1101
|
Safety and immunogenicity of neoadjuvant treatment using WT1-immunotherapeutic in combination with standard therapy in patients with WT1-positive Stage II/III breast cancer: a randomized Phase I study. Breast Cancer Res Treat 2017; 162:479-488. [PMID: 28176175 PMCID: PMC5332485 DOI: 10.1007/s10549-017-4130-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 12/23/2022]
Abstract
Purpose This Phase I, multicenter, randomized study (ClinicalTrials.gov NCT01220128) evaluated the safety and immunogenicity of recombinant Wilms’ tumor 1 (WT1) protein combined with the immunostimulant AS15 (WT1-immunotherapeutic) as neoadjuvant therapy administered concurrently with standard treatments in WT1-positive breast cancer patients. Methods Patients were treated in 4 cohorts according to neoadjuvant treatment (A: post-menopausal, hormone receptor [HR]-positive patients receiving aromatase inhibitors; B: patients receiving chemotherapy; C: HER2-overexpressing patients on trastuzumab–chemotherapy combination; D: HR-positive/HER2-negative patients on chemotherapy). Patients (cohorts A–C) were randomized (2:1) to receive 6 or 8 doses of WT1-immunotherapeutic or placebo together with standard neoadjuvant treatment in a double-blind manner; cohort D patients received WT1-immunotherapeutic in an open manner. Safety was assessed throughout the study. WT1-specific antibodies were assessed pre- and post-vaccination. Results Sixty-two patients were randomized; 60 received ≥ one dose of WT1-immunotherapeutic. Two severe toxicities were reported: diarrhea (cohort C; also reported as a grade 3 serious adverse event) and decreased left ventricular ejection fraction (cohort B; also reported as a grade 2 adverse event). Post-dose 4 of WT1-immunotherapeutic, 10/10 patients from cohort A, 0/8 patients from cohort B, 6/11 patients from cohort C, and 2/3 patients from cohort D were humoral responders. The sponsor elected to close the trial prematurely. Conclusions Concurrent administration of WT1-immunotherapeutic and standard neoadjuvant therapy was well tolerated and induced WT1-specific antibodies in patients receiving neoadjuvant aromatase inhibitors. In patients on neoadjuvant chemotherapy or trastuzumab–chemotherapy combination, the humoral response was impaired or blunted, likely due to either co-administration of corticosteroids and/or the chemotherapies themselves. Electronic supplementary material The online version of this article (doi:10.1007/s10549-017-4130-y) contains supplementary material, which is available to authorized users.
Collapse
|
1102
|
Ladoire S, Derangère V, Arnould L, Thibaudin M, Coudert B, Lorgis V, Desmoulins I, Chaix M, Fumoleau P, Ghiringhelli F. [The anti-tumor immune response in breast cancer: Update and therapeutic perspectives]. Ann Pathol 2017; 37:133-141. [PMID: 28159406 DOI: 10.1016/j.annpat.2016.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 11/18/2022]
Abstract
The role of the immune response in breast cancer is now well recognized and increasingly taken in account. The goal of this article is, in the first part, to underline its prognostic impact and to precise the immunosurvelliance, immunoselection and the immunosubversion concepts involved in the control and evasion of breast carcinoma. In the second part, therapeutic strategies for the restauration of anti-tumor immunity are developed. Vaccination strategies and checkpoints inhibitors blockade strategies are discussed as well as the immunogenic death linked to the conventional treatments of breast cancer.
Collapse
Affiliation(s)
- Sylvain Ladoire
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France; Plateforme de transfert en biologie cancérologique, centre Georges-François-Leclerc, 21000 Dijon, France; UMR Inserm U866, faculté de médecine de Dijon, 21000 Dijon, France; UFR des sciences de santé, 21000 Dijon, France; Université de Bourgogne, 21000 Dijon, France.
| | - Valentin Derangère
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France; Plateforme de transfert en biologie cancérologique, centre Georges-François-Leclerc, 21000 Dijon, France; UMR Inserm U866, faculté de médecine de Dijon, 21000 Dijon, France; Université de Bourgogne, 21000 Dijon, France; Département de pathologie et de biologie des tumeurs, centre Georges-François-Leclerc, 21000 Dijon, France
| | - Laurent Arnould
- Plateforme de transfert en biologie cancérologique, centre Georges-François-Leclerc, 21000 Dijon, France; Université de Bourgogne, 21000 Dijon, France; Département de pathologie et de biologie des tumeurs, centre Georges-François-Leclerc, 21000 Dijon, France
| | - Marion Thibaudin
- UMR Inserm U866, faculté de médecine de Dijon, 21000 Dijon, France
| | - Bruno Coudert
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France
| | - Veronique Lorgis
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France
| | - Isabelle Desmoulins
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France
| | - Marie Chaix
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France; UMR Inserm U866, faculté de médecine de Dijon, 21000 Dijon, France; UFR des sciences de santé, 21000 Dijon, France; Université de Bourgogne, 21000 Dijon, France
| | - Pierre Fumoleau
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France; Plateforme de transfert en biologie cancérologique, centre Georges-François-Leclerc, 21000 Dijon, France; UFR des sciences de santé, 21000 Dijon, France; Université de Bourgogne, 21000 Dijon, France
| | - François Ghiringhelli
- Département d'oncologie médicale, centre Georges-François-Leclerc, 21000 Dijon, France; Plateforme de transfert en biologie cancérologique, centre Georges-François-Leclerc, 21000 Dijon, France; UMR Inserm U866, faculté de médecine de Dijon, 21000 Dijon, France; UFR des sciences de santé, 21000 Dijon, France; Université de Bourgogne, 21000 Dijon, France
| |
Collapse
|
1103
|
Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, Chae YK, Cristofanilli M, Gradishar WJ, Giles FJ. Targeting Epidermal Growth Factor Receptor in triple negative breast cancer: New discoveries and practical insights for drug development. Cancer Treat Rev 2017; 53:111-119. [DOI: 10.1016/j.ctrv.2016.12.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 12/23/2016] [Accepted: 12/24/2016] [Indexed: 12/25/2022]
|
1104
|
Ross JS, Gay LM. Comprehensive genomic sequencing and the molecular profiles of clinically advanced breast cancer. Pathology 2017; 49:120-132. [DOI: 10.1016/j.pathol.2016.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 02/06/2023]
|
1105
|
Kelly CM, Komanyane L, Andreopoulou E. Should bone-targeted agents be standard of care in postmenopausal patients with early breast cancer? BREAST CANCER MANAGEMENT 2017. [DOI: 10.2217/bmt-2016-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of bone-targeted agents in the adjuvant treatment of early breast cancer has been debated for decades. A recent meta-analysis of individual patient data from 18,766 women provides compelling evidence supporting the use of these agents in postmenopausal women with early-stage breast cancer. Postmenopausal women who received a bisphosphonate had a significant reduction in breast cancer mortality (14.7 vs 18%; p = 0.002) and in overall survival (21.1 vs 23.5%; p = 0.005). Some questions remain such as; what bone-targeted agent is the best? What are the optimum doses, durations and schedules? Which patients benefit the most? The decision to include a bone-targeted agent should consider the absolute risk of distant recurrence and breast cancer mortality.
Collapse
Affiliation(s)
- Catherine M Kelly
- UCD School of Medicine, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Lore Komanyane
- UCD School of Medicine, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Eleni Andreopoulou
- Division of Breast Medical Oncology, Weill Cornell Medicine/New York Presbyterian Hospital, New York, NY 10128, USA
| |
Collapse
|
1106
|
Plimack ER, Bellmunt J, Gupta S, Berger R, Chow LQM, Juco J, Lunceford J, Saraf S, Perini RF, O'Donnell PH. Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE-012): a non-randomised, open-label, phase 1b study. Lancet Oncol 2017; 18:212-220. [PMID: 28081914 DOI: 10.1016/s1470-2045(17)30007-4] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/15/2016] [Accepted: 10/19/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND PD-1 and its ligands are expressed in urothelial cancer, and findings have shown that inhibition of the PD-1 pathway has clinical benefit. We aimed to assess the safety and activity of an anti-PD-1 antibody pembrolizumab in patients with locally advanced or metastatic urothelial cancer. METHODS This study was part of the non-randomised, multi-cohort, open-label, phase 1b KEYNOTE-012 basket trial. We enrolled patients aged 18 years and older with a histologically or cytologically confirmed diagnosis of locally advanced or metastatic urothelial cancer, including cancers of the renal pelvis, ureter, bladder, or urethra, from eight hospitals in the USA and Israel. Patients were required to have at least 1% PD-L1 expression detected on the tumour cells or in tumour stroma, as determined by immunohistochemistry. Patients were given 10 mg/kg intravenous pembrolizumab every 2 weeks until disease progression, unacceptable toxic effects, or the end of the study (ie, 24 months of treatment). Primary endpoints were safety and overall response (defined by Response Evaluation Criteria In Solid Tumors [RECIST] version 1.1), as assessed by a masked, independent central review. Safety was assessed in patients who received one or more doses of pembrolizumab (all-patients-as-treated population); activity was assessed in patients who received pembrolizumab, had measurable disease at baseline, and had one or more post-baseline scans, or discontinued because of progressive disease or treatment-related adverse events (full analysis set). This study is registered with ClinicalTrials.gov, number NCT01848834, and is no longer enrolling patients; follow-up is ongoing. FINDINGS Between May 14, 2013, and Dec 10, 2013, 115 patients were tissue pre-screened as part of a two-part consent process. 61 (53%) patients were PD-L1 positive, of whom 33 were enrolled in this study. All enrolled patients received at least one dose of pembrolizumab and were included in the safety analyses. 27 patients comprised the full analysis set and were deemed assessable for activity. Six patients were not assessable: three discontinued study drug because of a non-treatment-related adverse event before the first post-baseline scan, two withdrew before the first post-baseline scan, and one had no measurable disease at baseline. The most common treatment-related adverse events were fatigue (six [18%] of 33 patients) and peripheral oedema (4 [12%]). Five (15%) patients had 11 grade 3 treatment-related adverse events; no single event occurred in more than one patient. Three (9%) patients experienced five serious treatment-related adverse events. After median follow-up of 13 months (range 1-26, IQR 5-23), an overall response was achieved in seven (26% [95% CI 11-46]) of 27 assessable patients, with three (11% [2-29]) complete and four (15% [4-34]) partial responses. Of the four deaths that occurred during the study (cardiac arrest, pneumonia, sepsis, and subarachnoid haemorrhage), none were deemed treatment related. INTERPRETATION Pembrolizumab showed anti-tumour activity and acceptable safety in patients with advanced urothelial cancer, supporting ongoing phase 2 and 3 studies of pembrolizumab in this population. FUNDING Merck & Co., Inc.
