151
|
Gomes-Santos IL, Amoozgar Z, Kumar AS, Ho WW, Roh K, Talele NP, Curtis H, Kawaguchi K, Jain RK, Fukumura D. Exercise Training Improves Tumor Control by Increasing CD8 + T-cell Infiltration via CXCR3 Signaling and Sensitizes Breast Cancer to Immune Checkpoint Blockade. Cancer Immunol Res 2021; 9:765-778. [PMID: 33839688 PMCID: PMC8295193 DOI: 10.1158/2326-6066.cir-20-0499] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/23/2020] [Accepted: 03/31/2021] [Indexed: 11/16/2022]
Abstract
The mechanisms behind the antitumor effects of exercise training (ExTr) are not fully understood. Using mouse models of established breast cancer, we examined here the causal role of CD8+ T cells in the benefit acquired from ExTr in tumor control, as well as the ability of ExTr to improve immunotherapy responses. We implanted E0771, EMT6, MMTV-PyMT, and MCa-M3C breast cancer cells orthotopically in wild-type or Cxcr3-/- female mice and initiated intensity-controlled ExTr sessions when tumors reached approximately 100 mm3 We characterized the tumor microenvironment (TME) using flow cytometry, transcriptome analysis, proteome array, ELISA, and immunohistochemistry. We used antibodies against CD8+ T cells for cell depletion. Treatment with immune checkpoint blockade (ICB) consisted of anti-PD-1 alone or in combination with anti-CTLA-4. ExTr delayed tumor growth and induced vessel normalization, demonstrated by increased pericyte coverage and perfusion and by decreased hypoxia. ExTr boosted CD8+ T-cell infiltration, with enhanced effector function. CD8+ T-cell depletion prevented the antitumor effect of ExTr. The recruitment of CD8+ T cells and the antitumor effects of ExTr were abrogated in Cxcr3-/- mice, supporting the causal role of the CXCL9/CXCL11-CXCR3 pathway. ExTr also sensitized ICB-refractory breast cancers to treatment. Our results indicate that ExTr can normalize the tumor vasculature, reprogram the immune TME, and enhance the antitumor activity mediated by CD8+ T cells via CXCR3, boosting ICB responses. Our findings and mechanistic insights provide a rationale for the clinical translation of ExTr to improve immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Igor L Gomes-Santos
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - William W Ho
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kangsan Roh
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nilesh P Talele
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hannah Curtis
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kosuke Kawaguchi
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
152
|
Zafar H, Raza F, Ma S, Wei Y, Zhang J, Shen Q. Recent progress on nanomedicine-induced ferroptosis for cancer therapy. Biomater Sci 2021; 9:5092-5115. [PMID: 34160488 DOI: 10.1039/d1bm00721a] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current treatment strategies for cancer therapy have posed many problems in achieving high efficacy. Therefore, an urgent step is needed to develop innovative therapies that can win beyond satisfactory results against tumor. Ferroptosis that is a kind of non-apoptotic based programmed cell death has played a crucial role in eradicating tumors by reactive oxygen species and iron-dependent pathways. Research shows a remarkable potential of ferroptosis in eliminating aggressive malignancies resistant to traditional therapies. The combination of nanomedicine and ferroptosis has revealed a close relationship for the treatment of various cancer types with high efficacy. This review introduces the basics of nanomedicine-based ferroptosis first to emphasize the feasibility and properties of ferroptosis in cancer therapy. Then, the current research on the applications of nanomedicine for the ferroptosis-based anticancer therapy is highlighted. Finally, conclusions and future research directions in perspective of various challenges in developing nanomedicine-based ferroptosis into clinical therapeutics are discussed.
Collapse
Affiliation(s)
- Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan, Road, Shanghai, 200240, China.
| | | | | | | | | | | |
Collapse
|
153
|
Davey MG, Ryan ÉJ, Davey MS, Lowery AJ, Miller N, Kerin MJ. Clinicopathological and prognostic significance of programmed cell death ligand 1 expression in patients diagnosed with breast cancer: meta-analysis. Br J Surg 2021; 108:622-631. [PMID: 33963374 PMCID: PMC10364926 DOI: 10.1093/bjs/znab103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/06/2021] [Accepted: 02/25/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Uncertainty exists regarding the clinical relevance of programmed cell death ligand 1 (PD-L1) expression in breast cancer. METHODS A systematic review was performed in accordance with PRISMA guidelines. Observational studies that compared high versus low expression of PD-L1 on breast cancer cells were identified. Log hazard ratios (HRs) for disease-free and overall survival and their standard errors were calculated from Kaplan-Meier curves or Cox regression analyses, and pooled using the inverse-variance method. Dichotomous variables were pooled as odds ratios (ORs) using the Mantel-Haenszel method. RESULTS Sixty-five studies with 19 870 patients were included; 14 404 patients were classified as having low and 4975 high PD-L1 expression. High PD-L1 was associated with achieving a pathological complete response following neoadjuvant chemotherapy (OR 3.30, 95 per cent confidence interval 1.19 to 9.16; P < 0.01; I2 = 85 per cent). Low PD-L1 expression was associated with human epidermal growth factor receptor 2 (OR 3.98, 1.81 to 8.75; P < 0.001; I2 = 96 per cent) and luminal (OR 14.93, 6.46 to 34.51; P < 0.001; I2 = 99 per cent) breast cancer subtypes. Those with low PD-L1 had favourable overall survival rates (HR 1.30, 1.05 to 1.61; P = 0.02; I2 = 85 per cent). CONCLUSION Breast cancers with high PD-L1 expression are associated with aggressive clinicopathological and immunohistochemical characteristics and are more likely to achieve a pathological complete response following neoadjuvant chemotherapy. These breast cancers are, however, associated with worse overall survival outcomes.
Collapse
Affiliation(s)
- M G Davey
- Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - É J Ryan
- Department of Surgery, Galway University Hospitals, Galway, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M S Davey
- Department of Surgery, Galway University Hospitals, Galway, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - A J Lowery
- Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - N Miller
- Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - M J Kerin
- Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| |
Collapse
|
154
|
Raiter A, Zlotnik O, Lipovetsky J, Mugami S, Dar S, Lubin I, Sharon E, Cohen CJ, Yerushalmi R. A novel role for an old target: CD45 for breast cancer immunotherapy. Oncoimmunology 2021; 10:1929725. [PMID: 34104545 PMCID: PMC8158046 DOI: 10.1080/2162402x.2021.1929725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Breast cancer subtypes have not shown significant response to current immunomodulatory therapies. Although most subtypes are treatable, triple negative breast cancer (TNBC), an aggressive highly metastatic cancer, comprising 10-20% of breast cancers, remains an unmet medical need. New strategies are needed in order to overcome flaws in the responsiveness to current TNBC therapies. Our aims were: first, to determine the efficacy of a novel immunomodulatory peptide, C24D, on TNBC and second, to elucidate the molecular mechanism by which C24D induces immune-modulating tumor killing. Using mass spectrometry analysis, we identified CD45 as the C24D binding receptor. In vitro and in vivo TNBC models were used to assess the efficacy of C24D in reversing TNBC-induced immunosuppression and in triggering immune-modulated tumor cell killing. The CD45 signal transduction pathway was evaluated by western blot and FACS analyses. We revealed that addition of PBMCs from healthy female donors to TNBC cells results in a cascade of suppressive CD45 intracellular signals. On binding to CD45's extra-cellular domain on TNBC-suppressed leukocytes, the C24D peptide re-activates the Src family of tyrosine kinases, resulting in specific tumor immune response. In vitro, immune reactivation by C24D results in an increase of CD69+ T and CD69+ NK cells, triggering specific killing of TNBC cells. In vivo, C24D induced CD8+ and activated CD56+ tumor infiltrated cells, resulting in tumor apoptosis. Our results should renew interest in molecules targeting CD45, such as the C24D peptide, as a novel strategy for TNBC immunotherapy.
Collapse
Affiliation(s)
- Annat Raiter
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
- CONTACT Annat Raiter Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva49100, Israel
| | - Oran Zlotnik
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
- Surgery Department, Breast Cancer Unit, Beilinson Hospital, Rabin Medical Center, Petach Tikva, Israel
| | - Julia Lipovetsky
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Shany Mugami
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Shira Dar
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Ido Lubin
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Eran Sharon
- Surgery Department, Breast Cancer Unit, Beilinson Hospital, Rabin Medical Center, Petach Tikva, Israel
| | - Cyrille J. Cohen
- Laboratory of Tumor Immunotherapy, the Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
| | - Rinat Yerushalmi
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
- Breast Cancer Unit, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
155
|
Shi Z, Shen J, Qiu J, Zhao Q, Hua K, Wang H. CXCL10 potentiates immune checkpoint blockade therapy in homologous recombination-deficient tumors. Theranostics 2021; 11:7175-7187. [PMID: 34158843 PMCID: PMC8210593 DOI: 10.7150/thno.59056] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/12/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Homologous recombination deficiency (HRD) is a common molecular characteristic of genomic instability, and has been proven to be a biomarker for target therapy. However, until now, no research has explored the changes in the transcriptomics landscape of HRD tumors. Methods: The HRD score was established from SNP array data of breast cancer patients from the cancer genome atlas (TCGA) database. The transcriptome data of patients with different HRD scores were analyzed to identify biomarkers associated with HRD. The candidate biomarkers were validated in the gene expression omnibus (GEO) database and immunotherapy cohorts. Results: Based on data from the gene expression profile and clinical characteristics from 1310 breast cancer patients, including TCGA database and GEO database, we found that downstream targets of the cGAS-STING pathway, such as CXCL10, were upregulated in HRD tumors and could be used as a predictor of survival outcome in triple-negative breast cancer (TNBC) patients. Further comprehensive analysis of the tumor immune microenvironment (TIME) revealed that the expression of CXCL10 was positively correlated with neoantigen load and infiltrating immune cells. Finally, in vivo experimental data and clinical trial data confirmed that the expression of CXCL10 could be used as a biomarker for anti-PD-1/PD-L1 therapy. Conclusions: Together, our study not only revealed that CXCL10 is associated with HRD but also introduced a potential new perspective for identifying prognostic biomarkers of immunotherapy.
Collapse
Affiliation(s)
- Zhiwen Shi
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regualtion, Shanghai Institute of Planned Parenthood Research, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
| | - Junjun Qiu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regualtion, Shanghai Institute of Planned Parenthood Research, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Qingguo Zhao
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regualtion, Shanghai Institute of Planned Parenthood Research, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
| | - Keqin Hua
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regualtion, Shanghai Institute of Planned Parenthood Research, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Hongyan Wang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regualtion, Shanghai Institute of Planned Parenthood Research, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
- Children's Hospital of Fudan University, Shanghai 201100, China
| |
Collapse
|
156
|
Bertucci F, Boudin L, Finetti P, Van Berckelaer C, Van Dam P, Dirix L, Viens P, Gonçalves A, Ueno NT, Van Laere S, Birnbaum D, Mamessier E. Immune landscape of inflammatory breast cancer suggests vulnerability to immune checkpoint inhibitors. Oncoimmunology 2021; 10:1929724. [PMID: 34104544 PMCID: PMC8158040 DOI: 10.1080/2162402x.2021.1929724] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background. Anti-PD1/PDL1 immune checkpoint inhibitors (ICIs) showed promising results in breast cancer, and exploration of additional actionable immune checkpoints is ongoing. Inflammatory breast cancer (IBC) is an aggressive form of disease, the immune tumor microenvironment (TME) of which is poorly known. We aimed at providing the first comprehensive immune portrait of IBCs. Methods. From the gene expression profiles of 137 IBC and 252 non-IBC clinical samples, we measured the fractions of 22 immune cell types, expression of signatures associated with tertiary lymphoid structures (TLS) and with the response to ICIs (T cell-inflamed signature: TIS) and of 18 genes coding for major actionable immune checkpoints. The IBC/non-IBC comparison was adjusted upon the clinicopathological variables. Results. The immune profiles of IBCs were heterogeneous. CIBERSORT analysis showed profiles rich in macrophages, CD8+ and CD4 + T-cells, with remarkable similarity with melanoma TME. The comparison with non-IBCs showed significant enrichment in M1 macrophages, γδ T-cells, and memory B-cells. IBCs showed higher expression of TLS and TIS signatures. The TIS signature displayed values in IBCs close to those observed in other cancers sensitive to ICIs. Two-thirds of actionable immune genes (HAVCR2/TIM3, CD27, CD70, CTLA4, ICOS, IDO1, LAG3, PDCD1, TNFRSF9, PVRIG, CD274/PDL1, and TIGIT) were overexpressed in IBCs as compared to normal breast and two-thirds were overexpressed in IBCs versus non-IBCs, with very frequent co-overexpression. For most of them, the overexpression was associated with better pathological response to chemotherapy. Conclusion. Our results suggest the potential higher vulnerability of IBC to ICIs. Clinical trials.
Collapse
Affiliation(s)
- François Bertucci
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France.,Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Laurys Boudin
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Christophe Van Berckelaer
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium
| | - Peter Van Dam
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium.,Department of Oncological Research, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Patrice Viens
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Anthony Gonçalves
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France.,Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Naoto T Ueno
- Breast Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium.,Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Emilie Mamessier
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| |
Collapse
|
157
|
Managing side effects of immune checkpoint inhibitors in breast cancer. Crit Rev Oncol Hematol 2021; 162:103354. [PMID: 34029683 DOI: 10.1016/j.critrevonc.2021.103354] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) represent a major development in cancer therapy. The indications for these agents continue to expand across malignancies and disease settings. For years breast cancer (BC) has been considered immunologically quiescent compared with other tumor types. However, recent findings highlighted the immunogenicity of some BCs and paved the way for clinical trials of immunotherapy in BC that led to recent landmark approvals. As a drawback, the safety profile of ICIs is shaped by a specific spectrum of immune-related adverse events (irAEs) that can vary according to ICI class and tumor histology. This review will discuss the epidemiology of these adverse events, their kinetics, risk factors and the most important aspects in their management. A particular focus will be put on BC as the current landscape of immunotherapy for this disease is rapidly increasing the number of people treated with ICIs, thus susceptible to irAEs.
