151
|
Simmons CE, Brezden-Masley C, McCarthy J, McLeod D, Joy AA. Positive progress: current and evolving role of immune checkpoint inhibitors in metastatic triple-negative breast cancer. Ther Adv Med Oncol 2020; 12:1758835920909091. [PMID: 33014143 PMCID: PMC7517981 DOI: 10.1177/1758835920909091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) represents an aggressive breast cancer subtype with historically poor overall outcomes, due primarily to a lack of effective targeted agents. Chemotherapy has been the primary treatment approach, although immune checkpoint inhibitors (ICIs) are currently being investigated to improve patient outcomes. This review examines the clinical implications of current evidence on the use of ICIs for the treatment of metastatic TNBC. Methods: Our systematic search identified two phase III and five phase I/II trials reporting on the efficacy of ICIs used as monotherapy or combined with chemotherapy for the treatment of metastatic TNBC. Results: The phase III IMpassion 130 trial showed a significant improvement in median progression-free survival in the intent-to-treat (net 1.7 months, p = 0.002) and PD-L1-positive populations (net 2.5 months, p < 0.001) for the addition of first-line atezolizumab versus placebo to nab-paclitaxel in metastatic TNBC. Although median overall survival was not significantly improved in patients receiving atezolizumab overall [net 2.3 months, hazard ratio (HR) 0.86, 95% confidence interval (CI) 0.72–1.02, p = 0.078], numerical improvements in the PD-L1-positive population were compelling (net 7.0 months, HR 0.71; 95% CI 0.54–0.93). Toxicity profiles were as expected, and no new safety signals were observed. Pembrolizumab monotherapy did not significantly improve overall survival in similar patients that had received prior treatment in KEYNOTE-119. Conclusions: Atezolizumab plus nab-paclitaxel represents a potential new first-line standard of care for patients with metastatic PD-L1-positive TNBC. Other ICIs used as monotherapy, or combined with chemotherapy for advanced TNBC, as well as their use for earlier stage disease, are areas of ongoing investigation.
Collapse
Affiliation(s)
- Christine E Simmons
- Division of Medical Oncology, BC Cancer Agency-Vancouver, 600 West 10th Avenue, Vancouver, British Columbia, V5Z 4E6, Canada
| | | | - Joy McCarthy
- Dr H. Bliss Murphy Cancer Centre, St. John's, Newfoundland, Canada
| | | | | |
Collapse
|
152
|
Tan R, Li H, Huang Z, Zhou Y, Tao M, Gao X, Xu Y. Neural Functions Play Different Roles in Triple Negative Breast Cancer (TNBC) and non-TNBC. Sci Rep 2020; 10:3065. [PMID: 32080331 PMCID: PMC7033128 DOI: 10.1038/s41598-020-60030-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/04/2020] [Indexed: 11/09/2022] Open
Abstract
Triple negative breast cancer (TNBC) represents the most malignant subtype of breast cancer, and yet our understanding about its unique biology remains elusive. We have conducted a comparative computational analysis of transcriptomic data of TNBC and non-TNBC (NTNBC) tissue samples from the TCGA database, focused on genes involved in neural functions. Our main discoveries are: (1) while both subtypes involve neural functions, TNBC has substantially more up-regulated neural genes than NTNBC, suggesting that TNBC is more complex than NTNBC; (2) non-neural functions related to cell-microenvironment interactions and intracellular damage processing are key inducers of the neural genes in both TNBC and NTNBC, but the inducer-responder relationships are different in the two cancer subtypes; (3) key neural functions such as neural crest formation are predicted to enhance adaptive immunity in TNBC while glia development, along with a few other neural functions, induce both innate and adaptive immunity in NTNBC. These results reveal key differences in the biology between the two cancer subtypes, particularly in terms of the roles that neural functions play. Our findings may open new doors for further investigation of the distinct biology of TNBC vs. NTNBC.
Collapse
Affiliation(s)
- Renbo Tan
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Cancer Systems Biology Center, The China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Haoyang Li
- Cancer Systems Biology Center, The China-Japan Union Hospital of Jilin University, Changchun, 130033, China.,College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Zhenyu Huang
- Cancer Systems Biology Center, The China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Yi Zhou
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, 30602, USA
| | - Mingxin Tao
- Cancer Systems Biology Center, The China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xin Gao
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, 23955, Saudi Arabia
| | - Ying Xu
- Cancer Systems Biology Center, The China-Japan Union Hospital of Jilin University, Changchun, 130033, China. .,College of Computer Science and Technology, Jilin University, Changchun, 130012, China. .,Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, 30602, USA.
| |
Collapse
|
153
|
Kagihara JA, Andress M, Diamond JR. Nab-paclitaxel and atezolizumab for the treatment of PD-L1-positive, metastatic triple-negative breast cancer: review and future directions. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:59-65. [PMID: 32190733 DOI: 10.1080/23808993.2020.1730694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction Breast cancer is the most common malignancy in women in the United States and triple-negative breast cancer (TNBC) accounts for 15-20%. The standard of care for metastatic TNBC has been limited to cytotoxic chemotherapy with modest efficacy. TNBC is associated with high levels of tumor-infiltrating lymphocytes and PD-L1 expression, supporting the investigation of immune checkpoint inhibitors in this breast cancer subtype. Areas Covered This review summarizes the clinical data supporting the use of atezolizumab and nab-paclitaxel in the treatment of metastatic PD-L1-positive TNBC. It examines the pharmacology and toxicity profile of the combination in patients with metastatic TNBC. Expert Opinion The addition of atezolizumab to nab-paclitaxel prolonged progression-free survival in both the intention-to-treat and PD-L1-positive subgroups in the first line setting in patients with metastatic TNBC. The IMpassion 130 trial led to FDA-approval of this combination in patients with PD-L1-positive, metastatic TNBC and represents the first approval of immunotherapy for TNBC. This work supports ongoing investigations of other immunotherapy combinations in TNBC, predictive biomarker development and immunotherapy in patients with early stage TNBC. Immunotherapy combinations in TNBC have the potential to lead to improved survival in this group of patients with high risk disease.
Collapse
Affiliation(s)
- Jodi A Kagihara
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| | - Michelle Andress
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| | - Jennifer R Diamond
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| |
Collapse
|
154
|
Hodge J, Wang F, Wang J, Liu Q, Saaoud F, Wang Y, Singh UP, Chen H, Luo M, Ai W, Fan D. Overexpression of microRNA-155 enhances the efficacy of dendritic cell vaccine against breast cancer. Oncoimmunology 2020; 9:1724761. [PMID: 32117588 PMCID: PMC7028336 DOI: 10.1080/2162402x.2020.1724761] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
MicroRNA 155 (miR-155) plays important roles in the regulation of the development and functions of a variety of immune cells. We previously revealed a vital role of miR-155 in regulating the function of dendritic cells (DCs) in breast cancer. miR-155 deficiency in DCs impaired their maturation, migration, cytokine production, and ability to activate T cells. In the current study, to exploit the therapeutic value of miR-155 for breast cancer, we examined the impact of overexpression of miR-155 on antitumor responses generated by DC vaccines. We boosted miR-155 expression in DCs by generating a miR-155 transgenic mouse strain (miR-155tg) or using lentivirus transduction. DCs overexpressing miR-155 exhibited enhanced functions in response to tumor antigens. Using miR-155 overexpressing DCs, we generated a DC vaccine and found that the vaccine resulted in enhanced antitumor immunity against established breast cancers in mice, demonstrated by increased effector T cells in the mice, suppressed tumor growth, and drastically reduced lung metastasis. Our current study suggests that in future DC vaccine development for breast cancer or other solid tumors, introducing forced miR155 overexpression in DCs via various approaches such as viral transduction or nanoparticle delivery, as well as including other adjuvant agents such as TLR ligands or immune stimulating cytokines, may unleash the full therapeutic potential of the DC vaccines.
