801
|
Adams S, Diamond JR, Hamilton E, Pohlmann PR, Tolaney SM, Chang CW, Zhang W, Iizuka K, Foster PG, Molinero L, Funke R, Powderly J. Atezolizumab Plus nab-Paclitaxel in the Treatment of Metastatic Triple-Negative Breast Cancer With 2-Year Survival Follow-up: A Phase 1b Clinical Trial. JAMA Oncol 2019; 5:334-342. [PMID: 30347025 PMCID: PMC6439843 DOI: 10.1001/jamaoncol.2018.5152] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/05/2018] [Indexed: 12/30/2022]
Abstract
Importance The humanized monoclonal antibody atezolizumab targets programmed death-ligand 1 and has demonstrated durable single-agent activity in a subset of metastatic triple-negative breast cancers. To extend the observed activity, combinatorial approaches are being tested with standard cytotoxic chemotherapies known to induce immunogenic tumor cell death. Objective To examine the safety, tolerability, and preliminary clinical activity of atezolizumab plus nab-paclitaxel in metastatic triple-negative breast cancers. Design, Setting, and Participants This phase 1b multicohort study enrolled 33 women with stage IV or locally recurrent triple-negative breast cancers and 0 to 2 lines of prior chemotherapy in the metastatic setting from December 8, 2014, to April 30, 2017, at 11 sites in the United States. The median follow-up was 24.4 months (95% CI, 22.1-28.8 months). Interventions Patients received concurrent intravenous atezolizumab and intravenous nab-paclitaxel (minimum 4 cycles). Main Outcomes and Measures The primary end point was safety and tolerability. Secondary end points included best overall response rate by Response Evaluation Criteria in Solid Tumors, version 1.1; objective response rate; duration of response; disease control rate; progression-free survival; overall survival; and biomarker analyses. Results The 33 women had a median age of 55 years (range, 32-84 years) and received 1 or more doses of atezolizumab. All patients (100%) experienced at least 1 treatment-related adverse event, 24 patients (73%) experienced grade 3/4 adverse events, and 7 patients (21%) had grade 3/4 adverse events of special interest. No deaths were related to study treatment. The objective response rate was 39.4% (95% CI, 22.9%-57.9%), and the median duration of response was 9.1 months (95% CI, 2.0-20.9 months). The disease control rate was 51.5% (95% CI, 33.5%-69.2%). Median progression-free survival and overall survival were 5.5 months (95% CI, 5.1-7.7 months) and 14.7 months (95% CI, 10.1-not estimable), respectively. Concurrent nab-paclitaxel neither significantly changed biomarkers of the tumor immune microenvironment (programmed death-ligand 1, tumor-infiltrating lymphocytes, CD8) nor impaired atezolizumab systemic immune activation (expansion of proliferating CD8+ T cells, increase of CXCL10 chemokine). Conclusions and Relevance In this phase 1b trial for metastatic triple-negative breast cancers, the combination of atezolizumab plus nab-paclitaxel had a manageable safety profile. Antitumor responses were observed, including in patients previously treated with a taxane. Trial Registration ClinicalTrials.gov identifier: NCT01633970.
Collapse
Affiliation(s)
- Sylvia Adams
- New York University Perlmutter Cancer Center, New York
| | | | - Erika Hamilton
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville
| | - Paula R. Pohlmann
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | | | | | - Wei Zhang
- Genentech Inc, South San Francisco, California
| | - Koho Iizuka
- Genentech Inc, South San Francisco, California
| | | | | | - Roel Funke
- Genentech Inc, South San Francisco, California
| | - John Powderly
- Carolina BioOncology Institute, Huntersville, North Carolina
| |
Collapse
|
802
|
Adams S, Schmid P, Rugo HS, Winer EP, Loirat D, Awada A, Cescon DW, Iwata H, Campone M, Nanda R, Hui R, Curigliano G, Toppmeyer D, O'Shaughnessy J, Loi S, Paluch-Shimon S, Tan AR, Card D, Zhao J, Karantza V, Cortés J. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann Oncol 2019; 30:397-404. [PMID: 30475950 DOI: 10.1093/annonc/mdy517] [Citation(s) in RCA: 568] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Treatment options for previously treated metastatic triple-negative breast cancer (mTNBC) are limited. In cohort A of the phase II KEYNOTE-086 study, we evaluated pembrolizumab as second or later line of treatment for patients with mTNBC. PATIENTS AND METHODS Eligible patients had centrally confirmed mTNBC, ≥1 systemic therapy for metastatic disease, prior treatment with anthracycline and taxane in any disease setting, and progression on or after the most recent therapy. Patients received pembrolizumab 200 mg intravenously every 3 weeks for up to 2 years. Primary end points were objective response rate in the total and PD-L1-positive populations, and safety. Secondary end points included duration of response, disease control rate (percentage of patients with complete or partial response or stable disease for ≥24 weeks), progression-free survival, and overall survival. RESULTS All enrolled patients (N = 170) were women, 61.8% had PD-L1-positive tumors, and 43.5% had received ≥3 previous lines of therapy for metastatic disease. ORR (95% CI) was 5.3% (2.7-9.9) in the total and 5.7% (2.4-12.2) in the PD-L1-positive populations. Disease control rate (95% CI) was 7.6% (4.4-12.7) and 9.5% (5.1-16.8), respectively. Median duration of response was not reached in the total (range, 1.2+-21.5+) and in the PD-L1-positive (range, 6.3-21.5+) populations. Median PFS was 2.0 months (95% CI, 1.9-2.0), and the 6-month rate was 14.9%. Median OS was 9.0 months (95% CI, 7.6-11.2), and the 6-month rate was 69.1%. Treatment-related adverse events occurred in 103 (60.6%) patients, including 22 (12.9%) with grade 3 or 4 AEs. There were no deaths due to AEs. CONCLUSIONS Pembrolizumab monotherapy demonstrated durable antitumor activity in a subset of patients with previously treated mTNBC and had a manageable safety profile. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, NCT02447003.
Collapse
Affiliation(s)
- S Adams
- Department of Medicine, Perlmutter Cancer Center, New York University School of Medicine, New York, USA.
| | - P Schmid
- Centre for Experimental Cancer Medicin, Barts Cancer Institute, Queen Mary University London, London, UK
| | - H S Rugo
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco
| | - E P Winer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | | | - A Awada
- Oncology Medicine Departmen, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - D W Cescon
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - H Iwata
- Aichi Cancer Center Hospital, Nagoya, Japan
| | - M Campone
- Institut de Cancerologie de l'Ouest, Nantes, France
| | - R Nanda
- Department of Medicin, Section of Hematology/Oncology, The University of Chicago, Chicago, USA
| | - R Hui
- Westmead Hospital and the University of Sydney, Sydney, Australia
| | - G Curigliano
- Department of Oncology and Hematology, University of Milano, Milan; IEO, European Institute of Oncology IRCCS, Milano, Milan, Italy
| | - D Toppmeyer
- Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, USA
| | - J O'Shaughnessy
- Baylor University Medical Center, Dallas; Texas Oncology, Dallas; US Oncology, Dallas, USA
| | - S Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - S Paluch-Shimon
- Breast Cancer Service for Young Women, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - A R Tan
- Levine Cancer Institute, Atrium Health, Charlotte
| | - D Card
- Merck & Co., Inc., Kenilworth, USA
| | - J Zhao
- Merck & Co., Inc., Kenilworth, USA
| | | | - J Cortés
- Breast Cancer Program, Vall d'Hebron Institute of Oncology, Barcelona; Ramon y Cajal University Hospital, Madrid; IOB Institute of Oncology, Quiron Group, Barcelona, Spain
| |
Collapse
|
803
|
Adams S, Loi S, Toppmeyer D, Cescon DW, De Laurentiis M, Nanda R, Winer EP, Mukai H, Tamura K, Armstrong A, Liu MC, Iwata H, Ryvo L, Wimberger P, Rugo HS, Tan AR, Jia L, Ding Y, Karantza V, Schmid P. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of the phase II KEYNOTE-086 study. Ann Oncol 2019; 30:405-411. [PMID: 30475947 DOI: 10.1093/annonc/mdy518] [Citation(s) in RCA: 442] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Standard first-line treatment of metastatic triple-negative breast cancer (mTNBC) is chemotherapy. However, outcomes are poor, and new treatment options are needed. In cohort B of the phase II KEYNOTE-086 study, we evaluated pembrolizumab as first-line therapy for patients with PD-L1-positive mTNBC. PATIENTS AND METHODS Eligible patients had centrally confirmed mTNBC, no prior systemic anticancer therapy for metastatic disease, measurable disease at baseline per RECIST v1.1 by central review, no radiographic evidence of central nervous system metastases, and a tumor PD-L1 combined positive score ≥1. Patients received pembrolizumab 200 mg intravenously every 3 weeks for up to 2 years. The primary end point was safety. Secondary end points included objective response rate, disease control rate (percentage of patients with complete or partial response or stable disease for ≥24 weeks), duration of response, progression-free survival and overall survival. RESULTS All 84 patients enrolled were women, and 73 (86.9%) received prior (neo)adjuvant therapy. Fifty-three (63.1%) patients had treatment-related adverse events (AEs), including 8 patients (9.5%) with grade 3 severity; no patients experienced grade 4 AEs or died because of treatment-related AEs. Four patients had a complete response and 14 had a partial response, for an objective response rate of 21.4% (95% CI 13.9-31.4). Of the 13 patients with stable disease, 2 had stable disease lasting ≥24 weeks, for a disease control rate of 23.8% (95% CI 15.9-34.0). At data cut-off, 8 of 18 (44.4%) responses were ongoing, and median duration of response was 10.4 months (range 4.2 to 19.2+). Median progression-free survival was 2.1 months (95% CI 2.0-2.2), and median overall survival was 18.0 months (95% CI 12.9-23.0). CONCLUSIONS Pembrolizumab monotherapy had a manageable safety profile and showed durable antitumor activity as first-line therapy for patients with PD-L1-positive mTNBC. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, NCT02447003.
