1051
|
Yuan Y, Kos FJ, He TF, Yin HH, Li M, Hardwick N, Zurcher K, Schmolze D, Lee P, Pillai RK, Chung V, Diamond DJ. Complete regression of cutaneous metastases with systemic immune response in a patient with triple negative breast cancer receiving p53MVA vaccine with pembrolizumab. Oncoimmunology 2017; 6:e1363138. [PMID: 29209571 DOI: 10.1080/2162402x.2017.1363138] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022] Open
Abstract
A heavily pretreated patient with triple negative breast cancer distinguished by cutaneous metastases received p53MVA vaccine in combination with pembrolizumab. Her cutaneous metastases regressed and after 2 cycles of therapy, a skin biopsy showed a complete pathological response. Systemic response was confirmed with restaging CT and bone scans. Activation of p53-specific T cell responses and elevation of multiple immune response genes in peripheral blood correlated with the rapid clinical response which lasted for 6 months after the initiation of combined therapy.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Ferdynand J Kos
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Ting-Fang He
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Hongwei H Yin
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Mengsha Li
- Research Operations, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Nicola Hardwick
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Kathryn Zurcher
- Research Operations, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Peter Lee
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Raju K Pillai
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Vincent Chung
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Don J Diamond
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
1052
|
Costa R, Santa-Maria CA, Rossi G, Carneiro BA, Chae YK, Gradishar WJ, Giles FJ, Cristofanilli M. Developmental therapeutics for inflammatory breast cancer: Biology and translational directions. Oncotarget 2017; 8:12417-12432. [PMID: 27926493 PMCID: PMC5355355 DOI: 10.18632/oncotarget.13778] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/07/2016] [Indexed: 12/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer, which accounts for approximately 3% of cases of breast malignancies. Diagnosis relies largely on its clinical presentation, and despite a characteristic phenotype, underlying molecular mechanisms are poorly understood. Unique clinical presentation indicates that IBC is a distinct clinical and biological entity when compared to non-IBC. Biological understanding of non-IBC has been extrapolated into IBC and targeted therapies for HER2 positive (HER2+) and hormonal receptor positive non-IBC led to improved patient outcomes in the recent years. This manuscript reviews recent discoveries related to the underlying biology of IBC, clinical progress to date and suggests rational approaches for investigational therapies.
Collapse
Affiliation(s)
- Ricardo Costa
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America
| | - Cesar A Santa-Maria
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Giovanna Rossi
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Benedito A Carneiro
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - William J Gradishar
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Francis J Giles
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Massimo Cristofanilli
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| |
Collapse
|
1053
|
Abstract
PURPOSE OF REVIEW The purpose of the review is to summarize the data regarding PD-L1 expression in breast cancer and the results of first clinical trials with PD-1 or PD-L1 inhibitors in patients with metastatic breast cancer. RECENT FINDINGS PD-L1 expression is heterogeneous across primary breast cancers, and is generally associated with the presence of tumor-infiltrating lymphocytes and the presence of poor-prognosis features such as high grade, and aggressive molecular subtypes (triple-negative (TN), basal, HER2-enriched). Early phase clinical trials using PD-1 or PD-L1 inhibitors alone or in combination have shown objective tumor responses and durable long-term disease control, in heavily pre-treated patients, notably in the TN subtype. Blockade of PD-1 or PD-L1 shows impressive antitumor activity in some subsets of breast cancer patients. Many clinical trials are ongoing in the metastatic and neoadjuvant setting, alone and in combination with chemotherapy, targeted therapy, radiotherapy, and/or other immune therapy. The identification of biomarkers predictive for a clinical benefit is warranted.
Collapse
Affiliation(s)
- François Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Ste-Marguerite, 13009, Marseille, France. .,Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS U7258, INSERM U1068, Marseille, France. .,Faculty of Medicine, Aix-Marseille University, Marseille, France.
