Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation
Abstract
:Simple Summary
Abstract
1. Introduction
2. Two Roles of TME in TNBC Immunomodulation
2.1. Immunosuppressive TME in TNBC
2.1.1. PD-1/PD-L1 Axis
2.1.2. Foxp3+ Tregs
2.1.3. M2 Macrophages
2.1.4. MDSCs
2.2. Immunoreactive TME in TNBC
2.2.1. NK Cells
2.2.2. CD8+ TILs
2.2.3. M1 Macrophages
Items | Clinical Significance | Involved Mechanisms | References |
---|---|---|---|
PD-1/PD-L1 | Paradoxical role in prognosis | microRNA-200/ZEB1 axis, WNT signaling, loss of PTEN, PI3K signaling, and MUC1-C/MYC/NF-κB pathway | [31,34,35,36,37,38,39,40,41,44,45,46] |
Foxp3+ Tregs | Paradoxical role in prognosis | Notch pathway, IL-35/STAT1/STAT3, secretion of anti-inflammatory mediators such as interleukin | [50,51,52,53,54,56,57,58,59,115] |
M2 macrophages | Adverse prognostic indicator | MAPK pathway, NF-κB/PD-L1, release of immunosuppressive chemokines, JAK2/STAT3 signaling pathway, S100A4 activation, angiogenic program, HIF-1α, STAT5, NF-κB and ERK signaling | [61,62,63,64,116] |
MDSCs | Risk factor for PD plus SD | ΔNp63-dependent activation of the chemokines CXCL2 and CCL22, Glycolysis, hypoxia, secretion of inflammatory cytokines, Rb1 activation | [73,74,75,76,81,82] |
NK cells | Positive prognostic indicator | ADCC, Lectin-like Transcript-1 activation, bispecific antibody (MesobsFab) modulating chemorepellent inside tumor cell clusters | [84,85,92,94,95,96,97,98,117] |
CD8+ TILs | Favorable prognostic indicator | Inhibition of PDGFRβ/MEK1/2/JAK2 signal pathway, distinct metasignatures of CD8+ TILs, DHX37/PDCD11/NF-κB | [17,99,100,101,108,109,110] |
M1 macrophages | Favorable prognostic indicator | M1 polarization by FAD, CoREST and exposure to cell vaccine (ICV), release of CCL5 | [112,113,114] |
3. The Composition of TME Contributes to TNBC Subtype Classification
Subtype of TNBC | Subtype Number | Basis of Classification | Clinical Significance | References |
---|---|---|---|---|
BL1, BL2, IM, M, MSL, LAR | 6 | Gene expression profiles | IM subtype was associated with favorable prognosis. | [118] |
4 stroma axes (T,B,E,D) | 4 | Transcriptome of stroma | Better evaluated patient benefit from therapeutics. | [119] |
C1, C2, C3 | 3 | Gene expression profiling | C2 and C3 subtypes were sensitive to drugs combating immunosuppression. | [120] |
LAR, IM, BLIS, MES | 4 | Clinical, genomic, and transcriptomic data | Elevated immune cells and signaling in IM subtype. | [121] |
BL, IM, M, MSL, LAR | 5 | Gene expression profiles | Interaction between IM and MSL subtype suggested involvement of TME. | [122] |
MC1, MC2, MC3, MC4, MC5, MC6 | 6 | Transcriptomic and genomic data | High level of CD8+ and CD4+ immune signatures in MC6 subtype. | [123] |
Immunity_H, Immunity_M, Immunity_L | 3 | Immunogenomic profiling | Immunity_H subtype was correlated with immune cell expression and good prognosis | [124] |
LAR, basal, claudin-low, claudin-high and two immune subtypes | 6 | Clinical and gene expression data | Claudin-h and immune-positive subtype was associated with low pCR and favorable prognosis separately. | [125] |
Epi-CL-A, Epi-CL-B, Epi-CLC, Epi-CL-D | 4 | Genome-wide DNA methylation profiles | Positive regulation of T lymphocyte cytotoxicity and associated genes in Epi-CL-D subtype. | [126] |
Immune phenotype A and B | 2 | Density of five prognosis-related immune cells | Enriched immune-related pathways and molecules in phenotype A. | [127] |
ImA, ImB and ImC | 3 | Immune-related gene expression signatures | Platelet to lymphocyte ratio (PLR) was associated with tumor immune infiltration in TNBC. | [128] |
4. Chemotherapy-Induced TME Remodeling Modulates TNBC Immune Response
4.1. Chemotherapy-Induced HMGB1 Release Participates in TNBC Immunomodulation
4.1.1. Chemotherapy-Induced HMGB1 Enhances Anti-Tumor Immune Response
4.1.2. HMGB1 Is Related to High Recurrence Risk and Progressive Disease after Neoadjuvant Chemotherapy
4.2. Chemotherapy-Induced Exosomes Secretion Interconnects TME and TNBC Immune Response
4.2.1. Chemotherapy-Induced Exosomes Are Released to TME
4.2.2. Exosomes Are Related to TNBC Tumor Progression and Provide Therapy Options
4.3. S1P/SPHK1/S1PR1 Link TME Changes to TNBC Immunomodulation
4.3.1. S1P/SPHK1/S1PR1 Is Associated with TME Changes
4.3.2. S1P/SPHK1/S1PR1 Is Associated with TNBC Tumor Progression
Items | Clinical Significance | Involved Mechanisms | References |
---|---|---|---|
HMGB1 | Predict recurrence risk of residual tumor after neoadjuvant chemotherapy | TLR4 signal pathway, immune molecules such as TGF-β, IK12p7, and IFN-γ, p38/NFκB/Erk1/2 pathway, RAGE/IRF3/NF-κB | [19,20,139,140,142,144,145,146,170] |
Exosome | pCR prediction and distinct prognosis value in different subtype of breast cancer | HMGB1/TLR4/NF-κB signaling | [150,152,171,172] |
S1P/SPHK1/S1PR1 | Paradoxical role in tumor progression of TNBC | PPARγ signal pathway, STAT3/IL-6, IL-22, TCR activation | [158,160,163,169,173,174,175] |
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Perou, C.M.; Sørlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
- Prat, A.; Adamo, B.; Cheang, M.C.; Anders, C.K.; Carey, L.A.; Perou, C.M. Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist 2013, 18, 123–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, Y.; Ma, D.; Zhao, S.; Suo, C.; Shi, J.; Xue, M.Z.; Ruan, M.; Wang, H.; Zhao, J.; Li, Q.; et al. Multi-Omics Profiling Reveals Distinct Microenvironment Characterization and Suggests Immune Escape Mechanisms of Triple-Negative Breast Cancer. Clin. Cancer Res. 2019, 25, 5002–5014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- Dieci, M.V.; Mathieu, M.C.; Guarneri, V.; Conte, P.; Delaloge, S.; Andre, F.; Goubar, A. Prognostic and predictive value of tumor-infiltrating lymphocytes in two phase III randomized adjuvant breast cancer trials. Ann. Oncol. 2015, 26, 1698–1704. [Google Scholar] [CrossRef] [PubMed]
- Callari, M.; Cappelletti, V.; D’Aiuto, F.; Musella, V.; Lembo, A.; Petel, F.; Karn, T.; Iwamoto, T.; Provero, P.; Daidone, M.G.; et al. Subtype-Specific Metagene-Based Prediction of Outcome after Neoadjuvant and Adjuvant Treatment in Breast Cancer. Clin. Cancer Res. 2016, 22, 337–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loi, S.; Sirtaine, N.; Piette, F.; Salgado, R.; Viale, G.; Van Eenoo, F.; Rouas, G.; Francis, P.; Crown, J.P.; Hitre, E.; et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J. Clin. Oncol. 2013, 31, 860–867. [Google Scholar] [CrossRef]
- 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]