Collapse
Affiliation(s)
- S Adams
- Department of Medicine, Perlmutter Cancer Center, New York University School of Medicine, New York, USA.
| | - S Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - D Toppmeyer
- Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, USA
| | - D W Cescon
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - M De Laurentiis
- Dipartimento di Senologia, Istituto Nazionale Tumori - "Fondazione Pascale," Naples, Italy
| | - R Nanda
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago
| | - E P Winer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - H Mukai
- Department of Breast and Medical Oncology, National Cancer Center Hospital East, Kashiwa
| | - K Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - A Armstrong
- Breast Disease Research Group, The Christie NHS Foundation Trust, Manchester, UK
| | - M C Liu
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - H Iwata
- Aichi Cancer Center Hospital, Nagoya, Japan
| | - L Ryvo
- Division of Oncology, Sourasky Medical Center (Ichilov), Tel Aviv, Israel
| | - P Wimberger
- Department of Gynecology and Obstetric, University Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - H S Rugo
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco
| | - A R Tan
- Levine Cancer Institute, Atrium Health, Charlotte
| | - L Jia
- Merck & Co., Inc., Kenilworth, USA
| | - Y Ding
- Merck & Co., Inc., Kenilworth, USA
| | | | - P Schmid
- Centre for Experimental Cancer Medicin, Barts Cancer Institute, Queen Mary University London, London, UK
| |
Collapse
|
804
|
Gupta S, Vanderbilt CM, Cotzia P, Arias-Stella JA, Chang JC, Zehir A, Benayed R, Nafa K, Razavi P, Hyman DM, Baselga J, Berger MF, Ladanyi M, Arcila ME, Ross DS. Next-Generation Sequencing-Based Assessment of JAK2, PD-L1, and PD-L2 Copy Number Alterations at 9p24.1 in Breast Cancer: Potential Implications for Clinical Management. J Mol Diagn 2019; 21:307-317. [PMID: 30576871 PMCID: PMC6432425 DOI: 10.1016/j.jmoldx.2018.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/28/2018] [Accepted: 10/24/2018] [Indexed: 01/01/2023] Open
Abstract
Genomic amplification at 9p24.1, including the loci for JAK2, PD-L1, and PD-L2, has recently been described as a mechanism of resistance in postchemotherapy, triple-negative breast cancer. This genomic signature holds significant promise as a prognostic biomarker and has implications for targeted therapy with JAK2 inhibitors, as well as with immunotherapy. To guide future screening strategies, the frequency of these alterations was determined. A total of 5399 cases were included in the study. This encompassed 2890 institutional cases tested by the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets assay and 2509 cases from The Cancer Genome Atlas (TCGA). The combined incidence of 9p24.1 amplifications in both the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets and TCGA cohorts was 1.0% (56/5399 cases) and showed a >10-fold higher incidence in triple-negative breast cancer (triple-negative: 5.1%; non-triple-negative: 0.5%). Tumor mutation burden and stromal tumor infiltrating lymphocytes, parameters used to assess response to immunotherapy, were not significantly higher for these cases. The significance of genomic losses at 9p24.1 is unclear, and further studies are needed. Herein, we studied the spectrum of copy number alterations in breast cancer cases within our institutional clinical sequencing cohort and those profiled by TCGA to determine the frequency of genomic alterations that may predict response or resistance to JAK2 inhibitors and/or immunotherapy.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paolo Cotzia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Khedouja Nafa
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pedram Razavi
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David M Hyman
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - José Baselga
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
805
|
Telli ML, Vinayak S. Future of checkpoint blockade in triple-negative breast cancer: combination strategies to lead the way. Ann Oncol 2019; 30:347-348. [PMID: 30753266 DOI: 10.1093/annonc/mdz040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- M L Telli
- Department of Medicine, Division of Medical Oncology, Stanford University School of Medicine, Stanford.
| | - S Vinayak
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, USA
| |
Collapse
|
806
|
Fremd C, Hlevnjak M, Zapatka M, Zoernig I, Halama N, Fejzibegovic N, Thewes V, Lichter P, Schirmacher P, Kloor M, Marmé F, Schütz F, Kosaloglu Z, Sinn HP, Jäger D, Schneeweiss A. Mismatch Repair Deficiency Drives Durable Complete Remission by Targeting Programmed Death Receptor 1 in a Metastatic Luminal Breast Cancer Patient. Breast Care (Basel) 2019; 14:53-59. [PMID: 31019444 PMCID: PMC6465703 DOI: 10.1159/000492580] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In the field of breast cancer tumor biology, triple-negative breast cancer patients are the main focus of current clinical trials exploring the use of immune checkpoint inhibitors due to higher frequencies of somatic mutations, neoantigens, and resulting tumor-specific T-cell reactivity. CASE REPORT Here, we present the case of a 66-year-old woman with metastatic luminal breast cancer that rapidly responded to monotherapy with pembrolizumab, a monoclonal anti-PD-1 antibody. This patient obtained a partial clinical response within the first cycle of treatment and an ongoing durable complete remission after 12 weeks. Except for a transient immune-related thyreoiditis, there were no side effects observed offering remarkable quality of life to the patient. To evaluate the underlying mechanisms, we performed immunohistochemistry, explored the mutational landscape by whole-exome sequencing, and identified potential T-cell epitopes by prediction of neoantigens with high affinity binding to one of the patient's HLA. Briefly, we found a strong infiltration of CD8+ T cells without staining for PD-L1 in the tumor stroma. Exome sequencing revealed an enormous frequency of somatic and tumor-specific alterations, mainly C>T/G>A transitions. The mutational pattern was further linked to genome instability and deficient mismatch repair supported by the loss of MSH6 protein expression and therefore leading to susceptibility to immune checkpoint blockade. CONCLUSION Within the overall goal to establish operating procedures for breast cancer immunotherapy, we propose to re-evaluate testing for deficient mismatch repair and to further intensify the search for biomarkers predictive for the success of immune checkpoint modulation including all tumor biologic subtypes of breast cancer.
Collapse
Affiliation(s)
- Carlo Fremd
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg, Heidelberg, Germany
| | - Mario Hlevnjak
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Inka Zoernig
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg, Heidelberg, Germany
| | - Nino Fejzibegovic
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg, Heidelberg, Germany
| | - Verena Thewes
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Peter Schirmacher
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Florian Schütz
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Zeynep Kosaloglu
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Hans Peter Sinn
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg, Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
807
|
Scimeca M, Bonfiglio R, Urbano N, Cerroni C, Anemona L, Montanaro M, Fazi S, Schillaci O, Mauriello A, Bonanno E. Programmed death ligand 1 expression in prostate cancer cells is associated with deep changes of the tumor inflammatory infiltrate composition. Urol Oncol 2019; 37:297.e19-297.e31. [PMID: 30827759 DOI: 10.1016/j.urolonc.2019.02.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/12/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The main aim of this study was to investigate the putative correlation between the composition of intratumoral inflammatory infiltrate and the expression of programmed death ligand 1 (PD-L1) by prostate cancer cells. In addition, we evaluated the correlation between the expression of PD-L1 and PTX3. METHODS We enrolled 100 patients from which we collected one surgical sample each. Paraffin serial sections were obtained to perform histological classifications and tissues microarray construction. Serial tissues microarray paraffin sections were also used for PD-L1 analysis and intratumoral inflammatory infiltrate characterization (CD4, CD8, CD57, CD3, PD1, PSGL-1, TIGIT, CD20, CD38, CD68, CD163, and PTX3) by immunohistochemistry . RESULTS Our result showed a significant increase of the number of both PD-L1 and PTX3 positive cells in prostate tumors respect to benign lesions. Inflammatory infiltrate of PD-L1 positive prostate cancer lesions was characterized by a decrease of both PD1 positive lymphocytes and tumor-infiltrated macrophages, mainly M2 subpopulation. Also, PTX3 expression showed an inverse correlation with the number of PD-L1 positive prostate cancer cells. CONCLUSIONS If confirmed, our data could be useful to predict the variations of the inflammatory population related to PD-L1 expression in prostate cancer. This can lay the foundation to establish therapeutic protocols able to inhibit the PD-L1 activity and, at the same time, to reactivate the antitumor inflammatory process.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; San Raffaele University, Rome, Italy; OrchideaLab S.r.l., Rome, Italy
| | - Rita Bonfiglio
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Rome, Italy
| | | | - Chiara Cerroni
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Lucia Anemona
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Manuela Montanaro
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Sara Fazi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Orazio Schillaci
- San Raffaele University, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Mauriello
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Elena Bonanno
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; "Diagnostica Medica" and "Villa dei Platani", Avellino, Italy.
| |
Collapse
|
808
|
Caparica R, Lambertini M, Pondé N, Fumagalli D, de Azambuja E, Piccart M. Post-neoadjuvant treatment and the management of residual disease in breast cancer: state of the art and perspectives. Ther Adv Med Oncol 2019; 11:1758835919827714. [PMID: 30833989 PMCID: PMC6393951 DOI: 10.1177/1758835919827714] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/04/2019] [Indexed: 12/14/2022] Open
Abstract
Achieving a pathologic complete response after neoadjuvant treatment is associated with improved prognosis in breast cancer. The CREATE-X trial demonstrated a significant survival improvement with capecitabine in patients with residual invasive disease after neoadjuvant chemotherapy, and the KATHERINE trial showed a significant benefit of trastuzumab-emtansine (TDM1) in human epidermal growth factor receptor 2 (HER2)-positive patients who did not achieve a pathologic complete response after neoadjuvant treatment, creating interesting alternatives of post-neoadjuvant treatments for high-risk patients. New agents are arising as therapeutic options for metastatic breast cancer such as the cyclin-dependent kinase inhibitors and the immune-checkpoint inhibitors, but none has been incorporated into the post-neoadjuvant setting so far. Evolving techniques such as next-generation sequencing and gene expression profiles have improved our knowledge regarding the biology of residual disease, and also on the mechanisms involved in treatment resistance. The present manuscript reviews the current available strategies, the ongoing trials, the potential biomarker-guided approaches and the perspectives for the post-neoadjuvant treatment and the management of residual disease after neoadjuvant treatment in breast cancer.