Collapse
Affiliation(s)
- Johnie Hodge
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Fang Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.,Department of Microbiology, Air Force Medical University, Xi'an, China
| | - Junfeng Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Qing Liu
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Fatma Saaoud
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Udai P Singh
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Hexin Chen
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Ming Luo
- Center for Diagnostics and Therapeutics, Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - Walden Ai
- Department of Biology and Environmental Health Science, Benedict College, Columbia, SC, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
155
|
Immune checkpoint expression, microsatellite instability, and mutational burden: Identifying immune biomarker phenotypes in uterine cancer. Gynecol Oncol 2019; 156:393-399. [PMID: 31882243 DOI: 10.1016/j.ygyno.2019.11.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/06/2019] [Accepted: 11/26/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Increasing grade of endometrioid endometrial cancer (EEC) is associated with aggressive behavior and poor prognosis. The traditional classification system is limited in its ability to guide treatment planning and prognostication. We identify distinct immune biomarker phenotypes using known markers of immunogenicity to identify patients who may benefit from immune therapy (IT). METHODS 621 tumors were analyzed by multiplatform profiling. NextGen sequencing was performed on 592 genes. Tumor mutational burden (TMB) was defined as high (H) ≥10mutations/megabase. Microsatellite Instability (MSI) by NGS was ≥46 loci. PD-L1 positivity was ≥2+, >5% by IHC. Chi-square tests were used. RESULTS Overall, MSI-H was found in 33% of EECs, most frequent in grade 3 (G3), followed by grade 2 (G2) and grade 1 (G1) tumors (G3: 37%, G2: 32%, G1: 22%, p = 0.007). TMB-H was identified in 25% of EECs. TMB-H was most common in G3, followed by G2 and G1 tumors (G3: 34%, G2: 23%, G1: 13%, p = 0.006). Overall, PD-L1 expression was found in 5.5% of EECs. G3 EECs had the most frequent PD-L1 expression, followed by G2 and G1 tumors (G3: 12%, G2: 3.0%, G1: 0.9%, p < 0.0001). We identified POLE mutations in 4.5% (28/618). All POLE mutated tumors harbored TMB-H phenotypes but MSI-H and PD-L1 were only present in 10.7% and 14.8% of tumors respectively, suggesting upregulation of T-cell immune response in only a fraction of POLE mutated EECs. Triple negative (TN) biomarker phenotype (ER-/PR-/Her2-) was evaluated as a potential surrogate marker of tumor immunogenicity. We identified TN phenotype in 4% of G1 EEC compared with 9% in G2 and 33% in G3, suggesting loss of hormone expression and possible greater immunogenicity with increasing tumor grade. CONCLUSIONS High grade tumors appear to be more immunogenic than low grade tumors and may preferentially benefit from IT.
Collapse
|
156
|
Fabbri F, Salvi S, Bravaccini S. Know your enemy: Genetics, aging, exposomic and inflammation in the war against triple negative breast cancer. Semin Cancer Biol 2019; 60:285-293. [PMID: 31669505 DOI: 10.1016/j.semcancer.2019.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 01/11/2023]
Abstract
Triple negative breast cancer (TNBC) is one of the most biologically aggressive and very often lethal breast disease. It is one of the most puzzling women malignancies, and it currently appears not to be a good candidate to a standardized, unanimously accepted and sufficiently active therapeutic strategy. Fast proliferating and poorly differentiated, it is histopathologically heterogeneous, and even more ambiguous at the molecular level, offering few recurrent actionable targets to the clinicians. It is a formidable and vicious enemy that requires a huge investigational effort to find its vital weak spots. Here, we provide a broad review of "old but gold" biological aspects that taken together may help in finding new TNBC management strategies. A better and updated knowledge of the origins, war-like tactics, refueling mechanisms and escape routes of TNBC, will help in moving the decisive steps towards its final defeat.
Collapse
Affiliation(s)
- Francesco Fabbri
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Samanta Salvi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
157
|
Stamm H, Oliveira-Ferrer L, Grossjohann EM, Muschhammer J, Thaden V, Brauneck F, Kischel R, Müller V, Bokemeyer C, Fiedler W, Wellbrock J. Targeting the TIGIT-PVR immune checkpoint axis as novel therapeutic option in breast cancer. Oncoimmunology 2019; 8:e1674605. [PMID: 31741778 DOI: 10.1080/2162402x.2019.1674605] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/30/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022] Open
Abstract
Immune checkpoints are intensively investigated as targets in cancer therapy. T-cell immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT) and its ligand poliovirus receptor (PVR) are recently emerging as novel promising targets in immunotherapy. Here, we show that high expression of PVR represents an independent prognostic marker being associated with poor outcome for breast cancer patients. Furthermore, PVR mRNA, as well as protein expression, is associated with more aggressive breast cancer subtypes such as HER2 positive and triple-negative breast cancer. In vitro, blocking TIGIT or PVR resulted in enhanced immune cell-mediated lysis of breast cancer cell lines SKBR-3, MDA-MB-231, MDA-MB-468, and BT549 and additionally increased the cytotoxic effects of a bispecific T cell engager BiTE® antibody construct targeting EGFR. Taken together, our data identify the immune checkpoint factor PVR as a novel prognostic marker in breast cancer and indicate that blocking the TIGIT-PVR axis might represent a novel therapeutic option for the treatment of breast cancer patients.
Collapse
Affiliation(s)
- Hauke Stamm
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Eva-Maria Grossjohann
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Muschhammer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Thaden
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Brauneck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
158
|
Mangia A, Saponaro C, Vagheggini A, Opinto G, Centonze M, Vicenti C, Popescu O, Pastena M, Giotta F, Silvestris N. Should Tumor Infiltrating Lymphocytes, Androgen Receptor, and FOXA1 Expression Predict the Clinical Outcome in Triple Negative Breast Cancer Patients? Cancers (Basel) 2019; 11:cancers11091393. [PMID: 31540486 PMCID: PMC6769726 DOI: 10.3390/cancers11091393] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/28/2019] [Accepted: 09/09/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are a valuable indicator of the immune microenvironment that plays the central role in new anticancer drugs. TILs have a strong prognostic role in triple negative breast cancer (TNBC). Little is known about the interaction with the androgen receptor (AR) and forkhead box A1 (FOXA1). We analyzed the relationships between TIL levels, AR, and FOXA1 expression and their clinical significance in TNBC patients. Further, we investigated their interaction with other biomarkers like programmed cell death ligand-1 (PD-L1), breast cancer type 1 susceptibility protein (BRCA1), poly (ADP-Ribose) polymerase 1 (PARP1), and Na+/H+ exchanger regulatory factor 1 (NHERF1). The expression of the proteins was evaluated by immunohistochemistry in 124 TNBC samples. TILs were performed adhering to International TILs Working Group 2014 criteria. Cox proportional hazards models were also used to identify risk factors associated with poor prognosis. Multivariate analysis identified TILs as independent prognostic factor of disease free survival (DFS; p = 0.045). A Kaplan-Meyer analysis revealed that the patients with high TILs had a better DFS compared to patients with low TILs (p = 0.037), and the phenotypes TILs-/AR+ and TILs-/FOXA1- had a worse DFS (p = 0.032, p = 0.001 respectively). AR was associated with FOXA1 expression (p = 0.007), and the tumors FOXA1+ presented low levels of TILs (p = 0.028). A poor DFS was observed for AR+/FOXA1+ tumors compared to other TNBCs (p = 0.0117). Low TILs score was associated with poor patients' survival, and TILs level in combination with AR or FOXA1 expression affected patient's clinical outcome. In addition, AR+/FOXA1+ phenotype identified a specific subgroup of TNBC patients with poor prognosis. These data may suggest new ways of therapeutic intervention to support current treatments.
Collapse
Affiliation(s)
- Anita Mangia
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Concetta Saponaro
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Alessandro Vagheggini
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola (FC), Italy.
| | - Giuseppina Opinto
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Matteo Centonze
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Chiara Vicenti
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Ondina Popescu
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Maria Pastena
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Francesco Giotta
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II" of Bari, 70124 Bari, Italy.
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro' of Bari, 70124 Bari, Italy.
| |
Collapse
|
159
|
Zeindler J, Soysal SD, Piscuoglio S, Ng CKY, Mechera R, Isaak A, Weber WP, Muenst S, Kurzeder C. Nectin-4 Expression Is an Independent Prognostic Biomarker and Associated With Better Survival in Triple-Negative Breast Cancer. Front Med (Lausanne) 2019; 6:200. [PMID: 31572728 PMCID: PMC6753375 DOI: 10.3389/fmed.2019.00200] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/27/2019] [Indexed: 12/29/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) represents about 10-20% of all invasive breast cancers and is associated with a poor prognosis. The nectin cell adhesion protein 4 (Nectin-4) is a junction protein involved in the formation and maintenance of cell junctions. Nectin-4 has previously shown to be expressed in about 60% of TNBC as well as in TNBC metastases, but to be absent in normal breast tissue, which makes it a potential specific target for TNBC therapy. Previous studies have shown an association of Nectin-4 protein expression with worse prognosis in TNBC in a small patient cohort. The aim of our study was to explore the role of Nectin-4 in TNBC and confirm its impact on survival in a larger TNBC patient cohort. Material and Methods: We performed immunohistochemical staining for Nectin-4 on a tissue microarray encompassing 148 TNBC cases with detailed clinical annotation and outcomes data. Results: A high expression of Nectin-4 was present in 86 (58%) of the 148 TNBC cases. In multivariate survival analysis, high expression of Nectin-4 was associated with a significantly better overall survival when compared with low expression of Nectin-4 (p < 0.001). Nectin-4-high expression was also significantly associated with a lower tumor stage (p = 0.025) and pN0 lymph node stage (p = 0.034). Conclusion: Our results confirm that expression of Nectin-4 serves as a potential prognostic marker in TNBC and is associated with a significantly better overall survival. In addition, Nectin-4 represents a potential target in TNBC, and its role in molecular defined breast cancer subtype should be investigated in larger patient cohorts.