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Ste-Marguerite, 13009, Marseille, France.,Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS U7258, INSERM U1068, Marseille, France.,Faculty of Medicine, Aix-Marseille University, Marseille, France
| |
Collapse
|
1054
|
Effect of neoadjuvant chemotherapy on tumor-infiltrating lymphocytes and PD-L1 expression in breast cancer and its clinical significance. Breast Cancer Res 2017; 19:91. [PMID: 28784153 PMCID: PMC5547502 DOI: 10.1186/s13058-017-0884-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022] Open
Abstract
Background The effects of neoadjuvant chemotherapy on immune markers remain largely unknown. The specific aim of this study was to assess stromal tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) protein expression in a cohort of breast cancer patients treated with neoadjuvant chemotherapy. Methods Using quantitative immunofluorescence, we investigated stromal TILs and PD-L1 protein expression in pre-treatment and residual breast cancer tissue from a Yale Cancer Center patient cohort of 58 patients diagnosed with breast cancer from 2003 to 2009 and treated with neoadjuvant chemotherapy. We compared the TIL count and PD-L1 status in paired pre-treatment and residual cancer tissues and correlated changes and baseline levels with survival. Results Of the 58 patients, 46 (79.3%) had hormone-positive and 34 (58.6%) had node-positive breast cancer. Eighty-six percent of residual cancer tissues had TIL infiltration and 17% had PD-L1 expression. There was a trend for higher TIL counts in postchemotherapy compared to prechemotherapy samples (p = 0.09). Increase in TIL count was associated with longer 5-year recurrence-free survival (p = 0.02, HR = 3.9, 95% CI = 1.179–15.39). PD-L1 expression (both stromal and tumor cells) was significantly lower in post-treatment samples (p = 0.001). Change in PD-L1 expression after therapy or TILs and PD-L1 expression in the posttreatment samples did not correlate with survival. Conclusions Increase in stromal TILs in residual cancer compared to pretreatment tissue is associated with improved recurrence-free survival. Despite a trend for increasing TIL counts, PD-L1 expression decreased in residual disease compared to pretreatment samples. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0884-8) contains supplementary material, which is available to authorized users.
Collapse
|
1055
|
Betof AS, Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K, Rubinstein SM, Flaherty KT, Lawrence DP, Johnson DB, Sullivan RJ. Impact of Age on Outcomes with Immunotherapy for Patients with Melanoma. Oncologist 2017; 22:963-971. [PMID: 28476944 PMCID: PMC5553960 DOI: 10.1634/theoncologist.2016-0450] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 03/03/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Monoclonal antibodies (mAb) targeting PD-1/PD-L1 have revolutionized melanoma treatment, yet data regarding effectiveness and tolerability across age groups is limited. We sought to determine the impact of age on overall survival (OS), progression-free survival (PFS), and rates of immune-mediated toxicities in patients treated with anti-PD-1/anti-PD-L1 mAb at two academic medical centers. METHODS We retrospectively collected data on all patients with metastatic melanoma treated with anti-PD-1/PD-L1 mAb between May 2009 and April 2015. We used Kaplan-Meier and Cox regression analyses to assess OS and PFS and identify factors associated with these outcomes. We also compared rates of autoimmune toxicity across age groups. RESULTS Of 254 patients, 57 (22.4%) were <50 years old, 85 (33.5%) were age 50-64, 65 (25.6%) were age 65-74, and 47 (18.5%) were ≥75 years. Across age groups, no differences existed in median OS (age <50: 22.9 months, age 50-64: 25.3 months, age 65-74: 22.0 months, age ≥75: 24.3 months) or PFS (age <50: 4.1 months, age 50-64: 6.5 months, age 65-74: 5.4 months, age ≥75: 7.9 months). The presence of liver metastases and elevated pre-treatment lactate dehydrogenase (LDH) were associated with reduced OS. Presence of liver metastasis, pretreatment LDH, BRAF mutation, and type of melanoma correlated with PFS. Overall, 110 patients (43.3%) experienced immune-mediated toxicities; 25 (9.8%) had colitis and 26 (10.2%) had endocrine toxicity. Rates of colitis, hepatitis, and pneumonitis did not differ across age groups. CONCLUSION We demonstrated that patients could safely tolerate anti-PD1/PDL-1 mAb therapy and achieve similar outcomes regardless of their age. IMPLICATIONS FOR PRACTICE Immunotherapy has revolutionized treatment for patients with metastatic melanoma, yet data are lacking regarding the effectiveness and tolerability of these treatments for older patients. In this study, we demonstrated that patients with melanoma safely tolerate immunotherapy and achieve similar outcomes regardless of their age. Specifically, we utilized data from two academic cancer centers and found no significant difference in overall survival, progression free survival, or immune-related toxicities, other than arthritis, across age groups. As the population ages, studies such as this will become critical to help us understand how best to treat older adults with cancer.