Collapse
Affiliation(s)
| | | | | | - Raanan Berger
- Chaim Sheba Medical Center at Tel Hashomer, Tel-Hashomer, Ramat-Gan, Israel
| | | | | | | | | | | | | |
Collapse
|
1107
|
Catakovic K, Klieser E, Neureiter D, Geisberger R. T cell exhaustion: from pathophysiological basics to tumor immunotherapy. Cell Commun Signal 2017; 15:1. [PMID: 28073373 PMCID: PMC5225559 DOI: 10.1186/s12964-016-0160-z] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
The immune system is capable of distinguishing between danger- and non-danger signals, thus inducing either an appropriate immune response against pathogens and cancer or inducing self-tolerance to avoid autoimmunity and immunopathology. One of the mechanisms that have evolved to prevent destruction by the immune system, is to functionally silence effector T cells, termed T cell exhaustion, which is also exploited by viruses and cancers for immune escape In this review, we discuss some of the phenotypic markers associated with T cell exhaustion and we summarize current strategies to reinvigorate exhausted T cells by blocking these surface marker using monoclonal antibodies.
Collapse
Affiliation(s)
- Kemal Catakovic
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Müllner Hauptstrasse 48, Salzburg, 5020, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Eckhard Klieser
- Salzburg Cancer Research Institute, Salzburg, Austria.,Department of Pathology, Paracelsus Medical University, Müllner Hauptstrasse 48, Salzburg, 5020, Austria
| | - Daniel Neureiter
- Salzburg Cancer Research Institute, Salzburg, Austria.,Department of Pathology, Paracelsus Medical University, Müllner Hauptstrasse 48, Salzburg, 5020, Austria
| | - Roland Geisberger
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Müllner Hauptstrasse 48, Salzburg, 5020, Austria. .,Salzburg Cancer Research Institute, Salzburg, Austria.
| |
Collapse
|
1108
|
Molecular Biomarkers for Prediction of Targeted Therapy Response in Metastatic Breast Cancer: Trick or Treat? Int J Mol Sci 2017; 18:ijms18010085. [PMID: 28054957 PMCID: PMC5297719 DOI: 10.3390/ijms18010085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/23/2016] [Accepted: 12/26/2016] [Indexed: 01/01/2023] Open
Abstract
In recent years, the study of genomic alterations and protein expression involved in the pathways of breast cancer carcinogenesis has provided an increasing number of targets for drugs development in the setting of metastatic breast cancer (i.e., trastuzumab, everolimus, palbociclib, etc.) significantly improving the prognosis of this disease. These drugs target specific molecular abnormalities that confer a survival advantage to cancer cells. On these bases, emerging evidence from clinical trials provided increasing proof that the genetic landscape of any tumor may dictate its sensitivity or resistance profile to specific agents and some studies have already showed that tumors treated with therapies matched with their molecular alterations obtain higher objective response rates and longer survival. Predictive molecular biomarkers may optimize the selection of effective therapies, thus reducing treatment costs and side effects. This review offers an overview of the main molecular pathways involved in breast carcinogenesis, the targeted therapies developed to inhibit these pathways, the principal mechanisms of resistance and, finally, the molecular biomarkers that, to date, are demonstrated in clinical trials to predict response/resistance to targeted treatments in metastatic breast cancer.
Collapse
|
1109
|
Tumor Associated Macrophages as Therapeutic Targets for Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:331-370. [PMID: 29282692 DOI: 10.1007/978-981-10-6020-5_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor-associated macrophages (TAMs) are the most abundant inflammatory infiltrates in the tumor stroma. TAMs promote tumor growth by suppressing immunocompetent cells, including neovascularization and supporting cancer stem cells. In the chapter, we discuss recent efforts in reprogramming or inhibiting tumor-protecting properties of TAMs, and developing potential strategies to increase the efficacy of breast cancer treatment.
Collapse
|
1110
|
Bu X, Yao Y, Li X. Immune Checkpoint Blockade in Breast Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:383-402. [PMID: 29282694 DOI: 10.1007/978-981-10-6020-5_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is emerging as the most promising novel strategy for cancer treatment. Cancer immunotherapy is broadly categorized into three forms: immune checkpoint modulation, adoptive cell transfer, and cancer vaccine. Immune checkpoint blockade is demonstrated as the most clinically effective treatment with low immune-related adverse events (irAE). Blockade of PD-1/PD-L1 and CTLA-4 has achieved remarkable success in treating various types of tumors, which sparks great interests in this therapeutic strategy and expands the role of immune checkpoint blockade in treating tumors including breast cancer. Based on the notable results obtained from clinical trials, the United States' Food and Drug Administration (FDA) has approved multiple CTLA-4 monoclonal antibodies as well as the PD-1/PD-L1 monoclonal antibodies for treatment of different types of tumors. The theories of immunoediting, T-cell exhaustions, and co-stimulatory/co-inhibitory pathways are immunological foundations for immune checkpoint blockade therapy. Breast cancers such as triple negative breast cancer and HER-2 negative breast cancer respond to immune checkpoint blockade therapy due to their high immunogenicity. PD-1/PD-L1 blockade has just received FDA approval as a standard cancer therapy for solid tumors such as breast cancer. Development of immune checkpoint blockade focuses on two directions: one is to identify proper biomarkers of immune checkpoint blockade in breast cancer, and the other is to combine therapies with PD-1/PD-L1 blockade antibodies to achieve optimal clinical outcomes.
Collapse
Affiliation(s)
- Xia Bu
- Department of Medical Oncology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China.
| | - Yihui Yao
- Department of Medical Oncology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China
| | - Xiaoyu Li
- Department of Hematology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China
| |
Collapse
|
1111
|
Costa R, Carneiro B, Wainwright D, Santa-Maria C, Kumthekar P, Chae Y, Gradishar W, Cristofanilli M, Giles F. Developmental therapeutics for patients with breast cancer and central nervous system metastasis: current landscape and future perspectives. Ann Oncol 2017; 28:44-56. [PMID: 28177431 PMCID: PMC7360139 DOI: 10.1093/annonc/mdw532] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is the second-leading cause of metastatic disease in the central nervous system (CNS). Recent advances in the biological understanding of breast cancer have facilitated an unprecedented increase of survival in a subset of patients presenting with metastatic breast cancer. Patients with HER2 positive (HER2+) or triple negative breast cancer are at highest risk of developing CNS metastasis, and typically experience a poor prognosis despite treatment with local and systemic therapies. Among the obstacles ahead in the realm of developmental therapeutics for breast cancer CNS metastasis is the improvement of our knowledge on its biological nuances and on the interaction of the blood–brain barrier with new compounds. This article reviews recent discoveries related to the underlying biology of breast cancer brain metastases, clinical progress to date and suggests rational approaches for investigational therapies.
Collapse
Affiliation(s)
- R. Costa
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| | - B.A. Carneiro
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| | - D.A. Wainwright
- Department of Pathology
- Department of Neurology
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - C.A. Santa-Maria
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| | | | - Y.K. Chae
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| | - W.J. Gradishar
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| | - M. Cristofanilli
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| | - F.J. Giles
- Developmental Therapeutics Program, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago
| |
Collapse
|
1112
|
Varn FS, Mullins DW, Arias‐Pulido H, Fiering S, Cheng C. Adaptive immunity programmes in breast cancer. Immunology 2017; 150:25-34. [PMID: 27564847 PMCID: PMC5341497 DOI: 10.1111/imm.12664] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/12/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022] Open
Abstract
The role of the immune system in shaping cancer development and patient prognosis has recently become an area of intense focus in industry and academia. Harnessing the adaptive arm of the immune system for tumour eradication has shown great promise in a variety of tumour types. Differences between tissues, however, necessitate a greater understanding of the adaptive immunity programmes that are active within each tumour type. In breast cancer, adaptive immune programmes play diverse roles depending on the cellular infiltration found in each tumour. Cytotoxic T lymphocytes and T helper type 1 cells can induce tumour eradication, whereas regulatory T cells and T helper type 2 cells are known to be involved in tumour-promoting immunosuppressive responses. Complicating these matters, heterogeneous expression of hormone receptors and growth factors in different tumours leads to disparate, patient-specific adaptive immune responses. Despite this non-conformity in adaptive immune behaviours, encouraging basic and clinical results have been observed that suggest a role for immunotherapeutic approaches in breast cancer. Here, we review the literature pertaining to the adaptive immune response in breast cancer, summarize the primary findings relating to the breast tumour's biology, and discuss potential clinical immunotherapies.