Collapse
Affiliation(s)
- Rafael Caparica
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Matteo Lambertini
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Noam Pondé
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | - Martine Piccart
- Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 121, 1000 Bruxelles, Belgium
| |
Collapse
|
809
|
Niyomnaitham S, Parinyanitikul N, Roothumnong E, Jinda W, Samarnthai N, Atikankul T, Suktitipat B, Thongnoppakhun W, Limwongse C, Pithukpakorn M. Tumor mutational profile of triple negative breast cancer patients in Thailand revealed distinctive genetic alteration in chromatin remodeling gene. PeerJ 2019; 7:e6501. [PMID: 30828495 PMCID: PMC6394341 DOI: 10.7717/peerj.6501] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 01/22/2019] [Indexed: 12/28/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) is a breast cancer subtype characterized by absence of both hormonal receptors and human epithelial growth factor receptor 2 (HER2). TNBC accounts for 15–20% of breast cancer. TNBC is associated with more aggressive disease and worse clinical outcome. Though the underlying mechanism of TNBC is currently unclear, the heterogeneity of clinical characteristics in various population may relate to the difference in tumor mutational profile. There were studies on TNBC gene mutations in various ethnic groups but the tumor genome data on Thai TNBC patients is currently unknown. This study aims to investigate mutational profile of Thai TNBC. Methods The patients were Thai individuals who were diagnosed with primary breast carcinoma between 2014 and 2017. All surgically removed primary tumor tissues were carefully examined by pathologists and archived as formalin-fixed paraffin-embedded tumor. TNBC was defined by absence of hormonal receptors and HER2 by immunohistochemistry. Genomic DNA was extracted, enriched and sequenced of all exomes on the Illumina HiSeq. Genomic data were then processed through bioinformatics platform to identify genomic alterations and tumor mutational burden. Results A total of 116 TNBC patients were recruited. Genomic analysis of TNBC samples identified 81,460 variants, of which 5,906 variants were in cancer-associated genes. The result showed that Thai TNBC has higher tumor mutation burden than previously reported data. The most frequently mutated cancer-associated gene was TP53 similar to other TNBC cohorts. Meanwhile KMT2C was found to be more commonly mutated in Thai TNBC than previous studies. Mutational profile of Thai TNBC patients also revealed difference in many frequently mutated genes when compared to other Western TNBC cohorts. Conclusion This result supported that TNBC breast cancer patients from various ethnic background showed diverse genome alteration pattern. Although TP53 is the most commonly mutated gene across all cohorts, Thai TNBC showed different gene mutation frequencies, especially in KMT2C. In particular, the cancer gene mutations are more prevalent in Thai TNBC patients. This result provides important insight on diverse underlying genetic and epigenetic mechanisms of TNBC that could translate to a new treatment strategy for patients with this disease.
Collapse
Affiliation(s)
- Suvimol Niyomnaitham
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Napa Parinyanitikul
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ekkapong Roothumnong
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Worapoj Jinda
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Norasate Samarnthai
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Taywin Atikankul
- Department of Pathology, Queen Savang Vadhana Memorial Hospital, Thai Red Cross Society, Chonburi, Thailand
| | - Bhoom Suktitipat
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Integrative Computational Bioscience Center, Mahidol University, Bangkok, Thailand
| | - Wanna Thongnoppakhun
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanin Limwongse
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
810
|
Salmaninejad A, Valilou SF, Shabgah AG, Aslani S, Alimardani M, Pasdar A, Sahebkar A. PD-1/PD-L1 pathway: Basic biology and role in cancer immunotherapy. J Cell Physiol 2019; 234:16824-16837. [PMID: 30784085 DOI: 10.1002/jcp.28358] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
Over the course of past few years, cancer immunotherapy has been accompanied with promising results. However, preliminary investigations with respect to immunotherapy concentrated mostly on targeting the immune checkpoints, nowadays, emerge as the most efficient strategy to raise beneficial antitumor immune responses. Programmed cell death protein 1 (PD-1) plays an important role in subsiding immune responses and promoting self-tolerance through suppressing the activity of T cells and promoting differentiation of regulatory T cells. PD-1 is considered as an immune checkpoint and protects against autoimmune responses through both induction of apoptosis in antigen-specific T cells and inhibiting apoptosis in regulatory T cells. Several clinical trials exerting PD-1 monoclonal antibodies as well as other immune-checkpoint blockades have had prosperous outcomes and opened new horizons in tumor immunotherapy. Nonetheless, a bulk of patients have failed to respond to these newly emerging immune-based approach and the survival rate was not satisfying. Additional strategies, especially combination therapies, has been initiated and been further promising. Attempts to identify novel and well-suited predictive biomarkers are also sensed. In this review, the promotion of cancer immunotherapy targeting PD-1 immunoinhibitory pathway is discussed.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Department of Medical Genetics, Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Farajzadeh Valilou
- Medical Genetics Network (MeGeNe), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Malihe Alimardani
- Student Research Committee, Department of Medical Genetics, Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Pasdar
- Student Research Committee, Department of Medical Genetics, Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Applied Medicine, Medical School, University of Aberdeen, Aberdeen, UK
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
811
|
Mutational and Antigenic Landscape in Tumor Progression and Cancer Immunotherapy. Trends Cell Biol 2019; 29:396-416. [PMID: 30765144 DOI: 10.1016/j.tcb.2019.01.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/18/2022]
Abstract
Evolving neoplasms accumulate non-synonymous mutations at a high rate, potentially enabling the expression of antigenic epitopes that can be recognized by the immune system. Since they are not covered by central tolerance, such tumor neoantigens (TNAs) should be under robust immune control as they surge. However, genetic defects that impair cancer cell eradication by the immune system coupled with the establishment of local immunosuppression can enable TNA accumulation, which is generally associated with improved clinical sensitivity to various immunotherapies. Here, we explore how tumor-intrinsic factors and immunological processes shape the mutational and antigenic landscape of evolving neoplasms to influence clinical responses to immunotherapy, and propose strategies to achieve robust immunological control of the disease despite disabled immunosurveillance.
Collapse
|
812
|
Li M, Li A, Zhou S, Lv H, Yang W. SPAG5 upregulation contributes to enhanced c-MYC transcriptional activity via interaction with c-MYC binding protein in triple-negative breast cancer. J Hematol Oncol 2019; 12:14. [PMID: 30736840 PMCID: PMC6367803 DOI: 10.1186/s13045-019-0700-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that lacks effective therapeutic targets. Sperm-associated antigen 5 (SPAG5) is a mitotic spindle-associated protein that is involved in various biological processes in cervical cancer and bladder urothelial carcinoma. However, the role of SPAG5 in TNBC remains undefined. METHODS The expression of SPAG5 was examined in TNBC patients via quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry (IHC). The biological functions of SPAG5 in TNBC and the underlying mechanisms were investigated in vitro and in vivo. RESULTS SPAG5 expression was significantly upregulated in TNBC tissues compared with that in paired adjacent noncancerous tissues (ANTs). High SPAG5 expression was associated with increased lymph node metastasis and high risk of local recurrence. SPAG5 protein expression was significantly associated with poor disease-free survival in TNBC. Gene set enrichment analysis of TNBC data from The Cancer Genome Atlas (TCGA) indicated that high SPAG5 expression was significantly associated with cell cycle and the ATR-BRCA pathway. Functional assays demonstrated that SPAG5 expression promoted tumor growth in vitro and in vivo. In addition, SPAG5-silenced cells were more sensitive to the PARP inhibitor (PARPi) olaparib. Mechanistically, SPAG5 interacted with c-MYC binding protein (MYCBP), thereby increasing MYCBP protein levels and leading to increased c-MYC transcriptional activity, which promoted the expression of the c-MYC target genes: CDC20, CDC25C, BRCA1, BRCA2, and RAD51.Knockdown of MYCBP or c-MYC abolished the SPAG5-induced cell-cycle progression and cell proliferation of TNBC. CONCLUSIONS Collectively, our results indict that SPAG5 is an efficient prognostic factor in TNBC, and that SPAG5 knockdown increases the sensitivity of TNBC to the PARPi olaparib. SPAG5 promotes tumor growth and DNA repair by increasing c-MYC transcriptional activity via interaction with MYCBP. The SPAG5/MYCBP/c-MYC axis may represent a potential therapeutic target for TNBC treatment.
Collapse
Affiliation(s)
- Ming Li
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Anqi Li
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Shuling Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China. .,Institute of Pathology, Fudan University, Shanghai, China.
| |
Collapse
|
813
|
Yeong J, Lim JCT, Lee B, Li H, Ong CCH, Thike AA, Yeap WH, Yang Y, Lim AYH, Tay TKY, Liu J, Wong SC, Chen J, Lim EH, Iqbal J, Dent R, Newell EW, Tan PH. Prognostic value of CD8 + PD-1+ immune infiltrates and PDCD1 gene expression in triple negative breast cancer. J Immunother Cancer 2019; 7:34. [PMID: 30728081 PMCID: PMC6366051 DOI: 10.1186/s40425-019-0499-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
The role of programmed cell death protein-1 (PD-1)/programmed cell death ligand 1 (PD-L1) in triple negative breast cancer (TNBC) remains to be fully understood. In this study, we investigated the role of PD-1 as a prognostic marker for TNBC in an Asian cohort (n = 269). Samples from patients with TNBC were labeled with antibodies against PD-L1 and PD-1, and subjected to NanoString assays to measure the expression of immune-related genes. Associations between disease-free survival (DFS), overall survival (OS) and biomarker expression were investigated. Multivariate analysis showed that tumors with high PD-1+ immune infiltrates harbored significantly increased DFS, and this increase was significant even after controlling for clinicopathological parameters (HR 0.95; P = 0.030). In addition, the density of cells expressing both CD8 and PD-1, but not the density of CD8−PD-1+ immune infiltrates, was associated with improved DFS. Notably, this prognostic significance was independent of clinicopathological parameters and the densities of total CD8+ cell (HR 0.43, P = 0.011). At the transcriptional level, high expression of PDCD1 within the tumor was significantly associated with improved DFS (HR 0.38; P = 0.027). In line with these findings, high expression of IFNG (HR 0.38; P = 0.001) and IFN signaling genes (HR 0.46; p = 0.027) was also associated with favorable DFS. Inclusion of PD-1 immune infiltrates and PDCD1 gene expression added significant prognostic value for DFS (ΔLRχ2 = 6.35; P = 0.041) and OS (ΔLRχ2 = 9.53; P = 0.008), beyond that provided by classical clinicopathological variables. Thus, PD-1 mRNA and protein expression status represent a promising, independent indicator of prognosis in TNBC.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Jeffrey Chun Tatt Lim
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Huihua Li
- Division of Medicine, Singapore General Hospital, Singapore, Singapore
| | - Clara Chong Hui Ong
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Wei Hseun Yeap
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Yi Yang
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore.,Shanghai University of Finance and Economics, Shanghai, China
| | - Ansel Yi Herh Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Timothy Kwang Yong Tay
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore
| | - Jin Liu
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Siew-Cheng Wong
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Elaine Hsuen Lim
- National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Rebecca Dent
- National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore.
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore.