Collapse
|
158
|
Zhang Y, Asad S, Weber Z, Tallman D, Nock W, Wyse M, Bey JF, Dean KL, Adams EJ, Stockard S, Singh J, Winer EP, Lin NU, Jiang YZ, Ma D, Wang P, Shi L, Huang W, Shao ZM, Cherian M, Lustberg MB, Ramaswamy B, Sardesai S, VanDeusen J, Williams N, Wesolowski R, Obeng-Gyasi S, Sizemore GM, Sizemore ST, Verschraegen C, Stover DG. Genomic features of rapid versus late relapse in triple negative breast cancer. BMC Cancer 2021; 21:568. [PMID: 34006255 PMCID: PMC8130400 DOI: 10.1186/s12885-021-08320-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a heterogeneous disease and we have previously shown that rapid relapse of TNBC is associated with distinct sociodemographic features. We hypothesized that rapid versus late relapse in TNBC is also defined by distinct clinical and genomic features of primary tumors. Methods Using three publicly-available datasets, we identified 453 patients diagnosed with primary TNBC with adequate follow-up to be characterized as ‘rapid relapse’ (rrTNBC; distant relapse or death ≤2 years of diagnosis), ‘late relapse’ (lrTNBC; > 2 years) or ‘no relapse’ (nrTNBC: > 5 years no relapse/death). We explored basic clinical and primary tumor multi-omic data, including whole transcriptome (n = 453), and whole genome copy number and mutation data for 171 cancer-related genes (n = 317). Association of rapid relapse with clinical and genomic features were assessed using Pearson chi-squared tests, t-tests, ANOVA, and Fisher exact tests. We evaluated logistic regression models of clinical features with subtype versus two models that integrated significant genomic features. Results Relative to nrTNBC, both rrTNBC and lrTNBC had significantly lower immune signatures and immune signatures were highly correlated to anti-tumor CD8 T-cell, M1 macrophage, and gamma-delta T-cell CIBERSORT inferred immune subsets. Intriguingly, lrTNBCs were enriched for luminal signatures. There was no difference in tumor mutation burden or percent genome altered across groups. Logistic regression mModels that incorporate genomic features significantly outperformed standard clinical/subtype models in training (n = 63 patients), testing (n = 63) and independent validation (n = 34) cohorts, although performance of all models were overall modest. Conclusions We identify clinical and genomic features associated with rapid relapse TNBC for further study of this aggressive TNBC subset. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08320-7.
Collapse
Affiliation(s)
- Yiqing Zhang
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA
| | - Sarah Asad
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA
| | - Zachary Weber
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - David Tallman
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - William Nock
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA
| | - Meghan Wyse
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Jerome F Bey
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Kristin L Dean
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Elizabeth J Adams
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA
| | - Sinclair Stockard
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Jasneet Singh
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Eric P Winer
- Department of Medical Oncology, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Nancy U Lin
- Department of Medical Oncology, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, P.R. China
| | - Ding Ma
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, P.R. China
| | - Peng Wang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, P.R. China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 2005 Songhu Road, Shanghai, 200438, P.R. China
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), 250 Bibo Road, Shanghai, 201203, P.R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, P.R. China
| | - Mathew Cherian
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Maryam B Lustberg
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Bhuvaneswari Ramaswamy
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Sagar Sardesai
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Jeffrey VanDeusen
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Nicole Williams
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Robert Wesolowski
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Samilia Obeng-Gyasi
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA
| | - Gina M Sizemore
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Steven T Sizemore
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Claire Verschraegen
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA.,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA
| | - Daniel G Stover
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA. .,Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, 460 W 10th Ave, Columbus, OH, 43210, USA. .,Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA. .,Stefanie Spielman Comprehensive Breast Center, 1145 Olentangy River Rd, Columbus, OH, 43212, USA. .,Stefanie Spielman Comprehensive Breast Center, Ohio State University Comprehensive Cancer Center, Biomedical Research Tower, Room 512, Columbus, OH, 43210, USA.
| |
Collapse
|
159
|
Li D, Zhao W, Zhang X, Lv H, Li C, Sun L. NEFM DNA methylation correlates with immune infiltration and survival in breast cancer. Clin Epigenetics 2021; 13:112. [PMID: 34001208 PMCID: PMC8130356 DOI: 10.1186/s13148-021-01096-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 05/02/2021] [Indexed: 11/25/2022] Open
Abstract
Background This study aims to determine whether NEFM (neurofilament medium) DNA methylation correlates with immune infiltration and prognosis in breast cancer (BRCA) and to explore NEFM-connected immune gene signature. Methods NEFM transcriptional expression was analyzed in BRCA and normal breast tissues using Oncomine and Tumor Immune Estimation Resource (TIMER) databases. The relationship between NEFM DNA methylation and NEFM transcriptional expression was investigated in TCGA. Potential influence of NEFM DNA methylation/expression on clinical outcome was evaluated using TCGA BRCA, The Human Protein Atlas and Kaplan–Meier plotter databases. Association of NEFM transcriptional expression/DNA methylation with cancer immune infiltration was investigated using TIMER and TISIDB databases. Results High expression of NEFM correlated with better overall survival (OS) and recurrence-free survival (RFS) in TCGA BRCA and Kaplan–Meier plotter, whereas NEFM DNA methylation with worse OS in TCGA BRCA. NEFM transcriptional expression negatively correlated with DNA methylation. NEFM DNA methylation significantly negatively correlated with infiltrating levels of B, CD8+ T/CD4+ T cells, macrophages, neutrophils and dendritic cells in TIMER and TISIDB. NEFM expression positively correlated with macrophage infiltration in TIMER and TISIDB. After adjusted with tumor purity, NEFM expression weekly negatively correlated with infiltration level of B cells, whereas positively correlated with CD8+ T cell infiltration in TIMER gene modules. NEFM expression/DNA methylation correlated with diverse immune markers in TCGA and TISIDB. Conclusions NEFM low-expression/DNA methylation correlates with poor prognosis. NEFM expression positively correlates with macrophage infiltration. NEFM DNA methylation strongly negatively correlates with immune infiltration in BRCA. Our study highlights novel potential functions of NEFM expression/DNA methylation in regulation of tumor immune microenvironment. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01096-4.
Collapse
Affiliation(s)
- Dandan Li
- Department of Radiotherapy Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenhao Zhao
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China
| | - Xinyu Zhang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China
| | - Hanning Lv
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China
| | - Chunhong Li
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China.
| | - Lichun Sun
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
160
|
Blood Immunosenescence Signatures Reflecting Age, Frailty and Tumor Immune Infiltrate in Patients with Early Luminal Breast Cancer. Cancers (Basel) 2021; 13:cancers13092185. [PMID: 34063210 PMCID: PMC8125302 DOI: 10.3390/cancers13092185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/24/2021] [Accepted: 04/30/2021] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Treating older patients with (breast) cancer is a major challenge. On the one hand, older persons are more vulnerable to side effects of therapy, and over-treatment should be avoided. On the other hand, under-treatment (which is common in the elderly) can lead to worse survival and quality of life as well. Benefits of therapy and risk of (sometimes life threatening) toxicity should be carefully balanced. There is an urgent need for robust markers that reflect the body’s biological age and could aid in outlining optimal individual treatment regimens. Here we investigated whether age/frailty and characteristics of the tumor immune infiltrate are mirrored in specific blood biomarker combinations. Several three-biomarker panels were able to categorize patients quite efficiently, especially in terms of their clinical frailty status. Abstract Background: Immune/senescence-related host factors play a pivotal role in numerous biological and pathological process like aging, frailty and cancer. The assessment of these host factors via robust, non-invasive, and easy-to-measure blood biomarkers could improve insights in these processes. Here, we investigated in a series of breast cancer patients in which way single circulating biomarkers or biomarker panels relate to chronological age, frailty status, and tumor-associated inflammatory microenvironment. Methods: An extensive panel of blood immune/senescence markers and the tumor immune infiltrate was studied in young, middle-aged, and old patients with an early invasive hormone-sensitive, HER2-negative breast cancer. In the old group, clinical frailty was estimated via the G8-scores. Results: Several three-blood biomarker panels proved to be able to separate old chronological age from young age very efficiently. Clinically more important, several three-blood biomarker panels were strongly associated with clinical frailty. Performance of blood biomarker panels for prediction of the tumor immune infiltrate was lower. Conclusion: Immune/senescence blood biomarker panels strongly correlate with chronological age, and clinically more importantly with frailty status in early breast cancer patients. They require further investigation on their ability to provide a more complete picture on clinical frailty status and direct personalized therapy in older persons.
Collapse
|
161
|
Wang JH, Wahid KA, van Dijk LV, Farahani K, Thompson RF, Fuller CD. Radiomic biomarkers of tumor immune biology and immunotherapy response. Clin Transl Radiat Oncol 2021; 28:97-115. [PMID: 33937530 PMCID: PMC8076712 DOI: 10.1016/j.ctro.2021.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies are leading to improved outcomes for many cancers, including those with devastating prognoses. As therapies like immune checkpoint inhibitors (ICI) become a mainstay in treatment regimens, many concurrent challenges have arisen - for instance, delineating clinical responders from non-responders. Predicting response has proven to be difficult given a lack of consistent and accurate biomarkers, heterogeneity of the tumor microenvironment (TME), and a poor understanding of resistance mechanisms. For the most part, imaging data have remained an untapped, yet abundant, resource to address these challenges. In recent years, quantitative image analyses have highlighted the utility of medical imaging in predicting tumor phenotypes, prognosis, and therapeutic response. These studies have been fueled by an explosion of resources in high-throughput mining of image features (i.e. radiomics) and artificial intelligence. In this review, we highlight current progress in radiomics to understand tumor immune biology and predict clinical responses to immunotherapies. We also discuss limitations in these studies and future directions for the field, particularly if high-dimensional imaging data are to play a larger role in precision medicine.
Collapse
Affiliation(s)
- Jarey H. Wang
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Kareem A. Wahid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lisanne V. van Dijk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Keyvan Farahani
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, MD, United States
| | - Reid F. Thompson
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Clifton David Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
162
|
Werner LR, Gibson KA, Goodman ML, Helm DE, Walter KR, Holloran SM, Trinca GM, Hastings RC, Yang HH, Hu Y, Wei J, Lei G, Yang XY, Madan R, Molinolo AA, Markiewicz MA, Chalise P, Axelrod ML, Balko JM, Hunter KW, Hartman ZC, Lange CA, Hagan CR. Progesterone promotes immunomodulation and tumor development in the murine mammary gland. J Immunother Cancer 2021; 9:e001710. [PMID: 33958486 PMCID: PMC8103939 DOI: 10.1136/jitc-2020-001710] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Clinical studies have linked usage of progestins (synthetic progesterone [P4]) to breast cancer risk. However, little is understood regarding the role of native P4, signaling through the progesterone receptor (PR), in breast tumor formation. Recently, we reported a link between PR and immune signaling pathways, showing that P4/PR can repress type I interferon signaling pathways. Given these findings, we sought to investigate whether P4/PR drive immunomodulation in the mammary gland and promote tumor formation. METHODS To determine the effect of P4 on immune cell populations in the murine mammary gland, mice were treated with P4 or placebo pellets for 21 days. Immune cell populations in the mammary gland, spleen, and inguinal lymph nodes were subsequently analyzed by flow cytometry. To assess the effect of PR overexpression on mammary gland tumor development as well as immune cell populations in the mammary gland, a transgenic mouse model was used in which PR was overexpressed throughout the entire mouse. Immune cell populations were assessed in the mammary glands, spleens, and inguinal lymph nodes of 6-month-old transgenic and control mice by flow cytometry. Transgenic mice were also monitored for mammary gland tumor development over a 2-year time span. Following development of mammary gland tumors, immune cell populations in the tumors and spleens of transgenic and control mice were analyzed by flow cytometry. RESULTS We found that mice treated with P4 exhibited changes in the mammary gland indicative of an inhibited immune response compared with placebo-treated mice. Furthermore, transgenic mice with PR overexpression demonstrated decreased numbers of immune cell populations in their mammary glands, lymph nodes, and spleens. On long-term monitoring, we determined that multiparous PR-overexpressing mice developed significantly more mammary gland tumors than control mice. Additionally, tumors from PR-overexpressing mice contained fewer infiltrating immune cells. Finally, RNA sequencing analysis of tumor samples revealed that immune-related gene signatures were lower in tumors from PR-overexpressing mice as compared with control mice. CONCLUSION Together, these findings offer a novel mechanism of P4-driven mammary gland tumor development and provide rationale in investigating the usage of antiprogestin therapies to promote immune-mediated elimination of mammary gland tumors.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Animals
- Breast Neoplasms/chemically induced
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Drug Implants
- Female
- Galectin 4/genetics
- Galectin 4/metabolism
- Immunity, Innate/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mice, Transgenic
- Ovariectomy
- Progesterone/administration & dosage
- Receptors, Progesterone/agonists
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Signal Transduction
- Time Factors
- Tumor Burden/drug effects
- Tumor Escape/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Lauryn R Werner
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Katelin A Gibson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Merit L Goodman
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Dominika E Helm
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Katherine R Walter
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sean M Holloran
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Gloria M Trinca
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Richard C Hastings
- Flow Cytometry Core Laboratory, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ying Hu
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Junping Wei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Rashna Madan
- Division of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Alfredo A Molinolo
- Department of Pathology, University of California San Diego Moores Cancer Center, La Jolla, California, USA
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Margaret L Axelrod
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kent W Hunter
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Carol A Lange
- Department of Medicine (Hematology, Oncology, and Transplantation), University of Minnesota Cancer Center, Minneapolis, Minnesota, USA
- Department of Pharmacology, University of Minnesota Cancer Center, Minneapolis, Minnesota, USA
| | - Christy R Hagan
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
163
|
Fei W, Zhang Y, Ye Y, Li C, Yao Y, Zhang M, Li F, Zheng C. Bioactive metal-containing nanomaterials for ferroptotic cancer therapy. J Mater Chem B 2021; 8:10461-10473. [PMID: 33231601 DOI: 10.1039/d0tb02138e] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The clinical performance of the current cancer therapies is still far from satisfactory. The emerging ferroptosis-driven therapy strategies reignite the hope of chemotherapy in tumor treatment due to their incredible tumor suppression. Among ferroptosis-based cancer therapies, metal elements have attracted remarkable attention due to their inherent physicochemical properties in inducing ferroptosis of tumor cells quickly and strongly without complex cellular signal transduction. Although the discovery and applications of ferroptosis for tumor treatment have been discussed in many reviews, the unique advantages of metal-containing nanomaterials interfering ferroptotic cancer therapies (MIFCT) have seldom been mentioned. Here, we outline the latest advances of MIFCT comprehensively. Firstly, the functions of different kinds of metal elements or their ions are introduced to illustrate their advantages in MIFCT. Secondly, the emerging metal-containing nanomaterials that are designed to achieve ferroptosis-driven therapy are overviewed, including their ability to boost the Fenton or Fenton-like reaction for reactive oxygen species generation, act as hydrogen peroxide self-providers, damage the reducing system, and disturb cellular communication. Moreover, metal-containing nanomaterials with external energy conversion features for MIFCT are discussed. Finally, the future expectations and challenges of MIFCT for clinical cancer therapy are spotlighted.