- Gupta, S.; Roy, A.; Dwarakanath, B.S. Metabolic Cooperation and Competition in the Tumor Microenvironment: Implications for Therapy. Front. Oncol 2017, 7, 68. [Google Scholar] [CrossRef] [Green Version]
- Kim, I.S.; Zhang, X.H. One microenvironment does not fit all: Heterogeneity beyond cancer cells. Cancer Metastasis Rev. 2016, 35, 601–629. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Varn, F.S.; Mullins, D.W.; Arias-Pulido, H.; Fiering, S.; Cheng, C. Adaptive immunity programmes in breast cancer. Immunology 2017, 150, 25–34. [Google Scholar] [CrossRef] [Green Version]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
- Nguyen-Ngoc, K.V.; Cheung, K.J.; Brenot, A.; Shamir, E.R.; Gray, R.S.; Hines, W.C.; Yaswen, P.; Werb, Z.; Ewald, A.J. ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proc. Natl. Acad. Sci. USA 2012, 109, E2595–E2604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, W.; Lira, V.; Hudson, T.E.; Lemmens, E.E.; Hanson, W.G.; Flores, R.; Barajas, G.; Katibah, G.E.; Desbien, A.L.; Lauer, P.; et al. Recombinant Listeria promotes tumor rejection by CD8+ T cell-dependent remodeling of the tumor microenvironment. Proc. Natl. Acad. Sci. USA 2018, 115, 8179–8184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marra, A.; Viale, G.; Curigliano, G. Recent advances in triple negative breast cancer: The immunotherapy era. BMC Med. 2019, 17, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gruosso, T.; Gigoux, M.; VSK, M.; Bertos, N.; Zuo, D.; Perlitch, I.; SMI, S.; Zhao, H.; Souleimanova, M.; Johnson, R.M.; et al. Spatially distinct tumor immune microenvironments stratify triple-negative breast cancers. J. Clin. Investig. 2019, 129, 1785–1800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galluzzi, L.; Senovilla, L.; Zitvogel, L.; Kroemer, G. The secret ally: Immunostimulation by anticancer drugs. Nat. Rev. Drug Discov. 2012, 11, 215–233. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.Y.; Chiang, S.F.; Ke, T.W.; Chen, T.W.; Lan, Y.C.; You, Y.S.; Shiau, A.C.; Chen, W.T.; KSC, C. Cytosolic high-mobility group box protein 1 (HMGB1) and/or PD-1+ TILs in the tumor microenvironment may be contributing prognostic biomarkers for patients with locally advanced rectal cancer who have undergone neoadjuvant chemoradiotherapy. Cancer Immunol. Immunother. 2018, 67, 551–562. [Google Scholar] [CrossRef]
- Zhang, Y.; Yang, S.; Yang, Y.; Liu, T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect. Agents Cancer 2019, 14, 27. [Google Scholar] [CrossRef] [Green Version]
- Savas, P.; Salgado, R.; Denkert, C.; Sotiriou, C.; Darcy, P.K.; Smyth, M.J.; Loi, S. Clinical relevance of host immunity in breast cancer: From TILs to the clinic. Nat. Rev. Clin. Oncol. 2016, 13, 228–241. [Google Scholar] [CrossRef]
- Bense, R.D.; Sotiriou, C.; Piccart-Gebhart, M.J.; Haanen, J.B.A.G.; van Vugt, M.A.T.M.; de Vries, E.G.E.; Schröder, C.P.; Fehrmann, R.S.N. Relevance of Tumor-Infiltrating Immune Cell Composition and Functionality for Disease Outcome in Breast Cancer. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef] [Green Version]
- Sharpe, A.H.; Wherry, E.J.; Ahmed, R.; Freeman, G.J. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat. Immunol. 2007, 8, 239–245. [Google Scholar] [CrossRef]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
- Bertucci, F.; Finetti, P.; Birnbaum, D.; Mamessier, E. The PD1/PDL1 axis, a promising therapeutic target in aggressive breast cancers. Oncoimmunology 2016, 5, e1085148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGee, H.S.; Yagita, H.; Shao, Z.; Agrawal, D.K. Programmed Death-1 antibody blocks therapeutic effects of T-regulatory cells in cockroach antigen-induced allergic asthma. Am. J. Respir. Cell Mol. Biol. 2010, 43, 432–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, L.; Liu, F.; Li, R.; Li, Z.; Chen, X.; Zhou, Z.; Zhang, X.; Hu, T.; Zhang, Y.; Young, K.; et al. Role of programmed death ligands in effective T-cell interactions in extranodal natural killer/T-cell lymphoma. Oncol. Lett. 2014, 8, 1461–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Własiuk, P.; Putowski, M.; Giannopoulos, K. PD1/PD1L pathway, HLA-G and T regulatory cells as new markers of immunosuppression in cancers. Postepy Hig. Med. Dosw. 2016, 70, 1044–1058. [Google Scholar] [CrossRef] [PubMed]
- Brockwell, N.K.; Owen, K.L.; Zanker, D.; Spurling, A.; Rautela, J.; Duivenvoorden, H.M.; Baschuk, N.; Caramia, F.; Loi, S.; Darcy, P.K.; et al. Neoadjuvant Interferons: Critical for Effective PD-1-Based Immunotherapy in TNBC. Cancer Immunol. Res. 2017, 5, 871–884. [Google Scholar] [CrossRef] [Green Version]
- Mittendorf, E.A.; Philips, A.V.; Meric-Bernstam, F.; Qiao, N.; Wu, Y.; Harrington, S.; Su, X.; Wang, Y.; Gonzalez-Angulo, A.M.; Akcakanat, A.; et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol. Res. 2014, 2, 361–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adams, S.; Schmid, P.; Rugo, H.S.; Winer, E.P.; Loirat, D.; Awada, A.; Cescon, D.W.; Iwata, H.; Campone, M.; Nanda, R.; et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: Cohort A of the phase II KEYNOTE-086 study. Ann. Oncol. 2019, 30, 397–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.A.; Shaw, W.G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef] [PubMed]
- Baptista, M.Z.; Sarian, L.O.; Derchain, S.F.; Pinto, G.A.; Vassallo, J. Prognostic significance of PD-L1 and PD-L2 in breast cancer. Hum. Pathol. 2016, 47, 78–84. [Google Scholar] [CrossRef]
- Muenst, S.; Schaerli, A.R.; Gao, F.; Däster, S.; Trella, E.; Droeser, R.A.; Muraro, M.G.; Zajac, P.; Zanetti, R.; Gillanders, W.E.; et al. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res. Treat. 2014, 146, 15–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, M.; Li, A.; Zhou, S.; Xu, Y.; Xiao, Y.; Bi, R.; Yang, W. Heterogeneity of PD-L1 expression in primary tumors and paired lymph node metastases of triple negative breast cancer. BMC Cancer 2018, 18, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, W.Y.; Lee, Y.K.; Koo, J.S. Expression of PD-L1 in triple-negative breast cancer based on different immunohistochemical antibodies. J. Transl. Med. 2016, 14, 173. [Google Scholar] [CrossRef] [Green Version]
- Zhu, X.; Zhang, Q.; Wang, D.; Liu, C.; Han, B.; Yang, J.M. Expression of PD-L1 Attenuates the Positive Impacts of High-level Tumor-infiltrating Lymphocytes on Prognosis of Triple-negative Breast Cancer. Cancer Biol. Ther. 2019, 20, 1105–1112. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.H.; Chang, J.S.; Koo, J.S.; Park, J.W.; Sohn, J.H.; Keum, K.C.; Suh, C.O.; Kim, Y.B. Differential Prognostic Impact of Strong PD-L1 Expression and 18F-FDG Uptake in Triple-negative Breast Cancer. Am. J. Clin. Oncol. 2018, 41, 1049–1057. [Google Scholar] [CrossRef]