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore. .,Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
814
|
Heinhuis KM, Ros W, Kok M, Steeghs N, Beijnen JH, Schellens JHM. Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors. Ann Oncol 2019; 30:219-235. [PMID: 30608567 DOI: 10.1093/annonc/mdy551] [Citation(s) in RCA: 378] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Cancer immunotherapy has changed the standard of care for a subgroup of patients with advanced disease. Immune checkpoint blockade (ICB) in particular has shown improved survival compared with previous standards of care for several tumor types. Although proven to be successful in more immunogenic tumors, ICB is still largely ineffective in patients with tumors that are not infiltrated by immune cells, the so-called cold tumors. PATIENTS AND METHODS This review describes the effects of different chemotherapeutic agents on the immune system and the potential value of these different types of chemotherapy as combination partners with ICB in patients with solid tumors. Both preclinical data and currently ongoing clinical trials were evaluated. In addition, we reviewed findings regarding different dosing schedules, including the effects of an induction phase and applying metronomic doses of chemotherapy. RESULTS Combining ICB with other treatment modalities may lead to improved immunological conditions in the tumor microenvironment and could thereby enhance the antitumor immune response, even in tumor types that are so far unresponsive to ICB monotherapy. Chemotherapy, that was originally thought to be solely immunosuppressive, can exert immunomodulatory effects which may be beneficial in combination with immunotherapy. Each chemotherapeutic drug impacts the tumor microenvironment differently, and in order to determine the most suitable combination partners for ICB it is crucial to understand these mechanisms. CONCLUSION Preclinical studies demonstrate that the majority of chemotherapeutic drugs has been shown to exert immunostimulatory effects, either by inhibiting immunosuppressive cells and/or activating effector cells, or by increasing immunogenicity and increasing T-cell infiltration. However, for certain chemotherapeutic agents timing, dose and sequence of administration of chemotherapeutic agents and ICB is important. Further studies should focus on determining the optimal drug combinations, sequence effects and optimal concentration-time profiles in representative preclinical models.
Collapse
Affiliation(s)
- K M Heinhuis
- Divisions of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - W Ros
- Divisions of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - M Kok
- Medical Oncology and Molecular Oncology & Immunology, Utrecht University, Utrecht, The Netherlands
| | - N Steeghs
- Medical Oncology, Department of Clinical Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - J H Beijnen
- Divisions of Pharmacology, Utrecht University, Utrecht, The Netherlands; Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands; MC Slotervaart, Amsterdam, The Netherlands; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - J H M Schellens
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
815
|
Translational highlights in breast cancer research and treatment: recent developments with clinical impact. Curr Opin Obstet Gynecol 2019; 31:67-75. [DOI: 10.1097/gco.0000000000000510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
816
|
Garrido-Castro AC, Lin NU, Polyak K. Insights into Molecular Classifications of Triple-Negative Breast Cancer: Improving Patient Selection for Treatment. Cancer Discov 2019; 9:176-198. [PMID: 30679171 PMCID: PMC6387871 DOI: 10.1158/2159-8290.cd-18-1177] [Citation(s) in RCA: 891] [Impact Index Per Article: 148.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/15/2022]
Abstract
Triple-negative breast cancer (TNBC) remains the most challenging breast cancer subtype to treat. To date, therapies directed to specific molecular targets have rarely achieved clinically meaningful improvements in outcomes of patients with TNBC, and chemotherapy remains the standard of care. Here, we seek to review the most recent efforts to classify TNBC based on the comprehensive profiling of tumors for cellular composition and molecular features. Technologic advances allow for tumor characterization at ever-increasing depth, generating data that, if integrated with clinical-pathologic features, may help improve risk stratification of patients, guide treatment decisions and surveillance, and help identify new targets for drug development. SIGNIFICANCE: TNBC is characterized by higher rates of relapse, greater metastatic potential, and shorter overall survival compared with other major breast cancer subtypes. The identification of biomarkers that can help guide treatment decisions in TNBC remains a clinically unmet need. Understanding the mechanisms that drive resistance is key to the design of novel therapeutic strategies to help prevent the development of metastatic disease and, ultimately, to improve survival in this patient population.
Collapse
Affiliation(s)
- Ana C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
817
|
WNT signaling modulates PD-L1 expression in the stem cell compartment of triple-negative breast cancer. Oncogene 2019; 38:4047-4060. [PMID: 30705400 PMCID: PMC6755989 DOI: 10.1038/s41388-019-0700-2] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023]
Abstract
Triple-negative breast cancers (TNBCs) are characterized by a poor prognosis and lack of targeted treatments, and thus, new therapeutic strategies are urgently needed. Inhibitors against programmed death-1 (PD-1)/PD-1 ligand (PD-L1) have shown significant efficacy in various solid cancers, but their activity against TNBCs remains limited. Here, we report that human TNBCs molecularly stratified for high levels of PD-L1 (PD-L1High) showed significantly enriched expression of immune and cancer stemness pathways compared with those with low PD-L1 expression (PD-L1Low). In addition, the PD-L1High cases were significantly associated with a high stemness score (SSHigh) signature. TNBC cell lines gated for aldehyde dehydrogenase (ALDH) and CD44 stemness markers exhibited increased levels of PD-L1 versus their ALDH-negative and CD44Low counterparts, and PD-L1High cells generated significantly more mammospheres than PD-L1Low cells. Murine mammary SCA-1-positive tumor cells with PD-L1High expression generated tumors in vivo with higher efficacy than PD-L1Low cells. Furthermore, treatment of TNBC cells with selective WNT inhibitors or activators downregulated or upregulated PD-L1 expression, respectively, implying a functional cross-talk between WNT activity and PD-L1 expression. Remarkably, human TNBC samples contained tumor elements co-expressing PD-L1 with ALDH1A1 and/or CD44v6. Additionally, both PD-L1-/SCA1-positive and ALDH1A1-positive tumor elements were found in close contact with CD3-, and PD-1-positive T cells in murine and human tumor samples. Overall, our study suggests that PD-L1-positive tumor elements with a stemness phenotype may participate in the complex dynamics of TNBC-related immune evasion, which might be targeted through WNT signaling inhibition.
Collapse
|
818
|
Wang K, Li HL, Xiong YF, Shi Y, Li ZY, Li J, Zhang X, Li HY. Development and validation of nomograms integrating immune-related genomic signatures with clinicopathologic features to improve prognosis and predictive value of triple-negative breast cancer: A gene expression-based retrospective study. Cancer Med 2019; 8:686-700. [PMID: 30677255 PMCID: PMC6382728 DOI: 10.1002/cam4.1880] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/06/2018] [Accepted: 10/27/2018] [Indexed: 02/05/2023] Open
Abstract
Purpose Accumulating evidence indicated that triple‐negative breast cancer (TNBC) can stimulate stronger immune responses than other subtypes of breast cancer. We hypothesized that integrating immune‐related genomic signatures with clinicopathologic factors may yield a predictive accuracy exceeding that of the currently available system. Methods Ten signatures that reflect specific immunogenic or immune microenvironmental features of TNBC were identified and re‐analyzed using bioinformatic methods. Then, clinically annotated TNBC (n = 711) with the corresponding expression profiles, which predicted a patient's probability of disease‐free survival (DFS) and overall survival (OS), was pooled to evaluate their prognostic values and establish a clinicopathologic‐genomic nomogram. Three and two immune features were, respectively, selected out of 10 immune features to construct nomogram for DFS and OS prediction based on multivariate backward stepwise Cox regression analyses. Results By integrating the above immune expression signatures with prognostic clinicopathologic features, clinicopathologic‐genomic nomograms were cautiously constructed, which showed reasonable prediction accuracies (DFS: HR, 1.79; 95% CI, 1.46‐2.18, P < 0.001; AUC, 0.71; OS: HR, 1.96; 95% CI, 1.54‐2.49; P < 0.001; AUC, 0.73). The nomogram showed low‐risk subgroup had higher immune checkpoint molecules (PD‐L1, PD‐1, CTLA‐4, LAG‐3) expression and benefited from radiotherapy (HR, 0.2, 95% CI, 0.05‐0.89; P = 0.034) rather than chemotherapy (HR, 1.26, 95% CI, 0.66‐2.43; P = 0.485). Conclusions These findings offer evidence that immune‐related genomic data provide independent and complementary prognostic information for TNBC, and the nomogram might be a practical predictive tool to identify TNBC patients who would benefit from chemotherapy, radiotherapy, and upcoming popularity of immunotherapy.
Collapse
Affiliation(s)
- Kang Wang
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Hai-Lin Li
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yong-Fu Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yang Shi
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, Georgia.,West China School of Public Health, Sichuan University, Chengdu, China
| | - Zhu-Yue Li
- Institute of Hospital Management, West China Hospital, Sichuan University, Chengdu, China.,West China Hospital/West China School of Nursing, Sichuan University, Chengdu, China
| | - Jie Li
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xiang Zhang
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Hong-Yuan Li
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
819
|
Jia YQ, Yang B, Wen LL, Mu WX, Wang Z, Cheng B. Prognostic value of immune checkpoint molecules in head and neck cancer: a meta-analysis. Aging (Albany NY) 2019; 11:501-522. [PMID: 30668545 PMCID: PMC6366990 DOI: 10.18632/aging.101756] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 01/01/2019] [Indexed: 04/13/2023]
Abstract
Immune checkpoint molecules are important targets in cancer immunotherapy, but their association with prognosis in patients with head and neck cancer is controversial. In this meta-analysis, we searched for 12 immune checkpoint molecules in the PubMed, Embase and Cochrane Library databases and retrieved 52 studies with 7127 participants. Among the molecules included in the search, indoleamine 2, 3-dioxygenase (IDO), programmed death ligand 1 (PD-L1), and programmed death 1 (PD-1) met the inclusion criteria for further analysis. Higher expression of IDO was associated with poorer overall survival in head and neck cancer patients (P = 0.011), but higher expression of PD-L1 correlated with better overall survival specifically in nasopharyngeal carcinoma patients (P = 0.01). In a sensitivity analysis, higher PD-L1 expression correlated with better progression-free survival (P = 0.043), and was associated with better overall survival in Caucasian subjects (P = 0.02), nasopharyngeal carcinoma patients (P = 0.015), and studies with small sample sizes (P = 0.001). PD-1 had no prognostic significance. There was no publication bias affecting the results. Thus, among the immune checkpoint molecules, IDO and PD-L1 are potential prognostic predictors in head and neck cancer.
Collapse
Affiliation(s)
- Yi-Qun Jia
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Equal contribution
| | - Bo Yang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Equal contribution
| | - Li-Ling Wen
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Wen-Xin Mu
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Zhi Wang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Bin Cheng
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| |
Collapse
|
820
|
Hierro C, Matos I, Martin-Liberal J, Ochoa de Olza M, Garralda E. Agnostic-Histology Approval of New Drugs in Oncology: Are We Already There? Clin Cancer Res 2019; 25:3210-3219. [DOI: 10.1158/1078-0432.ccr-18-3694] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/23/2018] [Accepted: 01/18/2019] [Indexed: 11/16/2022]
|
821
|
Ayoub NM, Al-Shami KM, Yaghan RJ. Immunotherapy for HER2-positive breast cancer: recent advances and combination therapeutic approaches. BREAST CANCER-TARGETS AND THERAPY 2019; 11:53-69. [PMID: 30697064 PMCID: PMC6340364 DOI: 10.2147/bctt.s175360] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer immunotherapy has evolved dramatically with improved understanding of immune microenvironment and immunosurveillance. The immunogenicity of breast cancer is rather heterogeneous. Specific subtypes of breast cancer such as estrogen receptor (ER)-negative, human EGF receptor 2 (HER2)-positive, and triple-negative breast cancer (TNBC) have shown evidence of immunogenicity based on tumor–immune interactions. Several preclinical and clinical studies have explored the potential for immunotherapy to improve the clinical outcomes for different subtypes of breast cancer. This review describes the immune microenvironment of HER2-positive breast cancer and summarizes recent clinical advances of immunotherapeutic treatments in this breast cancer subtype. The review provides rationale and ongoing clinical evidence to the use of immune checkpoint inhibitors, therapeutic vaccines, and adoptive T cell immunotherapy in breast cancer. In addition, the present paper describes the most relevant clinical progress of strategies for the combination of immunotherapy with standard treatment modalities in HER2-positive breast cancer including chemotherapy, targeted therapy, and radiotherapy.