Collapse
Affiliation(s)
- Frederick S. Varn
- Department of Molecular and Systems BiologyGeisel School of Medicine at DartmouthHanoverNHUSA
| | - David W. Mullins
- Department of Medical EducationGeisel School of Medicine at DartmouthHanoverNHUSA
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthLebanonNHUSA
- Norris Cotton Cancer CenterLebanonNHUSA
| | - Hugo Arias‐Pulido
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthLebanonNHUSA
| | - Steven Fiering
- Department of Molecular and Systems BiologyGeisel School of Medicine at DartmouthHanoverNHUSA
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthLebanonNHUSA
- Norris Cotton Cancer CenterLebanonNHUSA
| | - Chao Cheng
- Department of Molecular and Systems BiologyGeisel School of Medicine at DartmouthHanoverNHUSA
- Norris Cotton Cancer CenterLebanonNHUSA
- Department of Biomedical Data ScienceGeisel School of Medicine at DartmouthLebanonNHUSA
| |
Collapse
|
1113
|
Tumour-infiltrating lymphocytes and the emerging role of immunotherapy in breast cancer. Pathology 2016; 49:141-155. [PMID: 28049579 DOI: 10.1016/j.pathol.2016.10.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/31/2016] [Accepted: 11/02/2016] [Indexed: 12/23/2022]
Abstract
Breast cancer has not previously been considered a highly immunogenic cancer. Observations of tumour-infiltrating lymphocytes (TILs) in and around neoplastic cells in tumour samples, and associations with improved pathological complete response and clinical survival end points have changed our perspective on this. Lymphocytic infiltrates have long been observed in breast cancer; however, more recently, retrospective analysis of prospectively collected tissue samples from clinical trials has demonstrated the potential role of host immunosurveillance in influencing the biology of breast cancer. This association appears to be strongest in triple negative and HER2 positive breast cancer subtypes. Contrastingly, the association in luminal tumours is less clear, and is potentially limited by substantial tumoural heterogeneity. Several methodologies have been employed to quantify, and describe the composition of TILs, each with its own advantages and disadvantages. The results of these analyses have been generally consistent, and valuable efforts are currently underway to standardise the evaluation of TILs toward a universal approach. More technical methods of TIL characterisation remain important in the research setting. The evaluation of TILs becomes increasingly relevant with the emerging role of immunotherapy in breast cancer. Early phase trials of checkpoint blockade show promising results; however, it is likely that some patients will require combination treatments to maximise therapeutic benefits. Equally, some patients may not derive any benefit from immunotherapies. This underscores the importance of the development of relevant predictive biomarkers. As a key representative of the immune interaction between host and tumour, lymphocytic infiltrates are ideally placed for continued research into the determinants of immunogenicity, and response to immunotherapeutic approaches. In this review, we will discuss the current methodologies of evaluation, and the clinical relevance of TILs. Additionally, we discuss the emerging role of immunotherapy in breast cancer, and the future of TIL characterisation in this context.
Collapse
|
1114
|
Radosevic-Robin N, Béguinot M, Penault-Llorca F. [Evaluation of the immune infiltrate in breast cancer]. Bull Cancer 2016; 104:52-68. [PMID: 27986267 DOI: 10.1016/j.bulcan.2016.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/10/2016] [Indexed: 01/21/2023]
Abstract
Tumour-infiltrating lymphocytes (TIL) are major components of the immune/"inflammatory" infiltrate found in tumour microenvironment. They reflect the intensity and the quality of the immune reaction to cancer. In breast cancer, TIL density and phenotypic profile have been demonstrated to be predictive of response to neoadjuvant treatment and of patient outcome. TIL density, currently the best-developed TIL-related biomarker, is defined as the percentage of tumour stroma surface occupied by TIL. The baseline TIL density of 50% and higher is associated with particularly high rates of pathological complete response to neoadjuvant therapy in triple negative and HER2+ breast cancer, as well as with significantly better recurrence-free and overall survival. Similar predictive and prognostic value has been demonstrated for the ratio between the numbers of CD8+ and FoxP3+ TIL. TIL density and the CD8+/FoxP3+ ratio are promising biomarkers in breast cancer, which could be used in tailoring of neoadjuvant and adjuvant systemic therapy and in selection of patients for different immunotherapy modalities. This article reviews elements of the immune response to cancer, methods of TIL analysis, evidence of TIL' prognostic and predictive value in the current breast cancer management as well as the perspectives for use of TIL' characteristics as biomarkers in breast cancer immunotherapy.
Collapse
Affiliation(s)
- Nina Radosevic-Robin
- Centre Jean Perrin, département d'anatomie et de cytologie pathologiques, 58, rue Montalembert, 63011 Clermont-Ferrand, France; Inserm U1240 « Imagerie moléculaire et stratégies théranostiques (IMoST) », 58, rue Montalembert, 63011 Clermont-Ferrand, France.
| | - Marie Béguinot
- Centre Jean-Perrin, département de chirurgie oncologique, 58, rue Montalembert, 63011 Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Centre Jean Perrin, département d'anatomie et de cytologie pathologiques, 58, rue Montalembert, 63011 Clermont-Ferrand, France; Inserm U1240 « Imagerie moléculaire et stratégies théranostiques (IMoST) », 58, rue Montalembert, 63011 Clermont-Ferrand, France; Université d'Auvergne, faculté de médecine, 28, place Henri-Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
1115
|
Buisseret L, Garaud S, de Wind A, Van den Eynden G, Boisson A, Solinas C, Gu-Trantien C, Naveaux C, Lodewyckx JN, Duvillier H, Craciun L, Veys I, Larsimont D, Piccart-Gebhart M, Stagg J, Sotiriou C, Willard-Gallo K. Tumor-infiltrating lymphocyte composition, organization and PD-1/ PD-L1 expression are linked in breast cancer. Oncoimmunology 2016; 6:e1257452. [PMID: 28197375 PMCID: PMC5283629 DOI: 10.1080/2162402x.2016.1257452] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/30/2016] [Accepted: 11/01/2016] [Indexed: 12/13/2022] Open
Abstract
The clinical relevance of tumor-infiltrating lymphocytes (TIL) in breast cancer (BC) has been clearly established by their demonstrated correlation with long-term positive outcomes. Nevertheless, the relationship between protective immunity, observed in some patients, and critical features of the infiltrate remains unresolved. This study examined TIL density, composition and organization together with PD-1 and PD-L1 expression in freshly collected and paraffin-embedded tissues from 125 patients with invasive primary BC. Tumor and normal breast tissues were analyzed using both flow cytometry and immunohistochemistry. TIL density distribution is a continuum with 25% of tumors identified as TIL-negative at a TIL density equivalent to normal breast tissues. TIL-positive tumors (75%) were equally divided into TIL-intermediate and TIL-high. Tumors had higher mean frequencies of CD4+ T cells and CD19+ B cells and a lower mean frequency of CD8+ T cells compare with normal tissues, increasing the CD4+/CD8+ T-cell ratio. Tertiary lymphoid structures (TLS), principally located in the peri-tumoral stroma, were detected in 60% of tumors and correlated with higher TIL infiltration. PD-1 and PD-L1 expression were also associated with higher TIL densities and TLS. TIL density, TLS and PD-L1 expression were correlated with more aggressive tumor characteristics, including higher proliferation and hormone receptor negativity. Our findings reveal an important relationship between PD-1/PD-L1 expression, increased CD4+ T and B-cell infiltration, TIL density and TLS, suggesting that evaluating not only the extent but also the nature and location of the immune infiltrate should be considered when evaluating antitumor immunity and the potential for benefit from immunotherapies.