Collapse
Affiliation(s)
- Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Yue Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China.
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Chaoqun Li
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China.
| | - Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Fanzhu Li
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China.
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
164
|
Yuan C, Chang K, Xu C, Li Q, Du Z. High expression of DLL3 is associated with a poor prognosis and immune infiltration in invasive breast cancer patients. Transl Oncol 2021; 14:101080. [PMID: 33915517 PMCID: PMC8093948 DOI: 10.1016/j.tranon.2021.101080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Previous studies have shown the prognostic value of delta like canonical Notch ligand 3 (DLL3) in patients with different types of tumors, but the role and predictive value of DLL3 in invasive breast cancer (IBC) have not been reported. In this study, we explored the prognostic ability and potential ways of DLL3 in IBC patients. METHODS We retrospectively enrolled 130 IBC patients from a single institution from 2004 to 2019 for bioinformatics and statistical analysis. The Cancer Genome Atlas breast invasive carcinoma (TCGA-BRCA) cohort was used for verification. RESULTS High expression of DLL3 was associated with overall survival (OS) in IBC patients (P = 0.023). Multivariate analysis further showed that DLL3 expression was an independent prognostic factor (hazard ratio [HR]: 1.08; 95% confidence interval [CI]: 1.01-1.15; P = 0.017). Time-dependent receiver operating characteristic (ROC) with the area under the curve (0.786) demonstrated that DLL3 expression can predict the survival outcome of IBC patients. Furthermore, the expression of DLL3 was related to a variety of tumor infiltrating immune cells (TIICs), particularly T cells regulatory (Tregs). Gene set enrichment analysis (GSEA) and immunohistochemistry (IHC) results indicated that DLL3 was closely related to p53 signaling pathway. CONCLUSIONS High expression of DLL3 was associated with poor prognosis and immune cell infiltration in IBC patients. Moreover, P53 signaling pathway may be the key pathway.
Collapse
Affiliation(s)
- Chong Yuan
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Kaili Chang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chenyue Xu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qingquan Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Zunguo Du
- Department of pathology, Huashan hospital, Fudan university, No.12 Middle Urumqi road, Shanghai 200032, China.
| |
Collapse
|
165
|
Gao C, Li H, Liu C, Xu X, Zhuang J, Zhou C, Liu L, Feng F, Sun C. Tumor Mutation Burden and Immune Invasion Characteristics in Triple Negative Breast Cancer: Genome High-Throughput Data Analysis. Front Immunol 2021; 12:650491. [PMID: 33968045 PMCID: PMC8097167 DOI: 10.3389/fimmu.2021.650491] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
In recent years, the emergence of immunotherapy has provided a new perspective for the treatment and management of triple-negative breast cancer (TNBC). However, the relationship between tumor mutation burden (TMB) and immune infiltration and the prognosis of TNBC remains unclear. In this study, to explore the immunogenicity of TNBC, we divided patients with TNBC into high and low TMB groups based on the somatic mutation data of TNBC in The Cancer Genome Atlas (TCGA), and screened out genes with mutation rate ≥10. Then, Kaplan-Meier survival analysis revealed that the 5-year survival rate of the high TMB group was much higher than that of the low TMB group and the two groups also showed differences in immune cell infiltration. Further exploration found that the FAT3 gene, which displays significant difference and a higher mutation rate between the two groups, is not only significantly related to the prognosis of TNBC patients but also exhibits difference in immune cell infiltration between the wild group and the mutant group of the FAT3 gene. The results of gene set enrichment analysis and drug sensitivity analysis further support the importance of the FAT3 gene in TNBC. This study reveals the characteristics of TMB and immune cell infiltration in triple-negative breast cancer and their relationship with prognosis, to provide new biomarkers and potential treatment options for the future treatment of TNBC. The FAT3 gene, as a risk predictor gene of TNBC, is considered a potential biological target and may provide new insight for the treatment of TNBC.
Collapse
Affiliation(s)
- Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaowei Xu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Chao Zhou
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijuan Liu
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fubin Feng
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
166
|
Montaño MA, Chagomerana MB, Borok M, Painschab M, Uldrick TS, Bender Ignacio RA. Impact of Antiretroviral Therapy on Cancer Treatment Outcomes among People Living with HIV in Low- and Middle-Income Countries: a Systematic Review. Curr HIV/AIDS Rep 2021; 18:105-116. [PMID: 33528741 PMCID: PMC10227634 DOI: 10.1007/s11904-021-00542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW The relationship between antiretroviral therapy (ART) and cancer treatment outcomes among people living with HIV (PLWH) in low- and middle-income countries (LMICs) is complex and poorly understood for many cancers. We aimed to summarize existing evidence from LMICs regarding the benefit of ART on cancer treatment-related outcomes. RECENT FINDINGS We included twelve observational studies that reported associations between ART status and cancer treatment outcomes among HIV-positive patients in LMICs. Most confirmed ART was associated with improved cancer treatment outcomes. Heterogeneity in cancers under study, outcome measurement, categorization of ART status, and reporting of HIV-related immune function made formal comparison between studies untenable. Where evaluated, ART generally has a positive effect on cancer outcomes in people with HIV in LMICs. However, there remains a substantial gap in the literature regarding the impact of ART on treatment outcomes for most cancer types. Future research should focus on the optimal timing and integration of ART and cancer treatment for PLWH with strategies applicable to constrained-resource settings.
Collapse
Affiliation(s)
- Michalina A Montaño
- Department of Biochemistry, University of Washington, Box 357236, Seattle, WA, 98195, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Maganizo B Chagomerana
- UNC Project-Malawi, Tidziwe Centre, Private Bag A-104, Lilongwe, Malawi
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Margaret Borok
- Department of Medicine, University of Zimbabwe College of Health Sciences, MP 167, Mount Pleasant, Harare, Zimbabwe
| | - Matthew Painschab
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, 101 Manning Drive, 7305, Chapel Hill, NC, 27514, USA
| | - Thomas S Uldrick
- Global Oncology Program, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., D5-310, Seattle, WA, 98109, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rachel A Bender Ignacio
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
- Harborview Medical Center, 325 9th Ave, 359929, Seattle, WA, 98104, USA
| |
Collapse
|
167
|
Abstract
IMPORTANCE Coping with the current and future burden of cancer requires an in-depth understanding of trends in cancer incidences and deaths. Estimated projections of cancer incidences and deaths will be important to guide future research funding allocations, health care planning, and health policy efforts. OBJECTIVE To estimate cancer incidences and deaths in the United States to the year 2040. DESIGN AND SETTING This cross-sectional study's estimated projection analysis used population growth projections and current population-based cancer incidence and death rates to calculate the changes in incidences and deaths to the year 2040. Cancer-specific incidences and deaths in the US were estimated for the most common cancer types. Demographic cancer-specific delay-adjusted incidence rates from the Surveillance, Epidemiology, and End Results Program were combined with US Census Bureau population growth projections (2016) and average annual percentage changes in incidence and death rates. Statistical analyses were performed from July 2020 to February 2021. MAIN OUTCOMES AND MEASURES Total cancer incidences and deaths to the year 2040. RESULTS This study estimated that the most common cancers in 2040 will be breast (364 000 cases) with melanoma (219 000 cases) becoming the second most common cancer; lung, third (208 000 cases); colorectal remaining fourth (147 000 cases); and prostate cancer dropping to the fourteenth most common cancer (66 000 cases). Lung cancer (63 000 deaths) was estimated to continue as the leading cause of cancer-related death in 2040, with pancreatic cancer (46 000 deaths) and liver and intrahepatic bile duct cancer (41 000 deaths) surpassing colorectal cancer (34 000 deaths) to become the second and third most common causes of cancer-related death, respectively. Breast cancer (30 000 deaths) was estimated to decrease to the fifth most common cause of cancer death. CONCLUSIONS AND RELEVANCE These findings suggest that there will be marked changes in the landscape of cancer incidence and deaths by 2040.
Collapse
Affiliation(s)
- Lola Rahib
- Cancer Commons, Mountain View, California
- Pancreatic Cancer Action Network, Manhattan Beach, California
| | - Mackenzie R. Wehner
- Department of Health Services Research, Department of Dermatology, MD Anderson Cancer Center, Houston, Texas
| | | | - Kevin T. Nead
- Department of Epidemiology, Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
168
|
Schreiber AR, Kagihara JA, Weiss JA, Nicklawsky A, Gao D, Borges VF, Kabos P, Diamond JR. Clinical Outcomes for Patients With Metastatic Breast Cancer Treated With Immunotherapy Agents in Phase I Clinical Trials. Front Oncol 2021; 11:640690. [PMID: 33816286 PMCID: PMC8010246 DOI: 10.3389/fonc.2021.640690] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/01/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immuno-oncology (IO) agents have demonstrated efficacy across many tumor types and have led to change in standard of care. In breast cancer, atezolizumab and pembrolizumab were recently FDA-approved in combination with chemotherapy specifically for patients with PD-L1-positive metastatic triple-negative breast cancer (TNBC). However, the single agent PD-1/PD-L1 inhibitors demonstrate only modest single agent efficacy in breast cancer. The purpose of this study was to investigate the efficacy of novel IO agents in patients with metastatic breast cancer (MBC), beyond TNBC, treated in phase I clinical trials at the University of Colorado. METHODS We performed a retrospective analysis using a database of patients with MBC who received treatment with IO agents in phase I/Ib clinical trials at the University of Colorado Hospital from January 1, 2012 to July 1, 2018. Patient demographics, treatments and clinical outcomes were obtained. RESULTS We identified 43 patients treated with an IO agent either as a single agent or in combination. The average age was 53 years; 55.8% had hormone receptor-positive/HER2-negative breast cancer, 39.5% TNBC and 4.7% HER2-positive. Patients received an average of 2 prior lines of chemotherapy (range 0-7) in the metastatic setting. Most patients (72.1%) received IO alone and 27.9% received IO plus chemotherapy. Median progression-free survival (PFS) was 2.3 months and median overall survival (OS) was 12.1 months. Patients remaining on study ≥ 6 months (20.9%) were more likely to be treated with chemotherapy plus IO compared to patients with a PFS < 6 months (77.8% v. 14.7%). No differences in number of metastatic sites, prior lines of chemotherapy, breast cancer subtype, absolute lymphocyte count, or LDH were identified between patients with a PFS ≥ 6 months vs. < 6 months. CONCLUSIONS Our phase I experience demonstrates benefit from IO therapy that was not limited to patients with TNBC and confirms improved efficacy from IO agents in combination with chemotherapy. A subset of patients with MBC treated in phase I clinical trials with an IO agent derived prolonged clinical benefit. Predictors of response to immunotherapy in breast cancer remain uncharacterized and further research is needed to identify these factors.