- AiErken, N.; Shi, H.J.; Zhou, Y.; Shao, N.; Zhang, J.; Shi, Y.; Yuan, Z.Y.; Lin, Y. High PD-L1 Expression Is Closely Associated with Tumor-Infiltrating Lymphocytes and Leads to Good Clinical Outcomes in Chinese Triple Negative Breast Cancer Patients. Int. J. Biol. Sci. 2017, 13, 1172–1179. [Google Scholar] [CrossRef] [Green Version]
- Botti, G.; Collina, F.; Scognamiglio, G.; Rao, F.; Peluso, V.; De Cecio, R.; Piezzo, M.; Landi, G.; De Laurentiis, M.; Cantile, M.; et al. Programmed Death Ligand 1 (PD-L1) Tumor Expression Is Associated with a Better Prognosis and Diabetic Disease in Triple Negative Breast Cancer Patients. Int. J. Mol. Sci. 2017, 18, 459. [Google Scholar] [CrossRef]
- Schmid, P.; Rugo, H.S.; Adams, S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Henschel, V.; Molinero, L.; Chui, S.Y.; et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): Updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 44–59. [Google Scholar] [CrossRef]
- Nanda, R.; Chow, L.Q.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat. Commun. 2014, 5, 5241. [Google Scholar] [CrossRef] [PubMed]
- Castagnoli, L.; Cancila, V.; Cordoba-Romero, S.L.; Faraci, S.; Talarico, G.; Belmonte, B.; Iorio, M.V.; Milani, M.; Volpari, T.; Chiodoni, C.; et al. WNT signaling modulates PD-L1 expression in the stem cell compartment of triple-negative breast cancer. Oncogene 2019, 38, 4047–4060. [Google Scholar] [CrossRef] [Green Version]
- Maeda, T.; Hiraki, M.; Jin, C.; Rajabi, H.; Tagde, A.; Alam, M.; Bouillez, A.; Hu, X.; Suzuki, Y.; Miyo, M.; et al. MUC1-C Induces PD-L1 and Immune Evasion in Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 205–215. [Google Scholar] [CrossRef] [Green Version]
- Voorwerk, L.; Slagter, M.; Horlings, H.M.; Sikorska, K.; van de Vijver, K.K.; de Maaker, M.; Nederlof, I.; Kiuin, R.J.C.; Warren, S.; Ong, S.; et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: The TONIC trial. Nat. Med. 2019, 25, 920–928. [Google Scholar] [CrossRef]
- Plitas, G.; Konopacki, C.; Wu, K.; Bos, P.D.; Morrow, M.; Putintseva, E.V.; Chudakov, D.M.; Rudensky, A.Y. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity 2016, 45, 1122–1134. [Google Scholar] [CrossRef] [Green Version]
- Hashemi, V.; Maleki, L.A.; Esmaily, M.; Masjedi, A.; Ghalamfarsa, G.; Namdar, A.; Yousefi, M.; Yousefi, B.; Jadidi-Niaragh, F. Regulatory T cells in breast cancer as a potent anti-cancer therapeutic target. Int. Immunopharmacol. 2020, 78, 106087. [Google Scholar] [CrossRef]
- Taylor, N.A.; Vick, S.C.; Iglesia, M.D.; Brickey, W.J.; Midkiff, B.R.; McKinnon, K.P.; Reisdorf, S.; Anders, C.K.; Carey, L.A.; Parker, J.S.; et al. Treg depletion potentiates checkpoint inhibition in claudin-low breast cancer. J. Clin. Investig. 2017, 127, 3472–3483. [Google Scholar] [CrossRef] [PubMed]
- Jamiyan, T.; Kuroda, H.; Yamaguchi, R.; Nakazato, Y.; Noda, S.; Onozaki, M.; Abe, A.; Hayashi, M. Prognostic impact of a tumor-infiltrating lymphocyte subtype in triple negative cancer of the breast. Breast Cancer 2020, 27, 880–892. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Wang, X.I.; Ding, J.; Sun, Q.; Zhang, S. The predictive and prognostic value of Foxp3+/CD25+ regulatory T cells and PD-L1 expression in triple negative breast cancer. Ann. Diagn. Pathol. 2019, 40, 143–151. [Google Scholar] [CrossRef]
- Yeong, J.; Thike, A.A.; Lim, J.C.; Lee, B.; Li, H.; Wong, S.C.; Hue, S.S.; Tan, P.H.; Iqbal, J. Higher densities of Foxp3+ regulatory T cells are associated with better prognosis in triple-negative breast cancer. Breast Cancer Res. Treat. 2017, 163, 21–35. [Google Scholar] [CrossRef]
- Tsiatas, M.; Kalogeras, K.T.; Manousou, K.; Wirtz, R.M.; Gogas, H.; Veltrup, E.; Zagouri, F.; Lazaridis, G.; Koutras, A.; Christodoulou, C.; et al. Evaluation of the prognostic value of CD3, CD8, and FOXP3 mRNA expression in early-stage breast cancer patients treated with anthracycline-based adjuvant chemotherapy. Cancer Med. 2018, 7, 5066–5082. [Google Scholar] [CrossRef] [PubMed]
- Ge, Y.; Böhm, H.H.; Rathinasamy, A.; Xydia, M.; Hu, X.; Pincha, M.; Umansky, L.; Breyer, C.; Hillier, M.; Bonertz, A.; et al. Tumor-Specific Regulatory T Cells from the Bone Marrow Orchestrate Antitumor Immunity in Breast Cancer. Cancer Immunol. Res. 2019, 7, 1998–2012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ortiz-Martínez, F.; Gutiérrez-Aviñó, F.J.; Sanmartín, E.; Pomares-Navarro, E.; Villalba-Riquelme, C.; García-Martínez, A.; Lerma, E.; Peiró, G. Association of Notch pathway down-regulation with Triple Negative/Basal-like breast carcinomas and high tumor-infiltrating FOXP3+ Tregs. Exp. Mol. Pathol. 2016, 100, 460–468. [Google Scholar] [CrossRef] [PubMed]
- Martinez, L.M.; Robila, V.; Clark, N.M.; Du, W.; Idowu, M.O.; Rutkowski, M.R.; Bos, P.D. Regulatory T Cells Control the Switch From in situ to Invasive Breast Cancer. Front. Immunol. 2019, 10, 1942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watanabe, M.A.; Oda, J.M.; Amarante, M.K.; Cesar, V.J. Regulatory T cells and breast cancer: Implications for immunopathogenesis. Cancer Metastasis Rev. 2010, 29, 569–579. [Google Scholar] [CrossRef] [PubMed]
- Hao, S.; Chen, X.; Wang, F.; Shao, Q.; Liu, J.; Zhao, H.; Yuan, C.; Ren, H.; Mao, H. Breast cancer cell-derived IL-35 promotes tumor progression via induction of IL-35-producing induced regulatory T cells. Carcinogenesis 2018, 39, 1488–1496. [Google Scholar] [CrossRef] [Green Version]
- Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, J.; Wang, X.H.; Zhao, Y.X.; Chen, C.; Xu, X.Y.; Sun, Q.; Wu, H.Y.; Chen, M.; Sang, J.F.; Su, L.; et al. Cancer-Associated Fibroblasts Correlate with Tumor-Associated Macrophages Infiltration and Lymphatic Metastasis in Triple Negative Breast Cancer Patients. J. Cancer 2018, 9, 4635–4641. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.J.; Wang, X.H.; Gao, S.T.; Chen, C.; Xu, X.Y.; Sun, Q.; Zhou, Z.H.; Wu, G.Z.; Yu, Q.; Xu, G.; et al. Tumor-associated macrophages correlate with phenomenon of epithelial-mesenchymal transition and contribute to poor prognosis in triple-negative breast cancer patients. J. Surg. Res. 2018, 222, 93–101. [Google Scholar] [CrossRef]