Collapse
Affiliation(s)
- Laurence Buisseret
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium; Breast Cancer Translational Research Laboratory J-C Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | | | - Gert Van den Eynden
- Department of Pathology, GZA Ziekenhuizen, Sint-Augustinus campus , Wilrijk, Belgium
| | - Anais Boisson
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | - Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | - Chunyan Gu-Trantien
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | - Céline Naveaux
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | - Jean-Nicolas Lodewyckx
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | - Hugues Duvillier
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium; Flow Cytometry Facility, Institut Jules Bordet, Brussels, Belgium
| | - Ligia Craciun
- Department of Pathology, Institut Jules Bordet , Brussels, Belgium
| | - Isabelle Veys
- Department of Surgery, Institut Jules Bordet , Brussels, Belgium
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet , Brussels, Belgium
| | | | - John Stagg
- Université de Montréal, Centre de Recherche du CHUM , Montreal, Quebec, Canada
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory J-C Heuson, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles , Brussels, Belgium
| |
Collapse
|
1116
|
Gombos A, Awada A. Advances in chemical pharmacotherapy to manage advanced breast cancer. Expert Opin Pharmacother 2016; 18:95-103. [PMID: 27927046 DOI: 10.1080/14656566.2016.1269748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Advanced breast cancer is still incurable. However, patients diagnosed with this fatal disease live longer. The selection of systemic therapy is mainly based on molecular subtype. The aim of management in these patients is to not only improve outcome, but also to maintain quality of life. Areas covered: In this paper we focus on available treatments and drugs under late development in the three main subtypes of breast cancer: luminal (hormone receptor positive), HER2 positive and triple negative disease. Main advances during the last years have been made in the treatment of HER2 positive breast cancer with the approval of several new targeted agents. Luminal breast cancer is also a field of active clinical research. So far triple negative breast cancer remains the subtype with the worse prognosis, even though new discoveries have been made to better understand the huge heterogeneity of this type of breast cancer. Expert opinion: Several new treatment options have recently been established in metastatic breast cancer. Side effects are sometimes cumbersome for the patient and are difficult to manage easily. Thus, identification of patients who derive the most benefit is needed. In addition, collaborative efforts should integrate the genotypic fragmentation in the management and future clinical research strategies of metastatic breast cancer patients.
Collapse
Affiliation(s)
- Andrea Gombos
- a Medical Oncology Clinic, Institut Jules Bordet , Université Libre de Bruxelles , Brussels , Belgium
| | - Ahmad Awada
- a Medical Oncology Clinic, Institut Jules Bordet , Université Libre de Bruxelles , Brussels , Belgium
| |
Collapse
|
1117
|
Hartkopf AD, Taran FA, Wallwiener M, Walter CB, Krämer B, Grischke EM, Brucker SY. PD-1 and PD-L1 Immune Checkpoint Blockade to Treat Breast Cancer. Breast Care (Basel) 2016; 11:385-390. [PMID: 28228704 DOI: 10.1159/000453569] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibition represents a major recent breakthrough in the treatment of malignant diseases including breast cancer. Blocking the programmed death receptor-1 (PD-1) and its ligand, PD-L1, has shown impressive antitumor activity and may lead to durable long-term disease control, especially in the triple-negative subtypes of breast cancer (TNBC). Although immune checkpoint blockade is generally well tolerated, specific immune-related adverse events (irAEs) may occur. This review summarizes the clinical efficacy, perspectives, and future challenges of using PD-1/PD-L1-directed antibodies in the treatment of breast cancer.
Collapse
Affiliation(s)
- Andreas D Hartkopf
- Department of Women's Health, University of Tübingen, Tübingen, Germany, Heidelberg, Germany
| | - Florin-Andrei Taran
- Department of Women's Health, University of Tübingen, Tübingen, Germany, Heidelberg, Germany
| | - Markus Wallwiener
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Christina B Walter
- Department of Women's Health, University of Tübingen, Tübingen, Germany, Heidelberg, Germany
| | - Bernhard Krämer
- Department of Women's Health, University of Tübingen, Tübingen, Germany, Heidelberg, Germany
| | - Eva-Maria Grischke
- Department of Women's Health, University of Tübingen, Tübingen, Germany, Heidelberg, Germany
| | - Sara Y Brucker
- Department of Women's Health, University of Tübingen, Tübingen, Germany, Heidelberg, Germany
| |
Collapse
|
1118
|
de la Cruz-Merino L, Chiesa M, Caballero R, Rojo F, Palazón N, Carrasco FH, Sánchez-Margalet V. Breast Cancer Immunology and Immunotherapy: Current Status and Future Perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 331:1-53. [PMID: 28325210 DOI: 10.1016/bs.ircmb.2016.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer immunology has gained renewed interest in the past few years due to emerging findings on mechanisms involved in tumoral immune evasion. Indisputably, immune edition is currently considered a critical hallmark of cancer. Basic research has revealed new targets which can be modulated in the clinical setting with new compounds and strategies. As recent evidence confirms, breast cancer (BC) is a complex and heterogeneous disease in which host immune responses play a substantial role. T-infiltrating lymphocytes measurement is suggested as a powerful new tool necessary to predict early BC evolution, especially in HER2-positive and triple negative subtypes. However, T-infiltrating lymphocytes, genomic platforms, and many other biomarkers in tissue and peripheral blood (e.g., regulatory T cells and myeloid-derived suppressor cells) are not the only factors being evaluated regarding their potential role as prognostic and/or predictive factors. Many ongoing clinical trials are exploring the activity of immune checkpoint modulators in BC treatment, both in the advanced and neoadjuvant setting. Although this field is expanding with exciting new discoveries and promising clinical results-and creating great expectations-there remain many uncertainties yet to be addressed satisfactorily before this long awaited therapeutic promise can come to fruition.
Collapse
Affiliation(s)
| | - M Chiesa
- GEICAM (Spanish Breast Cancer Research Group), Madrid, Spain
| | - R Caballero
- GEICAM (Spanish Breast Cancer Research Group), Madrid, Spain
| | - F Rojo
- Fundación Jiménez Díaz, Madrid, Spain
| | - N Palazón
- GEICAM (Spanish Breast Cancer Research Group), Madrid, Spain
| | - F H Carrasco
- GEICAM (Spanish Breast Cancer Research Group), Madrid, Spain
| | | |
Collapse
|
1119
|
Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, Hodi FS, Joshua AM, Kefford R, Hersey P, Joseph R, Gangadhar TC, Dronca R, Patnaik A, Zarour H, Roach C, Toland G, Lunceford JK, Li XN, Emancipator K, Dolled-Filhart M, Kang SP, Ebbinghaus S, Hamid O. Programmed Death-Ligand 1 Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J Clin Oncol 2016; 34:4102-4109. [PMID: 27863197 PMCID: PMC5562434 DOI: 10.1200/jco.2016.67.2477] [Citation(s) in RCA: 509] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Expression of programmed death-ligand 1 (PD-L1) is a potential predictive marker for response and outcome after treatment with anti-programmed death 1 (PD-1). This study explored the relationship between anti-PD-1 activity and PD-L1 expression in patients with advanced melanoma who were treated with pembrolizumab in the phase Ib KEYNOTE-001 study (clinical trial information: NCT01295827). Patients and Methods Six hundred fifty-five patients received pembrolizumab10 mg/kg once every 2 weeks or once every 3 weeks, or 2 mg/kg once every 3 weeks. Tumor response was assessed every 12 weeks per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 by independent central review. Primary outcome was objective response rate. Secondary outcomes included progression-free survival (PFS) and overall survival (OS). Membranous PD-L1 expression in tumor and tumor-associated immune cells was assessed by a clinical trial immunohistochemistry assay (22C3 antibody) and scored on a unique melanoma (MEL) scale of 0 to 5 by one of three pathologists who were blinded to clinical outcome; a score ≥ 2 (membranous staining in ≥ 1% of cells) was considered positive. Results Of 451 patients with evaluable PD-L1 expression, 344 (76%) had PD-L1-positive tumors. Demographic and staging variables were equally distributed among PD-L1-positive and -negative patients. An association between higher MEL score and higher response rate and longer PFS (hazard ratio, 0.76; 95% CI, 0.71 to 0.82) and OS (hazard ratio, 0.76; 95% CI, 0.69 to 0.83) was observed ( P < .001 for each). Objective response rate was 8%, 12%, 22%, 43%, 57%, and 53% for MEL 0, 1, 2, 3, 4, and 5, respectively. Conclusion PD-L1 expression in pretreatment tumor biopsy samples was correlated with response rate, PFS, and OS; however, patients with PD-L1-negative tumors may also achieve durable responses.