Collapse
Affiliation(s)
- Anna R. Schreiber
- Department of Medicine, University of Colorado Anschutz, Aurora, CO, United States
| | - Jodi A. Kagihara
- Department of Medicine, University of Colorado Cancer Center, Aurora, CO, United States
| | - Jennifer A. Weiss
- Department of Medicine, University of Colorado Anschutz, Aurora, CO, United States
| | - Andrew Nicklawsky
- University of Colorado School of Medicine, Aurora, CO, United States
| | - Dexiang Gao
- University of Colorado School of Medicine, Aurora, CO, United States
| | - Virginia F. Borges
- Department of Medicine, University of Colorado Cancer Center, Aurora, CO, United States
| | - Peter Kabos
- Department of Medicine, University of Colorado Cancer Center, Aurora, CO, United States
| | - Jennifer R. Diamond
- Department of Medicine, University of Colorado Cancer Center, Aurora, CO, United States
| |
Collapse
|
169
|
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: 6] [Impact Index Per Article: 2.0] [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
|
170
|
Gatti-Mays ME, Gameiro SR, Ozawa Y, Knudson KM, Hicks KC, Palena C, Cordes LM, Steinberg SM, Francis D, Karzai F, Lipkowitz S, Donahue RN, Jochems C, Schlom J, Gulley JL. Improving the Odds in Advanced Breast Cancer With Combination Immunotherapy: Stepwise Addition of Vaccine, Immune Checkpoint Inhibitor, Chemotherapy, and HDAC Inhibitor in Advanced Stage Breast Cancer. Front Oncol 2021; 10:581801. [PMID: 33747894 PMCID: PMC7977003 DOI: 10.3389/fonc.2020.581801] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/23/2020] [Indexed: 01/05/2023] Open
Abstract
Breast tumors commonly harbor low mutational burden, low PD-L1 expression, defective antigen processing/presentation, and an immunosuppressive tumor microenvironment (TME). In a malignancy mostly refractory to checkpoint blockade, there is an unmet clinical need for novel combination approaches that increase tumor immune infiltration and tumor control. Preclinical data have guided the development of this clinical trial combining 1) BN-Brachyury (a poxvirus vaccine platform encoding the tumor associated antigen brachyury), 2) bintrafusp alfa (a bifunctional protein composed of the extracellular domain of the TGF-βRII receptor (TGFβ "trap") fused to a human IgG1 anti-PD-L1), 3), entinostat (a class I histone deacetylase inhibitor), and 4) T-DM1 (ado-trastuzumab emtansine, a standard of care antibody-drug conjugate targeting HER2). We hypothesize that this tetratherapy will induce a robust immune response against HER2+ breast cancer with improved response rates through 1) expanding tumor antigen-specific effector T cells, natural killer cells, and immunostimulatory dendritic cells, 2) improving antigen presentation, and 3) decreasing inhibitory cytokines, regulatory T cells, and myeloid-derived suppressor cells. In an orthotopic HER2+ murine breast cancer model, tetratherapy induced high levels of antigen-specific T cell responses, tumor CD8+ T cell/Treg ratio, and augmented the presence of IFNγ- or TNFα-producing CD8+ T cells and IFNγ/TNFα bifunctional CD8+ T cells with increased cytokine production. Similar effects were observed in tumor CD4+ effector T cells. Based on this data, a phase 1b clinical trial evaluating the stepwise addition of BN-Brachyury, bintrafusp alfa, T-DM1 and entinostat in advanced breast cancer was designed. Arm 1 (TNBC) receives BN-Brachyury + bintrafusp alfa. Arm 2 (HER2+) receives T-DM1 + BN-Brachyury + bintrafusp alfa. After safety is established in Arm 2, Arm 3 (HER2+) will receive T-DM1 + BN-Brachyury + bintrafusp alfa + entinostat. Reimaging will occur every 2 cycles (1 cycle = 21 days). Arms 2 and 3 undergo research biopsies at baseline and after 2 cycles to evaluate changes within the TME. Peripheral immune responses will be evaluated. Co-primary objectives are response rate and safety. All arms employ a safety assessment in the initial six patients and a 2-stage Simon design for clinical efficacy (Arm 1 if ≥ three responses of eight then expand to 13 patients; Arms 2 and 3 if ≥ four responses of 14 then expand to 19 patients per arm). Secondary objectives include progression-free survival and changes in tumor infiltrating lymphocytes. Exploratory analyses include changes in peripheral immune cells and cytokines. To our knowledge, the combination of a vaccine, an anti-PD-L1 antibody, entinostat, and T-DM1 has not been previously evaluated in the preclinical or clinical setting. This trial (NCT04296942) is open at the National Cancer Institute (Bethesda, MD).
Collapse
Affiliation(s)
- Margaret E. Gatti-Mays
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sofia R. Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yohei Ozawa
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Karin M. Knudson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kristin C. Hicks
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Lisa M. Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Deneise Francis
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Stanley Lipkowitz
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Renee N. Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - James L. Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
171
|
Zhang Y, Xie X, Yeganeh PN, Lee DJ, Valle-Garcia D, Meza-Sosa KF, Junqueira C, Su J, Luo HR, Hide W, Lieberman J. Immunotherapy for breast cancer using EpCAM aptamer tumor-targeted gene knockdown. Proc Natl Acad Sci U S A 2021; 118:e2022830118. [PMID: 33627408 PMCID: PMC7936362 DOI: 10.1073/pnas.2022830118] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
New strategies for cancer immunotherapy are needed since most solid tumors do not respond to current approaches. Here we used epithelial cell adhesion molecule EpCAM (a tumor-associated antigen highly expressed on common epithelial cancers and their tumor-initiating cells) aptamer-linked small-interfering RNA chimeras (AsiCs) to knock down genes selectively in EpCAM+ tumors with the goal of making cancers more visible to the immune system. Knockdown of genes that function in multiple steps of cancer immunity was evaluated in aggressive triple-negative and HER2+ orthotopic, metastatic, and genetically engineered mouse breast cancer models. Gene targets were chosen whose knockdown was predicted to promote tumor neoantigen expression (Upf2, Parp1, Apex1), phagocytosis, and antigen presentation (Cd47), reduce checkpoint inhibition (Cd274), or cause tumor cell death (Mcl1). Four of the six AsiC (Upf2, Parp1, Cd47, and Mcl1) potently inhibited tumor growth and boosted tumor-infiltrating immune cell functions. AsiC mixtures were more effective than individual AsiC and could synergize with anti-PD-1 checkpoint inhibition.
Collapse
MESH Headings
- Animals
- Antigen Presentation/drug effects
- Antineoplastic Agents, Immunological/chemistry
- Antineoplastic Agents, Immunological/pharmacology
- Aptamers, Nucleotide/chemistry
- Aptamers, Nucleotide/immunology
- Aptamers, Nucleotide/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- CD47 Antigen/antagonists & inhibitors
- CD47 Antigen/genetics
- CD47 Antigen/immunology
- DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors
- DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics
- DNA-(Apurinic or Apyrimidinic Site) Lyase/immunology
- Epithelial Cell Adhesion Molecule/genetics
- Epithelial Cell Adhesion Molecule/immunology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/immunology
- Immunoconjugates/pharmacology
- Immunotherapy/methods
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Molecular Targeted Therapy
- Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/immunology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Phagocytosis/drug effects
- Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors
- Poly (ADP-Ribose) Polymerase-1/genetics
- Poly (ADP-Ribose) Polymerase-1/immunology
- RNA-Binding Proteins/antagonists & inhibitors
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/immunology
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/immunology
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/therapy
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Ying Zhang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Xuemei Xie
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Department of Lab Medicine and The Stem Cell Program, Boston Children's Hospital, Boston, MA 02115
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 300020 Tianjin, China
| | | | - Dian-Jang Lee
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - David Valle-Garcia
- Divison of Newborn Medicine and Epigenetics Program, Department of Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62210 Cuernavaca, México
| | - Karla F Meza-Sosa
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62210 Cuernavaca, México
| | - Caroline Junqueira
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
- René Rachou Institute, Oswaldo Cruz Foundation, 30190-002 Belo Horizonte, Brazil
| | - Jiayu Su
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Department of Lab Medicine and The Stem Cell Program, Boston Children's Hospital, Boston, MA 02115
- School of Life Sciences, Center for Bioinformatics, Peking University, 100871 Beijing, China
- Center for Statistical Science, Peking University, 100871 Beijing, China
| | - Hongbo R Luo
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Department of Lab Medicine and The Stem Cell Program, Boston Children's Hospital, Boston, MA 02115
| | - Winston Hide
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115;
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
172
|
O’Meara TA, Tolaney SM. Tumor mutational burden as a predictor of immunotherapy response in breast cancer. Oncotarget 2021; 12:394-400. [PMID: 33747355 PMCID: PMC7939529 DOI: 10.18632/oncotarget.27877] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 01/12/2023] Open
Abstract
Tumor mutational burden (TMB) is a promising tool to help define patients with triple-negative breast cancer (TNBC) most likely to benefit from immune checkpoint blockade (ICB) therapies. Roughly reflecting the degree of neo-antigens that tumors present to immune cells, TMB associates with multiple measures of tumoral immunogenicity and has proven clinically useful in cancers with relatively high mutation burden. TNBC carries higher TMB than other breast cancer subtypes, and recent data suggest that high-TMB TNBC cases may derive particular benefit from ICB in combination with chemotherapy (GeparNuevo, IMpassion130) or even ICB alone (KEYNOTE-119, TAPUR). Given the recent approval of pembrolizumab and atezolizumab in combination with chemotherapy for PD-L1-positive, metastatic TNBC, standardizing TMB calculation methods and cut-off values is of critical importance to deploy this clinical biomarker.
Collapse
Affiliation(s)
- Tess A. O’Meara
- Department of Internal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
173
|
Ciarametaro M, Houghton K, Wamble D, Dubois R. The Dollar or Disease Burden: Caps on Healthcare Spending May Save Money, but at What "Cost" to Patients? VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2021; 24:388-396. [PMID: 33641773 DOI: 10.1016/j.jval.2020.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/22/2020] [Accepted: 10/05/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Various strategies to address healthcare spending and medical costs continue to be debated and implemented in the United States. To date, these efforts have failed to adequately contain the growth of healthcare cost. An alternative strategy that has elicited rising interest among policymakers is budget caps. As budget caps become more prevalent, it is important to identify which features are needed to ensure success, both in terms of cost reduction and health improvement. METHODS We explored the impacts of different features of budget caps by comparing hypothetical service level and global budget caps across 3 annual budget cap growth strategies over a 10-year timeframe in 2005-2015 for 8 of the most commonly occurring conditions in the United States. Health was assessed by a measure of disease burden (disability-adjusted life years). RESULTS The results indicate that budget caps have the potential for creating savings but can also result in patient harm if not designed well. As a result of these findings, 5 principles were developed for designing budget caps and should guide the use of budget caps to address medical spending. CONCLUSIONS As public discussion grows about the use of budget caps to constrain health spending, it is critical to recognize that the budget cap design and the resulting healthcare provider behavior will determine whether there is potential harm to public health. Budget cap design should consider variability at the condition level, including patient population, improvements in health, treatment costs, and the innovations available, to both create savings and maximize patient health. In assessing the impact of healthcare spending caps on costs and disease burden, we demonstrate that budget cap design determines potential harm to public health.
Collapse
Affiliation(s)
| | - Katherine Houghton
- Research Triangle Institute Health Solutions (RTI-HS), Research Triangle Park, North Carolina, USA.
| | - David Wamble
- Research Triangle Institute Health Solutions (RTI-HS), Research Triangle Park, North Carolina, USA; Bristol Myers Squibb, Lawrence Township, New Jersey, USA
| | - Robert Dubois
- National Pharmaceutical Council, Washington, DC, USA
| |
Collapse
|
174
|
Zhang JQ, Plitas G. Immunotherapeutic strategies in breast cancer: A clinical update. J Surg Oncol 2021; 123:710-717. [PMID: 33155281 PMCID: PMC7889634 DOI: 10.1002/jso.26287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022]
Abstract
Immunotherapy has been incorporated into the standard of care for a wide range of malignancies. The study of tumor-infiltrating lymphocytes has emphasized the importance of the host antitumor immune response in the natural history of breast cancer. Recent clinical trials have used immunotherapeutic approaches to augment this response and improve outcomes for patients with breast cancer. Here, we review several current clinical trial data that indicate checkpoint blockade may mediate clinically significant responses.
Collapse
Affiliation(s)
- Jennifer Q. Zhang
- Department of Surgery, Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - George Plitas
- Department of Surgery, Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Ludwig Center at Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
175
|
Sequeira GR, Sahores A, Dalotto-Moreno T, Perrotta RM, Pataccini G, Vanzulli SI, Polo ML, Radisky DC, Sartorius CA, Novaro V, Lamb CA, Rabinovich GA, Salatino M, Lanari C. Enhanced Antitumor Immunity via Endocrine Therapy Prevents Mammary Tumor Relapse and Increases Immune Checkpoint Blockade Sensitivity. Cancer Res 2021; 81:1375-1387. [PMID: 33268529 DOI: 10.1158/0008-5472.can-20-1441] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/28/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
The role of active antitumor immunity in hormone receptor-positive (HR+) breast cancer has been historically underlooked. The aim of this study was to determine the contribution of the immune system to antiprogestin-induced tumor growth inhibition using a hormone-dependent breast cancer model. BALB/c-GFP+ bone marrow (BM) cells were transplanted into immunodeficient NSG mice to generate an immunocompetent NSG/BM-GFP+ (NSG-R) mouse model. Treatment with the antiprogestin mifepristone (MFP) inhibited growth of 59-2-HI tumors with similar kinetics in both animal models. Interestingly, MFP treatment reshaped the tumor microenvironment, enhancing the production of proinflammatory cytokines and chemokines. Tumors in MFP-treated immunocompetent mice showed increased infiltration of F4/80+ macrophages, natural killer, and CD8 T cells, displaying a central memory phenotype. Mechanistically, MFP induced immunogenic cell death (ICD) in vivo and in vitro, as depicted by the expression and subcellular localization of the alarmins calreticulin and HMGB-1 and the induction of an ICD gene program. Moreover, MFP-treated tumor cells efficiently activated immature dendritic cells, evidenced by enhanced expression of MHC-II and CD86, and induced a memory T-cell response, attenuating tumor onset and growth after re-challenge. Finally, MFP treatment increased the sensitivity of HR+ 59-2-HI tumor to PD-L1 blockade, suggesting that antiprogestins may improve immunotherapy response rates. These results contribute to a better understanding of the mechanisms underlying the antitumor effect of hormonal treatment and the rational design of therapeutic combinations based on endocrine and immunomodulatory agents in HR+ breast cancer. SIGNIFICANCE: Antiprogestin therapy induces immunogenic tumor cell death in PRA-overexpressing tumors, eliciting an adaptive immune memory response that protects mice from future tumor recurrence and increases sensitivity to PD-L1 blockade. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/5/1375/F1.large.jpg.
