK-cell cytotoxicity towards NPC cells. CONCLUSION The addition of an anti-PD-1 antibody to chemotherapy in patients with NPC could increase the efficacy of induction chemotherapy. If confirmed in a clinical trial, more efficient induction therapy could allow the dose of radiotherapy to be reduced and thereby diminish severe late effects of such therapy.
Collapse
Affiliation(s)
- Anna Makowska
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, Rhenish-Westphalian Technical University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Selina Meier
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, Rhenish-Westphalian Technical University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Lian Shen
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, Rhenish-Westphalian Technical University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Pierre Busson
- CNRS UMR 8126, Gustave Roussy and Université Paris-Sud/Paris-Saclay, Villejuif, France
| | - Valentin Baloche
- CNRS UMR 8126, Gustave Roussy and Université Paris-Sud/Paris-Saclay, Villejuif, France
| | - Udo Kontny
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, Rhenish-Westphalian Technical University, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
18
|
Arianfar E, Shahgordi S, Memarian A. Natural Killer Cell Defects in Breast Cancer: A Key Pathway for Tumor Evasion. Int Rev Immunol 2020; 40:197-216. [PMID: 33258393 DOI: 10.1080/08830185.2020.1845670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
As the most important innate immune component cancers invader, natural killer (NK) cells have a magnificent role in antitumor immunity without any prior sensitization. Different subsets of NK cells have distinct responses during tumor cell exposure, according to their phenotypes and environments. Their function is induced mainly by the activity of both inhibitory and activating receptors against cancerous cells. Since the immunosuppression in the tumor microenvironment of breast cancer patients has directly deteriorated the phenotype and disturbed the function of NK cells, recruiting compensatory mechanisms indicate promising outcomes for immunotherapeutic approaches. These evidences accentuate the importance of NK cell distinct features in protection against breast tumors. In this review, we discuss the several mechanisms involved in NK cells suppression which consequently promote tumor progression and disease recurrence in patients with breast cancer.
Collapse
Affiliation(s)
- Elaheh Arianfar
- Student Research Committee, Faculty of Medicine, Department of Immunology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sanaz Shahgordi
- Student Research Committee, Faculty of Medicine, Department of Immunology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Memarian
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran.,Immunology department, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
19
|
Kim R, Kawai A, Wakisaka M, Sawada S, Shimoyama M, Yasuda N, Hidaka M, Morita Y, Ohtani S, Ito M, Kawasaki K, Kin T, Arihiro K. Immune factors associated with the pathological and therapeutic effects of preoperative chemotherapy in patients with breast cancer. Transl Oncol 2020; 14:100927. [PMID: 33157515 PMCID: PMC7649526 DOI: 10.1016/j.tranon.2020.100927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022] Open
Abstract
The immune factors that determine the pathological and therapeutic effects of preoperative chemotherapy in patients with breast cancer are associated with local and systemic immune responses in the presence of tumor-infiltrating lymphocytes, in collaboration with downregulation of immunosuppressive factors mediated by vascular endothelial growth factor (VEGF) and cytotoxic T lymphocyte antigen 4 (CTLA-4) in regulatory T cells (Tregs) in the tumor microenvironment. Multivariate analysis showed that grade 2 and better therapeutic effects tended to be associated with higher natural killer cell levels after preoperative chemotherapy (odds ratio = 1.02; 95% confidence interval, 0.99–1.05; p = 0.07). Therapy targeting VEGF and CTLA-4 in Tregs to overcome tumor-derived immunosuppression may enhance the pathological and therapeutic responses to preoperative chemotherapy in patients with breast cancer.