Collapse
Affiliation(s)
- Allison S Betof
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan D Nipp
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Anita Giobbie-Hurder
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Krista Rubin
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | | | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Donald P Lawrence
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | | | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
1056
|
Curigliano G, Burstein HJ, Winer EP, Gnant M, Dubsky P, Loibl S, Colleoni M, Regan MM, Piccart-Gebhart M, Senn HJ, Thürlimann B, André F, Baselga J, Bergh J, Bonnefoi H, Brucker SY, Cardoso F, Carey L, Ciruelos E, Cuzick J, Denkert C, Di Leo A, Ejlertsen B, Francis P, Galimberti V, Garber J, Gulluoglu B, Goodwin P, Harbeck N, Hayes DF, Huang CS, Huober J, Khaled H, Jassem J, Jiang Z, Karlsson P, Morrow M, Orecchia R, Osborne KC, Pagani O, Partridge AH, Pritchard K, Ro J, Rutgers EJT, Sedlmayer F, Semiglazov V, Shao Z, Smith I, Toi M, Tutt A, Viale G, Watanabe T, Whelan TJ, Xu B. De-escalating and escalating treatments for early-stage breast cancer: the St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol 2017; 28:1700-1712. [PMID: 28838210 PMCID: PMC6246241 DOI: 10.1093/annonc/mdx308] [Citation(s) in RCA: 777] [Impact Index Per Article: 97.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The 15th St. Gallen International Breast Cancer Conference 2017 in Vienna, Austria reviewed substantial new evidence on loco-regional and systemic therapies for early breast cancer. Treatments were assessed in light of their intensity, duration and side-effects, seeking where appropriate to escalate or de-escalate therapies based on likely benefits as predicted by tumor stage and tumor biology. The Panel favored several interventions that may reduce surgical morbidity, including acceptance of 2 mm margins for DCIS, the resection of residual cancer (but not baseline extent of cancer) in women undergoing neoadjuvant therapy, acceptance of sentinel node biopsy following neoadjuvant treatment of many patients, and the preference for neoadjuvant therapy in HER2 positive and triple-negative, stage II and III breast cancer. The Panel favored escalating radiation therapy with regional nodal irradiation in high-risk patients, while encouraging omission of boost in low-risk patients. The Panel endorsed gene expression signatures that permit avoidance of chemotherapy in many patients with ER positive breast cancer. For women with higher risk tumors, the Panel escalated recommendations for adjuvant endocrine treatment to include ovarian suppression in premenopausal women, and extended therapy for postmenopausal women. However, low-risk patients can avoid these treatments. Finally, the Panel recommended bisphosphonate use in postmenopausal women to prevent breast cancer recurrence. The Panel recognized that recommendations are not intended for all patients, but rather to address the clinical needs of the majority of common presentations. Individualization of adjuvant therapy means adjusting to the tumor characteristics, patient comorbidities and preferences, and managing constraints of treatment cost and access that may affect care in both the developed and developing world.