Collapse
Affiliation(s)
- Gonzalo R Sequeira
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
- Hospital Público de Gestión Descentralizada Dr. Arturo Oñativia, Ciudad de Salta, Salta, Argentina
| | - Ana Sahores
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tomás Dalotto-Moreno
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Ramiro M Perrotta
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Gabriela Pataccini
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | | | - María L Polo
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Derek C Radisky
- Mayo Clinic Comprehensive Cancer Center, Department of Cancer Biology, Jacksonville, Florida
| | - Carol A Sartorius
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Virginia Novaro
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Caroline A Lamb
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Salatino
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina.
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
176
|
Yu C, Zhou L, Zhang Q. Identification of unanimous immune subtypes for different hormone receptor phenotypes of human breast cancer with potential prognostic significance. Int Immunopharmacol 2021; 94:107473. [PMID: 33611062 DOI: 10.1016/j.intimp.2021.107473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/06/2021] [Accepted: 01/28/2021] [Indexed: 12/19/2022]
Abstract
The immunogenicity of the breast tumor microenvironment is clinically heterogeneous. The insight into the role of tumor-infiltrating lymphocytes (TILs) might serve as a biomarker to predict a survival benefit and enable optimal patient selection for immunotherapy. In this study, we aimed at characterizing the breast cancer immune subtypes linked to CD8 T cells and associating with the patient characteristics and clinical outcomes. We analyzed the immune gene signatures of human breast cancer using The Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) database profiling of 1092 breast tumor patients. We performed hierarchical clustering to the immune gene expression by applying hormone receptor status including triple negative breast cancer to categorize 66 immune-related genes in breast tumors. The separation was characterized by dividing the breast tumors into two major immune subtypes: predominant immune (PI) subtype and low immune subtype (LI). Our results showed that both PI and LI subtypes can be observed in the different hormone receptor phenotypes of breast tumors, and PI subtype accounted for 16% and LI subtype 20% in the breast tumor patients. The estimated odds for LI subtype breast tumors were significantly higher than PI subtype breast tumors in primary tumor stages. Our data demonstrated that the PI subtype breast tumors have significantly improved survival compared with LI subtype. Our findings provide a novel perspective of breast cancer immune subtypes linked to CD8 T cells. The immune subtypes will be a valuable resource for future research to identify clinically relevant biomarkers for precision immunotherapy.
Collapse
Affiliation(s)
- ChenWei Yu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Linlin Zhou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Qiuyu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
177
|
Sinn BV, Loibl S, Hanusch CA, Zahm DM, Sinn HP, Untch M, Weber K, Karn T, Becker C, Marmé F, Schmitt WD, Müller V, Schem C, Treue D, Stickeler E, Klauschen F, Burchardi N, Furlanetto J, van Mackelenbergh M, Fasching PA, Schneeweiss A, Denkert C. Immune-related Gene Expression Predicts Response to Neoadjuvant Chemotherapy but not Additional Benefit from PD-L1 Inhibition in Women with Early Triple-negative Breast Cancer. Clin Cancer Res 2021; 27:2584-2591. [PMID: 33593886 DOI: 10.1158/1078-0432.ccr-20-3113] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/05/2020] [Accepted: 02/11/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE We evaluated mRNA signatures to predict response to neoadjuvant PD-L1 inhibition in combination with chemotherapy in early triple-negative breast cancer. EXPERIMENTAL DESIGN Targeted mRNA sequencing of 2,559 transcripts was performed in formalin-fixed, paraffin-embedded samples from 162 patients of the GeparNuevo trial. We focused on validation of four predefined gene signatures and differential gene expression analyses for new predictive markers. RESULTS Two signatures [GeparSixto signature (G6-Sig) and IFN signature (IFN-Sig)] were predictive for treatment response in a multivariate model including treatment arm [G6-Sig: OR, 1.558; 95% confidence interval (CI), 1.130-2.182; P = 0.008 and IFN-Sig: OR, 1.695; 95% CI, 1.234-2.376; P = 0.002), while the CYT metric predicted pathologic complete response (pCR) in the durvalumab arm, and the proliferation-associated gene signature in the placebo arm. Expression of PD-L1 mRNA was associated with better response in both arms, indicating that increased levels of PD-L1 are a general predictor of neoadjuvant therapy response. In an exploratory analysis, we identified seven genes that were higher expressed in responders in the durvalumab arm, but not the placebo arm: HLA-A, HLA-B, TAP1, GBP1, CXCL10, STAT1, and CD38. These genes were associated with cellular antigen processing and presentation and IFN signaling. CONCLUSIONS Immune-associated signatures are associated with pCR after chemotherapy, but might be of limited use for the prediction of response to additional immune checkpoint blockade. Gene expressions related to antigen presentation and IFN signaling might be interesting candidates for further evaluation.
Collapse
Affiliation(s)
- Bruno V Sinn
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institut of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Sibylle Loibl
- German Breast Group Forschungs GmbH, Neu-Isenburg, Germany
| | - Claus A Hanusch
- Department of Gynecology, Rotkreuzklinikum München, Munich, Germany
| | - Dirk-Michael Zahm
- Department of Gynecology and Obstetrics, SRH Waldklinikum Gera GmbH, Gera, Germany
| | - Hans-Peter Sinn
- Department of Pathology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Michael Untch
- Department of Gynecology, Helios Kliniken Berlin-Buch, Berlin, Germany
| | - Karsten Weber
- German Breast Group Forschungs GmbH, Neu-Isenburg, Germany
| | - Thomas Karn
- Department of Gynecology and Obstetrics, Goethe-University, Frankfurt, Germany
| | - Clemens Becker
- Department of Pathology, Rotkreuzklinikum München, Munich, Germany
| | - Frederik Marmé
- Department of Gynecology, Universitätsklinikum Mannheim, Mannheim, Germany
| | - Wolfgang D Schmitt
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institut of Health, Berlin, Germany
| | - Volkmar Müller
- Department of Gynecology, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | - Denise Treue
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institut of Health, Berlin, Germany
| | - Elmar Stickeler
- Department of Gynecology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Frederik Klauschen
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institut of Health, Berlin, Germany
| | | | | | | | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Andreas Schneeweiss
- Nationales Centrum für Tumorerkrankungen, Universitätsklinikum und Deutsches Krebsforschungszentrum Heidelberg, Heidelberg, Germany
| | - Carsten Denkert
- Department of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), Marburg, Germany.
| |
Collapse
|
178
|
Wang R, Yang Y, Ye WW, Xiang J, Chen S, Zou WB, Wang XJ, Chen T, Cao WM. Case Report: Significant Response to Immune Checkpoint Inhibitor Camrelizumab in a Heavily Pretreated Advanced ER+/HER2- Breast Cancer Patient With High Tumor Mutational Burden. Front Oncol 2021; 10:588080. [PMID: 33634015 PMCID: PMC7900143 DOI: 10.3389/fonc.2020.588080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/18/2020] [Indexed: 12/28/2022] Open
Abstract
Endocrine treatment plus CDK4/6 inhibitors have become standard of care for estrogen receptor positive (ER+) breast cancer. Although immune checkpoint inhibitors (ICIs) have shown promising antitumor activity in a variety of cancer types, only limited success has been achieved for metastatic breast cancer (mBC) patients, especially the ER+ subtype, which usually exhibit lower tumor mutation burden (TMB) compared with other subtypes and therefore perceived as immunologically quiescent. Here we present a case of an ER+/HER2- but TMB-high mBC patient who had significant response to combination therapy with anti-PD-1 antibody camrelizumab and vinorelbine and obtained partial response (PR) with a progression-free survival (PFS) of 5 months after failure of multiple lines of therapy. Our case indicates that TMB may serve as a potential biomarker in immunotherapy selection for normally immunologically "cold" tumors such as ER+ mBC, also molecular monitoring during the whole treatment course plays an important role in patient management.
Collapse
Affiliation(s)
- Rong Wang
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | | | - Wei-Wu Ye
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | | | | | - Wei-Bin Zou
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiao-Jia Wang
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Tianhui Chen
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Department of Cancer Prevention, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
179
|
Goff SL, Danforth DN. The Role of Immune Cells in Breast Tissue and Immunotherapy for the Treatment of Breast Cancer. Clin Breast Cancer 2021; 21:e63-e73. [PMID: 32893093 PMCID: PMC7775885 DOI: 10.1016/j.clbc.2020.06.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022]
Abstract
Immune cells are present in normal breast tissue and in breast carcinoma. The nature and distribution of the immune cell subtypes in these tissues are reviewed to promote a better understanding of their important role in breast cancer prevention and treatment. We conducted a review of the literature to define the type, location, distribution, and role of immune cells in normal breast tissue and in in situ and invasive breast cancer. Immune cells in normal breast tissue are located predominantly within the epithelial component in breast ductal lobules. Immune cell subtypes representing innate immunity (NK, CD68+, and CD11c+ cells) and adaptive immunity (most commonly CD8+, but CD4+ and CD20+ as well) are present; CD8+ cells are the most common subtype and are primarily effector memory cells. Immune cells may recognize neoantigens and endogenous and exogenous ligands and may serve in chronic inflammation and immunosurveillance. Progression to breast cancer is characterized by increased immune cell infiltrates in tumor parenchyma and stroma, including CD4+ and CD8+ granzyme B+ cytotoxic T cells, B cells, macrophages and dendritic cells. Tumor-infiltrating lymphocytes in breast cancer may serve as prognostic indicators for response to chemotherapy and for survival. Experimental strategies of adoptive transfer of breast tumor-infiltrating lymphocyte may allow regression of metastatic breast cancer and encourage development of innovative T-cell strategies for the immunotherapy of breast cancer. In conclusion, immune cells in breast tissues play an important role throughout breast carcinogenesis. An understanding of these roles has important implications for the prevention and the treatment of breast cancer.
Collapse
Affiliation(s)
- Stephanie L Goff
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
180
|
Arora M, Kumari S, Singh J, Chopra A, Chauhan SS. Expression pattern, regulation, and clinical significance of TOX in breast cancer. Cancer Immunol Immunother 2021; 70:349-363. [PMID: 32757053 PMCID: PMC10992774 DOI: 10.1007/s00262-020-02689-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
Thymocyte selection-associated high mobility group box protein (TOX) is a transcription factor implicated in the regulation of T cell exhaustion during chronic infection and cancer. While TOX is being targeted for cancer immunotherapy, limited information is available about its significance in breast cancer and other solid tumors. We performed a comprehensive analysis of TOX gene expression, its epigenetic regulation, protein localization, relation to tumor infiltrating immune cell composition, and prognostic significance in breast cancer using publicly available datasets. Our results suggest an inverse correlation between TOX expression and DNA methylation in tumor cells. However, its expression is elevated in tumor infiltrating immune cells (TIICs), which may compensates for the total TOX levels in the tumor as a whole. Furthermore, higher TOX levels in tumors are associated with T cell exhaustion signatures along with presence of active inflammatory response, including elevated levels of T cell effector cytokines. Survival analysis also confirmed that higher expression of TOX is associated with better prognosis in breast cancer. Therefore, expression of TOX may serve as a novel prognostic marker for this malignancy.
Collapse
Affiliation(s)
- Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sarita Kumari
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Jay Singh
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
181
|
Yamashita N, Long M, Fushimi A, Yamamoto M, Hata T, Hagiwara M, Bhattacharya A, Hu Q, Wong KK, Liu S, Kufe D. MUC1-C integrates activation of the IFN-γ pathway with suppression of the tumor immune microenvironment in triple-negative breast cancer. J Immunother Cancer 2021; 9:jitc-2020-002115. [PMID: 33495298 PMCID: PMC7839859 DOI: 10.1136/jitc-2020-002115] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2020] [Indexed: 01/09/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have had a profound impact on the treatment of many tumors; however, their effectiveness against triple-negative breast cancers (TNBCs) has been limited. One factor limiting responsiveness of TNBCs to ICIs is a lack of functional tumor-infiltrating lymphocytes (TILs) in ‘non-inflamed’ or ‘cold’ tumor immune microenvironments (TIMEs), although by unknown mechanisms. Targeting MUC1-C in a mouse transgenic TNBC tumor model increases cytotoxic tumor-infiltrating CD8+ T cells (CTLs), supporting a role for MUC1-C in immune evasion. The basis for these findings and whether they extend to human TNBCs are not known. Methods Human TNBC cells silenced for MUC1-C using short hairpin RNAs (shRNAs) were analyzed for the effects of MUC1-C on global transcriptional profiles. Differential expression and rank order analysis was used for gene set enrichment analysis (GSEA). Gene expression was confirmed by quantitative reverse-transcription PCR and immunoblotting. The The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets were analyzed for effects of MUC1 on GSEA, cell-type enrichment, and tumor immune dysfunction and exclusion. Single-cell scRNA-seq datasets of TNBC samples were analyzed for normalized expression associations between MUC1 and selected genes within tumor cells. Results Our results demonstrate that MUC1-C is a master regulator of the TNBC transcriptome and that MUC1-C-induced gene expression is driven by STAT1 and IRF1. We found that MUC1-C activates the inflammatory interferon (IFN)-γ-driven JAK1→STAT1→IRF1 pathway and induces the IDO1 and COX2/PTGS2 effectors, which play key roles in immunosuppression. Involvement of MUC1-C in activating the immunosuppressive IFN-γ pathway was extended by analysis of human bulk and scRNA-seq datasets. We further demonstrate that MUC1 associates with the depletion and dysfunction of CD8+ T cells in the TNBC TIME. Conclusions These findings demonstrate that MUC1-C integrates activation of the immunosuppressive IFN-γ pathway with depletion of TILs in the TNBC TIME and provide support for MUC1-C as a potential target for improving TNBC treatment alone and in combination with ICIs. Of translational significance, MUC1-C is a druggable target with chimeric antigen receptor (CAR) T cells, antibody-drug conjugates (ADCs) and a functional inhibitor that are under clinical development.