Immune activation plays an important role in achieving the pathological and therapeutic effects of preoperative chemotherapy in patients with breast cancer. We evaluated how the immune response contributes to various therapeutic effects. This study was conducted on 43 patients with stages II–IV breast cancer who received preoperative chemotherapy followed by surgery. Peripheral natural killer (pNK) cell activity and the neutrophil-lymphocyte ratio, lymphocyte-monocyte ratio, and platelet-lymphocyte ratio (PLR) were assessed before and after chemotherapy. Tumor-infiltrating lymphocytes (TILs) and levels of 14 tumor microenvironmental factors, analyzed by next-generation sequencing, were assessed in formalin-fixed, paraffin-embedded sections of preoperative biopsy samples and surgical specimens. Univariate analysis showed that grade 2 (G2) and better therapeutic effects were significantly associated with human epidermal growth factor receptor 2 (HER-2)-positive cancer, lower PLRs, and higher NK cell and interleukin-6 levels after chemotherapy. The disappearance of axillary lymph-node metastasis was significantly associated with HER-2-positive cancer; increased pNK cell activity and lower PLRs and vascular endothelial growth factor (VEGF) levels after chemotherapy; and increased cytotoxic T lymphocyte antigen 4 (CTLA-4) levels in regulatory T cells (Tregs) and ≥5% TILs before chemotherapy. Multivariate analysis showed that G2 and better therapeutic effects tended to be associated with higher NK cell levels after chemotherapy (odds ratio = 1.02; 95% confidence interval, 0.99–1.05; P = 0.07). The activation of local and systemic immune responses by downregulation of immunosuppressive factors, such as VEGF and CTLA-4 in Tregs, had variable pathological and therapeutic effects after preoperative chemotherapy in patients with breast cancer.
Collapse
Affiliation(s)
- Ryungsa Kim
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima, Japan.
| | - Ami Kawai
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima, Japan
| | - Megumi Wakisaka
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima, Japan
| | - Sayaka Sawada
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima, Japan
| | - Mika Shimoyama
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima, Japan
| | - Naomi Yasuda
- Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima, Japan
| | | | | | - Shoichiro Ohtani
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Mitsuya Ito
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Kensuke Kawasaki
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Takanori Kin
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
20
|
Henriksen JR, Nederby L, Donskov F, Waldstrøm M, Adimi P, Jakobsen A, Dahl Steffensen K. Blood natural killer cells during treatment in recurrent ovarian cancer. Acta Oncol 2020; 59:1365-1373. [PMID: 32692270 DOI: 10.1080/0284186x.2020.1791358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Recent research indicated favorable prognostic impact of intratumoral natural killer (NK) cells in ovarian carcinoma (OC). The role of NK cells during chemotherapy in OC is unknown. We investigated impact of NK cells in OC patients treated with palliative chemotherapy. METHODS Participants receiving palliative chemotherapy for recurrent OC (N = 72) had prospectively blood samples at baseline and before cycle 2. NK cell counts were quantified by flow cytometry. NK cell activity was measured by the NK Vue® assay, estimating interferon-gamma production. Overall survival (OS) was the primary endpoint. Cutoffs were predefined, NK numbers (≥184 × 106 cells/L vs. <184 × 106 cells/L) and NK activity (<200 pg/mL vs. ≥200 pg/mL). RESULTS Median OS in patients with low vs. high NK cell count at baseline was 7.1 months vs. 15.6 months (p = .028), respectively, and before cycle 2 was 5.7 vs. 17.3 months, p < .001, respectively. The difference in restricted mean survival (ΔRMST) was 5.7 months (95% CI: 3.3-8.0) at cycle 2 vs. 2.5 months (95% CI: -0.6 to 5.6) at baseline, showing a significant difference with no overlap of confidence intervals. In multivariate analyses, low NK cell count remained significant with a hazard ratio (HR)=2.83, 95% CI: 1.53-5.22, p = .001 (baseline) and HR = 3.34, 95% CI: 1.67-6.71, p = .001 (before cycle 2). Patients with both low NK count and NK activity at baseline (N = 20) had median OS 6.5 months vs. 11.5 months in patients with either high activity, high count or both (p = .007). In parallel, patients with both low NK activity and count at cycle 2 (N = 18) had a median survival of 4.0 months vs. 15.4 months (p < .001). CONCLUSIONS A low blood NK cell count in recurrent metastatic ovarian cancer during chemotherapy is associated with unfavorable prognostic impact. Early increase in survival difference based on NK cell status suggests an association between NK cell count and treatment benefit.