Collapse
Affiliation(s)
- G Curigliano
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | - H J Burstein
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - E P Winer
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M Gnant
- Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - P Dubsky
- Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
- Klinik St. Anna, Luzern, Switzerland
| | - S Loibl
- German Breast Group, Neu-Isenburg, Germany
| | - M Colleoni
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | - M M Regan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M Piccart-Gebhart
- Department of Medical Oncology, Institut Jules Bordet, UniversitÕ Libre de Bruxelles, Brussels, Belgium
| | - H-J Senn
- Tumor and Breast Center ZeTuP, St. Gallen
| | - B Thürlimann
- Breast Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - F André
- Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - J Baselga
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - J Bergh
- Karolinska Institute and University Hospital, Stockholm, Sweden
| | - H Bonnefoi
- University of Bordeaux, Bordeaux, France
| | - S Y Brucker
- Universitäts-Frauenklinik Tübingen, Tübingen, Germany
| | - F Cardoso
- Champalimaud Cancer Centre, Lisbon, Portugal
| | - L Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | - E Ciruelos
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - J Cuzick
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - C Denkert
- Institut für Pathologie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - A Di Leo
- Azienda Usl Toscana Centro, Prato, Italy
| | | | - P Francis
- Peter McCallum Cancer Centre, Melbourne, Australia
| | - V Galimberti
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | - J Garber
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - B Gulluoglu
- Marmara University School of Medicine, Istanbul, Turkey
| | - P Goodwin
- University of Toronto, Mount Sinai Hospital, Toronto, Canada
| | - N Harbeck
- University of Munich, München, Germany
| | - D F Hayes
- Comprehensive Cancer Center, University of Michigan, Ann-Arbor, USA
| | - C-S Huang
- National Taiwan University Hospital, Taipei, Taiwan
| | | | - H Khaled
- The National Cancer Institute, Cairo University, Cairo, Egypt
| | - J Jassem
- Medical University of Gdansk, Gdansk, Poland
| | - Z Jiang
- Hospital Affiliated to Military Medical Science, Beijing, China
| | - P Karlsson
- Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrensky University Hospital, Gothenburg, Sweden
| | - M Morrow
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - R Orecchia
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | | | - O Pagani
- Institute of Oncology Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - A H Partridge
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - K Pritchard
- Sunnybrook Odette Cancer Center, University of Toronto, Toronto, Canada
| | - J Ro
- National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - E J T Rutgers
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - F Sedlmayer
- LKH Salzburg, Paracelsus Medical University Clinics, Salzburg, Austria
| | - V Semiglazov
- N.N. Petrov Research Institute of Oncology, St. Petersburg, Russian Federation
| | - Z Shao
- Fudan University Cancer Hospital, Shanghai, China
| | - I Smith
- The Royal Marsden, Sutton, Surrey, UK
| | - M Toi
- Graduate School of Medicine Kyoto University, Sakyo-ku, Kyoto City, Japan
| | - A Tutt
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
| | - G Viale
- University of Milan, Milan, Italy
- Istituto Europeo di Oncologia, Milan, Italy
| | - T Watanabe
- Hamamatsu Oncology Center, Hamamatsu, Japan
| | | | - B Xu
- National Cancer Center, Chaoyang District, Beijing, China
| |
Collapse
|
1057
|
Wein L, Loi S. Mechanisms of resistance of chemotherapy in early-stage triple negative breast cancer (TNBC). Breast 2017; 34 Suppl 1:S27-S30. [DOI: 10.1016/j.breast.2017.06.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
1058
|
Turajlic S, Litchfield K, Xu H, Rosenthal R, McGranahan N, Reading JL, Wong YNS, Rowan A, Kanu N, Al Bakir M, Chambers T, Salgado R, Savas P, Loi S, Birkbak NJ, Sansregret L, Gore M, Larkin J, Quezada SA, Swanton C. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol 2017; 18:1009-1021. [PMID: 28694034 DOI: 10.1016/s1470-2045(17)30516-8] [Citation(s) in RCA: 695] [Impact Index Per Article: 86.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND The focus of tumour-specific antigen analyses has been on single nucleotide variants (SNVs), with the contribution of small insertions and deletions (indels) less well characterised. We investigated whether the frameshift nature of indel mutations, which create novel open reading frames and a large quantity of mutagenic peptides highly distinct from self, might contribute to the immunogenic phenotype. METHODS We analysed whole-exome sequencing data from 5777 solid tumours, spanning 19 cancer types from The Cancer Genome Atlas. We compared the proportion and number of indels across the cohort, with a subset of results replicated in two independent datasets. We assessed in-silico tumour-specific neoantigen predictions by mutation type with pan-cancer analysis, together with RNAseq profiling in renal clear cell carcinoma cases (n=392), to compare immune gene expression across patient subgroups. Associations between indel burden and treatment response were assessed across four checkpoint inhibitor datasets. FINDINGS We observed renal cell carcinomas to have the highest proportion (0·12) and number of indel mutations across the pan-cancer cohort (p<2·2 × 10-16), more than double the median proportion of indel mutations in all other cancer types examined. Analysis of tumour-specific neoantigens showed that enrichment of indel mutations for high-affinity binders was three times that of non-synonymous SNV mutations. Furthermore, neoantigens derived from indel mutations were nine times enriched for mutant specific binding, as compared with non-synonymous SNV derived neoantigens. Immune gene expression analysis in the renal clear cell carcinoma cohort showed that the presence of mutant-specific neoantigens was associated with upregulation of antigen presentation genes, which correlated (r=0·78) with T-cell activation as measured by CD8-positive expression. Finally, analysis of checkpoint inhibitor response data revealed frameshift indel count to be significantly associated with checkpoint inhibitor response across three separate melanoma cohorts (p=4·7 × 10-4). INTERPRETATION Renal cell carcinomas have the highest pan-cancer proportion and number of indel mutations. Evidence suggests indels are a highly immunogenic mutational class, which can trigger an increased abundance of neoantigens and greater mutant-binding specificity. FUNDING Cancer Research UK, UK National Institute for Health Research (NIHR) at the Royal Marsden Hospital National Health Service Foundation Trust, Institute of Cancer Research and University College London Hospitals Biomedical Research Centres, the UK Medical Research Council, the Rosetrees Trust, Novo Nordisk Foundation, the Prostate Cancer Foundation, the Breast Cancer Research Foundation, the European Research Council.