Collapse
Affiliation(s)
- Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan,
| | - Kamal M Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Rami J Yaghan
- Department of General Surgery and Urology, Faculty of Medicine, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
822
|
Kurozumi S, Matsumoto H, Kurosumi M, Inoue K, Fujii T, Horiguchi J, Shirabe K, Oyama T, Kuwano H. Prognostic significance of tumour-infiltrating lymphocytes for oestrogen receptor-negative breast cancer without lymph node metastasis. Oncol Lett 2019; 17:2647-2656. [PMID: 30867728 PMCID: PMC6396218 DOI: 10.3892/ol.2019.9938] [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: 03/29/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022] Open
Abstract
Tumour-infiltrating lymphocytes (TILs) are regarded as significant prognostic markers in patients with breast cancer. However, the prognostic utility of TIL expression based on the intrinsic subtypes has just been identified. The present study investigated the relationship between TIL grades and prognosis in 294 Japanese paitents with breast cancer stratified based on the intrinsic subtypes and clinicopathological characteristics. Stromal TIL status was evaluated using haematoxylin and eosin staining, and TIL grades were categorised into low (<10%), intermediate (≥10 and ≤40%) and high (>40%) groups. The relationship between TIL expression and the intrinsic subtypes, clinicopathological characteristics and patient prognosis was analyzed. It was revealed that high TIL expression was correlated with negative oestrogen receptor (ER) expression and high histological grade (P<0.001). Among the ER-negative patients, the relapse-free survival (RFS) rate of the high-grade TIL group was significantly higher than that of the low-grade TIL group (P=0.04). Among the ER-negative patients without lymph node metastasis, RFS and cancer-specific survival (CSS) rates of patients with high-grade TILs were significantly higher than the RFS and CSS rates of patients with low-grade TILs (P=0.01). However, among ER-positive patients, RFS was significantly higher in the low-grade TIL group than in the high-grade TIL group (P=0.02). In conclusion, TIL expression correlated with ER status and tumour proliferation. High TIL expression was a poor prognostic marker in ER-positive patients but was a good prognostic marker in ER-negative patients. Therefore, the biological association between TILs and primary breast tumours may differ between ER-positive and ER-negative breast cancer.
Collapse
Affiliation(s)
- Sasagu Kurozumi
- Division of Breast Surgery, Saitama Cancer Center, Saitama 362-0806, Japan.,Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hiroshi Matsumoto
- Division of Breast Surgery, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Masafumi Kurosumi
- Department of Pathology, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Kenichi Inoue
- Division of Breast Oncology, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Takaaki Fujii
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Narita, Chiba 286-8686, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hiroyuki Kuwano
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
823
|
McCart Reed AE, Kalita-De Croft P, Kutasovic JR, Saunus JM, Lakhani SR. Recent advances in breast cancer research impacting clinical diagnostic practice. J Pathol 2019; 247:552-562. [PMID: 30426489 DOI: 10.1002/path.5199] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/29/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
During the last decade, the genomics revolution has driven critical advances in molecular oncology and pathology, and a deeper appreciation of heterogeneity that is beginning to reshape our thinking around diagnostic classification. Recent developments have seen existing classification systems modified and improved where possible, gene-based diagnostics implemented and tumour-immune interactions modulated. We present a detailed discussion of this progress, including advances in the understanding of breast tumour classification, e.g. mixed ductal-lobular tumours and the spectrum of triple-negative breast cancer. The latest information on clinical trials and the implementation of gene-based diagnostics, including MammaPrint and Oncotype Dx and others, is synthesised, and emerging targeted therapies, as well as the burgeoning immuno-oncology field, and their relevance in breast cancer, are discussed. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Amy E McCart Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Priyakshi Kalita-De Croft
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Jamie R Kutasovic
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Jodi M Saunus
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Pathology Queensland, The Royal Brisbane & Women's Hospital, Brisbane, Australia
| |
Collapse
|
824
|
Reguera-Nuñez E, Xu P, Chow A, Man S, Hilberg F, Kerbel RS. Therapeutic impact of Nintedanib with paclitaxel and/or a PD-L1 antibody in preclinical models of orthotopic primary or metastatic triple negative breast cancer. J Exp Clin Cancer Res 2019; 38:16. [PMID: 30635009 PMCID: PMC6330500 DOI: 10.1186/s13046-018-0999-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/06/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is an aggressive malignancy with poor prognosis, in part because of the current lack of any approved molecularly targeted therapy. We evaluated various combinations of three different drugs: nintedanib, an antiangiogenic TKI targeting VEGF receptors, paclitaxel (PTX), or a PD-L1 antibody, using models of orthotopic primary or advanced metastatic TNBC involving a metastatic variant of the MDA-MB-231 human cell line (called LM2-4) in SCID mice and two mouse lines (EMT-6 and a drug-resistant variant, EMT-6/CDDP) in immunocompetent mice. These drugs were selected based on the following: PTX is approved for TNBC; nintedanib combined with docetaxel has shown phase III clinical trial success, albeit in NSCLC; VEGF can act as local immunosuppressive factor; and PD-L1 antibody plus taxane therapy was recently reported to have encouraging phase III trial benefit in TNBC. METHODS Statistical analyses were performed with ANOVA followed by Tukey's Multiple Comparison Test or with Kruskal-Wallis test followed by Dunn's Multiple Comparison Test. Survival curves were analyzed using a Log-rank (Mantel Cox) test. Differences were considered statistically significant when p values were < 0.05. RESULTS Toxicity analyses showed that nintedanib is well tolerated when administered 5-days ON 2-days OFF; PTX toxicity differed in mice, varied with cell lines used and may have influenced median survival in the metastatic EMT6/CDDP model; while toxicity of PD-L1 therapy depended on the cell lines and treatment settings tested. In the LM2-4 system, combining nintedanib with PTX enhanced overall antitumor efficacy in both primary and metastatic treatment settings. In immunocompetent mice, combining nintedanib or PTX with the PD-L1 antibody improved overall antitumor efficacy. Using the advanced metastatic EMT-6/CDDP model, optimal efficacy results were obtained using the triple combination. CONCLUSIONS These results suggest circumstances where nintedanib plus PTX may be potentially effective in treating TNBC, and nintedanib with PTX may improve PD-L1 therapy of metastatic TNBC.
Collapse
Affiliation(s)
- Elaine Reguera-Nuñez
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario Canada
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Ave, room S-217, Toronto, Ontario M4N 3M5 Canada
| | - Ping Xu
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Ave, room S-217, Toronto, Ontario M4N 3M5 Canada
| | - Annabelle Chow
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Ave, room S-217, Toronto, Ontario M4N 3M5 Canada
| | - Shan Man
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Ave, room S-217, Toronto, Ontario M4N 3M5 Canada
| | | | - Robert S. Kerbel
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario Canada
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Ave, room S-217, Toronto, Ontario M4N 3M5 Canada
| |
Collapse
|
825
|
Satram-Hoang S, Bajaj P, Stein A, Cortazar P, Momin F, Reyes C. Treatment Patterns and Mortality Risk among Elderly Patients with Metastatic Triple Negative Breast Cancer in the United States: An Observational Cohort Study Using SEER-Medicare Data. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/jct.2019.102009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
826
|
Abstract
Precision medicine approaches have found applications in the treatment of several tumor types and have led to rapid advancement in the number of available therapies for some difficult-to-treat diseases. In comparison to tumors like EGFR-mutated lung cancer, and BRAF-mutated melanoma for example, precision medicine in breast cancer is still in its infancy despite the much earlier identification of targets like ER and HER2. Though significant progress has been made in new therapies for hormone-receptor-positive and HER2-positive breast cancers, identification of molecular heterogeneity and lack of other valid reproducible targets in triple-negative breast cancer remain a challenge. In this chapter, we outline the recent advances in technology and targeted treatments for breast cancer, the remaining challenges and ongoing efforts to address these to make precision medicine a reality for all breast cancer patients.
Collapse
Affiliation(s)
- Jasgit C Sachdev
- HonorHealth Research Institute, Scottsdale, AZ, USA. .,Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Ana C Sandoval
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Mohammad Jahanzeb
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| |
Collapse
|
827
|
Systemic Treatment of HER2-Negative Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
828
|
Guha A, Armanious M, Fradley MG. Update on cardio-oncology: Novel cancer therapeutics and associated cardiotoxicities. Trends Cardiovasc Med 2019; 29:29-39. [DOI: 10.1016/j.tcm.2018.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/22/2018] [Accepted: 06/03/2018] [Indexed: 02/08/2023]
|
829
|
Diaz Bessone MI, Gattas MJ, Laporte T, Tanaka M, Simian M. The Tumor Microenvironment as a Regulator of Endocrine Resistance in Breast Cancer. Front Endocrinol (Lausanne) 2019; 10:547. [PMID: 31440208 PMCID: PMC6694443 DOI: 10.3389/fendo.2019.00547] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor positive breast neoplasias represent over 70% of diagnosed breast cancers. Depending on the stage at which the tumor is detected, HER2 status and genomic risk, endocrine therapy is combined with either radio, chemo and/or targeted therapy. A growing amount of evidence supports the notion that components of the tumor microenvironment play specific roles in response to treatment and that strategies targeting these key interactions with tumor cells could pave the way to a new generation of therapies. In this review, we analyze the evidence suggesting different components of the tumor microenvironment play a role in hormone receptor positive breast cancer progression. In particular we focus on the immune system, carcinoma associated fibroblasts and the extracellular matrix. Further insight into the cross talk between these constituents of the microenvironment and the tumor cells may lead to therapies that eliminate disseminated metastatic cells early on, and thus reduce distant disease relapse which is the leading cause of death for patients who are diagnosed with this illness.