Collapse
Affiliation(s)
- Jon Røikjær Henriksen
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Line Nederby
- Department of Biochemistry and Immunology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Marianne Waldstrøm
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
| | - Parvin Adimi
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Karina Dahl Steffensen
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
21
|
Morgan MM, Schuler LA, Ciciliano JC, Johnson BP, Alarid ET, Beebe DJ. Modeling chemical effects on breast cancer: the importance of the microenvironment in vitro. Integr Biol (Camb) 2020; 12:21-33. [PMID: 32118264 PMCID: PMC7060306 DOI: 10.1093/intbio/zyaa002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/18/2019] [Accepted: 02/01/2020] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that our ability to predict chemical effects on breast cancer is limited by a lack of physiologically relevant in vitro models; the typical in vitro breast cancer model consists of the cancer cell and excludes the mammary microenvironment. As the effects of the microenvironment on cancer cell behavior becomes more understood, researchers have called for the integration of the microenvironment into in vitro chemical testing systems. However, given the complexity of the microenvironment and the variety of platforms to choose from, identifying the essential parameters to include in a chemical testing platform is challenging. This review discusses the need for more complex in vitro breast cancer models and outlines different approaches used to model breast cancer in vitro. We provide examples of the microenvironment modulating breast cancer cell responses to chemicals and discuss strategies to help pinpoint what components should be included in a model.
Collapse
Affiliation(s)
- Molly M Morgan
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jordan C Ciciliano
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Elaine T Alarid
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
22
|
Badr N, Berditchevski F, Shaaban A. The Immune Microenvironment in Breast Carcinoma: Predictive and Prognostic Role in the Neoadjuvant Setting. Pathobiology 2019; 87:61-74. [DOI: 10.1159/000504055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/09/2019] [Indexed: 11/19/2022] Open
|
23
|
Kim R, Kawai A, Wakisaka M, Sawada S, Shimoyama M, Yasuda N, Hidaka M, Morita Y, Ohtani S, Arihiro K. Immune correlates of the differing pathological and therapeutic effects of neoadjuvant chemotherapy in breast cancer. Eur J Surg Oncol 2019; 46:77-84. [PMID: 31563296 DOI: 10.1016/j.ejso.2019.09.146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/26/2019] [Accepted: 09/17/2019] [Indexed: 01/21/2023] Open
Abstract
PURPOSE To evaluate immune responses paralleling the pathological and therapeutic effects of neoadjuvant chemotherapy (NAC) in the tumor microenvironment of breast cancer. PATIENTS AND METHODS 38 patients with stages II and III breast cancer received NAC followed by surgery in 2012-2018. Peripheral natural killer (pNK) cell activity, tumor-infiltrating lymphocytes (TILs), and levels of tumor microenvironmental factors were assessed before and after NAC. RESULTS In univariate analysis, grade 2 (G2) and better therapeutic effects were significantly associated with high post-NAC levels of NK cells and interleukin-6, and tended to be associated with higher CD4, CD8 and CTLA-4 transcripts. Disappearance of axillary lymph node metastasis (Ax+) was significantly associated with 1) increased NK and pNK levels, 2) decreased vascular endothelial growth factor (VEGF) transcripts after NAC, 3) the presence of ≥5% TILs, and tended to be associated with higher CTLA-4 levels before NAC. Multivariate analysis showed that G2 and better therapeutic effects were significantly associated with higher NK levels after NAC (OR = 1.07, 95% CI 1.00-1.14; p = 0.0255), and that disappearance of Ax+ was significantly associated with the presence of ≥5% pre-NAC TILs (OR = 19.87, 95% CI 2.24-175.80; p = 0.0072). CONCLUSIONS Increased NK cells after NAC, together with increased CD4+ and CD8+ T-cells, and decreased CTLA-4+ T cells and VEGF correlate with beneficial therapeutic effects. Systemic activation of pNK cell activity and the presence of pre-NAC TILs may improve the elimination of Ax + together with decreased immunosuppression by VEGF in tumors.
Collapse
Affiliation(s)
- Ryungsa Kim
- Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan.
| | - Ami Kawai
- Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | | | - Sayaka Sawada
- Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | | | - Naomi Yasuda
- Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | | | | | - Shoichiro Ohtani
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|