Collapse
Affiliation(s)
- Samra Turajlic
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Renal and Skin Units, The Royal Marsden Hospital National Health Service Foundation Trust, London, UK
| | - Kevin Litchfield
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Hang Xu
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Rachel Rosenthal
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK
| | - Nicholas McGranahan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK
| | - James L Reading
- Cancer Immunology Unit, Research Department of Haematology, London, UK
| | - Yien Ning S Wong
- Cancer Immunology Unit, Research Department of Haematology, London, UK
| | - Andrew Rowan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK
| | - Maise Al Bakir
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Tim Chambers
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Roberto Salgado
- Department of Pathology, Gasthuiszusters, Antwerp, Belgium; Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Savas
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Sherene Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Nicolai J Birkbak
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Laurent Sansregret
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Martin Gore
- Renal and Skin Units, The Royal Marsden Hospital National Health Service Foundation Trust, London, UK
| | - James Larkin
- Renal and Skin Units, The Royal Marsden Hospital National Health Service Foundation Trust, London, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, Research Department of Haematology, London, UK
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK; Department of Medical Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
1059
|
Sikandar B, Qureshi MA, Naseem S, Khan S, Mirza T. Increased Tumour Infiltration of CD4+ and CD8+ T-Lymphocytes in Patients with Triple Negative Breast Cancer Suggests Susceptibility to Immune Therapy. Asian Pac J Cancer Prev 2017; 18:1827-1832. [PMID: 28749113 PMCID: PMC5648386 DOI: 10.22034/apjcp.2017.18.7.1827] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background: Patients with triple negative breast cancer (TNBC) have limited therapeutic options, largely because the complex tumour environment is not well-characterized. These patients are potential, but largely un-fathomed, candidates for immunotherapy. It is therefore highly relevant to characterize leukocyte complexity in TNBCs. Objective: To investigate leukocyte complexity in tumour environment of patients with TNBCs. Materials and methods: A total of 104 consecutive breast cancer patients undergoing mastectomy were recruited in the study after ethical approval. Clinico-pathological parameters were recorded and H and E staining was performed to investigate tumour morphology. Receptor status was investigated using antibodies against ER, PgR and Her-2, and patients were classified as having TNBC or non-TNBC tumours (including Luminal A, Luminal B and Her2 overexpressing tumours). Immune-cell infiltration was investigated using special stains and antibodies: α-CD3 (T-lymphocytes), α-CD20 (B-lymphocytes), α-CD4 (helper T-lymphocytes) and α-CD8 (cytotoxic T-lymphocytes). Immune cell densities were quantified as cell/mm2 using the CAP guidelines. Results: Of the 104 breast cancer patients investigated, a total of 27 (26%) had TNBC and 77(74%) non-TNBC. Patients with TNBC showed significantly increased tumour infiltration of lymphocytes (T and B-lymphocytes) compared to the patients with non-TNBC, while myelocytic infiltration was not significantly different in the two groups. Within the TNBC group, infiltration of T-lymphocytes (equal densities of CD4+ and CD8+ T-lymphocytes) was significantly higher compared to B-lymphocytes. Conclusion: Patients with TNBC show increased lymphocytic infiltration (more T-lymphocytes compared to B-lymphocytes). This suggests higher immunogenicity of TNBCs and may indicate a higher responsiveness of these cancers to immunotherapy.