Collapse
Affiliation(s)
- Nami Yamashita
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Atsushi Fushimi
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Masaaki Yamamoto
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Tsuyoshi Hata
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Masayuki Hagiwara
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Kwok-Kin Wong
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Donald Kufe
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
182
|
Subgroup analysis of nelipepimut-S plus GM-CSF combined with trastuzumab versus trastuzumab alone to prevent recurrences in patients with high-risk, HER2 low-expressing breast cancer. Clin Immunol 2021; 225:108679. [PMID: 33485895 DOI: 10.1016/j.clim.2021.108679] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
HER2-targeted therapy has not benefited patients with low levels of HER2 expression; however, combination therapy may be effective. Primary analysis of a phase IIb trial investigating the HER2-derived vaccine nelipepimut-S (NPS) did not benefit the intention-to-treat population, but subset analysis showed a benefit in triple-negative breast cancer (TNBC) patients. The subset analysis of this multicenter, randomized, single-blind, phase IIb trial identified significant improvement in 36-month disease-free survival (DFS) between NPS (n = 55) and placebo (n = 44) in TNBC (HR 0.25, p = 0.01) and those who express HLA-A24 (HR 0.41, p = 0.05). The TNBC cohort demonstrated improved 36-month DFS in those with HER2 1+ expression (HR 0.17, p = 0.01), HLA-A24 positivity (HR 0.08, p < 0.01), or in those who received neoadjuvant chemotherapy (HR 0.21, p < 0.01). NPS vaccination with trastuzumab was associated with improved 36-month DFS among patients with TNBC. The observed benefit to this high-risk subgroup warrants confirmation in a phase III trial.
Collapse
|
183
|
Sanchez K, Kim I, Chun B, Pucilowska J, Redmond WL, Urba WJ, Martel M, Wu Y, Campbell M, Sun Z, Grunkemeier G, Chang SC, Bernard B, Page DB. Multiplex immunofluorescence to measure dynamic changes in tumor-infiltrating lymphocytes and PD-L1 in early-stage breast cancer. Breast Cancer Res 2021; 23:2. [PMID: 33413574 PMCID: PMC7788790 DOI: 10.1186/s13058-020-01378-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/03/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The H&E stromal tumor-infiltrating lymphocyte (sTIL) score and programmed death ligand 1 (PD-L1) SP142 immunohistochemistry assay are prognostic and predictive in early-stage breast cancer, but are operator-dependent and may have insufficient precision to characterize dynamic changes in sTILs/PD-L1 in the context of clinical research. We illustrate how multiplex immunofluorescence (mIF) combined with statistical modeling can be used to precisely estimate dynamic changes in sTIL score, PD-L1 expression, and other immune variables from a single paraffin-embedded slide, thus enabling comprehensive characterization of activity of novel immunotherapy agents. METHODS Serial tissue was obtained from a recent clinical trial evaluating loco-regional cytokine delivery as a strategy to promote immune cell infiltration and activation in breast tumors. Pre-treatment biopsies and post-treatment tumor resections were analyzed by mIF (PerkinElmer Vectra) using an antibody panel that characterized tumor cells (cytokeratin-positive), immune cells (CD3, CD8, CD163, FoxP3), and PD-L1 expression. mIF estimates of sTIL score and PD-L1 expression were compared to the H&E/SP142 clinical assays. Hierarchical linear modeling was utilized to compare pre- and post-treatment immune cell expression, account for correlation of time-dependent measurement, variation across high-powered magnification views within each subject, and variation between subjects. Simulation methods (Monte Carlo, bootstrapping) were used to evaluate the impact of model and tissue sample size on statistical power. RESULTS mIF estimates of sTIL and PD-L1 expression were strongly correlated with their respective clinical assays (p < .001). Hierarchical linear modeling resulted in more precise estimates of treatment-related increases in sTIL, PD-L1, and other metrics such as CD8+ tumor nest infiltration. Statistical precision was dependent on adequate tissue sampling, with at least 15 high-powered fields recommended per specimen. Compared to conventional t-testing of means, hierarchical linear modeling was associated with substantial reductions in enrollment size required (n = 25➔n = 13) to detect the observed increases in sTIL/PD-L1. CONCLUSION mIF is useful for quantifying treatment-related dynamic changes in sTILs/PD-L1 and is concordant with clinical assays, but with greater precision. Hierarchical linear modeling can mitigate the effects of intratumoral heterogeneity on immune cell count estimations, allowing for more efficient detection of treatment-related pharmocodynamic effects in the context of clinical trials. TRIAL REGISTRATION NCT02950259 .
Collapse
Affiliation(s)
- Katherine Sanchez
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - Isaac Kim
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - Brie Chun
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - Joanna Pucilowska
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - Walter J Urba
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - Maritza Martel
- Department of Pathology, Providence Portland Medical Center, Portland, OR, USA
| | - Yaping Wu
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - Mary Campbell
- Department of Pathology, Providence Portland Medical Center, Portland, OR, USA
| | - Zhaoyu Sun
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
| | - Gary Grunkemeier
- Medical Data Research Center, Providence Health & Services, Portland, OR, USA
| | - Shu Ching Chang
- Medical Data Research Center, Providence Health & Services, Portland, OR, USA
| | - Brady Bernard
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA
- Providence Cancer Institute, Portland, OR, USA
| | - David B Page
- Earle A. Chiles Research Institute, 4805 N.E. Glisan St., North Tower, Suite 2N87, Portland, OR, 97213, USA.
- Providence Cancer Institute, Portland, OR, USA.
| |
Collapse
|
184
|
Comprehensive analysis reveals a prognostic and therapeutic biomarker CD3D in the breast carcinoma microenvironment. Biosci Rep 2021; 41:227413. [PMID: 33350431 PMCID: PMC7791551 DOI: 10.1042/bsr20202898] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/29/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
Breast carcinoma (BRCA) is the most common carcinoma among women worldwide. Despite the great progress achieved in early detection and treatment, morbidity and mortality rates remain high. In the present study, we make a systematic analysis of BRCA using TCGA database by applying CIBERSORT and ESTIMATE computational methods, uncovered CD3D as a prognostic biomarker by intersection analysis of univariate COX and protein–protein interaction (PPI). It revealed that high CD3D expression was strongly associated with poor survival of BRCA, based on The Cancer Genome Atlas (TCGA) database and online websites. Gene Set Enrichment Analysis (GSEA) revealed that the high CD3D expression group was mainly enriched for the immune-related pathways and the low CD3D expression group was mainly enriched for metabolic-related activities. Based on CIBERSORT analysis, the difference test and correlation test suggested that CD3D had a strong correlation with T cells, particularly CD8 + T cells, which indicated that CD3D up-regulation may increase T cell immune infiltration in the TME and induce antitumor immunity by activating T lymphocytes. Furthermore, the correlation analysis showed that CD3D expression had a strongly positive correlation with immune checkpoints, which indicating that the underlying mechanism involves CD3D mediated regulation of T cell functions in BRCA, and single cell RNA-seq analysis revealed that CD3D correlate with CD8 + T cells and it is itself highly expressed in CD8 + T cells. In summary, we identified a prognostic biomarker CD3D in BRCA, which was associated with lymphocyte infiltration, immune checkpoints and could be developed for innovative therapeutics of BRCA.
Collapse
|
185
|
Jiang M, Li W, Zhu C, Li X, Zhang J, Luo Z, Qin B, Du Y, Luo L, You J. Perdurable PD-1 blockage awakes anti-tumor immunity suppressed by precise chemotherapy. J Control Release 2021; 329:1023-1036. [DOI: 10.1016/j.jconrel.2020.10.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/10/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
|
186
|
Liang YK, Deng ZK, Chen MT, Qiu SQ, Xiao YS, Qi YZ, Xie Q, Wang ZH, Jia SC, Zeng D, Lin HY. CXCL9 Is a Potential Biomarker of Immune Infiltration Associated With Favorable Prognosis in ER-Negative Breast Cancer. Front Oncol 2021; 11:710286. [PMID: 34527583 PMCID: PMC8435794 DOI: 10.3389/fonc.2021.710286] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/12/2021] [Indexed: 02/05/2023] Open
Abstract
The chemokine CXCL9 (C-X-C motif chemokine ligand 9) has been reported to be required for antitumour immune responses following immune checkpoint blockade. In this study, we sought to investigate the potential value of CXCL9 according to immune responses in patients with breast cancer (BC). A variety of open-source databases and online tools were used to explore the expression features and prognostic significance of CXCL9 in BC and its correlation with immune-related biomarkers followed by subsequent verification with immunohistochemistry experiments. The CXCL9 mRNA level was found to be significantly higher in BC than in normal tissue and was associated with better survival outcomes in patients with ER-negative tumours. Moreover, CXCL9 is significantly correlated with immune cell infiltration and immune-related biomarkers, including CTLA4, GZMB, LAG3, PDCD1 and HAVCR2. Finally, we performed immunohistochemistry with breast cancer tissue samples and observed that CXCL9 is highly expressed in the ER-negative subgroup and positively correlated with the immune-related factors LAG3, PD1, PDL1 and CTLA4 to varying degrees. These findings suggest that CXCL9 is an underlying biomarker for predicting the status of immune infiltration in ER-negative breast cancer.
Collapse
Affiliation(s)
- Yuan-ke Liang
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), Shantou, China
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou, China
| | - Ze-kun- Deng
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), Shantou, China
| | | | - Si-qi Qiu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou, China
- Clinical Research Center, Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China
| | - Ying-sheng Xiao
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou, China
- Department of Thyroid Surgery Shantou Central Hospital, Shantou, China
| | - Yu-zhu Qi
- SUMC, Shantou, China
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | | | - De Zeng
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou, China
- Department of Medical Oncology, Cancer Hospital of SUMC, Shantou, China
- *Correspondence: Hao-yu Lin, ; De Zeng,
| | - Hao-yu Lin
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), Shantou, China
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou, China
- *Correspondence: Hao-yu Lin, ; De Zeng,
| |
Collapse
|
187
|
Abbasi A, Pakravan N, Hassan ZM. Hyaluronic Acid Improves Hydrogen Peroxide Modulatory Effects on Calcium Channel and Sodium-Potassium Pump in 4T1 Breast Cancer Cell Line. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8681349. [PMID: 33456676 PMCID: PMC7787766 DOI: 10.1155/2020/8681349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 11/19/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022]
Abstract
Maintaining homeostasis of ion concentrations is critical in cancer cells. Under hypoxia, the levels of channels and pumps in cancer cells are more active than normal cells suggesting ion channels as a suitable therapeutic target. One of the contemporary ways for cancer therapy is oxidative stress. However, the effective concentration of oxidative stress on tumor cells has been reported to be toxic for normal cells as well. In this study, we benefited from the modifying effects of hyaluronic acid (HA) on H2O2, as a free radical source, to make a gradual release of oxidative stress on cancer cells while preventing/decreasing damage to normal cells under normoxia and hypoxic conditions. To do so, we initially investigated the optimal concentration of HA antioxidant capacity by the DPPH test. In the next step, we found optimum H2O2 dose by treating the 4T1 breast cancer cell line with increasing concentrations (0, 10, 20, 50,100, 200, 500, and 1000 μM) of H2O2 alone or H2O2 + HA (83%) for 24 hrs. The calcium channel and the sodium-potassium pumps were then evaluated by measuring the levels of calcium, sodium, and potassium ions using an atomic absorption flame spectrophotometer. The results revealed that treatment with H2O2 or H2O2+ HA led to an intracellular increase of calcium, sodium, and potassium in the normoxic and hypoxic circumstances in a dose-dependent manner. It is noteworthy that H2O2 + HA treatment had more favorable and controllable effects compared with H2O2 alone. Moreover, HA optimizes the antitumor effect of oxidative stress exerted by H2O2 making H2O2 + HA suitable for clinical use in cancer treatment along with chemotherapy.