- Yuan, Z.Y.; Luo, R.Z.; Peng, R.J.; Wang, S.S.; Xue, C. High infiltration of tumor-associated macrophages in triple-negative breast cancer is associated with a higher risk of distant metastasis. Onco Targets Ther 2014, 7, 1475–1480. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Q.; Le, K.; Xu, M.; Zhou, J.; Xiao, Y.; Yang, W.; Jiang, Y.; Xi, Z.; Huang, T. Combined MEK inhibition and tumor-associated macrophages depletion suppresses tumor growth in a triple-negative breast cancer mouse model. Int. Immunopharmacol. 2019, 76, 105864. [Google Scholar] [CrossRef]
- Roux, C.; Jafari, S.M.; Shinde, R.; Duncan, G.; Cescon, D.W.; Silvester, J.; Chu, M.F.; Hodgson, K.; Berger, T.; Wakeham, A.; et al. Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1. Proc. Natl. Acad. Sci. USA 2019, 116, 4326–4335. [Google Scholar] [CrossRef] [Green Version]
- Jing, W.; Guo, X.; Wang, G.; Bi, Y.; Han, L.; Zhu, Q.; Qiu, C.; Tanaka, M.; Zhao, Y. Breast cancer cells promote CD169+ macrophage-associated immunosuppression through JAK2-mediated PD-L1 upregulation on macrophages. Int. Immunopharmacol. 2020, 78, 106012. [Google Scholar] [CrossRef] [PubMed]
- Prasmickaite, L.; Tenstad, E.M.; Pettersen, S.; Jabeen, S.; Egeland, E.V.; Nord, S.; Pandya, A.; Haugen, M.H.; Kristensen, V.N.; Børresen-Dale, A.L.; et al. Basal-like breast cancer engages tumor-supportive macrophages via secreted factors induced by extracellular S100A4. Mol. Oncol. 2018, 12, 1540–1558. [Google Scholar] [CrossRef] [Green Version]
- Lu, X.; Yang, R.; Zhang, L.; Xi, Y.; Zhao, J.; Wang, F.; Zhang, H.; Li, Z. Macrophage Colony-stimulating Factor Mediates the Recruitment of Macrophages in Triple negative Breast Cancer. Int. J. Biol. Sci. 2019, 15, 2859–2871. [Google Scholar] [CrossRef] [Green Version]
- Donzelli, S.; Milano, E.; Pruszko, M.; Sacconi, A.; Masciarelli, S.; Iosue, I.; Melucci, E.; Gallo, E.; Terrenato, I.; Mottolese, M.; et al. Expression of ID4 protein in breast cancer cells induces reprogramming of tumour-associated macrophages. Breast Cancer Res. 2018, 20, 59. [Google Scholar] [CrossRef] [PubMed]
- Donzelli, S.; Sacconi, A.; Turco, C.; Gallo, E.; Milano, E.; Iosue, I.; Blandino, G.; Fazi, F.; Fontemaggi, G. Paracrine Signaling from Breast Cancer Cells Causes Activation of ID4 Expression in Tumor-Associated Macrophages. Cells 2020, 9, 418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sami, E.; Paul, B.T.; Koziol, J.A.; ElShamy, W.M. The Immunosuppressive Microenvironment in BRCA1-IRIS-Overexpressing TNBC Tumors Is Induced by Bidirectional Interaction with Tumor-Associated Macrophages. Cancer Res. 2020, 80, 1102–1117. [Google Scholar] [CrossRef] [Green Version]
- Hamilton, M.J.; Banáth, J.P.; Lam, V.; Lepard, N.E.; Krystal, G.; Bennewith, K.L. Serum inhibits the immunosuppressive function of myeloid-derived suppressor cells isolated from 4T1 tumor-bearing mice. Cancer Immunol. Immunother. 2012, 61, 643–654. [Google Scholar] [CrossRef]
- Li, F.; Zhao, Y.; Wei, L.; Li, S.; Liu, J. Tumor-infiltrating Treg, MDSC, and IDO expression associated with outcomes of neoadjuvant chemotherapy of breast cancer. Cancer Biol. Ther. 2018, 19, 695–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, S.; Wilkes, D.W.; Samuel, N.; Blanco, M.A.; Nayak, A.; Alicea-Torres, K.; Gluck, C.; Sinha, S.; Gabrilovich, D.; Chakrabarti, R. ΔNp63-driven recruitment of myeloid-derived suppressor cells promotes metastasis in triple-negative breast cancer. J. Clin. Investig. 2018, 128, 5095–5109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, W.; Tanikawa, T.; Kryczek, I.; Xia, H.; Li, G.; Wu, K.; Wei, S.; Zhao, L.; Vatan, L.; Wen, B.; et al. Aerobic Glycolysis Controls Myeloid-Derived Suppressor Cells and Tumor Immunity via a Specific CEBPB Isoform in Triple-Negative Breast Cancer. Cell Metab. 2018, 28, 87–103.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qian, X.; Zhang, Q.; Shao, N.; Shan, Z.; Cheang, T.; Zhang, Z.; Su, Q.; Wang, S.; Lin, Y. Respiratory hyperoxia reverses immunosuppression by regulating myeloid-derived suppressor cells and PD-L1 expression in a triple-negative breast cancer mouse model. Am. J. Cancer Res. 2019, 9, 529–545. [Google Scholar] [PubMed]
- Dominguez, C.; McCampbell, K.K.; David, J.M.; Palena, C. Neutralization of IL-8 decreases tumor PMN-MDSCs and reduces mesenchymalization of claudin-low triple-negative breast cancer. JCI Insight 2017, 2, e94296. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Zhang, X.; Yang, W.; Dou, D.; Zhang, H.; Tang, Y.; Zhong, W.; Meng, J.; Bai, Y.; Liu, Y.; et al. Prim-O-glucosylcimifugin enhances the antitumour effect of PD-1 inhibition by targeting myeloid-derived suppressor cells. J. Immunother. Cancer 2019, 7, 231. [Google Scholar] [CrossRef]
- Piranlioglu, R.; Lee, E.; Ouzounova, M.; Bollag, R.J.; Vinyard, A.H.; Arbab, A.S.; Marasco, D.; Guzel, M.; Cowell, J.K.; Thangaraju, M.; et al. Primary tumor-induced immunity eradicates disseminated tumor cells in syngeneic mouse model. Nat. Commun. 2019, 10, 1430. [Google Scholar] [CrossRef] [Green Version]
- Wesolowski, R.; Duggan, M.C.; Stiff, A.; Markowitz, J.; Trikha, P.; Levine, K.M.; Schoenfield, L.; Abdel-Rasoul, M.; Layman, R.; Ramaswamy, B.; et al. Circulating myeloid-derived suppressor cells increase in patients undergoing neo-adjuvant chemotherapy for breast cancer. Cancer Immunol. Immunother. 2017, 66, 1437–1447. [Google Scholar] [CrossRef]
- Ban, Y.; Mai, J.; Li, X.; Mitchell-Flack, M.; Zhang, T.; Zhang, L.; Chouchane, L.; Ferrari, M.; Shen, H.; Ma, X. Targeting Autocrine CCL5-CCR5 Axis Reprograms Immunosuppressive Myeloid Cells and Reinvigorates Antitumor Immunity. Cancer Res. 2017, 77, 2857–2868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Lv, D.; Kim, H.J.; Kurt, R.A.; Bu, W.; Li, Y.; Ma, X. A novel role of hematopoietic CCL5 in promoting triple-negative mammary tumor progression by regulating generation of myeloid-derived suppressor cells. Cell Res. 2013, 23, 394–408. [Google Scholar] [CrossRef] [PubMed]
- Levy, E.M.; Roberti, M.P.; Mordoh, J. Natural killer cells in human cancer: From biological functions to clinical applications. J. Biomed. Biotechnol. 2011, 2011, 676198. [Google Scholar] [CrossRef] [PubMed]
- Tian, W.; Wang, L.; Yuan, L.; Duan, W.; Zhao, W.; Wang, S.; Zhang, Q. A prognostic risk model for patients with triple negative breast cancer based on stromal natural killer cells, tumor-associated macrophages and growth-arrest specific protein 6. Cancer Sci. 2016, 107, 882–889. [Google Scholar] [CrossRef] [PubMed]
- Park, I.H.; Yang, H.N.; Lee, K.J.; Kim, T.S.; Lee, E.S.; Jung, S.Y.; Kwon, Y.; Kong, S.Y. Tumor-derived IL-18 induces PD-1 expression on immunosuppressive NK cells in triple-negative breast cancer. Oncotarget 2017, 8, 32722–32730. [Google Scholar] [CrossRef] [Green Version]