Collapse
Affiliation(s)
- Jasmin Zeindler
- Breast Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Savas Deniz Soysal
- Visceral Surgery Research Laboratory, Clarunis, Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Visceral Surgery Research Laboratory, Clarunis, Department of Biomedicine, University of Basel, Basel, Switzerland.,Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Charlotte K Y Ng
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Robert Mechera
- Visceral Surgery Research Laboratory, Clarunis, Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Andrej Isaak
- Department of Vascular and Endovascular Surgery, University Hospital Basel, Basel, Switzerland
| | - Walter Paul Weber
- Breast Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Simone Muenst
- Breast Center, University Hospital Basel and University of Basel, Basel, Switzerland.,Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Christian Kurzeder
- Breast Center, University Hospital Basel and University of Basel, Basel, Switzerland.,Department of Obstetrics and Gynecology, University Hospital of Basel, Basel, Switzerland
| |
Collapse
|
160
|
Pinho MP, Patente TA, Flatow EA, Sallusto F, Barbuto JAM. Frequency determination of breast tumor-reactive CD4 and CD8 T cells in humans: unveiling the antitumor immune response. Oncoimmunology 2019; 8:1607674. [PMID: 31413917 DOI: 10.1080/2162402x.2019.1607674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/21/2019] [Accepted: 04/07/2019] [Indexed: 01/11/2023] Open
Abstract
As cancer immunotherapy gains importance, the determination of a patient's ability to react to his/her tumor is unquestionably relevant. Though the presence of T cells that recognize specific tumor antigens is well established, the total frequency of tumor-reactive T cells in humans is difficult to assess, especially due to the lack of broad analysis techniques. Here, we describe a strategy that allows this determination, in both CD4 and CD8 compartments, using T cell proliferation induced by tumor cell-lysate pulsed dendritic cells as the readout. All 12 healthy donor tested had circulating CD4 and CD8 tumor cell-reactive T cells. The detection of these T cells, not only in the naïve but also in the memory compartment, can be seen as an evidence of tumor immunosurveillance in humans. As expected, breast cancer patients had higher frequencies of blood tumor-reactive T cells, but with differences among breast cancer subtypes. Interestingly, the frequency of blood tumor-reactive T cells in patients did not correlate to the frequency of infiltrating tumor-reactive T cells, highlighting the danger of implying a local tumor response from blood obtained data. In conclusion, these data add T cell evidence to immunosurveillance in humans, confirm that immune parameters in blood may be misleading and describe a tool to follow the tumor-specific immune response in patients and, thus, to design better immunotherapeutic approaches.
Collapse
Affiliation(s)
- Mariana Pereira Pinho
- Department of Immunology, Institute of Biomedical Sciences of the University of Sao Paulo, Sao Paulo, Brazil
| | - Thiago Andrade Patente
- Department of Immunology, Institute of Biomedical Sciences of the University of Sao Paulo, Sao Paulo, Brazil
| | - Elizabeth Alexandra Flatow
- Department of Immunology, Institute of Biomedical Sciences of the University of Sao Paulo, Sao Paulo, Brazil
| | - Federica Sallusto
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland.,Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
161
|
Lyu H, Li M, Jiang Z, Liu Z, Wang X. Correlate the TP53 Mutation and the HRAS Mutation with Immune Signatures in Head and Neck Squamous Cell Cancer. Comput Struct Biotechnol J 2019; 17:1020-1030. [PMID: 31428295 PMCID: PMC6695281 DOI: 10.1016/j.csbj.2019.07.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Although immunotherapy has emerged as an effective therapeutic strategy for various cancers including head and neck squamous cell carcinomas (HNSCCs), only a subset of patients can benefit from such therapy. Hence, it is pressing to discover predictive biomarkers for cancer immunotherapy response. TP53 and HRAS mutations frequently occur in HNSCC and correlate with a worse prognosis in HNSCC. We extensively characterized the associations of TP53 mutations and HRAS mutations with HNSCC immunity based on multiple cancer genomics datasets. We compared the enrichment levels of 20 immune signatures between TP53-mutated and TP53-wildtype HNSCCs, and between HRAS-mutated and HRAS-wildtype HNSCCs, and found that TP53 mutations were associated with depressed immune signatures while HRAS mutations were associated with enhanced immune signatures in HNSCC. Moreover, we found multiple p53- and RAS-mediated pathways showing significant correlations with HNSCC immunity. Furthermore, we demonstrated that the association between TP53 mutation and tumor immunity was independent of the human papillomavirus (HPV) infection and smoking status in HNSCC. These data suggest that p53 and RAS may play important roles in regulating HNSCC immunity and that the TP53 and HRAS mutation status could be useful biomarkers for stratifying HNSCC patients responsive to immunotherapy.
Collapse
Key Words
- APC, Antigen-Presenting Cell
- BH, Benjami and Hochberg
- Cancer genomics
- DFS, Disease Free Survival
- EMT, Epithelial-Mesenchymal Transition
- FDR, False Discovery Rate
- GSEA, Gene-Set Enrichment Analysis
- HLA, Human Leukocyte Antigen
- HNSCC, Head and Neck Squamous Cell Carcinomas
- HPV, Human Papilloma Virus
- HRAS mutations
- Head and neck squamous cell carcinomas
- MHC, Major Histocompatibility Complex
- NK, Natural Killer
- OR, Odds Ratio
- OS, Overall Survival
- TILs, Tumor-Infiltrating Lymphocytes
- TIM, Tumor Immune Microenvironment
- TMB, Tumor Mutation Burden
- TP53 mutations
- Tumor immunity
- dMMR, Deficient Mismatch-Repair
- pDCs, Plasmacytoid Dendritic Cells
- ssGSEA, single-sample Gene-Set Enrichment Analysis
Collapse
Affiliation(s)
- Haoyu Lyu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Mengyuan Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Zehang Jiang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Zhixian Liu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
162
|
Notch Signaling Activation as a Hallmark for Triple-Negative Breast Cancer Subtype. JOURNAL OF ONCOLOGY 2019; 2019:8707053. [PMID: 31379945 PMCID: PMC6657611 DOI: 10.1155/2019/8707053] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subgroup of 15%-20% of diagnosed breast cancer patients. It is generally considered to be the most difficult breast cancer subtype to deal with, due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), which usually direct targeted therapies. In this scenario, the current treatments of TNBC-affected patients rely on tumor excision and conventional chemotherapy. As a result, the prognosis is overall poor. Thus, the identification and characterization of targets for novel therapies are urgently required. The Notch signaling pathway has emerged to act in the pathogenesis and tumor progression of TNBCs. Firstly, Notch receptors are associated with the regulation of tumor-initiating cells (TICs) behavior, as well as with the aetiology of TNBCs. Secondly, there is a strong evidence that Notch pathway is a relevant player in mammary cancer stem cells maintenance and expansion. Finally, Notch receptors expression and activation strongly correlate with the aggressive clinicopathological and biological phenotypes of breast cancer (e.g., invasiveness and chemoresistance), which are relevant characteristics of TNBC subtype. The purpose of this up-to-date review is to provide a detailed overview of the specific role of all four Notch receptors (Notch1, Notch2, Notch3, and Notch4) in TNBCs, thus identifying the Notch signaling pathway deregulation/activation as a pathognomonic feature of this breast cancer subtype. Furthermore, this review will also discuss recent information associated with different therapeutic options related to the four Notch receptors, which may be useful to evaluate prognostic or predictive indicators as well as to develop new therapies aimed at improving the clinical outcome of TNBC patients.