Collapse
Affiliation(s)
- María Inés Diaz Bessone
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - María José Gattas
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Tomás Laporte
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Max Tanaka
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
- Amsterdam UMC, VUmc School of Medical Sciences, University of Vrije, Amsterdam, Netherlands
| | - Marina Simian
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
- *Correspondence: Marina Simian
| |
Collapse
|
830
|
Quist J, Mirza H, Cheang MCU, Telli ML, O'Shaughnessy JA, Lord CJ, Tutt ANJ, Grigoriadis A. A Four-gene Decision Tree Signature Classification of Triple-negative Breast Cancer: Implications for Targeted Therapeutics. Mol Cancer Ther 2019; 18:204-212. [PMID: 30305342 PMCID: PMC6330084 DOI: 10.1158/1535-7163.mct-18-0243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/02/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022]
Abstract
The molecular complexity of triple-negative breast cancers (TNBCs) provides a challenge for patient management. We set out to characterize this heterogeneous disease by combining transcriptomics and genomics data, with the aim of revealing convergent pathway dependencies with the potential for treatment intervention. A Bayesian algorithm was used to integrate molecular profiles in two TNBC cohorts, followed by validation using five independent cohorts (n = 1,168), including three clinical trials. A four-gene decision tree signature was identified, which robustly classified TNBCs into six subtypes. All four genes in the signature (EXO1, TP53BP2, FOXM1, and RSU1) are associated with either genomic instability, malignant growth, or treatment response. One of the six subtypes, MC6, encompassed the largest proportion of tumors (∼50%) in early diagnosed TNBCs. In TNBC patients with metastatic disease, the MC6 proportion was reduced to 25%, and was independently associated with a higher response rate to platinum-based chemotherapy. In TNBC cell line data, platinum sensitivity was recapitulated, and a sensitivity to the inhibition of the phosphatase PPM1D was revealed. Molecularly, MC6-TNBCs displayed high levels of telomeric allelic imbalances, enrichment of CD4+ and CD8+ immune signatures, and reduced expression of genes negatively regulating the MAPK signaling pathway. These observations suggest that our integrative classification approach may identify TNBC patients with discernible and theoretically pharmacologically tractable features that merit further studies in prospective trials.
Collapse
Affiliation(s)
- Jelmar Quist
- Cancer Bioinformatics, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom
- Breast Cancer Now Research Unit, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, Cancer Research UK King's Health Partners Centre, King's College London, London, United Kingdom
| | - Hasan Mirza
- Cancer Bioinformatics, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom
- Breast Cancer Now Research Unit, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, Cancer Research UK King's Health Partners Centre, King's College London, London, United Kingdom
| | - Maggie C U Cheang
- Clinical Trials and Statistics Unit (ICR-CTSU), The Institute of Cancer Research, Surrey, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Melinda L Telli
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Christopher J Lord
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom
| | - Andrew N J Tutt
- Breast Cancer Now Research Unit, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, Cancer Research UK King's Health Partners Centre, King's College London, London, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Anita Grigoriadis
- Cancer Bioinformatics, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom.
- Breast Cancer Now Research Unit, Cancer Centre at Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, Cancer Research UK King's Health Partners Centre, King's College London, London, United Kingdom
| |
Collapse
|
831
|
Qin Y, Vasilatos SN, Chen L, Wu H, Cao Z, Fu Y, Huang M, Vlad AM, Lu B, Oesterreich S, Davidson NE, Huang Y. Inhibition of histone lysine-specific demethylase 1 elicits breast tumor immunity and enhances antitumor efficacy of immune checkpoint blockade. Oncogene 2019; 38:390-405. [PMID: 30111819 PMCID: PMC6336685 DOI: 10.1038/s41388-018-0451-5] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/18/2018] [Accepted: 07/21/2018] [Indexed: 01/03/2023]
Abstract
Immunotherapy strategies have been emerging as powerful weapons against cancer. Early clinical trials reveal that overall response to immunotherapy is low in breast cancer patients, suggesting that effective strategies to overcome resistance to immunotherapy are urgently needed. In this study, we investigated whether epigenetic reprograming by modulating histone methylation could enhance effector T lymphocyte trafficking and improve therapeutic efficacy of immune checkpoint blockade in breast cancer with focus on triple-negative breast cancer (TNBC) subtype. In silico analysis of The Cancer Genome Atlas (TCGA) data shows that expression of histone lysine-specific demethylase 1 (LSD1) is inversely associated with the levels of cytotoxic T cell-attracting chemokines (C-C motif chemokine ligand 5 (CCL5), C-X-C motif chemokine ligand 9 and 10 (CXCL9, CXCL10)) and programmed death-ligand 1 (PD-L1) in clinical TNBC specimens. Tiling chromatin immunoprecipitation study showed that re-expression of chemokines by LSD1 inhibition is associated with increased H3K4me2 levels at proximal promoter regions. Rescue experiments using concurrent treatment with small interfering RNA or inhibitor of chemokine receptors blocked LSD1 inhibitor-enhanced CD8+ T cell migration, indicating a critical role of key T cell chemokines in LSD1-mediated CD8+ lymphocyte trafficking to the tumor microenvironment. In mice bearing TNBC xenograft tumors, anti-PD-1 antibody alone failed to elicit obvious therapeutic effect. However, combining LSD1 inhibitors with PD-1 antibody significantly suppressed tumor growth and pulmonary metastasis, which was associated with reduced Ki-67 level and augmented CD8+ T cell infiltration in xenograft tumors. Overall, these results suggest that LSD1 inhibition may be an effective adjuvant treatment with immunotherapy as a novel management strategy for poorly immunogenic breast tumors.
Collapse
Affiliation(s)
- Ye Qin
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shauna N Vasilatos
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lin Chen
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hao Wu
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhishen Cao
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yumei Fu
- Allegheny General Hospital Pathology, Pittsburgh, PA, USA
| | - Min Huang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anda M Vlad
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, and University of Washington, Seattle, WA, USA
| | - Yi Huang
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
832
|
De Los Santos MC, Dragomir MP, Calin GA. The role of exosomal long non-coding RNAs in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:1178-1192. [PMID: 31867576 PMCID: PMC6924635 DOI: 10.20517/cdr.2019.74] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
One of the major challenges in oncology is drug resistance, which triggers relapse and shortens patients’ survival. In order to promote drug desensitization, cancer cells require the establishment of an ideal tumor microenvironment that accomplishes specific conditions. To achieve this objective, cellular communication is a key factor. Classically, cells were believed to restrictively communicate by ligand-receptor binding, physical cell-to-cell interactions and synapses. Nevertheless, the crosstalk between tumor cells and stroma cells has also been recently reported to be mediated through exosomes, the smallest extracellular vesicles, which transport a plethora of functionally active molecules, such as: proteins, lipids, messenger RNA, DNA, microRNA or long non-coding RNA (lncRNAs). LncRNAs are RNA molecules greater than 200 base pairs that are deregulated in cancer and other diseases. Exosomal lncRNAs are highly stable and can be found in several body fluids, being considered potential biomarkers for tumor liquid biopsy. Exosomal lncRNAs promote angiogenesis, cell proliferation and drug resistance. The role of exosomal lncRNAs in drug resistance affects the main treatment strategies in oncology: chemotherapy, targeted therapy, hormone therapy and immunotherapy. Overall, knowing the molecular mechanisms by which exosomal lncRNA induce pharmacologic resistance could improve further drug development and identify drug resistance biomarkers.
Collapse
Affiliation(s)
- Mireia Cruz De Los Santos
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Mihnea P Dragomir
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.,Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 40015, Romania.,Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest 022328, Romania
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.,Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
833
|
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States; Université Paris Descartes/Paris V, Paris, France.
| | - Nils-Petter Rudqvist
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
834
|
Chen H, Lu W, Zhang Y, Zhu X, Zhou J, Chen Y. A Bayesian network meta-analysis of the efficacy of targeted therapies and chemotherapy for treatment of triple-negative breast cancer. Cancer Med 2019; 8:383-399. [PMID: 30525293 PMCID: PMC6346255 DOI: 10.1002/cam4.1892] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/28/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease with poorer prognosis than other subtypes, yet effective therapies are still not available. We aimed to compare the efficacy of various targeted therapies with chemotherapy (CT) in TNBC patients using a network meta-analysis. A systematic literature search was performed in PubMed, EMBASE, and the Cochrane Library. A total of 27 randomized controlled trials (RCTs), involving 6924 TNBC patients, were included. Olaparib significantly improved PFS (0.43, 0.29-0.64) and ORR (2.57, 1.31-5.09) in comparison with CT. As for bevacizumab + CT, it showed a significant improvement of PFS (0.66, 0.55-0.80) and ORR (2.15, 1.16-4.05) compared with CT + placebo. It was also superior to CT alone in PFS (0.48, 0.35-0.65) and pCR (1.30, 1.13-1.49 for breast and axillary nodes and 1.26, 1.11-1.44 for breast). Other targeted agents like iniparib, sorafenib, cetuximab, and ipatasertib combined with CT showed significant superiority in PFS compared with CT alone, and the HRs were 0.75 (0.62-0.90), 0.44 (0.21-0.91), 0.67 (0.47-0.96), and 0.44 (0.24-0.81), respectively, while some other agents such as sunitinib and cetuximab had the lowest SUCRA in OS, PFS, or ORR without any benefits. In conclusion, our results indicated that the addition of bevacizumab to CT was beneficial for TNBC patients, and olaparib had a great effect in PFS and ORR, especially for those with BRCA mutations. When combined with CT, targeted agents including iniparib, sorafenib, cetuximab, and ipatasertib may have better efficacies for treating TNBC.
Collapse
Affiliation(s)
- Huihui Chen
- Department of Surgical Oncology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Wei Lu
- Department of Surgical Oncology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Yixin Zhang
- Department of Surgical Oncology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Department of Thyroid and Breast SurgeryYinzhou People HospitalNingboZhejiangChina
| | - Xuan Zhu
- Department of Surgical Oncology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- The Key Laboratory of Cancer Prevention and InterventionChina National Ministry of EducationHangzhouZhejiangChina
| | - Jiaojiao Zhou
- Department of Surgical Oncology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- The Key Laboratory of Cancer Prevention and InterventionChina National Ministry of EducationHangzhouZhejiangChina
| | - Yiding Chen
- Department of Surgical Oncology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- The Key Laboratory of Cancer Prevention and InterventionChina National Ministry of EducationHangzhouZhejiangChina
| |
Collapse
|
835
|
Aydiner A, Igci A, Cabioglu N, Ozer L, Sen F, Keskin S, Muslumanoglu M, Karanlik H, Arslan Ibis K, Kucucuk S, Dincer M, Yavuz E, Tuzlali S, Soran A. Decision Pathways in Breast Cancer Management. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
836
|
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: 366] [Impact Index Per Article: 52.3] [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
|
837
|
Collins JM, Gulley JL. Product review: avelumab, an anti-PD-L1 antibody. Hum Vaccin Immunother 2018; 15:891-908. [PMID: 30481100 PMCID: PMC6605872 DOI: 10.1080/21645515.2018.1551671] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/05/2018] [Accepted: 11/17/2018] [Indexed: 02/06/2023] Open
Abstract
Although immunotherapies have been employed for many decades, immune checkpoint inhibitors have only recently entered the oncologic landscape. Avelumab is a fully human monoclonal antibody that blocks the interaction between PD-L1 on tumor cells and PD-1 on T cells, thereby inhibiting immunosuppression in the tumor microenvironment and reducing tumor growth. Most early clinical trials of avelumab as monotherapy and in combination regimens were part of the international JAVELIN clinical trial program, which included more than 7000 patients in more than 30 trials with at least 15 tumor types. Avelumab has been approved by the U.S. FDA for the treatment of metastatic Merkel cell carcinoma and metastatic urothelial carcinoma that has progressed during or following treatment with a platinum-based regimen. Its acceptable safety profile and ability to induce durable responses in otherwise deadly tumors provide the rationale for its use in other tumor types and in combination with other therapies.