Collapse
Affiliation(s)
- Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nafiseh Pakravan
- Department of Immunology, Medical School, Alborz University of Medical Sciences, Karaj, Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
188
|
Miglietta F, Cona MS, Dieci MV, Guarneri V, La Verde N. An overview of immune checkpoint inhibitors in breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:452-472. [PMID: 36046385 PMCID: PMC9400749 DOI: 10.37349/etat.2020.00029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Although breast cancer is not traditionally considered an immunogenic type of tumor, the combination of immunotherapy and chemotherapy has recently emerged as a novel treatment option in triple-negative subtype in the advanced setting and other similar combinations of immune checkpoint inhibitors with chemotherapy are expected to become part of the neoadjuvant management in the near future. In addition, encouraging results have been observed with the combination of immune checkpoint blockade with diverse biological agents, including anti-HER2 agents, CDK 4/6 inhibitors, PARP-inhibitors. The present review summarized the available evidence coming from clinical trials on the role of immune checkpoint inhibitors in the management of breast cancer, both in advanced and early setting.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Maria Silvia Cona
- Department of Oncology, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Nicla La Verde
- Department of Oncology, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| |
Collapse
|
189
|
Gómez-Aleza C, Nguyen B, Yoldi G, Ciscar M, Barranco A, Hernández-Jiménez E, Maetens M, Salgado R, Zafeiroglou M, Pellegrini P, Venet D, Garaud S, Trinidad EM, Benítez S, Vuylsteke P, Polastro L, Wildiers H, Simon P, Lindeman G, Larsimont D, Van den Eynden G, Velghe C, Rothé F, Willard-Gallo K, Michiels S, Muñoz P, Walzer T, Planelles L, Penninger J, Azim HA, Loi S, Piccart M, Sotiriou C, González-Suárez E. Inhibition of RANK signaling in breast cancer induces an anti-tumor immune response orchestrated by CD8+ T cells. Nat Commun 2020; 11:6335. [PMID: 33303745 PMCID: PMC7728758 DOI: 10.1038/s41467-020-20138-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Most breast cancers exhibit low immune infiltration and are unresponsive to immunotherapy. We hypothesized that inhibition of the receptor activator of nuclear factor-κB (RANK) signaling pathway may enhance immune activation. Here we report that loss of RANK signaling in mouse tumor cells increases leukocytes, lymphocytes, and CD8+ T cells, and reduces macrophage and neutrophil infiltration. CD8+ T cells mediate the attenuated tumor phenotype observed upon RANK loss, whereas neutrophils, supported by RANK-expressing tumor cells, induce immunosuppression. RANKL inhibition increases the anti-tumor effect of immunotherapies in breast cancer through a tumor cell mediated effect. Comparably, pre-operative single-agent denosumab in premenopausal early-stage breast cancer patients from the Phase-II D-BEYOND clinical trial (NCT01864798) is well tolerated, inhibits RANK pathway and increases tumor infiltrating lymphocytes and CD8+ T cells. Higher RANK signaling activation in tumors and serum RANKL levels at baseline predict these immune-modulatory effects. No changes in tumor cell proliferation (primary endpoint) or other secondary endpoints are observed. Overall, our preclinical and clinical findings reveal that tumor cells exploit RANK pathway as a mechanism to evade immune surveillance and support the use of RANK pathway inhibitors to prime luminal breast cancer for immunotherapy. Receptor activator of nuclear factor-κB (RANK)/RANK-ligand (RANKL) signaling regulates the tumor-immune crosstalk. Here the authors show that systemic RANKL inhibition promotes CD8 + T cell infiltration in patients with early breast cancer and that loss of RANK signaling in tumor cells drives a T cell-dependent anti-tumor response in preclinical models.
Collapse
Affiliation(s)
- Clara Gómez-Aleza
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Bastien Nguyen
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Guillermo Yoldi
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Marina Ciscar
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.,Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alexandra Barranco
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.,Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Marion Maetens
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
| | - Maria Zafeiroglou
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Pasquale Pellegrini
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - David Venet
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Eva M Trinidad
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Sandra Benítez
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Peter Vuylsteke
- Department of Medical Oncology, Université Catholique de Louvain, CHU UCL, Namur, site Sainte-Elisabeth, Namur, Belgium
| | - Laura Polastro
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Hans Wildiers
- Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
| | - Philippe Simon
- Department of Obstetrics and Gynaecology, Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Geoffrey Lindeman
- Peter MacCallum Cancer Centre, The Walter and Eliza Hall Institute of Medical Research and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gert Van den Eynden
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Chloé Velghe
- Clinical Trial Supporting Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Françoise Rothé
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Stefan Michiels
- Service de Biostatistique et D'Epidémiologie, Gustave Roussy, CESP, U1018, Université Paris-Sud, Faculté de Médcine, Université Paris-Saclay, Villejuif, France
| | - Purificación Muñoz
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS, Université Claude Bernard, Lyon, France
| | - Lourdes Planelles
- BiOncotech Therapeutics, Parc Cientific Universitat, Valencia, Spain
| | - Josef Penninger
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Hatem A Azim
- Division of Hematology/Oncology, Department of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sherene Loi
- Peter MacCallum Cancer Centre, The Walter and Eliza Hall Institute of Medical Research and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Martine Piccart
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium. .,Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Eva González-Suárez
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain. .,Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|
190
|
Alfonso S, Jenner AL, Craig M. Translational approaches to treating dynamical diseases through in silico clinical trials. CHAOS (WOODBURY, N.Y.) 2020; 30:123128. [PMID: 33380031 DOI: 10.1063/5.0019556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
The primary goal of drug developers is to establish efficient and effective therapeutic protocols. Multifactorial pathologies, including dynamical diseases and complex disorders, can be difficult to treat, given the high degree of inter- and intra-patient variability and nonlinear physiological relationships. Quantitative approaches combining mechanistic disease modeling and computational strategies are increasingly leveraged to rationalize pre-clinical and clinical studies and to establish effective treatment strategies. The development of clinical trials has led to new computational methods that allow for large clinical data sets to be combined with pharmacokinetic and pharmacodynamic models of diseases. Here, we discuss recent progress using in silico clinical trials to explore treatments for a variety of complex diseases, ultimately demonstrating the immense utility of quantitative methods in drug development and medicine.
Collapse
Affiliation(s)
- Sofia Alfonso
- Department of Physiology, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Adrianne L Jenner
- Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Morgan Craig
- Department of Physiology, McGill University, Montreal, Quebec H3A 0G4, Canada
| |
Collapse
|
191
|
Wang Y, Zhang Y, Du Y, Zhou M, Hu Y, Zhang S. Emerging roles of N6-methyladenosine (m 6A) modification in breast cancer. Cell Biosci 2020; 10:136. [PMID: 33292526 PMCID: PMC7690038 DOI: 10.1186/s13578-020-00502-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
N6-Methyladenosine (m6A) is the most abundant, dynamic, and reversible epigenetic RNA modification that is found in coding and non-coding RNAs. Emerging studies have shown that m6A and its regulators affect multiple steps in RNA metabolism and play broad roles in various cancers. Worldwide, breast cancer is the most prevalent cancer in female. It is a very heterogeneous disease characterized by genetic and epigenetic variations in tumor cells. Increasing evidence has shown that the dysregulation of m6A-related effectors, as methyltransferases, demethylases, and m6A binding proteins, is pivotal in breast cancer pathogenesis. In this review, we have summarized the most up-to-date research on the biological functions of m6A modification in breast cancer and have discussed the potential clinical applications and future directions of m6A modification as a biomarker as well as a therapeutic target of breast cancer.
Collapse
Affiliation(s)
- Yanyan Wang
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| | - Yujie Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yushen Du
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Meiqi Zhou
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yue Hu
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Suzhan Zhang
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| |
Collapse
|
192
|
Xu X, Zhang M, Xu F, Jiang S. Wnt signaling in breast cancer: biological mechanisms, challenges and opportunities. Mol Cancer 2020; 19:165. [PMID: 33234169 PMCID: PMC7686704 DOI: 10.1186/s12943-020-01276-5] [Citation(s) in RCA: 263] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling is a highly conserved signaling pathway that plays a critical role in controlling embryonic and organ development, as well as cancer progression. Genome-wide sequencing and gene expression profile analyses have demonstrated that Wnt signaling is involved mainly in the processes of breast cancer proliferation and metastasis. The most recent studies have indicated that Wnt signaling is also crucial in breast cancer immune microenvironment regulation, stemness maintenance, therapeutic resistance, phenotype shaping, etc. Wnt/β-Catenin, Wnt-planar cell polarity (PCP), and Wnt-Ca2+ signaling are three well-established Wnt signaling pathways that share overlapping components and play different roles in breast cancer progression. In this review, we summarize the main findings concerning the relationship between Wnt signaling and breast cancer and provide an overview of existing mechanisms, challenges, and potential opportunities for advancing the therapy and diagnosis of breast cancer.
Collapse
Affiliation(s)
- Xiufang Xu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Miaofeng Zhang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Faying Xu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Shaojie Jiang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| |
Collapse
|
193
|
Wang J, Zhang X, Li J, Ma X, Feng F, Liu L, Wu J, Sun C. ADRB1 was identified as a potential biomarker for breast cancer by the co-analysis of tumor mutational burden and immune infiltration. Aging (Albany NY) 2020; 13:351-363. [PMID: 33234738 PMCID: PMC7835009 DOI: 10.18632/aging.104204] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
Breast cancer (BRCA) has traditionally been considered as having poor immunogenicity and is characterized by relatively low tumor mutational burden (TMB). Improving immunogenicity may improve the response to clinical immunotherapy of BRCA. However, the relationship between TMB, immune infiltration, and prognosis in BRCA remains unclear. We aimed to explore their interrelations and potential biomarkers. In this study, based on somatic mutation data of BRCA from The Cancer Genome Atlas (TCGA), patients were categorized into high and low TMB groups utilizing the TMB values. CIBERSOFT algorithm indicated significant infiltration of activated partial immune cells in high TMB group. Besides, ADRB1 had been identified as a prognosis-related immune gene in the mutant genes by the combination of the ImmPort database and the univariate Cox analysis. ADRB1 mutation was associated with lower TMB and manifested a satisfactory clinical prognosis. Various database applications (Gene Set Enrichment Analysis, Tumor IMmune Estimation Resource, Connectivity Map, KnockTF) supported the selection of treatment strategies targeting ADRB1. In conclusion, TMB was not an independent prognostic factor for BRCA and high TMB was more likely to activate a partial immune response. ADRB1 was identified as a potential biomarker and may provide new insights for co-therapy of BRCA.
Collapse
Affiliation(s)
- Jia Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, P. R. China
| | - Xiaolu Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, P. R. China
| | - Jie Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, P. R. China
| | - Xiaoran Ma
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, P. R. China
| | - Fubin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, Shandong, P. R. China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, Shandong, P. R. China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, P. R. China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, Shandong, P. R. China.,Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| |
Collapse
|
194
|
Quintela-Fandino M, Holgado E, Manso L, Morales S, Bermejo B, Colomer R, Apala JV, Blanco R, Muñoz M, Caleiras E, Iranzo V, Martinez M, Dominguez O, Hornedo J, Gonzalez-Cortijo L, Cortes J, Gasol Cudos A, Malon D, Lopez-Alonso A, Moreno-Ortíz MC, Mouron S, Mañes S. Immuno-priming durvalumab with bevacizumab in HER2-negative advanced breast cancer: a pilot clinical trial. Breast Cancer Res 2020; 22:124. [PMID: 33176887 PMCID: PMC7661209 DOI: 10.1186/s13058-020-01362-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/25/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Preclinical research suggests that the efficacy of immune checkpoint inhibitors in breast cancer can be enhanced by combining them with antiangiogenics, particularly in a sequential fashion. We sought to explore the efficacy and biomarkers of combining the anti-PD-L1 durvalumab plus the antiangiogenic bevacizumab after bevacizumab monotherapy for advanced HER2-negative breast cancer. METHODS Patients had advanced HER2-negative disease that progressed while receiving single-agent bevacizumab maintenance as a part of a previous chemotherapy plus bevacizumab regimen. Treatment consisted of bi-weekly durvalumab plus bevacizumab (10 mg/kg each i.v.). Peripheral-blood mononuclear cells (PBMCs) were obtained before the first durvalumab dose and every 4 weeks and immunophenotyped by flow-cytometry. A fresh pre-durvalumab tumor biopsy was obtained; gene-expression studies and immunohistochemical staining to assess vascular normalization and characterize the immune infiltrate were conducted. Patients were classified as "non-progressors" if they had clinical benefit (SD/PR/CR) at 4 months. The co-primary endpoints were the changes in the percentage T cell subpopulations in PBMCs in progressors versus non-progressors, and PFS/OS time. RESULTS Twenty-six patients were accrued. Median PFS and OS were 3.5 and 11 months; a trend for a longer OS was detected for the hormone-positive subset (19.8 versus 7.4 months in triple-negatives; P = 0.11). Clinical benefit rate at 2 and 4 months was 60% and 44%, respectively, without significant differences between hormone-positive and triple-negative (P = 0.73). Non-progressors' tumors displayed vascular normalization features as a result of previous bevacizumab, compared with generally abnormal patterns observed in progressors. Non-progressors also showed increased T-effector and T-memory signatures and decreased TREG signatures in gene expression studies in baseline-post-bevacizumab-tumors compared with progressors. Notably, analysis of PBMC populations before durvalumab treatment was concordant with the findings in tumor samples and showed a decreased percentage of circulating TREGs in non-progressors. CONCLUSIONS This study reporting on sequential bevacizumab+durvalumab in breast cancer showed encouraging activity in a heavily pre-treated cohort. The correlative studies agree with the preclinical rationale supporting an immunopriming effect exerted by antiangiogenic treatment, probably by reducing TREGs cells both systemically and in tumor tissue. The magnitude of this benefit should be addressed in a randomized setting. TRIAL REGISTRATION (www.clinicaltrials.gov): NCT02802098 . Registered on June 16, 2020.