Collapse
|
163
|
Decoding Immune Heterogeneity of Triple Negative Breast Cancer and Its Association with Systemic Inflammation. Cancers (Basel) 2019; 11:cancers11070911. [PMID: 31261762 PMCID: PMC6678607 DOI: 10.3390/cancers11070911] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive subtype with limited therapeutic options. New opportunities are emerging from current comprehensive characterization of tumor immune infiltration and fitness. Therefore, effectiveness of current chemotherapies and novel immunotherapies are partially dictated by host inflammatory and immune profiles. However, further progress in breast cancer immuno-oncology is required to reach a detailed awareness of the immune infiltrate landscape and to determine additional reliable and easily detectable biomarkers. In this study, by analyzing gene expression profiles of 54 TNBC cases we identified three TNBC clusters displaying unique immune features. Deep molecular characterization of immune cells cytolytic-activity and tumor-inflammation status reveled variability in the local composition of the immune infiltrate in the TNBC clusters, reconciled by tumor-infiltrating lymphocytes counts. Platelet-to-lymphocyte ratio (PLR), a blood systemic parameter of inflammation evaluated using pre-surgical blood test data, resulted negatively correlated with local tumoral cytolytic activity and T cell–inflamed microenvironment, whereas tumor aggressiveness score signature positively correlated with PLR values. These data highlighted that systemic inflammation parameters may represent reliable and informative markers of the local immune tumor microenvironment in TNBC patients and could be exploited to decipher tumor infiltrate properties and consequently to select the most appropriate therapies.
Collapse
|
164
|
Tan AHY, Tu W, McCuaig R, Hardy K, Donovan T, Tsimbalyuk S, Forwood JK, Rao S. Lysine-Specific Histone Demethylase 1A Regulates Macrophage Polarization and Checkpoint Molecules in the Tumor Microenvironment of Triple-Negative Breast Cancer. Front Immunol 2019; 10:1351. [PMID: 31249575 PMCID: PMC6582666 DOI: 10.3389/fimmu.2019.01351] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages play an important role in regulating the tumor microenvironment (TME). Here we show that classical (M1) macrophage polarization reduced expression of LSD1, nuclear REST corepressor 1 (CoREST), and the zinc finger protein SNAIL. The LSD1 inhibitor phenelzine targeted both the flavin adenine dinucleotide (FAD) and CoREST binding domains of LSD1, unlike the LSD1 inhibitor GSK2879552, which only targeted the FAD domain. Phenelzine treatment reduced nuclear demethylase activity and increased transcription and expression of M1-like signatures both in vitro and in a murine triple-negative breast cancer model. Overall, the LSD1 inhibitors phenelzine and GSK2879552 are useful tools for dissecting the contribution of LSD1 demethylase activity and the nuclear LSD1-CoREST complex to switching macrophage polarization programs. These findings suggest that inhibitors must have dual FAD and CoREST targeting abilities to successfully initiate or prime macrophages toward an anti-tumor M1-like phenotype in triple-negative breast cancer.
Collapse
Affiliation(s)
- Abel H Y Tan
- Epigenetics and Transcription Laboratory Melanie Swan Memorial Translational Centre, Sci-Tech, University of Canberra, Canberra, ACT, Australia
| | - WenJuan Tu
- Epigenetics and Transcription Laboratory Melanie Swan Memorial Translational Centre, Sci-Tech, University of Canberra, Canberra, ACT, Australia
| | - Robert McCuaig
- Epigenetics and Transcription Laboratory Melanie Swan Memorial Translational Centre, Sci-Tech, University of Canberra, Canberra, ACT, Australia
| | - Kristine Hardy
- Epigenetics and Transcription Laboratory Melanie Swan Memorial Translational Centre, Sci-Tech, University of Canberra, Canberra, ACT, Australia
| | - Thomasina Donovan
- Epigenetics and Transcription Laboratory Melanie Swan Memorial Translational Centre, Sci-Tech, University of Canberra, Canberra, ACT, Australia
| | - Sofiya Tsimbalyuk
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Jade K Forwood
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Sudha Rao
- Epigenetics and Transcription Laboratory Melanie Swan Memorial Translational Centre, Sci-Tech, University of Canberra, Canberra, ACT, Australia
| |
Collapse
|
165
|
TP53 Mutations Promote Immunogenic Activity in Breast Cancer. JOURNAL OF ONCOLOGY 2019; 2019:5952836. [PMID: 31275382 PMCID: PMC6582869 DOI: 10.1155/2019/5952836] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 03/14/2019] [Accepted: 04/15/2019] [Indexed: 12/25/2022]
Abstract
Background Although immunotherapy has recently achieved clinical successes in a variety of cancers, thus far there is no immunotherapeutic strategy for breast cancer (BC). Thus, it is important to discover biomarkers for identifying BC patients responsive to immunotherapy. TP53 mutations were often associated with worse clinical outcomes in BC whose triple-negative subtype has a high TP53 mutation rate (approximately 80%). To explore a potentially promising therapeutic option for the TP53-mutated BC subtype, we studied the association between TP53 mutations and immunogenic activity in BC. Methods We compared the enrichment levels of 26 immune signatures that indicated activities of diverse immune cells, functions, and pathways between TP53-mutated and TP53-wildtype BCs based on two large-scale BC multiomics datasets. Moreover, we explored the molecular cues associated with the differences in immunogenic activity between TP53-mutated and TP53-wildtype BCs. Furthermore, we performed experimental validation of the findings from bioinformatics analysis. Results Bioinformatics analysis showed that almost all analyzed immune signatures showed significantly higher enrichment levels in TP53-mutated BCs than in TP53-wildtype BCs. Moreover, in vitro experiments confirmed that mutant p53 could increase BC immunogenicity. Both computational and experimental results demonstrated that TP53 mutations could promote BC immunogenicity via regulation of the p53-mediated pathways including cell cycle, apoptosis, Wnt, Jak-STAT, NOD-like receptor, and glycolysis. Furthermore, we found that elevated immune activity was likely associated with a better survival prognosis in TP53-mutated BCs, but not necessarily in TP53-wildtype BCs. Conclusions TP53 mutations may promote immunogenic activity in BC, suggesting that the TP53 mutation status could be a useful biomarker for stratifying BC patients responsive to immunotherapy.
Collapse
|
166
|
Caparica R, Lambertini M, de Azambuja E. How I treat metastatic triple-negative breast cancer. ESMO Open 2019; 4:e000504. [PMID: 31231572 PMCID: PMC6555596 DOI: 10.1136/esmoopen-2019-000504] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with a high risk of recurrence and generally a bad prognosis. More than one-third of patients with TNBC will present distant metastases during the course of their disease. Although chemotherapy has been the main treatment option for metastatic TNBC for a long time, this scenario has changed recently with the advent of the polyadenosine diphosphate-ribose polymerase inhibitors (PARPis) for patients harbouring a mutation in the BRCA genes (BRCAmut) and also with the results of immunotherapy in patients with PD-L1-positive tumours. The present manuscript proposes a treatment algorithm for patients with metastatic TNBC based on the currently available, most relevant literature on the topic. For patients with a BRCAmut and able to tolerate chemotherapy, we recommend initiating treatment with platins (carboplatin/cisplatin) and to start PARPis at disease progression. For patients with PD-L1-positive tumours (PD-L1 expression on tumour-infiltrating immune cells ≥1%), we recommend first-line treatment with nab-paclitaxel and atezolizumab, when available. In patients without a BRCA mutation and with PD-L1-negative tumours, we recommend single-agent chemotherapy with taxanes (paclitaxel or docetaxel) as a first-line treatment. In patients with a high disease burden or who are very symptomatic, combinations such as anthracyclines plus cyclophosphamide or platins with taxanes are valid options. Chemotherapy should be maintained until the occurrence of disease progression or limiting toxicities. After progression to first-line chemotherapy, anthracyclines are an option for patients who received taxanes and vice versa. For patients who progressed to taxanes and anthracyclines, or who present contraindications to these agents, fluorouracil/capecitabine, eribulin, gemcitabine, cisplatin/carboplatin, vinorelbine and ixabepilone are alternatives. The treatment of TNBC is constantly evolving, and the inclusion of patients in ongoing trials evaluating new targeted agents, immunotherapy and predictive biomarkers should be encouraged, in an attempt to improve metastatic TNBC treatment outcomes.