Collapse
Affiliation(s)
- Adil I. Daud
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Jedd D. Wolchok
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Caroline Robert
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Wen-Jen Hwu
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Jeffrey S. Weber
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Antoni Ribas
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - F. Stephen Hodi
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Anthony M. Joshua
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Richard Kefford
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Peter Hersey
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Richard Joseph
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Tara C. Gangadhar
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Roxana Dronca
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Amita Patnaik
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Hassane Zarour
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Charlotte Roach
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Grant Toland
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Jared K. Lunceford
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Xiaoyun Nicole Li
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Kenneth Emancipator
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Marisa Dolled-Filhart
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - S. Peter Kang
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Scot Ebbinghaus
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| | - Omid Hamid
- Adil I. Daud, University of California, San Francisco, San Francisco; Antoni Ribas, University of California, Los Angeles; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles; Charlotte Roach and Grant Toland, Dako North America, Carpinteria, CA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Wen-Jen Hwu, The University of Texas MD Anderson Cancer Center, Houston; Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa; Richard Joseph, Mayo Clinic, Jacksonville, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Tara C. Gangadhar, Abramson Cancer Center at the University of Pennsylvania, Philadelphia; Hassane Zarour, University of Pittsburgh, Pittsburgh, PA; Roxana Dronca, Mayo Clinic, Rochester, MN; Jared K. Lunceford, Xiaoyun Nicole Li, Kenneth Emancipator, Marisa Dolled-Filhart, S. Peter Kang, and Scot Ebbinghaus, Merck & Co, Kenilworth, NJ; Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif, France; Anthony M. Joshua, Princess Margaret Cancer Centre, Toronto, ON, Canada; Richard Kefford, Crown Princess Mary Cancer Centre, Westmead Hospital and Melanoma Institute Australia; Richard Kefford, Macquarie University; and Richard Kefford and Peter Hersey, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
1120
|
Ali HR, Chlon L, Pharoah PDP, Markowetz F, Caldas C. Patterns of Immune Infiltration in Breast Cancer and Their Clinical Implications: A Gene-Expression-Based Retrospective Study. PLoS Med 2016; 13:e1002194. [PMID: 27959923 PMCID: PMC5154505 DOI: 10.1371/journal.pmed.1002194] [Citation(s) in RCA: 437] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/04/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immune infiltration of breast tumours is associated with clinical outcome. However, past work has not accounted for the diversity of functionally distinct cell types that make up the immune response. The aim of this study was to determine whether differences in the cellular composition of the immune infiltrate in breast tumours influence survival and treatment response, and whether these effects differ by molecular subtype. METHODS AND FINDINGS We applied an established computational approach (CIBERSORT) to bulk gene expression profiles of almost 11,000 tumours to infer the proportions of 22 subsets of immune cells. We investigated associations between each cell type and survival and response to chemotherapy, modelling cellular proportions as quartiles. We found that tumours with little or no immune infiltration were associated with different survival patterns according to oestrogen receptor (ER) status. In ER-negative disease, tumours lacking immune infiltration were associated with the poorest prognosis, whereas in ER-positive disease, they were associated with intermediate prognosis. Of the cell subsets investigated, T regulatory cells and M0 and M2 macrophages emerged as the most strongly associated with poor outcome, regardless of ER status. Among ER-negative tumours, CD8+ T cells (hazard ratio [HR] = 0.89, 95% CI 0.80-0.98; p = 0.02) and activated memory T cells (HR 0.88, 95% CI 0.80-0.97; p = 0.01) were associated with favourable outcome. T follicular helper cells (odds ratio [OR] = 1.34, 95% CI 1.14-1.57; p < 0.001) and memory B cells (OR = 1.18, 95% CI 1.0-1.39; p = 0.04) were associated with pathological complete response to neoadjuvant chemotherapy in ER-negative disease, suggesting a role for humoral immunity in mediating response to cytotoxic therapy. Unsupervised clustering analysis using immune cell proportions revealed eight subgroups of tumours, largely defined by the balance between M0, M1, and M2 macrophages, with distinct survival patterns by ER status and associations with patient age at diagnosis. The main limitations of this study are the use of diverse platforms for measuring gene expression, including some not previously used with CIBERSORT, and the combined analysis of different forms of follow-up across studies. CONCLUSIONS Large differences in the cellular composition of the immune infiltrate in breast tumours appear to exist, and these differences are likely to be important determinants of both prognosis and response to treatment. In particular, macrophages emerge as a possible target for novel therapies. Detailed analysis of the cellular immune response in tumours has the potential to enhance clinical prediction and to identify candidates for immunotherapy.
Collapse
Affiliation(s)
- H. Raza Ali
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Leon Chlon
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, United Kingdom
| | - Paul D. P. Pharoah
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, United Kingdom
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
1121
|
Targeting the host immune system: PD-1 and PD-L1 antibodies and breast cancer. Curr Opin Support Palliat Care 2016; 10:336-342. [DOI: 10.1097/spc.0000000000000243] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
1122
|
Abstract
Targeted immunotherapy based on PD-1/PD-L1 suppression has revolutionized the treatment of various solid tumors. A remarkable improvement has also been observed in the treatment of patients with refractory/relapsing classical Hodgkin lymphoma (cHL). We investigated PD-L1 status in a variety of treatment resistant lymphomas. Tumor samples from 78 patients with therapy resistant lymphomas were immunohistochemically (IHC) investigated for the expression of PD-L1 using two antibody clones (SP142 and SP263, Ventana). Thirteen PD-L1+ cases were further analyzed for gene copy number variations (CNV) by NGS and for PD-L1/JAK2/PD-L2 co-amplification using fluorescent in-situ hybridization assay (FISH). PD-L1 positivity (≥5% positive cancer cells, IHC) was present in 32/77 (42%) and 33/71 cases (46%) using SP142 and SP263 antibodies, respectively. Concordance between the two anti-PD-L1 clones was high with only three (4%) discrepant cases. The strongest and consistent (10/11 cases) expression was observed in cHL and primary mediastinal B-cell lymphomas (3/3). Diffuse large B-cell lymphomas (DLBCL) were frequently positive (13/26) irrespective of subtype. Follicular (1/8), peripheral T-cell (3/11) and mantle cell (1/8) lymphomas were rarely positive, while small lymphocytic lymphoma/CLL and marginal zone lymphomas were consistently negative (3/3). Co-amplification/CNVs of PD-L1/JAK2/PD-L2 were observed in 3 cases of DLBCL and cHL, respectively. Of note, all three cHL-amplified cases were positive by FISH, but not by NGS. Since only a fraction of the IHC positive lymphoma cases were positive by FISH and NGS assays, other mechanisms are involved in PD-L1 upregulation, especially in DLBCL. FISH assay may be more suitable than NGS assay for determination of PD-L1 alterations in cHL.
Collapse
|
1123
|
Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, O'Brien M, Rao S, Hotta K, Leiby MA, Lubiniecki GM, Shentu Y, Rangwala R, Brahmer JR. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016; 375:1823-1833. [PMID: 27718847 DOI: 10.1056/nejmoa1606774] [Citation(s) in RCA: 7430] [Impact Index Per Article: 825.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pembrolizumab is a humanized monoclonal antibody against programmed death 1 (PD-1) that has antitumor activity in advanced non-small-cell lung cancer (NSCLC), with increased activity in tumors that express programmed death ligand 1 (PD-L1). METHODS In this open-label, phase 3 trial, we randomly assigned 305 patients who had previously untreated advanced NSCLC with PD-L1 expression on at least 50% of tumor cells and no sensitizing mutation of the epidermal growth factor receptor gene or translocation of the anaplastic lymphoma kinase gene to receive either pembrolizumab (at a fixed dose of 200 mg every 3 weeks) or the investigator's choice of platinum-based chemotherapy. Crossover from the chemotherapy group to the pembrolizumab group was permitted in the event of disease progression. The primary end point, progression-free survival, was assessed by means of blinded, independent, central radiologic review. Secondary end points were overall survival, objective response rate, and safety. RESULTS Median progression-free survival was 10.3 months (95% confidence interval [CI], 6.7 to not reached) in the pembrolizumab group versus 6.0 months (95% CI, 4.2 to 6.2) in the chemotherapy group (hazard ratio for disease progression or death, 0.50; 95% CI, 0.37 to 0.68; P<0.001). The estimated rate of overall survival at 6 months was 80.2% in the pembrolizumab group versus 72.4% in the chemotherapy group (hazard ratio for death, 0.60; 95% CI, 0.41 to 0.89; P=0.005). The response rate was higher in the pembrolizumab group than in the chemotherapy group (44.8% vs. 27.8%), the median duration of response was longer (not reached [range, 1.9+ to 14.5+ months] vs. 6.3 months [range, 2.1+ to 12.6+]), and treatment-related adverse events of any grade were less frequent (occurring in 73.4% vs. 90.0% of patients), as were grade 3, 4, or 5 treatment-related adverse events (26.6% vs. 53.3%). CONCLUSIONS In patients with advanced NSCLC and PD-L1 expression on at least 50% of tumor cells, pembrolizumab was associated with significantly longer progression-free and overall survival and with fewer adverse events than was platinum-based chemotherapy. (Funded by Merck; KEYNOTE-024 ClinicalTrials.gov number, NCT02142738 .).
Collapse
Affiliation(s)
- Martin Reck
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Delvys Rodríguez-Abreu
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Andrew G Robinson
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Rina Hui
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Tibor Csőszi
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Andrea Fülöp
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Maya Gottfried
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Nir Peled
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Ali Tafreshi
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Sinead Cuffe
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Mary O'Brien
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Suman Rao
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Katsuyuki Hotta
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Melanie A Leiby
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Gregory M Lubiniecki
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Yue Shentu
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Reshma Rangwala
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| | - Julie R Brahmer
- From Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany (M.R.); Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain (D.R.-A.); Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston, ON, Canada (A.G.R.); Westmead Hospital and the University of Sydney, Sydney (R.H.), and Southern Medical Day Care Centre, Wollongong, NSW (A.T.) - both in Australia; Jász-Nagykun-Szolnok County Hospital, Szolnok (T.C.), and Országos Korányi TBC és Pulmonológiai Intézet, Budapest (A.F.) - both in Hungary; Meir Medical Center, Kfar-Saba (M.G.), and Davidoff Cancer Center, Tel Aviv University, Petah Tikva (N.P.) - both in Israel; St. James's Hospital and Cancer Trials Ireland, Dublin (S.C.); the Royal Marsden Hospital, Sutton, Surrey, United Kingdom (M.O.); MedStar Franklin Square Hospital (S.R.) and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins (J.R.B.) - both in Baltimore; Okayama University Hospital, Okayama, Japan (K.H.); and Merck, Kenilworth, NJ (M.A.L., G.M.L., Y.S., R.R.)