Collapse
Affiliation(s)
- Bushra Sikandar
- Department of Pathology, Dow International Medical College, Dow Diagnostic Research and Reference Laboratory, Dow
University of Health Sciences Karachi, Pakistan.
| | | | | | | | | |
Collapse
|
1060
|
Donovan MJ, Cordon-Cardo C. Implementation of a Precision Pathology Program Focused on Oncology-Based Prognostic and Predictive Outcomes. Mol Diagn Ther 2017; 21:115-123. [PMID: 28000172 DOI: 10.1007/s40291-016-0249-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Personalized or precision medicine as a diagnostic and therapeutic paradigm was introduced some 10-15 years ago, with the advent of biomarker discovery as a mechanism for identifying prognostic and predictive attributes associated with treatment indication and outcome. While the concept is not new, the successful development and implementation of novel 'companion diagnostics', especially in oncology, continues to represent a significant challenge and is currently at the forefront of smart trial design and therapeutic choice. The ability to determine patient selection for a specific therapy has broad implications including better chances for a positive outcome, limited exposure to potentially toxic drugs and improved health economics. Importantly, a significant step in this paradigm is the role of predictive pathology or the accurate assessment of morphology at the microscopic level. In breast cancer, this has been most useful where histologic attributes such as the classification of tubular and cribriform carcinoma dictates surgery while neoadjuvant studies suggest that patients with lobular carcinoma are not likely to benefit from chemotherapy. The next level of 'personalized pathology' at the tissue-cellular level is the use of 'protein biomarker panels' to classify the disease process and ultimately drive tumor characterization and treatment. The following review article will focus on the evolution of predictive pathology from a subjective, 'opinion-based' approach to a quantitative science. In addition, we will discuss the individual components of the precise pathology platform including advanced image analysis, biomarker quantitation with mathematical modeling and the integration with fluid-based (i.e. blood, urine) analytics as drivers of next generation precise patient phenotyping.
Collapse
|
1061
|
Trapani D, Esposito A, Criscitiello C, Mazzarella L, Locatelli M, Minchella I, Minucci S, Curigliano G. Entinostat for the treatment of breast cancer. Expert Opin Investig Drugs 2017; 26:965-971. [DOI: 10.1080/13543784.2017.1353077] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Dario Trapani
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| | - Angela Esposito
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| | - Luca Mazzarella
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| | - Marzia Locatelli
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| | - Ida Minchella
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| | - Saverio Minucci
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development, European Institute of Oncology, Milan, Italy
| |
Collapse
|
1062
|
Gallo S, Sangiolo D, Carnevale Schianca F, Aglietta M, Montemurro F. Treating breast cancer with cell-based approaches: an overview. Expert Opin Biol Ther 2017; 17:1255-1264. [PMID: 28728493 DOI: 10.1080/14712598.2017.1356816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Breast cancer is the most common malignancy in women. Despite there being considerable progress in the treatment of this disease, metastatic dissemination is still considered an incurable condition at the present time, causing 500,000 deaths worldwide every year. Although most of the research efforts have been focused on pharmacological approaches, over the last three decades, the use of bone marrow and peripheral blood-derived cell therapy approaches have been attempted and developed. Areas covered: This review will briefly address cell therapy for breast cancer, including autologous stem cell transplantations for overcoming the myelosuppressive effects of high-dose chemotherapy, allogeneic stem cell transplants and adoptive immunotherapy using bone-marrow derived T-cells. Expert opinion: The treatment of breast cancer using bone marrow or peripheral-blood derived cells has evolved from a supportive care approach to allow dose escalation of conventional chemotherapy to a therapeutic strategy aimed at eliciting immune cell mediated anticancer immunity. This latter principle has led to the development of adoptive immunotherapies, either with 'natural' or genetically engineered effectors, which are being intensively investigated for their great potential against several solid tumors, including breast cancer.