Collapse
Affiliation(s)
- Rafael Caparica
- Department of Medical Oncology, Institut Jules Bordet, Bruxelles, Belgium.
| | - Matteo Lambertini
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | | |
Collapse
|
167
|
Jørgensen N, Persson G, Hviid TVF. The Tolerogenic Function of Regulatory T Cells in Pregnancy and Cancer. Front Immunol 2019; 10:911. [PMID: 31134056 PMCID: PMC6517506 DOI: 10.3389/fimmu.2019.00911] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells, a subpopulation of suppressive T cells, are potent mediators of self-tolerance and essential for the suppression of triggered immune responses. The immune modulating capacity of these cells play a major role in both transplantation, autoimmune disease, allergy, cancer and pregnancy. During pregnancy, low numbers of regulatory T cells are associated with pregnancy failure and pregnancy complications such as pre-eclampsia. On the other hand, in cancer, low numbers of immunosuppressive T cells are correlated with better prognosis. Hence, maternal immune tolerance toward the fetus during pregnancy and the escape from host immunosurveillance by cancer seem to be based on similar immunological mechanisms being highly dependent on the balance between immune activation and suppression. As regulatory T cells hold a crucial role in several biological processes, they may also be promising subjects for therapeutic use. Especially in the field of cancer, cell therapy and checkpoint inhibitors have demonstrated that immune-based therapies have a very promising potential in treatment of human malignancies. However, these therapies are often accompanied by adverse autoimmune side effects. Therefore, expanding the knowledge to recognize the complexities of immune regulation pathways shared across different immunological scenarios is extremely important in order to improve and develop new strategies for immune-based therapy. The intent of this review is to highlight the functional characteristics of regulatory T cells in the context of mechanisms of immune regulation in pregnancy and cancer, and how manipulation of these mechanisms potentially may improve therapeutic options.
Collapse
Affiliation(s)
| | | | - Thomas Vauvert F. Hviid
- Department of Clinical Biochemistry, Centre for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Zealand University Hospital, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
168
|
Chrétien S, Zerdes I, Bergh J, Matikas A, Foukakis T. Beyond PD-1/PD-L1 Inhibition: What the Future Holds for Breast Cancer Immunotherapy. Cancers (Basel) 2019; 11:E628. [PMID: 31060337 PMCID: PMC6562626 DOI: 10.3390/cancers11050628] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy has altered the management of human malignancies, improving outcomes in an expanding list of diseases. Breast cancer - presumably due to its perceived low immunogenicity - is a late addition to this list. Furthermore, most of the focus has been on the triple negative subtype because of its higher tumor mutational load and lymphocyte-enriched stroma, although emerging data show promise on the other breast cancer subtypes as well. To this point the clinical use of immunotherapy is limited to the inhibition of two immune checkpoints, Programmed Cell Death Protein 1 (PD-1) and Cytotoxic T-lymphocyte-associated Protein 4 (CTLA-4). Consistent with the complexity of the regulation of the tumor - host interactions and their lack of reliance on a single regulatory pathway, combinatory approaches have shown improved efficacy albeit at the cost of increased toxicity. Beyond those two checkpoints though, a large number of co-stimulatory or co-inhibitory molecules play major roles on tumor evasion from immunosurveillance. These molecules likely represent future targets of immunotherapy provided that the promise shown in early data is translated into improved patient survival in randomized trials. The biological role, prognostic and predictive implications regarding breast cancer and early clinical efforts on exploiting these immune-related therapeutic targets are herein reviewed.
Collapse
Affiliation(s)
- Sebastian Chrétien
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Ioannis Zerdes
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Jonas Bergh
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Alexios Matikas
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Theodoros Foukakis
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| |
Collapse
|
169
|
Wang H, Milberg O, Bartelink IH, Vicini P, Wang B, Narwal R, Roskos L, Santa-Maria CA, Popel AS. In silico simulation of a clinical trial with anti-CTLA-4 and anti-PD-L1 immunotherapies in metastatic breast cancer using a systems pharmacology model. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190366. [PMID: 31218069 PMCID: PMC6549962 DOI: 10.1098/rsos.190366] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/24/2019] [Indexed: 05/10/2023]
Abstract
The low response rate of immune checkpoint blockade in breast cancer has highlighted the need for predictive biomarkers to identify responders. While a number of clinical trials are ongoing, testing all possible combinations is not feasible. In this study, a quantitative systems pharmacology model is built to integrate immune-cancer cell interactions in patients with breast cancer, including central, peripheral, tumour-draining lymph node (TDLN) and tumour compartments. The model can describe the immune suppression and evasion in both TDLN and the tumour microenvironment due to checkpoint expression, and mimic the tumour response to checkpoint blockade therapy. We investigate the relationship between the tumour response to checkpoint blockade therapy and composite tumour burden, PD-L1 expression and antigen intensity, including their individual and combined effects on the immune system, using model-based simulations. The proposed model demonstrates the potential to make predictions of tumour response of individual patients given sufficient clinical measurements, and provides a platform that can be further adapted to other types of immunotherapy and their combination with molecular-targeted therapies. The patient predictions demonstrate how this systems pharmacology model can be used to individualize immunotherapy treatments. When appropriately validated, these approaches may contribute to optimization of breast cancer treatment.
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Oleg Milberg
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Imke H. Bartelink
- Department of Medicine, University of California, San Francisco, CA, USA
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD), MedImmune, South San Francisco, CA, USA
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Paolo Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK, MedImmune, Cambridge, UK
| | - Bing Wang
- Amador Bioscience Inc, Pleasanton, CA 94588, USA
| | - Rajesh Narwal
- Clinical Pharmacology and DMPK (CPD), MedImmune, Gaithersburg, MD, USA
| | - Lorin Roskos
- Clinical Pharmacology and DMPK (CPD), MedImmune, Gaithersburg, MD, USA
| | - Cesar A. Santa-Maria
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
170
|
He Y, Liu ZX, Jiang ZH, Wang XS. Identification of genomic features associated with immunotherapy response in gastrointestinal cancers. World J Gastrointest Oncol 2019; 11:270-280. [PMID: 31040893 PMCID: PMC6475671 DOI: 10.4251/wjgo.v11.i4.270] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/29/2019] [Accepted: 03/28/2019] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal (GI) cancers prevail and account for an extremely high number of cancer deaths worldwide. The traditional treatment strategies, including surgery, chemotherapy, radiotherapy, and targeted therapy, have a limited therapeutic effect for advanced GI cancers. Recently, immunotherapy has shown promise in treating various refractory malignancies, including the GI cancers with mismatch repair deficiency (dMMR) or microsatellite instability (MSI). Thus, immunotherapy could be a promising treatment approach for GI cancers. Unfortunately, only a small proportion of GI cancer patients currently respond to immunotherapy. Therefore, it is important to discover predictive biomarkers for stratifying GI cancer patients response to immunotherapy. Certain genomic features, such as dMMR/MSI, tumor mutation burden (TMB), and tumor aneuploidy have been associated with tumor immunity and im-munotherapy response and may serve as predictive biomarkers for cancer immunotherapy. In this review, we examined the correlations between tumor immunity and three genomic features: dMMR/MSI, TMB, and tumor aneuploidy. We also explored their correlations using The Cancer Genome Atlas data and confirmed that the dMMR/MSI status, high TMB, and low tumor aneuploidy are associated with elevated tumor immunity in GI cancers. To improve the immunotherapeutic potential in GI cancers, more genetic or genomic features associated with tumor immune response need to be identified. Furthermore, it is worth exploring the combination of different immunotherapeutic methods and the combination of immunotherapy with other therapeutic approaches for cancer therapy.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| | - Zhi-Xian Liu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| | - Ze-Hang Jiang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| | - Xiao-Sheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing 211198, Jiangsu Province, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| |
Collapse
|
171
|
Increased glycolysis correlates with elevated immune activity in tumor immune microenvironment. EBioMedicine 2019; 42:431-442. [PMID: 30935888 PMCID: PMC6491961 DOI: 10.1016/j.ebiom.2019.03.068] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023] Open
Abstract
Background Prior studies showed that tumor glycolysis and tumor immune evasion are interdependent. However, a systematic investigation of the association between tumor glycolysis and tumor immunity in various cancers remains lacking. Methods Using the Cancer Genome Atlas (TCGA) datasets, we explored the association between glycolytic activity and immune signatures in 14 cancer types. We also explored the associations between glycolytic activity and tumor immunity associated genetic features, including PD-L1 expression, tumor mutation burden (TMB), and tumor aneuploidy. Moreover, we performed in vitro experiments to verify some findings from bioinformatics analysis. Furthermore, we explored the association between tumor glycolytic activity and immunotherapy response. Findings Glycolytic activity was likely correlated with active immune signatures in various cancers and highly glycolytic tumors presented an immune-stimulatory tumor microenvironment. Compared to TMB and aneuploidy, glycolytic activity was a stronger and more consistent predictor for immune signatures in diverse cancers. Both computational and experimental analyses showed that glycolysis could increase PD-L1 expression in tumor. Glycolytic activity had a strong correlation with apoptosis which was a strong positive predictor for immune signatures, suggesting that apoptosis could be an important medium connecting glycolytic activity with immune activity in cancer. Finally, highly glycolytic tumors exhibited a better immunotherapy response and a favorable survival in the immunotherapy setting. Interpretation Tumor glycolysis may increase tumor immunity in diverse cancers. Glycolytic activity enhances PD-L1 expression on tumor cells and thus promotes anti-PD-1/PD-L1 immunotherapy response. Thus, the tumor glycolytic activity could be a predictive biomarker for immunotherapy response in diverse cancers. Fund This work was supported by the China Pharmaceutical University (grant numbers 3150120001, 2632018YX01 to XW).