- Foulds, G.A.; Vadakekolathu, J.; TMA, A.; Nagarajan, D.; Reeder, S.; Johnson, C.; Hood, S.; Moseley, P.M.; SYT, C.; Pockley, A.G.; et al. Immune-Phenotyping and Transcriptomic Profiling of Peripheral Blood Mononuclear Cells from Patients with Breast Cancer: Identification of a 3 Gene Signature Which Predicts Relapse of Triple Negative Breast Cancer. Front. Immunol. 2018, 9, 2028. [Google Scholar] [CrossRef] [PubMed]
- Fregni, G.; Perier, A.; Avril, M.F.; Caignard, A. NK cells sense tumors, course of disease and treatments: Consequences for NK-based therapies. Oncoimmunology 2012, 1, 38–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choucair, K.; Duff, J.R.; Cassidy, C.S.; Albrethsen, M.T.; Kelso, J.D.; Lenhard, A.; Staats, H.; Patel, R.; Brunicardi, F.C.; Dworkin, L.; et al. Natural killer cells: A review of biology, therapeutic potential and challenges in treatment of solid tumors. Future Oncol. 2019, 15, 3053–3069. [Google Scholar] [CrossRef]
- Habif, G.; Crinier, A.; André, P.; Vivier, E.; Narni-Mancinelli, E. Targeting natural killer cells in solid tumors. Cell. Mol. Immunol. 2019, 16, 415–422. [Google Scholar] [CrossRef]
- Stojanovic, A.; Cerwenka, A. Natural killer cells and solid tumors. J. Innate Immun. 2011, 3, 355–364. [Google Scholar] [CrossRef]
- Shenouda, M.M.; Gillgrass, A.; Nham, T.; Hogg, R.; Lee, A.J.; Chew, M.V.; Shafaei, M.; Aarts, C.; Lee, D.A.; Hassell, J.; et al. Ex vivo expanded natural killer cells from breast cancer patients and healthy donors are highly cytotoxic against breast cancer cell lines and patient-derived tumours. Breast Cancer Res. 2017, 19, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nham, T.; Poznanski, S.M.; Fan, I.Y.; Vahedi, F.; Shenouda, M.M.; Lee, A.J.; Chew, M.V.; Hogg, R.T.; Lee, D.A.; Ashkar, A.A. Ex Vivo-expanded Natural Killer Cells Derived From Long-term Cryopreserved Cord Blood are Cytotoxic Against Primary Breast Cancer Cells. J. Immunother. 2018, 41, 64–72. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Ran, R.; Shao, B.; Rugo, H.S.; Yang, Y.; Hu, Z.; Wei, Z.; Wan, F.; Kong, W.; Song, G.; et al. Combined peripheral natural killer cell and circulating tumor cell enumeration enhance prognostic efficiency in patients with metastatic triple-negative breast cancer. Chin. J. Cancer Res. 2018, 30, 315–326. [Google Scholar] [CrossRef] [PubMed]
- Hu, Z. Tissue factor as a new target for CAR-NK cell immunotherapy of triple-negative breast cancer. Sci. Rep. 2020, 10, 2815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Juliá, E.P.; Amante, A.; Pampena, M.B.; Mordoh, J.; Levy, E.M. Avelumab, an IgG1 anti-PD-L1 Immune Checkpoint Inhibitor, Triggers NK Cell-Mediated Cytotoxicity and Cytokine Production Against Triple Negative Breast Cancer Cells. Front. Immunol. 2018, 9, 2140. [Google Scholar] [CrossRef]
- Roberti, M.P.; Juliá, E.P.; Rocca, Y.S.; Amat, M.; Bravo, A.I.; Loza, J.; Coló, F.; Loza, C.M.; Fabiano, V.; Maino, M.; et al. Overexpression of CD85j in TNBC patients inhibits Cetuximab-mediated NK-cell ADCC but can be restored with CD85j functional blockade. Eur. J. Immunol. 2015, 45, 1560–1569. [Google Scholar] [CrossRef]
- Del, B.J.; Florès-Florès, R.; Josselin, E.; Goubard, A.; Ganier, L.; Castellano, R.; Chames, P.; Baty, D.; Kerfelec, B. A Bispecific Antibody-Based Approach for Targeting Mesothelin in Triple Negative Breast Cancer. Front. Immunol. 2019, 10, 1593. [Google Scholar]
- Marrufo, A.M.; Mathew, S.O.; Chaudhary, P.; Malaer, J.D.; Vishwanatha, J.K.; Mathew, P.A. Blocking LLT1 (CLEC2D, OCIL)-NKRP1A (CD161) interaction enhances natural killer cell-mediated lysis of triple-negative breast cancer cells. Am. J. Cancer Res. 2018, 8, 1050–1063. [Google Scholar]
- Vihervuori, H.; Autere, T.A.; Repo, H.; Kurki, S.; Kallio, L.; Lintunen, M.M.; Talvinen, K.; Kronqvist, P. Tumor-infiltrating lymphocytes and CD8+ T cells predict survival of triple-negative breast cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 3105–3114. [Google Scholar] [CrossRef] [Green Version]
- Ali, H.R.; Provenzano, E.; Dawson, S.J.; Blows, F.M.; Liu, B.; Shah, M.; Earl, H.M.; Poole, C.J.; Hiller, L.; Dunn, J.A.; et al. Association between CD8+ T-cell infiltration and breast cancer survival in 12,439 patients. Ann. Oncol. 2014, 25, 1536–1543. [Google Scholar] [CrossRef]
- Miyashita, M.; Sasano, H.; Tamaki, K.; Hirakawa, H.; Takahashi, Y.; Nakagawa, S.; Watanabe, G.; Tada, H.; Suzuki, A.; Ohuchi, N.; et al. Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: A retrospective multicenter study. Breast Cancer Res. 2015, 17, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papaioannou, E.; Sakellakis, M.; Melachrinou, M.; Tzoracoleftherakis, E.; Kalofonos, H.; Kourea, E. A Standardized Evaluation Method for FOXP3+ Tregs and CD8+ T-cells in Breast Carcinoma: Association With Breast Carcinoma Subtypes, Stage and Prognosis. Anticancer Res. 2019, 39, 1217–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, H.; Li, X.; Chen, C.; Chen, J.; Sun, J.; Sun, S.; Jin, L.; Li, J.; Sun, S.; Wu, X. Quantum dot-based immunofluorescent imaging and quantitative detection of TOP2A and prognostic value in triple-negative breast cancer. Int. J. Nanomed. 2016, 11, 5519–5529. [Google Scholar] [CrossRef] [Green Version]
- Rao, N.; Qiu, J.; Wu, J.; Zeng, H.; Su, F.; Qiu, K.; Wu, J.; Yao, H. Significance of Tumor-Infiltrating Lymphocytes and the Expression of Topoisomerase IIα in the Prediction of the Clinical Outcome of Patients with Triple-Negative Breast Cancer after Taxane-Anthracycline-Based Neoadjuvant Chemotherapy. Chemotherapy 2017, 62, 246–255. [Google Scholar] [CrossRef]
- Racioppi, L.; Nelson, E.R.; Huang, W.; Mukherjee, D.; Lawrence, S.A.; Lento, W.; Masci, A.M.; Jiao, Y.; Park, S.; York, B.; et al. CaMKK2 in myeloid cells is a key regulator of the immune-suppressive microenvironment in breast cancer. Nat. Commun. 2019, 10, 2450. [Google Scholar] [CrossRef] [Green Version]
- Pantelidou, C.; Sonzogni, O.; De Oliveria Taveira, M.; Mehta, A.K.; Kothari, A.; Wang, D.; Visal, T.; Li, M.K.; Pinto, J.; Castrillon, J.A.; et al. PARP Inhibitor Efficacy Depends on CD8+ T-cell Recruitment via Intratumoral STING Pathway Activation in BRCA-Deficient Models of Triple-Negative Breast Cancer. Cancer Discov. 2019, 9, 722–737. [Google Scholar] [CrossRef] [Green Version]