Collapse
MESH Headings
- Adult
- Aged
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Bevacizumab/administration & dosage
- Bevacizumab/adverse effects
- Breast/pathology
- Breast Neoplasms/blood
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Disease Progression
- Female
- Humans
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Middle Aged
- Pilot Projects
- Progression-Free Survival
- Proof of Concept Study
- Receptor, ErbB-2/analysis
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain.
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain.
- Medical Oncology Department, Hospital Universitario Quiron, Pozuelo de Alarcon, Spain.
| | - Esther Holgado
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Luis Manso
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Serafin Morales
- Medical Oncology Department, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Hospital Clínico Universitario, Valencia, Spain
- INCLIVA, Valencia, Spain
- CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Ramon Colomer
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan V Apala
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Raquel Blanco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Manuel Muñoz
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit - Biotechnology Program, CNIO - Spanish National Cancer Research Center, Madrid, Spain
| | - Vega Iranzo
- CIBERONC, Instituto Carlos III, Madrid, Spain
- Medical Oncology Department, Hospital General Universitario de Valencia, Valencia, Spain
- Medicine Department, Universitat de Valencia, Valencia, Spain
| | - Mario Martinez
- Pathology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Orlando Dominguez
- Genomics Core Unit - Biotechnology Program, CNIO - Spanish National Cancer Research Center, Madrid, Spain
| | - Javier Hornedo
- Medical Oncology Department, Hospital Universitario Quiron, Pozuelo de Alarcon, Spain
| | | | - Javier Cortes
- ION Institute of Oncology, Quironsalud Group - Madrid & Barcelona, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Ariadna Gasol Cudos
- Medical Oncology Department, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Diego Malon
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Antonio Lopez-Alonso
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - María C Moreno-Ortíz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Silvana Mouron
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain.
| |
Collapse
|
195
|
Gil Del Alcazar CR, Alečković M, Polyak K. Immune Escape during Breast Tumor Progression. Cancer Immunol Res 2020; 8:422-427. [PMID: 32238387 DOI: 10.1158/2326-6066.cir-19-0786] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunotherapy using checkpoint inhibitors is one of the most promising current cancer treatment strategies. However, in breast cancer, its success has been limited to a subset of patients with triple-negative disease, whose durability of observed responses remain unclear. The lack of detailed understanding of breast tumor immune evasion mechanisms and the treatment of patients with highly heterogeneous metastatic disease contribute to these disappointing results. Here we discuss the current knowledge about immune-related changes during breast tumor progression, with special emphasis on the in situ-to-invasive breast carcinoma transition that may represent a key step of immunoediting in breast cancer. Comprehensive characterization of early-stage disease and better understanding of immunologic drivers of disease progression will likely expand the tools available for immunotherapy and improve patient stratification.
Collapse
Affiliation(s)
- Carlos R Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
196
|
Won KA, Spruck C. Triple‑negative breast cancer therapy: Current and future perspectives (Review). Int J Oncol 2020; 57:1245-1261. [PMID: 33174058 PMCID: PMC7646583 DOI: 10.3892/ijo.2020.5135] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for 10-15% of all breast cancer cases. TNBCs lack estrogen and progesterone receptors and express low levels of HER2, and therefore do not respond to hormonal or anti-HER2 therapies. TNBC is a particularly aggressive form of breast cancer that generally displays poorer prognosis compared to other breast cancer subtypes. TNBC is chemotherapy sensitive, and this treatment remains the standard of care despite its limited benefit. Recent advances with novel agents have been made for specific subgroups with PD-L1+ tumors or germline Brca-mutated tumors. However, only a fraction of these patients responds to immune checkpoint or PARP inhibitors and even those who do respond often develop resistance and relapse. Various new agents and combination strategies have been explored to further understand molecular and immunological aspects of TNBC. In this review, we discuss clinical trials in the management of TNBC as well as perspectives for potential future treatments.
Collapse
Affiliation(s)
| | - Charles Spruck
- Tumor Initiation and Maintenance Program, NCI‑Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
197
|
Wu ZZ, Wang S, Yang QC, Wang XL, Yang LL, Liu B, Sun ZJ. Increased Expression of SHMT2 Is Associated With Poor Prognosis and Advanced Pathological Grade in Oral Squamous Cell Carcinoma. Front Oncol 2020; 10:588530. [PMID: 33163414 PMCID: PMC7581701 DOI: 10.3389/fonc.2020.588530] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
This study focused on the expression of mitochondrial serine hydroxymethyltransferase (SHMT2) in oral squamous cell carcinoma (OSCC) and its correlation with clinical traits and the prognosis of OSCC patients. Immunochemical staining and Western blotting were used to quantify the expression of SHMT2 and related immune markers in OSCC. Using OSCC microarrays and The Cancer Genome Atlas (TCGA) database, we evaluated the association between SHMT2 and various clinical traits. We found that increased expression of SHMT2 was detected in OSCC and correlated with advanced pathological grade and recurrence of OSCC. By a multivariate Cox proportional hazard model, high expression of SHMT2 was shown to indicate a negative prognosis. In addition, in the OSCC microenvironment, increasing the expression of SHMT2 was associated with high expression levels of programmed cell death-ligand 1 (PD-L1), CKLF-like MARVEL transmembrane domain containing 6 (CMTM6), V-type immunoglobulin domain-containing suppressor (VISTA), B7-H4, Slug, and CD317. In the future, more effort will be required to investigate the role of SHMT2 in the OSCC microenvironment.
Collapse
Affiliation(s)
- Zhi-Zhong Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qi-Chao Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiao-Long Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lei-Lei Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
198
|
Xu Y, Rogers CJ. Physical Activity and Breast Cancer Prevention: Possible Role of Immune Mediators. Front Nutr 2020; 7:557997. [PMID: 33134306 PMCID: PMC7578403 DOI: 10.3389/fnut.2020.557997] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
There is strong evidence that physical activity (PA) reduces risk, recurrence, and mortality from breast cancer. Emerging data suggest that PA induces changes in inflammatory and immune mediators that may contribute to beneficial effects on breast cancer outcomes. Thus, the goal of this review was to evaluate the evidence linking the protective benefit of PA to modulation of immune responses in breast cancer. A literature search was conducted to identify studies that evaluated the impact of PA on tumor and immune outcomes in breast cancer patients and in mammary tumor models. Nineteen studies investigated the effect of PA interventions on cancer immune outcomes using preclinical breast cancer models. Tumor growth was reduced in 11 studies, unchanged in three studies, and increased in one study. Spontaneous metastasis was reduced in two studies and survival was improved in four studies. Frequently assessed immune outcomes include splenic cell number and function, circulating inflammatory cytokines, and intratumoral immune cells and inflammatory markers. Circulating inflammatory cytokine responses were heterogeneous in preclinical models. Within the tumor microenvironment (TME), several studies documented a change in the infiltration of immune cells with an increase in effector cells and a reduction in immune suppressive cells. Twenty-three studies investigated the effect of PA interventions on immune outcomes in breast cancer patients. Thirteen studies used aerobic PA interventions and 10 studies used a combination of aerobic and resistance exercise interventions. Cycling and treadmill activities were the most commonly used PA modalities. Circulating immune cells and inflammatory cytokines were the most frequently assessed immune outcomes in the clinical studies. Among the 19 studies that evaluated a PA intervention during the post treatment period, 10 reported a reduction in the levels of at least one inflammatory cytokine. No inflammatory cytokines were quantified in the three studies that evaluated a PA intervention during treatment with chemotherapy. Immune outcomes within the tumor were assessed in only one study performing a PA intervention prior to surgery. Results from preclinical and clinical studies suggest that PA exerts heterogeneous effects on inflammatory cytokines, but may alter the gene expression profile and immune infiltrates in the tumor which may result in a reduction in immunosuppressive factors. However, additional studies are needed to better understand the effect of PA on immune outcomes in the TME.
Collapse
Affiliation(s)
- Yitong Xu
- Intercollege Graduate Degree Program in Integrative and Biomedical Physiology, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Connie J Rogers
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States.,Center for Molecular Immunology and Infectious Disease, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States.,Penn State Cancer Institute, Hershey, PA, United States
| |
Collapse
|
199
|
Tolaney SM, Barroso-Sousa R, Keenan T, Li T, Trippa L, Vaz-Luis I, Wulf G, Spring L, Sinclair NF, Andrews C, Pittenger J, Richardson ET, Dillon D, Lin NU, Overmoyer B, Partridge AH, Van Allen E, Mittendorf EA, Winer EP, Krop IE. Effect of Eribulin With or Without Pembrolizumab on Progression-Free Survival for Patients With Hormone Receptor-Positive, ERBB2-Negative Metastatic Breast Cancer: A Randomized Clinical Trial. JAMA Oncol 2020; 6:1598-1605. [PMID: 32880602 PMCID: PMC7489368 DOI: 10.1001/jamaoncol.2020.3524] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022]
Abstract
Importance Prior studies have shown that only a small proportion of patients with hormone receptor (HR)-positive metastatic breast cancer (MBC) experience benefit from programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors given as monotherapy. There are data suggesting that activity may be greater with combination strategies. Objective To compare the efficacy of eribulin plus pembrolizumab vs eribulin alone in patients with HR-positive, ERBB2 (formerly HER2)-negative MBC. Design, Setting, and Participants Multicenter phase 2 randomized clinical trial of patients with HR-positive, ERBB2-negative MBC who had received 2 or more lines of hormonal therapy and 0 to 2 lines of chemotherapy. Interventions Patients were randomized 1:1 to eribulin, 1.4 mg/m2 intravenously, on days 1 and 8 plus pembrolizumab, 200 mg/m2 intravenously, on day 1 of a 21-day cycle or eribulin alone. At time of progression, patients in the eribulin monotherapy arm could cross over and receive pembrolizumab monotherapy. Main Outcomes and Measures The primary end point was progression-free survival (PFS). Secondary end points were objective response rate (ORR) and overall survival (OS). Exploratory analyses assessed the association between PFS and PD-L1 status, tumor-infiltrating lymphocytes (TILs), tumor mutational burden (TMB), and genomic alterations. Results Eighty-eight patients started protocol therapy; the median (range) age was 57 (30-76) years, median (range) number of prior lines of chemotherapy was 1 (0-2), and median (range) number of prior lines of hormonal therapy was 2 (0-5). Median follow-up was 10.5 (95% CI, 0.4-22.8) months. Median PFS and ORR were not different between the 2 groups (PFS, 4.1 vs 4.2 months; hazard ratio, 0.80; 95% CI, 0.50-1.26; P = .33; ORR, 27% vs 34%, respectively; P = .49). Fourteen patients started crossover treatment with pembrolizumab; 1 patient experienced stable disease. All-cause adverse events occurred in all patients (grade ≥3, 65%) including 2 treatment-related deaths in the combination group, both from immune-related colitis in the setting of sepsis, attributed to both drugs. The PD-L1 22C3 assay was performed on archival tumor samples in 65 patients: 24 (37%) had PD-L1-positive tumors. Analysis indicated that PD-L1 status, TILs, TMB, and genomic alterations were not associated with PFS. Conclusions and Relevance In this randomized clinical trial of patients with HR-positive, ERBB2-negative MBC, the addition of pembrolizumab to eribulin did not improve PFS, ORR, or OS compared with eribulin alone in either the intention-to-treat or PD-L1-positive populations. Further efforts to explore the benefits of adding checkpoint inhibition to chemotherapy among less heavily pretreated patients are needed. Trial Registration ClinicalTrials.gov Identifier: NCT03051659.
Collapse
Affiliation(s)
- Sara M. Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Romualdo Barroso-Sousa
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Oncology Center, Hospital Sírio-Libanês, Brasília, Brazil
| | - Tanya Keenan
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Boston, Massachusetts
| | - Tianyu Li
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lorenzo Trippa
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Gerburg Wulf
- Medical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Laura Spring
- Medical Oncology, Massachusetts General Hospital, Boston
| | | | - Chelsea Andrews
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jessica Pittenger
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Deborah Dillon
- Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Nancy U. Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Beth Overmoyer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ann H. Partridge
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eliezer Van Allen
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Boston, Massachusetts
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Eric P. Winer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ian E. Krop
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
200
|
Zagorulya M, Duong E, Spranger S. Impact of anatomic site on antigen-presenting cells in cancer. J Immunother Cancer 2020; 8:e001204. [PMID: 33020244 PMCID: PMC7537336 DOI: 10.1136/jitc-2020-001204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 12/24/2022] Open
Abstract
Checkpoint blockade immunotherapy (CBT) can induce long-term clinical benefits in patients with advanced cancer; however, response rates to CBT vary by cancer type. Cancers of the skin, lung, and kidney are largely responsive to CBT, while cancers of the pancreas, ovary, breast, and metastatic lesions to the liver respond poorly. The impact of tissue-resident immune cells on antitumor immunity is an emerging area of investigation. Recent evidence indicates that antitumor immune responses and efficacy of CBT depend on the tissue site of the tumor lesion. As myeloid cells are predominantly tissue-resident and can shape tumor-reactive T cell responses, it is conceivable that tissue-specific differences in their function underlie the tissue-site-dependent variability in CBT responses. Understanding the roles of tissue-specific myeloid cells in antitumor immunity can open new avenues for treatment design. In this review, we discuss the roles of tissue-specific antigen-presenting cells (APCs) in governing antitumor immune responses, with a particular focus on the contributions of tissue-specific dendritic cells. Using the framework of the Cancer-Immunity Cycle, we examine the contributions of tissue-specific APC in CBT-sensitive and CBT-resistant carcinomas, highlight how these cells can be therapeutically modulated, and identify gaps in knowledge that remain to be addressed.
Collapse
Affiliation(s)
- Maria Zagorulya
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ellen Duong
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Stefani Spranger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|