Collapse
|
172
|
Elnaggar J, Tsien F, Yates C, Davis M, Miele L, Hicks C. An Integrative Genomics Approach for Associating Genetic Susceptibility with the Tumor Immune Microenvironment in Triple Negative Breast Cancer. BIOMEDICAL JOURNAL OF SCIENTIFIC & TECHNICAL RESEARCH 2019; 15:11074-11085. [PMID: 38618278 PMCID: PMC11013954 DOI: 10.26717/bjstr.2019.15.002642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Background Triple-negative breast cancer (TNBC) is the most aggressive form of breast cancer. It is a heterogeneous disease with poor prognosis. Contributing to the worse prognosis in TNBC is the higher rates of relapse and rapid progression to metastatic disease which is often lethal. With the exception of cytotoxic chemotherapy, there is currently no effective targeted therapies. Immunotherapy such as vaccines offer new opportunities for treatment of TNBC. But realizing the potential of immunotherapy and vaccination may require understanding the association between the tumor immune microenvironment and genetic susceptibility to TNBC. The objective of this exploratory study was to investigate the potential association between genetic susceptibility and tumor immune microenvironment in TNBC. Methods We integrated information on genetic variants and genes associated with an increased risk of developing breast cancer with gene expression data from the Caucasian women diagnosed with the basal-like immune activated (N=54) and basal-like immune suppressed (N=60) subtypes of TNBC to discover and characterize immune modulated gene signatures, molecular networks and biological pathways enriched for genetic susceptibility variants. Results The investigation revealed immune modulated gene signatures, molecular networks and biological pathways enriched for genetic susceptibility variants. The discovered pathways included the role of BRCA1 in DNA damage response, hereditary breast cancer, aryl hydrocarbon receptor and molecular mechanisms of cancer signaling pathways. Conclusion The investigation suggests the link between genetic susceptibility and the tumor immune microenvironment in TNBC and establishes putative functional bridges between genetic predisposition and immune modulated pathways.
Collapse
Affiliation(s)
- Jacob Elnaggar
- Department of Genetics Louisiana State University Health Sciences Center-School of Medicine, 533 Bolivar Street, New Orleans, LA 70112
| | - Fern Tsien
- Department of Genetics Louisiana State University Health Sciences Center-School of Medicine, 533 Bolivar Street, New Orleans, LA 70112
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee AL, 36088
| | - Melisa Davis
- Henry Ford Health System, One Ford Place, 3CE, Detroit, MI 48202
| | - Lucio Miele
- Department of Genetics Louisiana State University Health Sciences Center-School of Medicine, 533 Bolivar Street, New Orleans, LA 70112
| | - Chindo Hicks
- Department of Genetics Louisiana State University Health Sciences Center-School of Medicine, 533 Bolivar Street, New Orleans, LA 70112
| |
Collapse
|
173
|
Velloso FJ, Campos AR, Sogayar MC, Correa RG. Proteome profiling of triple negative breast cancer cells overexpressing NOD1 and NOD2 receptors unveils molecular signatures of malignant cell proliferation. BMC Genomics 2019; 20:152. [PMID: 30791886 PMCID: PMC6385390 DOI: 10.1186/s12864-019-5523-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background Triple negative breast cancer (TNBC) is a malignancy with very poor prognosis, due to its aggressive clinical characteristics and lack of response to receptor-targeted drug therapy. In TNBC, immune-related pathways are typically upregulated and may be associated with a better prognosis of the disease, encouraging the pursuit for immunotherapeutic options. A number of immune-related molecules have already been associated to the onset and progression of breast cancer, including NOD1 and NOD2, innate immune receptors of bacterial-derived components which activate pro-inflammatory and survival pathways. In the context of TNBC, overexpression of either NOD1or NOD2 is shown to reduce cell proliferation and increase clonogenic potential in vitro. To further investigate the pathways linking NOD1 and NOD2 signaling to tumorigenesis in TNBC, we undertook a global proteome profiling of TNBC-derived cells ectopically expressing each one of these NOD receptors. Results We have identified a total of 95 and 58 differentially regulated proteins in NOD1- and NOD2-overexpressing cells, respectively. We used bioinformatics analyses to identify enriched molecular signatures aiming to integrate the differentially regulated proteins into functional networks. These analyses suggest that overexpression of both NOD1 and NOD2 may disrupt immune-related pathways, particularly NF-κB and MAPK signaling cascades. Moreover, overexpression of either of these receptors may affect several stress response and protein degradation systems, such as autophagy and the ubiquitin-proteasome complex. Interestingly, the levels of several proteins associated to cellular adhesion and migration were also affected in these NOD-overexpressing cells. Conclusions Our proteomic analyses shed new light on the molecular pathways that may be modulating tumorigenesis via NOD1 and NOD2 signaling in TNBC. Up- and downregulation of several proteins associated to inflammation and stress response pathways may promote activation of protein degradation systems, as well as modulate cell-cycle and cellular adhesion proteins. Altogether, these signals seem to be modulating cellular proliferation and migration via NF-κB, PI3K/Akt/mTOR and MAPK signaling pathways. Further investigation of altered proteins in these pathways may provide more insights on relevant targets, possibly enabling the immunomodulation of tumorigenesis in the aggressive TNBC phenotype. Electronic supplementary material The online version of this article (10.1186/s12864-019-5523-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fernando J Velloso
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo (USP), São Paulo, SP, 05360-130, Brazil
| | - Alexandre R Campos
- SBP Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Mari C Sogayar
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo (USP), São Paulo, SP, 05360-130, Brazil
| | - Ricardo G Correa
- SBP Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
174
|
Pinto JA, Saravia CH, Flores C, Araujo JM, Martínez D, Schwarz LJ, Casas A, Bravo L, Zavaleta J, Chuima B, Alvarado H, Fujita R, Gómez HL. Precision medicine for locally advanced breast cancer: frontiers and challenges in Latin America. Ecancermedicalscience 2019; 13:896. [PMID: 30792813 PMCID: PMC6372295 DOI: 10.3332/ecancer.2019.896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Indexed: 12/18/2022] Open
Abstract
Advances in high-throughput technologies and their involvement in the 'omics' of cancer have made possible the identification of hundreds of biomarkers and the development of predictive and prognostic platforms that model the management of cancer from evidence-based medicine to precision medicine. Latin America (LATAM) is a region characterised by fragmented healthcare, high rates of poverty and disparities to access to a basic standard of care not only for cancer but also for other complex diseases. Patients from the public setting cannot afford targeted therapy, the facilities offering genomic platforms are scarce and the use of high-precision radiotherapy is limited to few facilities. Despite the fact that LATAM oncologists are well-trained in the use of genomic platforms and constantly participate in genomic projects, a medical practice based in precision oncology is a great challenge and frequently limited to private practice. In breast cancer, we are waiting for the results of large basket trials to incorporate the detection of actionable mutations to select targeted treatments, in a similar way to the management of lung cancer. On the other hand and paradoxically, in the 'one fit is not for all' era, clinical and genomic studies continue grouping our patients under the single label 'Latin American' or 'Hispanic' despite the different ancestries and genomic backgrounds seen in the region. More regional cancer genomic initiatives and public availability of this data are needed in order to develop more precise oncology in locally advanced breast cancer.