Collapse
Affiliation(s)
- Julie M. Collins
- Medical Oncology Service, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James L. Gulley
- Medical Oncology Service, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
838
|
Kuehnemuth B, Piseddu I, Wiedemann GM, Lauseker M, Kuhn C, Hofmann S, Schmoeckel E, Endres S, Mayr D, Jeschke U, Anz D. CCL1 is a major regulatory T cell attracting factor in human breast cancer. BMC Cancer 2018; 18:1278. [PMID: 30572845 PMCID: PMC6302432 DOI: 10.1186/s12885-018-5117-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/20/2018] [Indexed: 01/03/2023] Open
Abstract
Background Regulatory T cells (Treg) suppress cytotoxic T cell anti-tumoral immune responses and thereby promote tumor progression. Prevention of intratumoral Treg accumulation by inhibition of their migration to the tumor microenvironment is a promising therapeutic strategy. The aim of this study was to identify the role of the two major Treg-attracting chemokines CCL1 and CCL22 in human breast cancer. Methods One hundred ninety-nine tissue samples of patients with invasive breast cancer were stained for CCL1 and CCL22 by immunohistochemistry. Chemokine expression and tumor infiltration by regulatory T cells, determined by expression of the transcription factor FoxP3, were quantified and their correlation to clinical features was statistically analyzed. Results Both CCL1 and CCL22 were expressed in most breast cancer tissues. CCL1 was significantly over-expressed in invasive breast cancer as compared to normal breast tissue. CCL1, but surprisingly not CCL22, showed a significant correlation with the number of tumor-infiltrating FoxP3+ Treg (p< 0.001). High numbers of intratumoral CCL1 expressing cells were related to high grade tumors (G4) and a positive estrogen receptor (ER) status whereas high CCL22 expression was generally seen in lower grade tumors. The median survival of 88 patients with high intratumoral CCL1 expression was 37 months compared to 50 months for the 87 patients with low CCL1 levels, this trend was however not statistically significant. Conclusions We found a high expression of CCL1 in human breast cancer. CCL1 significantly correlated with the infiltration of immunosuppressive FoxP3+ Treg, that are known to negatively affect survival. Thus, CCL1 may serve as prognostic marker and novel therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Benjamin Kuehnemuth
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Ignazio Piseddu
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Gabriela M Wiedemann
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany.,Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Lauseker
- Institut für medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Klinikum der Universität München, Munich, Germany
| | - Christina Kuhn
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Munich, Germany
| | - Simone Hofmann
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Munich, Germany
| | - Elisa Schmoeckel
- Pathologisches Institut, Medizinische Fakultät der Ludwig-Maximilians Universität München, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Doris Mayr
- Pathologisches Institut, Medizinische Fakultät der Ludwig-Maximilians Universität München, Munich, Germany
| | - Udo Jeschke
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Munich, Germany
| | - David Anz
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany. .,Medizinische Klinik und Poliklinik II, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
839
|
Pelekanou V, Villarroel-Espindola F, Schalper KA, Pusztai L, Rimm DL. CD68, CD163, and matrix metalloproteinase 9 (MMP-9) co-localization in breast tumor microenvironment predicts survival differently in ER-positive and -negative cancers. Breast Cancer Res 2018; 20:154. [PMID: 30558648 PMCID: PMC6298021 DOI: 10.1186/s13058-018-1076-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Abstract
Background The role of tumor-associated macrophages (TAMs) in the cancer immune landscape and their potential as treatment targets or modulators of response to treatment are gaining increasing interest. TAMs display high molecular and functional complexity. Therefore their objective assessment as breast cancer biomarkers is critical. The aims of this study were to objectively determine the in situ expression and significance of TAM biomarkers (CD68, CD163, and MMP-9) in breast cancer and to identify subclasses of patients who could benefit from TAM-targeting therapies. Methods We measured CD68, CD163, and MMP-9 protein expression in formalin-fixed paraffin-embedded tissues of breast carcinomas represented in tissue microarray format using multiplexed quantitative immunofluorescence (QIF) in two independent Yale cohorts: cohort A—n = 398, estrogen receptor–positive (ER+) and ER− cases—and the triple-negative breast cancer (TNBC)-only cohort B (n = 160). Associations between macrophage markers, ER status, and survival were assessed. Protein expression measured by QIF was compared with mRNA expression data from the METABRIC study. Results All three macrophage markers were co-expressed, displaying higher expression in ER− cancers. High pan-macrophage marker CD68 correlated with poorer overall survival (OS) only in ER− cases of cohort A (P = 0.02). High expression of CD163 protein in TAMs was associated with improved OS in ER− cases (cohort A, P = 0.03 and TNBC cohort B, P = 0.04, respectively) but not in ER+ cancers. MMP-9 protein was not individually associated with OS. High expression of MMP-9 in the CD68+/CD163+ TAMs was associated with worse OS in ER+ tumors (P <0.001) but not in ER− cancers. In the METABRIC dataset, mRNA levels followed the co-expression pattern observed in QIF but did not always show the same trend regarding OS. Conclusions Macrophage activity markers correlate with survival differently in ER+ and ER− cancers. The association between high co-expression and co-localization of MMP-9/CD163/CD68 and poor survival in ER+ cancers suggests that these cancers may be candidates for macrophage-targeted therapies. Electronic supplementary material The online version of this article (10.1186/s13058-018-1076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vasiliki Pelekanou
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA. .,Sanofi US Services Inc., Bridgewater Township, USA.
| | - Franz Villarroel-Espindola
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| | - Lajos Pusztai
- Department of Medical Oncology, Yale School of Medicine, 330 Cedar Street, New Haven, 06520, CT, USA
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| |
Collapse
|
840
|
Novel Approaches to Immunotherapy in Triple Negative Breast Cancer. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2018. [DOI: 10.5812/ijcm.87024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
841
|
Fusco N, Lopez G, Corti C, Pesenti C, Colapietro P, Ercoli G, Gaudioso G, Faversani A, Gambini D, Michelotti A, Despini L, Blundo C, Vaira V, Miozzo M, Ferrero S, Bosari S. Mismatch Repair Protein Loss as a Prognostic and Predictive Biomarker in Breast Cancers Regardless of Microsatellite Instability. JNCI Cancer Spectr 2018; 2:pky056. [PMID: 31360876 PMCID: PMC6649738 DOI: 10.1093/jncics/pky056] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/20/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Background Breast cancers that harbor mismatch-repair (MMR) deficiency and/or microsatellite instability (MSI) might be sensitive to immune checkpoint blockade, but there are currently no specific guidelines for assessing MMR status in breast cancer. Here, we sought to define the clinical value of MMR immunohistochemistry (IHC) and MSI analysis in breast cancers. Methods We subjected 444 breast cancers to MMR IHC and MSI analysis. Cases were classified as MMR-proficient (pMMR), MMR-deficient (dMMR), and MMR-heterogeneous (hMMR) based on the loss of immunoreactivity; MSI was defined by instability in the five indicators recommended by the National Cancer Institute for endometrial and colorectal cancers. Correlation of MMR status with patients' survival was assessed using the Kaplan-Meier estimator. Statistical tests were two-sided. Results Loss of MMR proteins was homogeneous (dMMR) in 75 patients (17%) and heterogeneous (hMMR) in 55 (12%). Among luminal breast cancers, there were similar frequencies of dMMR and hMMR tumors. Overall, the rate of discrepancy between IHC and MSI analysis was high (91%). Women with Luminal B-like dMMR carcinomas (n = 44) showed shorter overall survival (median = 77 months, range = 0-115 months) than those with pMMR (n = 205) or hMMR (n = 35) tumors (median = 84 months, range = 0-127 months) (P = .008). On the contrary, patients with estrogen receptor-negative breast cancers treated with chemotherapy lived longer in cases of dMMR (n = 9) than pMMR (n = 33) or hMMR (n = 7) tumors, with 87 months of median survival (range = 73-123 months) for the former compared with 79 months (range = 8-113 months) for the latter two categories (P < .001). Conclusions Immunohistochemistry and MSI are not interchangeable tests in breast carcinomas. MMR protein loss is a more common event than MSI and shows intra-tumor heterogeneity. MMR IHC allows the identification of clinically relevant subclasses of breast cancer patients, provided that multiple areas of the tumor are analyzed.
Collapse
Affiliation(s)
- Nicola Fusco
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical, and Dental Sciences
| | - Gianluca Lopez
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Corti
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Pesenti
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology & Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Patrizia Colapietro
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology & Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Giulia Ercoli
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Gaudioso
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Faversani
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical, and Dental Sciences
| | | | - Anna Michelotti
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Division of Medical Oncology (DG, AM)
| | - Luca Despini
- Division of Breast Surgery, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Concetta Blundo
- Division of Breast Surgery, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology & Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Monica Miozzo
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology & Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical, and Dental Sciences
| | - Silvano Bosari
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
842
|
Tsoutsou PG, Zaman K, Martin Lluesma S, Cagnon L, Kandalaft L, Vozenin MC. Emerging Opportunities of Radiotherapy Combined With Immunotherapy in the Era of Breast Cancer Heterogeneity. Front Oncol 2018; 8:609. [PMID: 30619749 PMCID: PMC6305124 DOI: 10.3389/fonc.2018.00609] [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: 05/25/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
The association of radiotherapy and immunotherapy has recently emerged as an exciting combination that might improve outcomes in many solid tumor settings. In the context of breast cancer, this opportunity is promising and under investigation. Given the heterogeneity of breast cancer, it might be meaningful to study the association of radiotherapy and immunotherapy distinctly among the various breast cancer subtypes. The use of biomarkers, such as tumor infiltrating lymphocytes, which are also associated to breast cancer heterogeneity, might provide an opportunity for tailored studies. This review highlights current knowledge of the association of radiotherapy and immunotherapy in the setting of breast cancer and attempts to highlight the therapeutic opportunities among breast cancer heterogeneity.