- Niavarani, S.R.; Lawson, C.; Boudaud, M.; Simard, C.; Tai, L.H. Oncolytic vesicular stomatitis virus-based cellular vaccine improves triple-negative breast cancer outcome by enhancing natural killer and CD8+ T-cell functionality. J. Immunother. Cancer 2020, 8, e000465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Gruosso, T.; Zuo, D.; Omeroglu, A.; Meterissian, S.; Guiot, M.C.; Salazar, A.; Park, M.; Levine, H. Infiltration of CD8+ T cells into tumor cell clusters in triple-negative breast cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 3678–3687. [Google Scholar] [CrossRef] [Green Version]
- Kalimutho, M.; Sinha, D.; Mittal, D.; Srihari, S.; Nanayakkara, D.; Shafique, S.; Raninga, P.; Nag, P.; Parsons, K.; Khanna, K.K. Blockade of PDGFRβ circumvents resistance to MEK-JAK inhibition via intratumoral CD8+ T-cells infiltration in triple-negative breast cancer. J. Exp. Clin. Cancer Res. 2019, 38, 85. [Google Scholar] [CrossRef] [Green Version]
- Dong, M.B.; Wang, G.; Chow, R.D.; Ye, L.; Zhu, L.; Dai, X.; Park, J.J.; Kim, H.R.; Errami, Y.; Guzman, C.D.; et al. Systematic Immunotherapy Target Discovery Using Genome-Scale In Vivo CRISPR Screens in CD8 T Cells. Cell 2019, 178, 1189–1204.e23. [Google Scholar] [CrossRef] [PubMed]
- Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [Green Version]
- AHY, T.; Tu, W.; McCuaig, R.; Hardy, K.; Donovan, T.; Tsimbalyuk, S.; Forwood, J.K.; Rao, S. Lysine-Specific Histone Demethylase 1A Regulates Macrophage Polarization and Checkpoint Molecules in the Tumor Microenvironment of Triple-Negative Breast Cancer. Front. Immunol. 2019, 10, 1351. [Google Scholar]
- Steenbrugge, J.; Breyne, K.; Demeyere, K.; De Wever, O.; Sanders, N.N.; Van Den Broeck, W.; Colpaert, C.; Vermeulen, P.; Van Laere, S.; Meyer, E. Anti-inflammatory signaling by mammary tumor cells mediates prometastatic macrophage polarization in an innovative intraductal mouse model for triple-negative breast cancer. J. Exp. Clin. Cancer Res. 2018, 37, 191. [Google Scholar] [CrossRef] [PubMed]
- Araujo, J.M.; Gomez, A.C.; Aguilar, A.; Salgado, R.; Balko, J.M.; Bravo, L.; Doimi, F.; Bretel, D.; Morante, Z.; Flores, C.; et al. Effect of CCL5 expression in the recruitment of immune cells in triple negative breast cancer. Sci. Rep. 2018, 8, 4899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, H.; Wang, S.H.; Chen, S.C.; Chen, C.Y.; Lin, T.M. Zoledronic acid blocks the interaction between breast cancer cells and regulatory T-cells. BMC Cancer 2019, 19, 176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tiwari, P.; Blank, A.; Cui, C.; Schoenfelt, K.Q.; Zhou, G.; Xu, Y.; Khramtsova, G.; Olopade, F.; Shah, A.M.; Khan, S.A.; et al. Metabolically activated adipose tissue macrophages link obesity to triple-negative breast cancer. J. Exp. Med. 2019, 216, 1345–1358. [Google Scholar] [CrossRef] [PubMed]
- TNT, U.; Lee, K.H.; Ahn, S.J.; Kim, K.W.; Min, J.J.; Hyun, H.; Yoon, M.S. Real-Time Tracking of Ex Vivo-Expanded Natural Killer Cells Toward Human Triple-Negative Breast Cancers. Front. Immunol. 2018, 9, 825. [Google Scholar]
- Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [Green Version]
- SMI, S.; Bertos, N.; Gruosso, T.; Gigoux, M.; Souleimanova, M.; Zhao, H.; Omeroglu, A.; Hallett, M.T.; Park, M. Identification of Interacting Stromal Axes in Triple-Negative Breast Cancer. Cancer Res. 2017, 77, 4673–4683. [Google Scholar]
- Jézéquel, P.; Kerdraon, O.; Hondermarck, H.; Guérin-Charbonnel, C.; Lasla, H.; Gouraud, W.; Canon, J.L.; Gombos, A.; Dalenc, F.; Delaloge, S.; et al. Identification of three subtypes of triple-negative breast cancer with potential therapeutic implications. Breast Cancer Res. 2019, 21, 65. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.Z.; Ma, D.; Suo, C.; Shi, J.; Xue, M.; Hu, X.; Xiao, Y.; Yu, K.D.; Liu, Y.R.; Yu, Y.; et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell 2019, 35, 428–440.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tseng, L.M.; Chiu, J.H.; Liu, C.Y.; Tsai, Y.F.; Wang, Y.L.; Yang, C.W.; Shyr, Y.M. A comparison of the molecular subtypes of triple-negative breast cancer among non-Asian and Taiwanese women. Breast Cancer Res. Treat. 2017, 163, 241–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Quist, J.; Mirza, H.; MCU, C.; Telli, M.L.; O’Shaughnessy, J.A.; Lord, C.J.; ANJ, T.; Grigoriadis, A. A Four-gene Decision Tree Signature Classification of Triple-negative Breast Cancer: Implications for Targeted Therapeutics. Mol. Cancer Ther. 2019, 18, 204–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, Y.; Jiang, Z.; Chen, C.; Wang, X. Classification of triple-negative breast cancers based on Immunogenomic profiling. J. Exp. Clin. Cancer Res. 2018, 37, 327. [Google Scholar] [CrossRef]
- Prado-Vázquez, G.; Gámez-Pozo, A.; Trilla-Fuertes, L.; Arevalillo, J.M.; Zapater-Moros, A.; Ferrer-Gómez, M.; Díaz-Almirón, M.; López-Vacas, R.; Navarro, H.; Maín, P.; et al. A novel approach to triple-negative breast cancer molecular classification reveals a luminal immune-positive subgroup with good prognoses. Sci. Rep. 2019, 9, 1538. [Google Scholar] [CrossRef]
- DiNome, M.L.; JIJ, O.; Matsuba, C.; Manughian-Peter, A.O.; Ensenyat-Mendez, M.; Chang, S.C.; Jalas, J.R.; Salomon, M.P.; Marzese, D.M. Clinicopathological Features of Triple-Negative Breast Cancer Epigenetic Subtypes. Ann. Surg. Oncol. 2019, 26, 3344–3353. [Google Scholar] [CrossRef]
- Zheng, S.; Zou, Y.; Xie, X.; Liang, J.Y.; Yang, A.; Yu, K.; Wang, J.; Tang, H.; Xie, X. Development and Validation of a Stromal Immune Phenotype Classifier for Predicting Immune Activity and Prognosis in Triple-Negative Breast Cancer. Int. J. Cancer 2020, 147, 542–553. [Google Scholar] [CrossRef]
- Romero-Cordoba, S.; Meneghini, E.; Sant, M.; Iorio, M.V.; Sfondrini, L.; Paolini, B.; Agresti, R.; Tagliabue, E.; Bianchi, F. Decoding Immune Heterogeneity of Triple Negative Breast Cancer and Its Association with Systemic Inflammation. Cancers 2019, 11, 911. [Google Scholar] [CrossRef] [Green Version]
- Zitvogel, L.; Kepp, O.; Kroemer, G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat. Rev. Clin. Oncol. 2011, 8, 151–160. [Google Scholar] [CrossRef]
- Kroemer, G.; Galluzzi, L.; Kepp, O.; Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 2013, 31, 51–72. [Google Scholar] [CrossRef] [PubMed]
- Voloshin, T.; Kaynan, N.; Davidi, S.; Porat, Y.; Shteingauz, A.; Schneiderman, R.S.; Zeevi, E.; Munster, M.; Blat, R.; Tempel, B.C.; et al. Tumor-treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti-PD-1 therapy. Cancer Immunol. Immunother. 2020, 69, 1191–1204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Golden, E.B.; Frances, D.; Pellicciotta, I.; Demaria, S.; Helen, B.M.; Formenti, S.C. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014, 3, e28518. [Google Scholar] [CrossRef] [Green Version]