Collapse
Affiliation(s)
- Joseph A Pinto
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - César H Saravia
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - Claudio Flores
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - Jhajaira M Araujo
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - David Martínez
- Departamento de Radioterapia, Oncosalud-AUNA, Lima 15036, Perú
| | - Luis J Schwarz
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Lima 15036, Perú
| | - Alberto Casas
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima 15067, Perú
| | - Leny Bravo
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima 15067, Perú
| | - Jenny Zavaleta
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima 15067, Perú
| | | | - Hober Alvarado
- Facultad de Ciencias Biológicas, Universidad Nacional San Luis Gonzaga de Ica, Ica 11004, Perú
| | - Ricardo Fujita
- Centro de Genética y Biología Molecular, Universidad de San Martín de Porres, Lima 15024, Perú
| | - Henry L Gómez
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Lima 15036, Perú.,Departamento de Medicina Oncológica, Instituto Nacional de Enfermedades Neoplásicas, 15038, Perú
| |
Collapse
|
175
|
Wang X, Li M. Correlate tumor mutation burden with immune signatures in human cancers. BMC Immunol 2019; 20:4. [PMID: 30634925 PMCID: PMC6329192 DOI: 10.1186/s12865-018-0285-5] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/28/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Tumor mutation burden (TMB) has been associated with cancer immunotherapeutic response and cancer prognosis. Although many explorations have revealed that high TMB may yield many neoantigens to incite antitumor immune response, a systematic exploration of the correlation between TMB and immune signatures in different cancer types is lacking. RESULTS We classified cancer into the lower-TMB subtype and the higher-TMB subtype for each of 32 cancer types based on their somatic mutation data from the Cancer Genome Atlas (TCGA), and compared the expression levels of immune-related genes and gene-sets between both subtypes of cancers in each cancer type. In some cancer types most of the immune signatures analyzed were upregulated in the lower-TMB subtype, while in some other cancer types the immune signatures were prone to be upregulated in the higher-TMB subtype. However, the regulatory T cells, immune cell infiltrate, tumor-infiltrating lymphocytes, and cytokine signatures tended to be upregulated in the lower-TMB subtype, and the cancer-testis antigen (CTA) and pro-inflammatory signatures were inclined to be upregulated in the higher-TMB subtype. Importantly, high TMB was associated with elevated expression of PD-L1 in diverse prevailing cancers. Furthermore, we found that higher TMB was associated with better survival prognosis in numerous cancer types while was associated with worse prognosis in a few cancer types. CONCLUSIONS High TMB may inhibit immune cell infiltrations while promote CTAs expression and inflammatory response in cancer. In many common cancer types, higher TMB may respond favorably to anti-PD-1/PD-L1 immunotherapy. Our data implicate that higher-TMB patients could gain a more favorable prognosis in diverse cancer types if treated with immunotherapy, otherwise would have a poorer prognosis compared to lower-TMB patients.
Collapse
Affiliation(s)
- Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198 China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198 China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198 China
| | - Mengyuan Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198 China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198 China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198 China
| |
Collapse
|
176
|
Al-Khadairi G, Naik A, Thomas R, Al-Sulaiti B, Rizly S, Decock J. PRAME promotes epithelial-to-mesenchymal transition in triple negative breast cancer. J Transl Med 2019; 17:9. [PMID: 30602372 PMCID: PMC6317205 DOI: 10.1186/s12967-018-1757-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Background The triple negative breast cancer (TNBC) paradox marks a major challenge in the treatment-decision making process. TNBC patients generally respond better to neoadjuvant chemotherapy compared to other breast cancer patients; however, they have a substantial higher risk of disease recurrence. We evaluated the expression of the tumor-associated antigen PReferentially Antigen expressed in MElanoma (PRAME) as a prognostic biomarker in breast cancer and explored its role in cell migration and invasion, key hallmarks of progressive and metastatic disease. Methods TCGA and GTeX datasets were interrogated to assess the expression of PRAME in relation to overall and disease-free survival. The role of PRAME in cell migration and invasion was investigated using gain- and loss-of-function TNBC cell line models. Results We show that PRAME promotes migration and invasion of TNBC cells through changes in expression of E-cadherin, N-cadherin, vimentin and ZEB1, core markers of an epithelial-to-mesenchymal transition. Mechanistic analysis of PRAME-overexpressing cells showed an upregulation of 11 genes (SNAI1, TCF4, TWIST1, FOXC2, IL1RN, MMP2, SOX10, WNT11, MMP3, PDGFRB, and JAG1) and downregulation of 2 genes (BMP7 and TSPAN13). Gene ontology analyses revealed enrichment of genes that are dysregulated in ovarian and esophageal cancer and are involved in transcription and apoptosis. In line with this, interrogation of TCGA and GTEx data demonstrated an increased PRAME expression in ovarian and esophageal tumor tissues in addition to breast tumors where it is associated with worse survival. Conclusions Our findings indicate that PRAME plays a tumor-promoting role in triple negative breast cancer by increasing cancer cell motility through EMT-gene reprogramming. Therefore, PRAME could serve as a prognostic biomarker and/or therapeutic target in TNBC.
Collapse
Affiliation(s)
- Ghaneya Al-Khadairi
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Doha, Qatar
| | - Remy Thomas
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Doha, Qatar
| | - Boshra Al-Sulaiti
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Doha, Qatar
| | - Shaheen Rizly
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Doha, Qatar
| | - Julie Decock
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, Qatar. .,Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Doha, Qatar.
| |
Collapse
|
177
|
He Y, Jiang Z, Chen C, Wang X. Classification of triple-negative breast cancers based on Immunogenomic profiling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:327. [PMID: 30594216 PMCID: PMC6310928 DOI: 10.1186/s13046-018-1002-1] [Citation(s) in RCA: 336] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022]
Abstract
Background Abundant evidence shows that triple-negative breast cancer (TNBC) is heterogeneous, and many efforts have been devoted to identifying TNBC subtypes on the basis of genomic profiling. However, few studies have explored the classification of TNBC specifically based on immune signatures that may facilitate the optimal stratification of TNBC patients responsive to immunotherapy. Methods Using four publicly available TNBC genomics datasets, we classified TNBC on the basis of the immunogenomic profiling of 29 immune signatures. Unsupervised and supervised machine learning methods were used to perform the classification. Results We identified three TNBC subtypes that we named Immunity High (Immunity_H), Immunity Medium (Immunity_M), and Immunity Low (Immunity_L) and demonstrated that this classification was reliable and predictable by analyzing multiple different datasets. Immunity_H was characterized by greater immune cell infiltration and anti-tumor immune activities, as well as better survival prognosis compared to the other subtypes. Besides the immune signatures, some cancer-associated pathways were hyperactivated in Immunity_H, including apoptosis, calcium signaling, MAPK signaling, PI3K–Akt signaling, and RAS signaling. In contrast, Immunity_L presented depressed immune signatures and increased activation of cell cycle, Hippo signaling, DNA replication, mismatch repair, cell adhesion molecule binding, spliceosome, adherens junction function, pyrimidine metabolism, glycosylphosphatidylinositol (GPI)-anchor biosynthesis, and RNA polymerase pathways. Furthermore, we identified a gene co-expression subnetwork centered around five transcription factor (TF) genes (CORO1A, STAT4, BCL11B, ZNF831, and EOMES) specifically significant in the Immunity_H subtype and a subnetwork centered around two TF genes (IRF8 and SPI1) characteristic of the Immunity_L subtype. Conclusions The identification of TNBC subtypes based on immune signatures has potential clinical implications for TNBC treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-1002-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Zehang Jiang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Cai Chen
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China. .,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China. .,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
178
|
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is associated with poor prognosis and limited treatment options. However, TNBC is known to be more immunogenic compared to other breast cancer subtypes, with tumor-infiltrating lymphocytes playing an important prognostic and predictive role. Furthermore, TNBC has a higher level of programmed cell death-ligand 1 (PD-L1) expression. Therapeutic blockade of PD-L1 using atezolizumab is thus expected to activate and enhance tumor-specific T-cell responses, resulting in improved anti-tumor activity. Areas covered: This review summarizes the development and the impact of the PD-L1 inhibitor atezolizumab in advanced TNBC; it examines the mechanism of action, pharmacokinetics and the available preclinical and clinical data. Expert opinion: Atezolizumab, a novel immune checkpoint inhibitors targeting PD-L1, is an effective and well-tolerated treatment option for metastatic TNBC. In general, TNBC has a high unmet medical need, hence the clinical development of atezolizumab should continue, particularly for TNBC. Indeed, atezolizumab has the potential to substantially augment the therapeutic armamentarium for TNBC. This should lead to improved immunotherapeutic strategies and the enhancement of the outcome for this group of breast cancer patients.