| |
Collapse
|
1124
|
Ikeda S, Goodman AM, Cohen PR, Jensen TJ, Ellison CK, Frampton G, Miller V, Patel SP, Kurzrock R. Metastatic basal cell carcinoma with amplification of PD-L1: exceptional response to anti-PD1 therapy. NPJ Genom Med 2016; 1. [PMID: 27942391 PMCID: PMC5142752 DOI: 10.1038/npjgenmed.2016.37] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Metastatic basal cell carcinomas are rare malignancies harbouring Hedgehog pathway alterations targetable by SMO antagonists (vismodegib/sonidegib). We describe, for the first time, the molecular genetics and response of a patient with Hedgehog inhibitor-resistant metastatic basal cell carcinoma who achieved rapid tumour regression (ongoing near complete remission at 4 months) with nivolumab (anti-PD1 antibody). He had multiple hallmarks of anti-PD1 responsiveness including high mutational burden (>50 mutations per megabase; 19 functional alterations in tissue next-generation sequencing (NGS; 315 genes)) as well as PDL1/PDL2/JAK2 amplification (as determined by both tissue NGS and by analysis of plasma-derived cell-free DNA). The latter was performed using technology originally developed for the genome-wide detection of sub-chromosomal copy-number alterations (CNAs) in noninvasive prenatal testing and showed numerous CNAs including amplification of the 9p24.3-9p22.2 region containing PD-L1, PD-L2 and JAK2. Of interest, PD-L1, PD-L2 and JAK2 amplification is a characteristic of Hodgkin lymphoma, which is exquisitely sensitive to nivolumab. In conclusion, selected SMO antagonist-resistant metastatic basal cell carcinomas may respond to nivolumab based on underlying molecular genetic mechanisms that include PD-L1 amplification and high tumour mutational burden.
Collapse
Affiliation(s)
- Sadakatsu Ikeda
- Department of Medicine, Division of Hematology/Oncology, University of California, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA; Cancer Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aaron M Goodman
- Department of Medicine, Division of Hematology/Oncology, University of California, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | - Philip R Cohen
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | - Sandip P Patel
- Department of Medicine, Division of Hematology/Oncology, University of California, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, University of California, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
1125
|
Perspectives in immunotherapy: meeting report from the “Immunotherapy Bridge”, Napoli, December 5th 2015. J Immunother Cancer 2016. [PMCID: PMC5067891 DOI: 10.1186/s40425-016-0168-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Harnessing the immune system and preventing immune escape, the immunotherapy of cancer provides great potential for clinical application, in broad patient populations, achieving both conventional and unconventional clinical responses. After the substantial advances in melanoma, the focus of cancer immunotherapy has expanded to include many other cancers. Targeting immune checkpoints and further mechanisms used by tumors to avoid anticancer immunity, different approaches are under evaluation, including combination therapies. The first Immunotherapy Bridge meeting focused on various cancer types including melanoma, non-small cell lung cancer, renal cell, breast and ovarian carcinoma, and discussed mechanisms of action of single agents and combination strategies, and the prediction of clinical responses.
Collapse
|
1126
|
Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer 2016; 4:59. [PMID: 27777769 PMCID: PMC5067916 DOI: 10.1186/s40425-016-0165-6] [Citation(s) in RCA: 566] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/13/2016] [Indexed: 12/16/2022] Open
Abstract
Tumor infiltrating lymphocytes (TIL) play an essential role in mediating response to chemotherapy and improving clinical outcomes in all subtypes of breast cancer. Triple negative breast cancers (TN) are most likely to have tumors with >50 % lymphocytic infiltrate, termed lymphocyte predominant breast cancer, and derive the greatest survival benefit from each 10 % increase in TIL. The majority of HER2+ breast cancers have similar level of immune infiltrate as TN breast cancer yet the presence of TILs has not shown the same survival benefit. For HER2+ breast cancers, type 1 T-cells, either increased TBET+ tumor infiltration or increased type 1 HER2-specific CD4+ T-cells in the peripheral blood, are associated with better outcomes. Hormone receptor positive HER2 negative tumors tend to have the least immune infiltrate yet are the only breast cancer subtype to show worse prognosis with increased FOXP3 regulatory T-cell infiltrate. Notably, all breast cancer subtypes have tumors with low, intermediate, or high TIL infiltrate. Tumors with high TILs may also have increased PD-L1 expression which might be the reason that TN breast cancer seems to demonstrate the most robust clinical response to immune checkpoint inhibitor therapy but further investigation is needed. Tumors with intermediate or low levels of pre-treatment immune infiltrate, on the other hand, may benefit from an intervention that may increase TIL, particularly type 1 T-cells. Examples of these interventions include specific types of cytotoxic chemotherapy, radiation, or vaccine therapy. Therefore, the systematic evaluation of TIL and specific populations of TIL may be able to both guide prognosis and the appropriate sequencing of therapies in breast cancer.
Collapse
Affiliation(s)
- Sasha E. Stanton
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, 850 Republican Street, 2nd Floor, Box 358050, Seattle, WA 98195-8050 USA
| | - Mary L. Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, 850 Republican Street, 2nd Floor, Box 358050, Seattle, WA 98195-8050 USA
| |
Collapse
|
1127
|
Bense RD, Sotiriou C, Piccart-Gebhart MJ, Haanen JBAG, van Vugt MATM, de Vries EGE, Schröder CP, Fehrmann RSN. Relevance of Tumor-Infiltrating Immune Cell Composition and Functionality for Disease Outcome in Breast Cancer. J Natl Cancer Inst 2016; 109:2905892. [PMID: 27737921 DOI: 10.1093/jnci/djw192] [Citation(s) in RCA: 276] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/25/2016] [Indexed: 12/20/2022] Open
Abstract
Background Not all breast cancer patients benefit from neoadjuvant or adjuvant therapy, resulting in considerable undertreatment or overtreatment. New insights into the role of tumor-infiltrating immune cells suggest that their composition, as well as their functionality, might serve as a biomarker to enable optimal patient selection for current systemic therapies and upcoming treatment options such as immunotherapy. Methods We performed several complementary unbiased in silico analyses on gene expression profiles of 7270 unrelated tumor samples of nonmetastatic breast cancer patients with known clinical follow-up. CIBERSORT was used to estimate the fraction of 22 immune cell types to study their relations with pathological complete response (pCR), disease-free survival (DFS), and overall survival (OS). In addition, we used four previously reported immune gene signatures and a CD8+ T-cell exhaustion signature to assess their relationships with breast cancer outcome. Multivariable binary logistic regression and multivariable Cox regression were used to assess the association of immune cell-type fractions and immune signatures with pCR and DFS/OS, respectively. Results Increased fraction of regulatory T-cells in human epidermal growth factor receptor 2 (HER2)-positive tumors was associated with a lower pCR rate (odds ratio [OR] = 0.15, 95% confidence interval [CI] = 0.03 to 0.69), as well as shorter DFS (hazard ratio [HR] = 3.13, 95% CI = 1.23 to 7.98) and OS (HR = 7.69, 95% CI = 3.43 to 17.23). A higher fraction of M0 macrophages in estrogen receptor (ER)-positive tumors was associated with worse DFS (HR = 1.66, 95% CI = 1.18 to 2.33) and, in ER-positive/HER2-negative tumors, with worse OS (HR = 1.71, 95% CI = 1.12 to 2.61). Increased fractions of γδ T-cells in all breast cancer patients related to a higher pCR rate (OR = 1.55, 95% CI = 1.01 to 2.38), prolonged DFS (HR = 0.68, 95% CI = 0.48 to 0.98), and, in HER2-positive tumors, with prolonged OS (HR = 0.27, 95% CI = 0.10 to 0.73). A higher fraction of activated mast cells was associated with worse DFS (HR = 5.85, 95% CI = 2.20 to 15.54) and OS (HR = 5.33, 95% CI = 2.04 to 13.91) in HER2-positive tumors. The composition of relevant immune cell types frequently differed per breast cancer subtype. Furthermore, a high CD8+ T-cell exhaustion signature score was associated with shortened DFS in patients with ER-positive tumors regardless of HER2 status (HR = 1.80, 95% CI = 1.07 to 3.04). Conclusions The main hypothesis generated in our unbiased in silico approach is that a multitude of immune cells are related to treatment response and outcome in breast cancer.