Collapse
Affiliation(s)
- Pelagia G Tsoutsou
- Division of Oncology, Radio-oncology Department, Vaudois University Hospital Centre (CHUV), Lausanne, Switzerland.,Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland.,Radiation Oncology Department, Hôpital Neuchâtelois, La Chaux-de-Fonds, Switzerland
| | - Khalil Zaman
- Department of Oncology, Breast Center, Vaudois University Hospital Centre (CHUV), Lausanne, Switzerland
| | - Silvia Martin Lluesma
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Laurene Cagnon
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana Kandalaft
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Marie-Catherine Vozenin
- Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland
| |
Collapse
|
843
|
Vikas P, Borcherding N, Zhang W. The clinical promise of immunotherapy in triple-negative breast cancer. Cancer Manag Res 2018; 10:6823-6833. [PMID: 30573992 PMCID: PMC6292225 DOI: 10.2147/cmar.s185176] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease with poorer outcomes compared to other breast cancer subtypes. Contributing to the worse prognosis in TNBC is the higher rates of relapse and rapid progression after relapse. Advances in targeted therapeutics and conventional chemotherapy for TNBC have been stymied due to the lack of specific targets. Moreover, the responses to chemotherapy in TNBC lack durability, partially accounting for the higher rates of relapse. Immunotherapy, notably immune-checkpoint blockade, has shown to improve survival and maintain robust antitumor responses in both hematologic and solid malignancies. Unlike lung cancer, melanoma, and bladder cancer, most breast cancers are not inherently immunogenic and typically have low T cell infiltration. However, among breast cancer subtypes, TNBC is characterized by greater tumor immune infiltrate and higher degree of stromal and intratumoral tumor-infiltrating lymphocytes (TILs), a predictive marker for responses to immunotherapy. Moreover, in TNBC, the high number of stromal TILs is predictive of more favorable survival outcomes and response to chemotherapy. Immunotherapy is being extensively explored in TNBC and clinical trials are showing some promising results. This article focuses on the rationale for immunotherapy in TNBC, to explore and discuss preclinical data, results from early clinical trials, and to summarize some ongoing trials. We will also discuss the potential application of immunotherapy in TNBC from a clinician's perspective.
Collapse
Affiliation(s)
- Praveen Vikas
- Department of Internal Medicine, College of Medicine, University of Iowa, Iowa City, IA, USA,
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA,
| | - Nicholas Borcherding
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA,
- Department of Pathology, College of Medicine, University of Iowa, Iowa City, IA, USA
- Cancer Biology Graduate Program, College of Medicine, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Weizhou Zhang
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA,
- Department of Pathology, College of Medicine, University of Iowa, Iowa City, IA, USA
- Cancer Biology Graduate Program, College of Medicine, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
844
|
Altan M, Kidwell KM, Pelekanou V, Carvajal-Hausdorf DE, Schalper KA, Toki MI, Thomas DG, Sabel MS, Hayes DF, Rimm DL. Association of B7-H4, PD-L1, and tumor infiltrating lymphocytes with outcomes in breast cancer. NPJ Breast Cancer 2018; 4:40. [PMID: 30564631 PMCID: PMC6288133 DOI: 10.1038/s41523-018-0095-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023] Open
Abstract
B7-H4 (VTCN1) is a member of the CD28/B7 family of immune co-inhibitory molecules. The relationship of tumor and stromal B7-H4 protein expression with PD-L1, tumor infiltrating lymphocytes (TILs) and its association with clinico-pathological variables are not well defined. Herein, we explore the expression level of B7-H4 protein in breast cancer and evaluate its association with TILs, levels of PD-L1 expression, and clinico-pathological characteristics in two independent populations. In this study, we used multiplexed automated quantitative immunofluorescence (QIF) to measure the levels of B7-H4 and PD-L1 protein and determined TILs through pathologist assessment of H&E-stained preparations in over a thousand breast cancer cases from two institutions represented in tissue microarray format. Associations between the marker levels, major clinico-pathological variables, and survival were analyzed. We detected B7-H4 protein was highly expressed in both breast cancer and stromal cells. Its expression was independent of breast cancer intrinsic subtypes. PD-L1 expression was higher in triple negative breast cancers. Neither B7-H4 nor PD-L1 were associated with survival in breast cancer. Our study shows there is a mutually exclusive pattern of B7-H4 with both tumor PD-L1 expression and TILs in all breast cancers, independent of breast cancer intrinsic subtype. This exclusive pattern suggests that some breast tumors may preferentially use one B7-related immune evasion mechanism/pathway. This could explain the clinical benefit that is seen only in a fraction of patients with immune checkpoint inhibitors directed exclusively towards PD-L1 in breast cancer.
Collapse
Affiliation(s)
- Mehmet Altan
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Kelley M. Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI USA
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | | | - Daniel E. Carvajal-Hausdorf
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
- Anatomic Pathology, Clinica Alemana-Facultad de Medicina Universidad de Desarrollo, Vitacura, Santiago Chile
| | - Kurt A. Schalper
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Maria I. Toki
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Dafydd G. Thomas
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Michael S. Sabel
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Daniel F. Hayes
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - David L. Rimm
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
845
|
Talhouk A, Derocher H, Schmidt P, Leung S, Milne K, Gilks CB, Anglesio MS, Nelson BH, McAlpine JN. Molecular Subtype Not Immune Response Drives Outcomes in Endometrial Carcinoma. Clin Cancer Res 2018; 25:2537-2548. [DOI: 10.1158/1078-0432.ccr-18-3241] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 11/16/2022]
|
846
|
Subgrouping breast cancer patients based on immune evasion mechanisms unravels a high involvement of transforming growth factor-beta and decoy receptor 3. PLoS One 2018; 13:e0207799. [PMID: 30513096 PMCID: PMC6279052 DOI: 10.1371/journal.pone.0207799] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/06/2018] [Indexed: 11/24/2022] Open
Abstract
In the era of immunotherapy and personalized medicine, there is an urgent need for advancing the knowledge of immune evasion in different cancer types and identifying reliable biomarkers that guide both therapy selection and patient inclusion in clinical trials. Given the differential immune responses and evasion mechanisms in breast cancer, we expect to identify different breast cancer groups based on their expression of immune-related genes. For that, we used the sequential biclustering method on The Cancer Genome Atlas RNA-seq breast cancer data and identified 7 clusters. We found that 77.4% of the clustered tumor specimens evade through transforming growth factor-beta (TGF-β) immunosuppression, 57.7% through decoy receptor 3 (DcR3) counterattack, 48.0% through cytotoxic T-lymphocyte-associated protein 4 (CTLA4), and 34.3% through programmed cell death-1 (PD-1). TGF-β and DcR3 are potential novel drug targets for breast cancer immunotherapy. Targeting TGF-β and DcR3 may provide a powerful approach for treating breast cancer because 57.7% of patients overexpressed these two molecules. Furthermore, triple-negative breast cancer (TNBC) patients clustered equally into two subgroups: one with impaired antigen presentation and another with high leukocyte recruitment but four different evasion mechanisms. Thus, different TNBC patients may be treated with different immunotherapy approaches. We identified biomarkers to cluster patients into subgroups based on immune evasion mechanisms and guide the choice of immunotherapy. These findings provide a better understanding of patients’ response to immunotherapies and shed light on the rational design of novel combination therapies.
Collapse
|
847
|
Abstract
Immunotherapy through immune checkpoint blockers (ICBs) is quickly transforming cancer treatment by improving patients' outcomes. However, innate and acquired resistance to ICBs remain a major challenge in clinical settings. Indoleamine 2,3-dioxygenases (IDOs) are enzymes involved in tryptophan catabolism with a central immunosuppressive function within the tumor microenvironment. IDOs are over-expressed in cancer patients and have increasingly been associated with worse outcomes and a poor prognosis. Preclinical data have shown that combining IDO and checkpoint inhibition might be a valuable strategy to improve the efficacy of immunotherapy. Currently, several IDO inhibitors have been evaluated in clinical trials, showing favorable pharmacokinetic profiles and promising efficacy. This review describes the mechanisms involved in IDO-mediated immune suppression and its role in cancer immune escape, focusing on the potential clinical application of IDO inhibitors as an immunotherapy strategy for cancer treatment.
Collapse
|
848
|
Hinchcliff E, Hong D, Le H, Chisholm G, Iyer R, Naing A, Hwu P, Jazaeri A. Characteristics and outcomes of patients with recurrent ovarian cancer undergoing early phase immune checkpoint inhibitor clinical trials. Gynecol Oncol 2018; 151:407-413. [PMID: 30366646 PMCID: PMC6281778 DOI: 10.1016/j.ygyno.2018.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To describe the clinical outcomes associated with the use of checkpoint inhibitor therapy in recurrent ovarian malignancy. METHODS Women with recurrent ovarian cancer treated with an immune checkpoint inhibitor between 1/2012 and 8/2017 were included. RECIST criteria determined disease status, and immune related adverse events (irAE) were graded per trial protocols. Predictors of response, irAE, progression free survival (PFS) and overall survival (OS) were investigated. RESULTS Forty-four women were included with a median age of 53 years, median of 4 prior lines of chemotherapy, and most commonly high grade serous pathology (59.1%). 3 patients had partial response and 3 had pseudoprogression, for a response rate of 14.2%. In subset analysis of high grade serous (HGSOC) pathology, platinum sensitivity at time of checkpoint inhibitor therapy was correlated with response (p = 0.01). There were 28 grade 3/4 irAEs in 21 patients (47.7%). Combination therapy rather than monotherapy predicted irAE (OR 5.7, CI 1.6-20.9, p = 0.02). The most common severe irAE was elevation in hepatic or pancreatic enzymes in 12 total patients (13.6% each). Interestingly, the number of genes mutated was protective from hepatic/pancreatic AE (p = 0.02). CONCLUSIONS While response rate was similar to prior literature, in patients with HGSOC platinum sensitivity at time of checkpoint inhibitor initiation was correlated to response. Grade 3/4 hepatic and pancreatic enzyme elevations were more common in ovarian cancer patients than has been previously reported in other tumor types. The number of genes mutated was inversely correlated to risk of this type of irAEs but not to total irAEs.
Collapse
Affiliation(s)
- Emily Hinchcliff
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.
| | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Hung Le
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Gary Chisholm
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Revathy Iyer
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
849
|
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
|
850
|
Shi Y, Yang F, Huang D, Guan X. Androgen blockade based clinical trials landscape in triple negative breast cancer. Biochim Biophys Acta Rev Cancer 2018; 1870:283-290. [DOI: 10.1016/j.bbcan.2018.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 01/12/2023]
|