Collapse
Affiliation(s)
- Anne-Sophie Heimes
- a Department of Obstetrics and Gynecology, Division of Molecular Oncology , University Medical Center , Mainz , Germany
| | - Marcus Schmidt
- a Department of Obstetrics and Gynecology, Division of Molecular Oncology , University Medical Center , Mainz , Germany
| |
Collapse
|
179
|
Valdes-Mora F, Handler K, Law AMK, Salomon R, Oakes SR, Ormandy CJ, Gallego-Ortega D. Single-Cell Transcriptomics in Cancer Immunobiology: The Future of Precision Oncology. Front Immunol 2018; 9:2582. [PMID: 30483257 PMCID: PMC6240655 DOI: 10.3389/fimmu.2018.02582] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Tumors are formed by cancer cells and a myriad of non-cancerous cell types that together with the extracellular matrix form the tumor microenvironment. These cancer-associated cells and components contribute to shape the progression of cancer and are deeply involved in patient outcome. The immune system is an essential part of the tumor microenvironment, and induction of cancer immunotolerance is a necessary step involved in tumor formation and growth. Immune mechanisms are intimately associated with cancer progression, invasion, and metastasis; as well as to tumor dormancy and modulation of sensitivity to drug therapy. Transcriptome analyses have been extensively used to understand the heterogeneity of tumors, classifying tumors into molecular subtypes and establishing signatures that predict response to therapy and patient outcomes. However, the classification of the tumor cell diversity and specially the identification of rare populations has been limited in these transcriptomic analyses of bulk tumor cell populations. Massively-parallel single-cell RNAseq analysis has emerged as a powerful method to unravel heterogeneity and to study rare cell populations in cancer, through unsupervised sampling and modeling of transcriptional states in single cells. In this context, the study of the role of the immune system in cancer would benefit from single cell approaches, as it will enable the characterization and/or discovery of the cell types and pathways involved in cancer immunotolerance otherwise missed in bulk transcriptomic information. Thus, the analysis of gene expression patterns at single cell resolution holds the potential to provide key information to develop precise and personalized cancer treatment including immunotherapy. This review is focused on the latest single-cell RNAseq methodologies able to agnostically study thousands of tumor cells as well as targeted single-cell RNAseq to study rare populations within tumors. In particular, we will discuss methods to study the immune system in cancer. We will also discuss the current challenges to the study of cancer at the single cell level and the potential solutions to the current approaches.
Collapse
Affiliation(s)
- Fatima Valdes-Mora
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kristina Handler
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andrew M K Law
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Robert Salomon
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Samantha R Oakes
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Christopher J Ormandy
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - David Gallego-Ortega
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
180
|
Boivin G, Kalambaden P, Faget J, Rusakiewicz S, Montay-Gruel P, Meylan E, Bourhis J, Lesec G, Vozenin MC. Cellular Composition and Contribution of Tertiary Lymphoid Structures to Tumor Immune Infiltration and Modulation by Radiation Therapy. Front Oncol 2018; 8:256. [PMID: 30038899 PMCID: PMC6046619 DOI: 10.3389/fonc.2018.00256] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/21/2018] [Indexed: 01/12/2023] Open
Abstract
Immune-based anti-cancer strategies combined with radiation therapy (RT) are actively being investigated but many questions remain, such as the ideal treatment scheme and whether a potent immune response can be generated both locally and systemically. In this context, tumor-associated tertiary lymphoid structures (TLS) have become a subject of research. While TLS are present in several types of cancer with strong similarities, they are especially relevant in medullary breast carcinoma (MBC). This suggests that MBC patients are ideally suited for investigating this question and may benefit from adapted therapeutic options. As RT is a corner-stone of MBC treatment, investigating interactions between RT and TLS composition is also clinically relevant. We thus first characterized the lymphoid structures associated with MBC in a patient case report and demonstrated that they closely resemble the TLS observed in a genetical mouse model. In this model, we quantitatively and qualitatively investigated the cellular composition of the tumor-associated TLS. Finally, we investigated TLS regulation after hypo-fractionated RT and showed that RT induced their acute and transient depletion, followed by a restoration phase. This study is the first work to bring a comprehensive and timely characterization of tumor-associated TLS in basal conditions and after RT. It highlights cellular targets (i.e., Tregs) that could be selectively modulated in subsequent studies to optimize anti-tumor immune response. The study of TLS modulation is worth further investigation in the context of RT and personalized medicine.
Collapse
Affiliation(s)
- Gaël Boivin
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Pradeep Kalambaden
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Julien Faget
- School of Life Sciences, Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sylvie Rusakiewicz
- Center of Experimental Therapies (CTE), Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Pierre Montay-Gruel
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Etienne Meylan
- School of Life Sciences, Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jean Bourhis
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Radio-Oncology Service, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | | | - Marie-Catherine Vozenin
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Radio-Oncology Service, Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
181
|
Xu X, Zhang L, He X, Zhang P, Sun C, Xu X, Lu Y, Li F. TGF-β plays a vital role in triple-negative breast cancer (TNBC) drug-resistance through regulating stemness, EMT and apoptosis. Biochem Biophys Res Commun 2018; 502:160-165. [PMID: 29792857 DOI: 10.1016/j.bbrc.2018.05.139] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 05/18/2018] [Indexed: 12/17/2022]
Abstract
Triple negative breast cancer (TNBC) is the most malignant subtype of breast cancer in which the cell surface lacks usual targets for drug to exhibit its effects. Epirubicin (Epi) is widely used for TNBC, but a substantial number of patients develop Epi resistance that is usually associated with poor prognosis. Transforming growth factor (TGF-β) is a multifunctional cytokine. In recent study, it appears that TGF-β influences the cancer stem cell population, thus, the drug resistance of cancer may also be affected. We used epirubicin to treat MDA-MB-231 (MB-231) cells and found that TGF-β and breast cancer stem cell markers CD44+CD24- were increased and were dose-dependent of epirubicin. We established drug-resistant cell line from parental MB-231 cells by chronic treatment with low-concentration epirubicin. The MB-231/Epi cell line showed relatively slow growth rate with varied morphology. Transwell assay and drug sensitivity assay revealed that the malignant cell behaviors in terms of migration, invasion and epirubicin-resistant properties were markedly increased in the MB-231/Epi cells. Western blot, immunofluorescence assay, and flow cytometry were used to analyze the expression levels of the breast cancer stem cell markers, CD44 and CD24. Mammospheres assay showed that the stemness of MB-231/Epi was increased compared to their parental cells. Interestingly, MB-231/Epi cells showed different expression levels of apoptosis-related markers: Bcl2, Bax; EMT-related markers E-cadherin, N-cadherin and cell cycle-related marker cyclinD1. These genes have all been shown to be regulated by the TGF-β pathway. Taken together, our findings suggest that TGF-β plays a vital role in TNBC epirubicin-resistance through regulating stemness, EMT and apoptosis.
Collapse
Affiliation(s)
- Xiaodan Xu
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Lu Zhang
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xiaogang He
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Ping Zhang
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Caihong Sun
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xiaojun Xu
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yaojuan Lu
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China; Shenzhen Academy of Peptide Targeting Technology at Pingshan, Shenzhen, 518118, China
| | - Feifei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
182
|
Thompson JA, Christensen BC, Marsit CJ. Methylation-to-Expression Feature Models of Breast Cancer Accurately Predict Overall Survival, Distant-Recurrence Free Survival, and Pathologic Complete Response in Multiple Cohorts. Sci Rep 2018; 8:5190. [PMID: 29581450 PMCID: PMC5979962 DOI: 10.1038/s41598-018-23494-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/13/2018] [Indexed: 12/03/2022] Open
Abstract
Prognostic biomarkers serve a variety of purposes in cancer treatment and research, such as prediction of cancer progression, and treatment eligibility. Despite growing interest in multi-omic data integration for defining prognostic biomarkers, validated methods have been slow to emerge. Given that breast cancer has been the focus of intense research, it is amenable to studying the benefits of multi-omic prognostic models due to the availability of datasets. Thus, we examined the efficacy of our methylation-to-expression feature model (M2EFM) approach to combining molecular and clinical predictors to create risk scores for overall survival, distant metastasis, and chemosensitivity in breast cancer. Gene expression, DNA methylation, and clinical variables were integrated via M2EFM to build models of overall survival using 1028 breast tumor samples and applied to validation cohorts of 61 and 327 samples. Models of distant recurrence-free survival and pathologic complete response were built using 306 samples and validated on 182 samples. Despite different populations and assays, M2EFM models validated with good accuracy (C-index or AUC ≥ 0.7) for all outcomes and had the most consistent performance compared to other methods. Finally, we demonstrated that M2EFM identifies functionally relevant genes, which could be useful in translating an M2EFM biomarker to the clinic.
Collapse
Affiliation(s)
- Jeffrey A Thompson
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, USA.
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, USA
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, USA
| |
Collapse
|