Collapse
Affiliation(s)
- Rico D Bense
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - Christos Sotiriou
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - Martine J Piccart-Gebhart
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - John B A G Haanen
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - Marcel A T M van Vugt
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - Elisabeth G E de Vries
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - Carolien P Schröder
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| | - Rudolf S N Fehrmann
- Affiliations of authors: Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (RDB, MATMvV, EGEdV, CPS, RSNF); Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium (CS, MJPG); Division of Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands (JBAGH)
| |
Collapse
|
1128
|
Fleisher B, Clarke C, Ait-Oudhia S. Current advances in biomarkers for targeted therapy in triple-negative breast cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2016; 8:183-197. [PMID: 27785100 PMCID: PMC5063595 DOI: 10.2147/bctt.s114659] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer (TNBC) is a complex heterogeneous disease characterized by the absence of three hallmark receptors: human epidermal growth factor receptor 2, estrogen receptor, and progesterone receptor. Compared to other breast cancer subtypes, TNBC is more aggressive, has a higher prevalence in African-Americans, and more frequently affects younger patients. Currently, TNBC lacks clinically accepted targets for tailored therapy, warranting the need for candidate biomarkers. BiomarkerBase, an online platform used to find biomarkers reported in clinical trials, was utilized to screen all potential biomarkers for TNBC and select only the ones registered in completed TNBC trials through clinicaltrials.gov. The selected candidate biomarkers were classified as surrogate, prognostic, predictive, or pharmacodynamic (PD) and organized by location in the blood, on the cell surface, in the cytoplasm, or in the nucleus. Blood biomarkers include vascular endothelial growth factor/vascular endothelial growth factor receptor and interleukin-8 (IL-8); cell surface biomarkers include EGFR, insulin-like growth factor binding protein, c-Kit, c-Met, and PD-L1; cytoplasm biomarkers include PIK3CA, pAKT/S6/p4E-BP1, PTEN, ALDH1, and the PIK3CA/AKT/mTOR-related metabolites; and nucleus biomarkers include BRCA1, the gluco-corticoid receptor, TP53, and Ki67. Candidate biomarkers were further organized into a "cellular protein network" that demonstrates potential connectivity. This review provides an inventory and reference point for promising biomarkers for breakthrough targeted therapies in TNBC.
Collapse
Affiliation(s)
- Brett Fleisher
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL
| | - Charlotte Clarke
- Department of Translational Research, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Sihem Ait-Oudhia
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL
| |
Collapse
|
1129
|
Sanchez K, Page D, McArthur HL. Immunotherapy in breast cancer: An overview of modern checkpoint blockade strategies and vaccines. Curr Probl Cancer 2016; 40:151-162. [PMID: 27855963 DOI: 10.1016/j.currproblcancer.2016.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/21/2016] [Indexed: 12/29/2022]
Abstract
Immune therapy has recently emerged as a standard-of-care strategy for the treatment of melanoma, lung cancer, bladder cancer, among other malignancies. However, the role of immune therapy in the treatment of breast cancer is still being determined. Two current strategies for harnessing the immune system to treat cancer include drugs that modulate key T cell inhibitory checkpoints and vaccines. Specifically, modern immune therapy strategies can facilitate T-cell mediated tumor regression by priming the immune system against specific tumor associated antigens, by modulating immunoregulatory signals, or both. In breast cancer, preliminary data from preclinical and early clinical studies are promising. In fact, clinical data with checkpoint blockade as monotherapy has been reported in multiple breast cancer subtypes to date, with durable responses observed in a significant proportion of women with chemotherapy resistant disease. However, because the number of genetic mutations and thus, the number of neoantigens available for immune response are modest in most breast cancers when compared with other cancers, most breast cancers may not be inherently sensitive to immune modulation and therefore may require strategies that enhance tumor associated antigen presentation if immune modulation strategies are to be effective. To that end, studies that combine checkpoint blockade with other strategies including established systemic therapies (including hormone therapy and chemotherapy), radiation therapy, and localized therapy including tumor freezing (cryoablation) are underway in breast cancer. Studies that combine checkpoint blockade with vaccines are also planned. Herein, we provide a brief summary of key components of the immune response against cancer, a rationale for the use of immune therapy in breast cancer, data from early clinical trials of checkpoint blockade and vaccine strategies in breast cancer, and future directions in the field.
Collapse
Affiliation(s)
- Katherine Sanchez
- Providence Cancer Center / Earle A. Chiles Research Institute, Portland, OR
| | - David Page
- Providence Cancer Center / Earle A. Chiles Research Institute, Portland, OR
| | | |
Collapse
|
1130
|
Afghahi A, Telli ML, Kurian AW. Genetics of triple-negative breast cancer: Implications for patient care. Curr Probl Cancer 2016; 40:130-140. [PMID: 28340968 DOI: 10.1016/j.currproblcancer.2016.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022]
Abstract
Patients with triple-negative breast cancer (TNBC), defined as lacking expression of the estrogen and progesterone receptors (ER/PR) and amplification of the HER2 oncogene, often have a more aggressive disease course than do patients with hormone receptor-positive breast cancer, including higher rates of visceral and central nervous system metastases, early cancer recurrences and deaths. Triple-negative breast cancer is associated with a young age at diagnosis and both African and Ashkenazi Jewish ancestry, the latter due to three common founder mutations in the highly penetrant cancer susceptibility genes BRCA1 and BRCA2 (BRCA1/2). In the past decade, there has been a surge both in genetic testing technology and in patient access to such testing. Advances in genetic testing have enabled more rapid and less expensive commercial sequencing than could be imagined only a few years ago. Massively parallel, next-generation sequencing allows the simultaneous analysis of many different genes. Studies of TNBC patients in the current era have revealed associations of TNBC with mutations in several moderate penetrance breast cancer susceptibility genes, including PALB2, BARD1, BRIP1, RAD51C and RAD51D. Interestingly, many of these genes, like BRCA1/2, are involved in homologous recombination DNA double-stranded repair. In this review, we summarize the current understanding of pathogenic germline gene mutations associated with TNBC and the early detection and prevention strategies for women at risk of developing this high-risk breast cancer subtype. Furthermore, we discuss recent the advances in targeted therapies for TNBC patients with a hereditary predisposition, including the role of poly (ADP-ribose) polymerase (PARP) inhibitors in BRCA1/2 mutation-associated breast cancers.
Collapse
Affiliation(s)
- Anosheh Afghahi
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, CO
| | - Melinda L Telli
- Stanford University School of Medicine, Department of Medicine, Stanford, CA
| | - Allison W Kurian
- Stanford University School of Medicine, Department of Medicine, Stanford, CA; Stanford University School of Medicine, Department of Health Research and Policy, Stanford, CA.
| |
Collapse
|
1131
|
|
1132
|
Saha P, Nanda R. Concepts and targets in triple-negative breast cancer: recent results and clinical implications. Ther Adv Med Oncol 2016; 8:351-9. [PMID: 27583027 PMCID: PMC4981296 DOI: 10.1177/1758834016657071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease in which tumors are defined by lack of expression of the estrogen receptor (ER), the progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) receptor. No targeted therapies are available for the treatment of TNBC, and chemotherapy remains the standard of care. Gene expression profiling has identified six distinct molecular subtypes of TNBC. The identification of novel targets, coupled with the development of therapies for different subsets of TNBC, holds great promise for the future treatment of this aggressive form of breast cancer. This review focuses on novel therapies in development for the treatment of TNBC.
Collapse
Affiliation(s)
- Poornima Saha
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Rita Nanda
- University of Chicago Medical Center, 5841 S. Maryland Ave., MC 2115, Chicago, IL 60637-1470, USA
| |
Collapse
|
1133
|
McArthur HL, Diab A, Page DB, Yuan J, Solomon SB, Sacchini V, Comstock C, Durack JC, Maybody M, Sung J, Ginsberg A, Wong P, Barlas A, Dong Z, Zhao C, Blum B, Patil S, Neville D, Comen EA, Morris EA, Kotin A, Brogi E, Wen YH, Morrow M, Lacouture ME, Sharma P, Allison JP, Hudis CA, Wolchok JD, Norton L. A Pilot Study of Preoperative Single-Dose Ipilimumab and/or Cryoablation in Women with Early-Stage Breast Cancer with Comprehensive Immune Profiling. Clin Cancer Res 2016; 22:5729-5737. [PMID: 27566765 DOI: 10.1158/1078-0432.ccr-16-0190] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 06/30/2016] [Accepted: 07/31/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE To assess the safety and tolerability of preoperative cryoablation-mediated tumor antigen presentation and/or ipilimumab-mediated immune modulation in women with operable breast cancer. EXPERIMENTAL DESIGN In this pilot study, 19 women with breast cancer for whom mastectomy was planned were treated with preoperative tumor cryoablation (n = 7), single-dose ipilimumab at 10 mg/kg (n = 6), or both (n = 6). The primary outcome for this pilot study was safety/tolerability as defined as freedom from delays in pre-planned, curative-intent mastectomy. Exploratory studies of immune activation were performed on peripheral blood and tumor. RESULTS Preoperative cryoablation and/or ipilimumab were safe and tolerable, with no delays in pre-planned surgery. Grade III toxicity was seen in 1 of 19 (unrelated rash after ipilimumab). Combination therapy was associated with sustained peripheral elevations in: Th1-type cytokines, activated (ICOS+) and proliferating (Ki67+) CD4+ and CD8+ T cells, and posttreatment proliferative T-effector cells relative to T-regulatory cells within tumor. CONCLUSIONS Preoperative cryoablation and single-dose ipilimumab are safe alone or in combination with no surgical delays incurred. Potentially favorable intratumoral and systemic immunologic effects were observed with the combination, suggesting the possibility for induced and synergistic antitumor immunity with this strategy. Clin Cancer Res; 22(23); 5729-37. ©2016 AACR.