- Nakasone, E.S.; Askautrud, H.A.; Kees, T.; Park, J.H.; Plaks, V.; Ewald, A.J.; Fein, M.; Rasch, M.G.; Tan, Y.X.; Qiu, J.; et al. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell 2012, 21, 488–503. [Google Scholar] [CrossRef] [Green Version]
- DeNardo, D.G.; Brennan, D.J.; Rexhepaj, E.; Ruffell, B.; Shiao, S.L.; Madden, S.F.; Gallagher, W.M.; Wadhwani, N.; Keil, S.D.; Junaid, S.A.; et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011, 1, 54–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kodumudi, K.N.; Woan, K.; Gilvary, D.L.; Sahakian, E.; Wei, S.; Djeu, J.Y. A novel chemoimmunomodulating property of docetaxel: Suppression of myeloid-derived suppressor cells in tumor bearers. Clin. Cancer Res. 2010, 16, 4583–4594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sims, G.P.; Rowe, D.C.; Rietdijk, S.T.; Herbst, R.; Coyle, A.J. HMGB1 and RAGE in inflammation and cancer. Annu. Rev. Immunol. 2010, 28, 367–388. [Google Scholar] [CrossRef]
- Bianchi, M.E.; Crippa, M.P.; Manfredi, A.A.; Mezzapelle, R.; Rovere, Q.P.; Venereau, E. High-mobility group box 1 protein orchestrates responses to tissue damage via inflammation, innate and adaptive immunity, and tissue repair. Immunol. Rev. 2017, 280, 74–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ladoire, S.; Enot, D.; Senovilla, L.; Ghiringhelli, F.; Poirier-Colame, V.; Chaba, K.; Semeraro, M.; Chaix, M.; Penault-Llorca, F.; Arnould, L.; et al. The presence of LC3B puncta and HMGB1 expression in malignant cells correlate with the immune infiltrate in breast cancer. Autophagy 2016, 12, 864–875. [Google Scholar] [CrossRef] [Green Version]
- Ladoire, S.; Penault-Llorca, F.; Senovilla, L.; Dalban, C.; Enot, D.; Locher, C.; Prada, N.; Poirier-Colame, V.; Chaba, K.; Arnould, L.; et al. Combined evaluation of LC3B puncta and HMGB1 expression predicts residual risk of relapse after adjuvant chemotherapy in breast cancer. Autophagy 2015, 11, 1878–1890. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.J.; Kim, A.; Song, I.H.; Park, I.A.; Yu, J.H.; Ahn, J.H.; Gong, G. Cytoplasmic expression of high mobility group B1 (HMGB1) is associated with tumor-infiltrating lymphocytes (TILs) in breast cancer. Pathol. Int. 2016, 66, 202–209. [Google Scholar] [CrossRef]
- Amornsupak, K.; Insawang, T.; Thuwajit, P.; O-Charoenrat, P.; Eccles, S.A.; Thuwajit, C. Cancer-associated fibroblasts induce high mobility group box 1 and contribute to resistance to doxorubicin in breast cancer cells. BMC Cancer 2014, 14, 955. [Google Scholar] [CrossRef] [PubMed]
- Amornsupak, K.; Jamjuntra, P.; Warnnissorn, M.; O-Charoenrat, P.; Sa-Nguanraksa, D.; Thuwajit, P.; Eccles, S.A.; Thuwajit, C. High ASMA+ Fibroblasts and Low Cytoplasmic HMGB1+ Breast Cancer Cells Predict Poor Prognosis. Clin. Breast Cancer 2017, 17, 441–452.e2. [Google Scholar] [CrossRef] [Green Version]
- Tanabe, Y.; Tsuda, H.; Yoshida, M.; Yunokawa, M.; Yonemori, K.; Shimizu, C.; Yamamoto, S.; Kinoshita, T.; Fujiwara, Y.; Tamura, K. Pathological features of triple-negative breast cancers that showed progressive disease during neoadjuvant chemotherapy. Cancer Sci. 2017, 108, 1520–1529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, B.F.; Tzeng, H.E.; Chen, P.C.; Wang, C.Q.; Su, C.M.; Wang, Y.; Hu, G.N.; Zhao, Y.M.; Wang, Q.; Tang, C.H. HMGB1 genetic polymorphisms are biomarkers for the development and progression of breast cancer. Int. J. Med. Sci. 2018, 15, 580–586. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Kang, F.B.; Wang, J.; Yang, C.; He, D.W. Downregulation of miR-205 contributes to epithelial-mesenchymal transition and invasion in triple-negative breast cancer by targeting HMGB1-RAGE signaling pathway. Anticancer Drugs 2019, 30, 225–232. [Google Scholar] [CrossRef]
- Su, Z.; Ni, P.; She, P.; Liu, Y.; Richard, S.A.; Xu, W.; Zhu, H.; Wang, J. Bio-HMGB1 from breast cancer contributes to M-MDSC differentiation from bone marrow progenitor cells and facilitates conversion of monocytes into MDSC-like cells. Cancer Immunol. Immunother. 2017, 66, 391–401. [Google Scholar] [CrossRef]
- Park, S.J.; Kim, J.M.; Kim, J.; Hur, J.; Park, S.; Kim, K.; Shin, H.J.; Chwae, Y.J. Molecular mechanisms of biogenesis of apoptotic exosome-like vesicles and their roles as damage-associated molecular patterns. Proc. Natl. Acad. Sci. USA 2018, 115, E11721. [Google Scholar] [CrossRef] [Green Version]
- Goh, C.Y.; Wyse, C.; Ho, M.; O’Beirne, E.; Howard, J.; Lindsay, S.; Kelly, P.; Higgins, M.; McCann, A. Exosomes in triple negative breast cancer: Garbage disposals or Trojan horses. Cancer Lett. 2020, 473, 90–97. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Gilkes, D.M.; Takano, N.; Xiang, L.; Luo, W.; Bishop, C.J.; Chaturvedi, P.; Green, J.J.; Semenza, G.L. Hypoxia-inducible factors and RAB22A mediate formation of microvesicles that stimulate breast cancer invasion and metastasis. Proc. Natl. Acad. Sci. USA 2014, 111, E3234–E3242. [Google Scholar] [CrossRef] [Green Version]
- Stevic, I.; Müller, V.; Weber, K.; Fasching, P.A.; Karn, T.; Marmé, F.; Schem, C.; Stickeler, E.; Denkert, C.; van Mackelenbergh, M.; et al. Specific microRNA signatures in exosomes of triple-negative and HER2-positive breast cancer patients undergoing neoadjuvant therapy within the GeparSixto trial. BMC Med. 2018, 16, 179. [Google Scholar] [CrossRef] [Green Version]
- PMM, O.; Alkhilaiwi, F.; Cavalli, I.J.; Malheiros, D.; de Souza Fonseca Ribeiro, E.M.; Cavalli, L.R. Extracellular vesicles from triple-negative breast cancer cells promote proliferation and drug resistance in non-tumorigenic breast cells. Breast Cancer Res. Treat. 2018, 172, 713–723. [Google Scholar]
- Ni, Q.; Stevic, I.; Pan, C.; Müller, V.; Oliveira-Ferrer, L.; Pantel, K.; Schwarzenbach, H. Different signatures of miR-16, miR-30b and miR-93 in exosomes from breast cancer and DCIS patients. Sci. Rep. 2018, 8, 12974. [Google Scholar] [CrossRef]
- Li, Y.; Liang, Y.; Sang, Y.; Song, X.; Zhang, H.; Liu, Y.; Jiang, L.; Yang, Q. MiR-770 suppresses the chemo-resistance and metastasis of triple negative breast cancer via direct targeting of STMN1. Cell Death Dis. 2018, 9, 14. [Google Scholar] [CrossRef] [PubMed]