Collapse
Affiliation(s)
- Susanna Gallo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| | - Dario Sangiolo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | | | - Massimo Aglietta
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | - Filippo Montemurro
- c Investigative Clinical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| |
Collapse
|
1063
|
Atypical responses in patients with advanced melanoma, lung cancer, renal-cell carcinoma and other solid tumors treated with anti-PD-1 drugs: A systematic review. Cancer Treat Rev 2017; 59:71-78. [PMID: 28756306 DOI: 10.1016/j.ctrv.2017.07.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 12/16/2022]
Abstract
Anti-programmed death receptor 1 (PD-1) drugs nivolumab and pembrolizumab were recently approved for the treatment of advanced melanoma and other solid tumors. Atypical patterns of response (i.e. tumor shrinkage or stabilization after initial progression) were observed in about 10% of metastatic melanoma patients treated with anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) drug ipilimumab and were associated with improved survival; however, the rate of atypical response patterns to anti-PD-1 therapy is not clear. An electronic search was performed to identify clinical trials evaluating response to anti-PD-1 therapy with nivolumab and pembrolizumab in patients with advanced solid tumors. Thirty-eight studies were included in our analysis for a total of 7069 patients with advanced cancer treated with anti-PD-1 therapy. Responses were evaluated by unconventional response criteria in 19 trials and were observed for all cancer types but tumors with mismatch-repair deficiency and head and neck squamous cell carcinoma. Overall, 151 atypical responses were observed in 2400 patients (6%) evaluated by unconventional response criteria. The results of our systematic review highlight the clinical relevance of unconventional responses to anti-PD-1 therapy and support further investigation into the development of tools that may assist evaluation of the antitumor activity of immunotherapy.
Collapse
|
1064
|
Sai J, Owens P, Novitskiy SV, Hawkins OE, Vilgelm AE, Yang J, Sobolik T, Lavender N, Johnson AC, McClain C, Ayers GD, Kelley MC, Sanders M, Mayer IA, Moses HL, Boothby M, Richmond A. PI3K Inhibition Reduces Mammary Tumor Growth and Facilitates Antitumor Immunity and Anti-PD1 Responses. Clin Cancer Res 2017; 23:3371-3384. [PMID: 28003307 PMCID: PMC5479746 DOI: 10.1158/1078-0432.ccr-16-2142] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/23/2016] [Accepted: 12/14/2016] [Indexed: 12/13/2022]
Abstract
Purpose: Metastatic breast cancers continue to elude current therapeutic strategies, including those utilizing PI3K inhibitors. Given the prominent role of PI3Kα,β in tumor growth and PI3Kγ,δ in immune cell function, we sought to determine whether PI3K inhibition altered antitumor immunity.Experimental Design: The effect of PI3K inhibition on tumor growth, metastasis, and antitumor immune response was characterized in mouse models utilizing orthotopic implants of 4T1 or PyMT mammary tumors into syngeneic or PI3Kγ-null mice, and patient-derived breast cancer xenografts in humanized mice. Tumor-infiltrating leukocytes were characterized by IHC and FACS analysis in BKM120 (30 mg/kg, every day) or vehicle-treated mice and PI3Kγnull versus PI3KγWT mice. On the basis of the finding that PI3K inhibition resulted in a more inflammatory tumor leukocyte infiltrate, the therapeutic efficacy of BKM120 (30 mg/kg, every day) and anti-PD1 (100 μg, twice weekly) was evaluated in PyMT tumor-bearing mice.Results: Our findings show that PI3K activity facilitates tumor growth and surprisingly restrains tumor immune surveillance. These activities could be partially suppressed by BKM120 or by genetic deletion of PI3Kγ in the host. The antitumor effect of PI3Kγ loss in host, but not tumor, was partially reversed by CD8+ T-cell depletion. Treatment with therapeutic doses of both BKM120 and antibody to PD-1 resulted in consistent inhibition of tumor growth compared with either agent alone.Conclusions: PI3K inhibition slows tumor growth, enhances antitumor immunity, and heightens susceptibility to immune checkpoint inhibitors. We propose that combining PI3K inhibition with anti-PD1 may be a viable therapeutic approach for triple-negative breast cancer. Clin Cancer Res; 23(13); 3371-84. ©2016 AACR.