Collapse
Affiliation(s)
| | - Adi Diab
- MD Anderson Cancer Center, Houston, Texas
| | - David B Page
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Jianda Yuan
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | | | | | | | - Jeremy C Durack
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Majid Maybody
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Janice Sung
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Arielle Ginsberg
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Phillip Wong
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Afsar Barlas
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Zhiwan Dong
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Chunjun Zhao
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Brian Blum
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Sujata Patil
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Deirdre Neville
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | | | | | - Alan Kotin
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Edi Brogi
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Y Hannah Wen
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Monica Morrow
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | | | | | | | - Clifford A Hudis
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| | - Larry Norton
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York
| |
Collapse
|
1134
|
Lin Z, Chen X, Li Z, Luo Y, Fang Z, Xu B, Han M. PD-1 Antibody Monotherapy for Malignant Melanoma: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0160485. [PMID: 27483468 PMCID: PMC4970765 DOI: 10.1371/journal.pone.0160485] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/20/2016] [Indexed: 12/31/2022] Open
Abstract
Antibodies targeting programmed death 1 (PD-1) help prevent tumor cells from escaping immune-mediated destruction. We conducted this systematic review and meta-analysis to gain insight into the efficacy of PD-1 antibodies for the treatment of melanoma. Five trials involving 2,828 adult patients were included in this meta-analysis. In patients with previously untreated or refractory melanoma, treatment with PD-1 antibodies significantly improved the six-month progression-free survival (PFS) (HR 0.55, 95% CI 0.50-0.60, P<0.00001) and the overall response rate (OR 3.89, 95% CI 3.12-4.83, P<0.00001). This meta-analysis indicated that anti-PD-1 treatment might provide a significant survival benefit in patients with melanoma. In addition, we found that patients treated with nivolumab reported significantly fewer treatment-related adverse events (OR 0.74, 95% CI 0.57-0.97, P = 0.03) than those treated with other agents, but there was a dose-dependent increase in the frequency of adverse events in patients treated with pembrolizumab.
Collapse
Affiliation(s)
- Zhijuan Lin
- Department of Hematology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Xing Chen
- Department of Nephrology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Zhifeng Li
- Department of Hematology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Yiming Luo
- Department of Hematology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Zhihong Fang
- Department of Hematology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Bing Xu
- Department of Hematology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Mingzhe Han
- Department of Hematology, the First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, People's Republic of China
| |
Collapse
|
1135
|
Miyan M, Schmidt-Mende J, Kiessling R, Poschke I, de Boniface J. Differential tumor infiltration by T-cells characterizes intrinsic molecular subtypes in breast cancer. J Transl Med 2016; 14:227. [PMID: 27473163 PMCID: PMC4966793 DOI: 10.1186/s12967-016-0983-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
Background Molecular subtypes of breast cancer and presence of tumor-infiltrating immune cells have both been implicated as important predictive and prognostic factors for improved risk stratification and treatment individualization of breast cancer patients. Their association, however, has not been studied in detail. The aim of this study was to evaluate the expression of the T cell markers CD8, FoxP3, CD3 and ζ-chain in molecular subtypes of the invasive margin and tumor center of breast cancer and corresponding sentinel nodes and to deduct prognostic information from these findings. Methods Tumor and sentinel node sections from 177 patients with primary, invasive, unilateral early-stage breast cancer were stained by immunohistochemistry and T-cell phenotypes quantified manually. Clinical data were collected from medical records. Results The degree of T-cell infiltration and expression of all markers differed significantly among the molecular subtypes, being highest in non-luminal, more aggressive tumors: more T-cell infiltration and higher expression of all markers were associated with hormone receptor negativity, higher proliferation and higher histological grades, but also with larger tumor size. Basal-like tumors, and most remarkably their tumor centers, hosted the highest number of FoxP3+ T-cells with an unfavorable ratio to cytotoxic CD8+ T-cells. T-cell infiltration was generally higher in the invasive margin than the tumor center. A scoring system based on densities of CD3 and CD8 could significantly separate molecular subtypes (p < 0.001). Conclusions Thus, immunological patterns with functional implications within each subtype are associated with prognostic factors. These findings should be further validated in studies using larger patient populations and longer follow-up. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0983-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Miyan
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - J Schmidt-Mende
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.,Department of Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - R Kiessling
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - I Poschke
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - J de Boniface
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Department of Breast and Endocrine Surgery, P9:03, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
1136
|
Puglisi F, Bisagni G, Ciccarese M, Fontanella C, Gamucci T, Leo L, Molino A, Silva RR, Marchetti P. A Delphi consensus and open debate on the role of first-line bevacizumab for HER2-negative metastatic breast cancer. Future Oncol 2016; 12:2589-2602. [PMID: 27443691 DOI: 10.2217/fon-2016-0295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
To gain consensus on the role of bevacizumab plus paclitaxel as first-line treatment for HER2-negative metastatic breast cancer, a panel of expert oncologists experienced in treating patients with metastatic breast cancer in Italy participated in a Delphi consensus study. The panel reached a full consensus on the efficacy of bevacizumab plus paclitaxel and the clinical meaningfulness of the progression-free survival benefit compared with paclitaxel alone, despite the lack of an overall survival effect in clinical trials. The participants agreed that real-world data support the effectiveness and well-defined safety profile of the regimen. Views on the use of bevacizumab plus paclitaxel in specific patient populations were not unanimous and clinical judgment remains important. Nevertheless, a high level of agreement was reached.
Collapse
Affiliation(s)
- Fabio Puglisi
- Department of Medical & Biological Sciences, University of Udine, Udine, Italy
| | - Giancarlo Bisagni
- Oncology Unit, Department of Oncology, Azienda Ospedaliera ASMN, Istituto di Ricovero e Cura a Carattere Scientifico, Reggio Emilia, Italy
| | | | - Caterina Fontanella
- Department of Medical & Biological Sciences, University of Udine, Udine, Italy
| | | | - Luigi Leo
- Oncology Unit, Azienda Ospedaliera dei Colli, Naples, Italy
| | | | - Rosa Rita Silva
- Medical Oncology Unit, ASUR Marche AV2 Fabriano, Fabriano, Italy
| | - Paolo Marchetti
- Medical Oncology, Sant'Andrea Hospital, Sapienza University of Rome & IDI-IRCCS, Rome, Italy
| |
Collapse
|
1137
|
Migali C, Milano M, Trapani D, Criscitiello C, Esposito A, Locatelli M, Minchella I, Curigliano G. Strategies to modulate the immune system in breast cancer: checkpoint inhibitors and beyond. Ther Adv Med Oncol 2016; 8:360-74. [PMID: 27583028 DOI: 10.1177/1758834016658423] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Is breast cancer (BC) immunogenic? Many data suggest that it is. Many observations demonstrated the prognostic role of tumor-infiltrating lymphocytes (TILs) in triple negative (TN) and human epidermal growth factor receptor 2 (HER-2)-positive BC. TNBCs are poorly differentiated tumors with high genetic instability and very high heterogeneity. This heterogeneity enhances the 'danger signals' and select clone variants that could be more antigenic or, in other words, that could more strongly stimulate a host immune antitumor response. The response to chemotherapy is at least partly dependent on an immunological reaction against those tumor cells that are dying during the chemotherapy. One of the mechanisms whereby chemotherapy can stimulate the immune system to recognize and destroy malignant cells is commonly known as immunogenic cell death (ICD). ICD elicits an adaptive immune response. Which are the clinical implications of all 'immunome' data produced in the last years? First, validate prognostic or predictive role of TILs. Second, validate immune genomic signatures that may be predictive and prognostic in patients with TN disease. Third, incorporate an 'immunoscore' into traditional classification of BC, thus providing an essential prognostic and potentially predictive tool in the pathology report. Fourth, implement clinical trials for BC in the metastatic setting with drugs that target immune-cell-intrinsic checkpoints. Blockade of one of these checkpoints, cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) or the programmed cell death 1 (PD-1) receptor may provide proof of concepts for the activity of an immune-modulation approach in the treatment of a BC.
Collapse
Affiliation(s)
- Cristina Migali
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Monica Milano
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Dario Trapani
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Carmen Criscitiello
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Angela Esposito
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Marzia Locatelli
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Ida Minchella
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Giuseppe Curigliano
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milano, Italy
| |
Collapse
|
1138
|
Lee KL, Kuo YC, Ho YS, Huang YH. Isolation and characterization of Pseudomonas aeruginosa PAO mutant that produces altered elastase. J Bacteriol 1980; 11:cancers11091334. [PMID: 31505803 PMCID: PMC6769912 DOI: 10.3390/cancers11091334] [Citation(s) in RCA: 165] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is cancer that tested as negative for estrogen receptors (ER), progesterone receptors (PR), and excess human epidermal growth factor receptor 2 (HER2) protein which accounts for 15%–20% of all breast cancer cases. TNBC is considered to be a poorer prognosis than other types of breast cancer, mainly because it involves more aggressive phenotypes that are similar to stem cell–like cancer cells (cancer stem cell, CSC). Thus, targeted treatment of TNBC remains a major challenge in clinical practice. This review article surveys the latest evidence concerning the role of genomic alteration in current TNBC treatment responses, current clinical trials and potential targeting sites, CSC and drug resistance, and potential strategies targeting CSCs in TNBC. Furthermore, the role of insulin-like growth factor 1 receptor (IGF-1R) and nicotinic acetylcholine receptors (nAChR) in stemness expression, chemoresistance, and metastasis in TNBC and their relevance to potential treatments are also discussed and highlighted.
Collapse
Affiliation(s)
- Kha-Liang Lee
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yung-Che Kuo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yuan-Soon Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan.
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan.
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|