- Kavanagh, E.L.; Lindsay, S.; Halasz, M.; Gubbins, L.C.; Weiner-Gorzel, K.; MHZ, G.; McGoldrick, A.; Collins, E.; Henry, M.; Blanco-Fernández, A.; et al. Protein and chemotherapy profiling of extracellular vesicles harvested from therapeutic induced senescent triple negative breast cancer cells. Oncogenesis 2017, 6, e388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gong, C.; Tian, J.; Wang, Z.; Gao, Y.; Wu, X.; Ding, X.; Qiang, L.; Li, G.; Han, Z.; Yuan, Y.; et al. Functional exosome-mediated co-delivery of doxorubicin and hydrophobically modified microRNA 159 for triple-negative breast cancer therapy. J. Nanobiotechnology 2019, 17, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, M.; Gai, C.; Li, Z.; Ding, D.; Zheng, J.; Zhang, W.; Lv, S.; Li, W. Targeted exosome-encapsulated erastin induced ferroptosis in triple negative breast cancer cells. Cancer Sci. 2019, 110, 3173–3182. [Google Scholar] [CrossRef] [Green Version]
- Tsuchida, J.; Nagahashi, M.; Takabe, K.; Wakai, T. Clinical Impact of Sphingosine-1-Phosphate in Breast Cancer. Mediat. Inflamm. 2017, 2017, 2076239. [Google Scholar] [CrossRef] [Green Version]
- Kim, E.Y.; Choi, B.; Kim, J.E.; Park, S.O.; Kim, S.M.; Chang, E.J. Interleukin-22 Mediates the Chemotactic Migration of Breast Cancer Cells and Macrophage Infiltration of the Bone Microenvironment by Potentiating S1P/SIPR Signaling. Cells 2020, 9, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sukocheva, O.A. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int. J. Mol. Sci. 2018, 19, 420. [Google Scholar] [CrossRef] [Green Version]
- Chakraborty, P.; Vaena, S.G.; Thyagarajan, K.; Chatterjee, S.; Al-Khami, A.; Selvam, S.P.; Nguyen, H.; Kang, I.; Wyatt, M.W.; Baliga, U.; et al. Pro-Survival Lipid Sphingosine-1-Phosphate Metabolically Programs T Cells to Limit Anti-tumor Activity. Cell Rep. 2019, 28, 1879–1893.e7. [Google Scholar] [CrossRef] [Green Version]
- Weichand, B.; Popp, R.; Dziumbla, S.; Mora, J.; Strack, E.; Elwakeel, E.; Frank, A.C.; Scholich, K.; Pierre, S.; Syed, S.N.; et al. S1PR1 on tumor-associated macrophages promotes lymphangiogenesis and metastasis via NLRP3/IL-1β. J. Exp. Med. 2017, 214, 2695–2713. [Google Scholar] [CrossRef]
- Liu, S.; Ni, C.; Zhang, D.; Sun, H.; Dong, X.; Che, N.; Liang, X.; Chen, C.; Liu, F.; Bai, J.; et al. S1PR1 regulates the switch of two angiogenic modes by VE-cadherin phosphorylation in breast cancer. Cell Death Dis. 2019, 10, 200. [Google Scholar] [CrossRef]
- Rathinasamy, A.; Domschke, C.; Ge, Y.; Böhm, H.H.; Dettling, S.; Jansen, D.; Lasitschka, F.; Umansky, L.; Gräler, M.H.; Hartmann, J.; et al. Tumor specific regulatory T cells in the bone marrow of breast cancer patients selectively upregulate the emigration receptor S1P1. Cancer Immunol. Immunother. 2017, 66, 593–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, S.; Yang, J. Preclinical study of the antitumor effect of sphingosine-1-phosphate receptor 1 antibody (S1PR1-antibody) against human breast cancer cells. Investig. New. Drugs 2019, 37, 57–64. [Google Scholar] [CrossRef] [PubMed]
- Jung, M.; Ören, B.; Mora, J.; Mertens, C.; Dziumbla, S.; Popp, R.; Weigert, A.; Grossmann, N.; Fleming, I.; Brüne, B. Lipocalin 2 from macrophages stimulated by tumor cell-derived sphingosine 1-phosphate promotes lymphangiogenesis and tumor metastasis. Sci. Signal. 2016, 9, ra64. [Google Scholar] [CrossRef] [PubMed]
- Acharya, S.; Yao, J.; Li, P.; Zhang, C.; Lowery, F.J.; Zhang, Q.; Guo, H.; Qu, J.; Yang, F.; Wistuba, I.I.; et al. Sphingosine Kinase 1 Signaling Promotes Metastasis of Triple-Negative Breast Cancer. Cancer Res. 2019, 79, 4211–4226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maiti, A.; Takabe, K.; Hait, N.C. Metastatic triple-negative breast cancer is dependent on SphKs/S1P signaling for growth and survival. Cell. Signal. 2017, 32, 85–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagahashi, M.; Yamada, A.; Katsuta, E.; Aoyagi, T.; Huang, W.C.; Terracina, K.P.; Hait, N.C.; Allegood, J.C.; Tsuchida, J.; Yuza, K.; et al. Targeting the SphK1/S1P/S1PR1 Axis That Links Obesity, Chronic Inflammation, and Breast Cancer Metastasis. Cancer Res. 2018, 78, 1713–1725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lei, F.J.; Cheng, B.H.; Liao, P.Y.; Wang, H.C.; Chang, W.C.; Lai, H.C.; Yang, J.C.; Wu, Y.C.; Chu, L.C.; Ma, W.L. Survival benefit of sphingosin-1-phosphate and receptors expressions in breast cancer patients. Cancer Med. 2018, 7, 3743–3754. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Chapman, N.M.; Zhang, B.; Li, M.; Fan, M.; Laribee, R.N.; Zaidi, M.R.; Pfeffer, L.M.; Chi, H.; Wu, Z.H. Upregulation of PD-L1 via HMGB1-Activated IRF3 and NF-κB Contributes to UV Radiation-Induced Immune Suppression. Cancer Res. 2019, 79, 2909–2922. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Shi, H.; Yuan, X.; Jiang, P.; Qian, H.; Xu, W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol. Cancer 2018, 17, 146. [Google Scholar] [CrossRef] [PubMed]
- Sheller-Miller, S.; Urrabaz-Garza, R.; Saade, G.; Menon, R. Damage-Associated molecular pattern markers HMGB1 and cell-Free fetal telomere fragments in oxidative-Stressed amnion epithelial cell-Derived exosomes. J. Reprod. Immunol. 2017, 123, 3–11. [Google Scholar] [CrossRef]
- Song, S.; Min, H.; Niu, M.; Wang, L.; Wu, Y.; Zhang, B.; Chen, X.; Liang, Q.; Wen, Y.; Wang, Y.; et al. S1PR1 predicts patient survival and promotes chemotherapy drug resistance in gastric cancer cells through STAT3 constitutive activation. EBioMedicine 2018, 37, 168–176. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.; Yang, C.; Yu, F.; Ding, W.; Hu, Y.; Cheng, F.; Zhang, F.; Guan, B.; Wang, X.; Lu, L.; et al. Endoplasmic reticulum resident oxidase ERO1-Lalpha promotes hepatocellular carcinoma metastasis and angiogenesis through the S1PR1/STAT3/VEGF-A pathway. Cell Death Dis. 2018, 9, 1105. [Google Scholar] [CrossRef] [PubMed]
- Lankadasari, M.B.; Aparna, J.S.; Mohammed, S.; James, S.; Aoki, K.; Binu, V.S.; Nair, S.; Harikumar, K.B. Targeting S1PR1/STAT3 loop abrogates desmoplasia and chemosensitizes pancreatic cancer to gemcitabine. Theranostics 2018, 8, 3824–3840. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zheng, H.; Siddharth, S.; Parida, S.; Wu, X.; Sharma, D. Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation. Cancers 2021, 13, 3357. https://doi.org/10.3390/cancers13133357
Zheng H, Siddharth S, Parida S, Wu X, Sharma D. Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation. Cancers. 2021; 13(13):3357. https://doi.org/10.3390/cancers13133357
Chicago/Turabian StyleZheng, Hongmei, Sumit Siddharth, Sheetal Parida, Xinhong Wu, and Dipali Sharma. 2021. "Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation" Cancers 13, no. 13: 3357. https://doi.org/10.3390/cancers13133357
APA StyleZheng, H., Siddharth, S., Parida, S., Wu, X., & Sharma, D. (2021). Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation. Cancers, 13(13), 3357. https://doi.org/10.3390/cancers13133357