Collapse
Affiliation(s)
- Jiqing Sai
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Philip Owens
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | | | - Oriana E Hawkins
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Anna E Vilgelm
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Jinming Yang
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Tammy Sobolik
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Nicole Lavender
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Andrew C Johnson
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Colt McClain
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | - Mark C Kelley
- Department of Surgical Oncology, Vanderbilt University, Nashville, Tennessee
| | - Melinda Sanders
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Ingrid A Mayer
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Harold L Moses
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Mark Boothby
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee.
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
1065
|
Corona SP, Sobhani N, Ianza A, Roviello G, Mustacchi G, Bortul M, Zanconati F, Generali D. Advances in systemic therapy for metastatic breast cancer: future perspectives. Med Oncol 2017; 34:119. [PMID: 28526922 DOI: 10.1007/s12032-017-0975-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 04/29/2017] [Indexed: 12/16/2022]
Abstract
Breast cancer (BC) is the most common cancer in women worldwide. One in eight women will develop the disease in her lifetime. Notwithstanding the incredible progress made in this field, BC still represents the second most common cause of cancer-related death in women. Targeted drugs have revolutionised breast cancer treatment and improved the prognosis as well as the life expectancy of millions of women. However, the phenomenon of primary and secondary pharmacological resistance is becoming increasingly evident, limiting the efficacy of these agents and calling for a better in-depth knowledge and understanding of the biology as well as the biochemical crosstalk underlying the disease. The advent of laboratory technologies in the clinical setting such as the routine use of next generation sequencing has allowed identification of new genetic alterations as well as providing a precise picture of the molecular landscapes of each tumour. Consequently, new specific therapeutic approaches are becoming available to minimise or delay the occurrence of resistance. In this review, we analyse the latest research and news from the clinical development side for each BC subtype.
Collapse
Affiliation(s)
- S P Corona
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - N Sobhani
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - A Ianza
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - G Roviello
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - G Mustacchi
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - M Bortul
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - F Zanconati
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - D Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy.
| |
Collapse
|
1066
|
Chen S, Wang RX, Liu Y, Yang WT, Shao ZM. PD-L1 expression of the residual tumor serves as a prognostic marker in local advanced breast cancer after neoadjuvant chemotherapy. Int J Cancer 2017; 140:1384-1395. [PMID: 27925176 DOI: 10.1002/ijc.30552] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/23/2016] [Indexed: 12/22/2022]
Abstract
This study sought to investigate the prevalence of programmed death ligand 1 (PD-L1) and its prognostic value in patients with residual tumors after neoadjuvant chemotherapy (NCT) for locally advanced breast cancer. A total of 309 patients considered as non-pathological complete responders (non-pCR) after NCT followed by mastectomy were selected. The expression of PD-L1 and tumor-infiltrating lymphocytes (TILs) in residual breast cancer cells was assessed by immunohistochemistry in surgical specimens. The median density was used to classify PD-L1 expression from low to high. The prognostic value of various clinicopathological factors was evaluated. The expression of PD-L1 was more commonly observed in patients with low levels of total TILs (p < 0.001), high levels of FOXP3+ TILs (p < 0.001) and low levels of CD8+ TILs (p < 0.001). This served as an independent prognostic factor for both relapse-free survival (Hazard ratio = 1.824, p = 0.013) and overall survival (OS) (Hazard ratio = 2.585, p = 0.001). High expression of PD-L1 was correlated to worse survival, which is most significantly observed in triple-negative patients. Patients classified as PD-L1-high/CD8-low exhibited relatively unfavorable survival, whereas patients with either low expression of PD-L1 or high expression of CD8 had similar outcomes. PD-L1 expression in residual tumor can be used as a prognostic marker in non-pCR patients after receiving NCT for breast cancer, which highlights the importance of immune evasion in the therapeutic vulnerability of chemoresistant cancer cells as well as the potential of anti-PD-L1 treatments in non-pCR responders.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ruo-Xi Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yin Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Wen-Tao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Department of Pathology, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institutes of Biomedical Science, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
1067
|
Abstract
Anti-tumor immunity is a new line of research for the treatment of patients with solid tumors. In this field, negative regulators of the immune system called immune checkpoints play a key role in limiting antitumor immunologic responses. For this reason, immune checkpoint-inhibiting agents, such as those directed against cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death-1 receptor (PD1) and its ligand PD-L1, have been developed as antitumor drugs, producing interesting results in preclinical and clinical studies. We present an updated review of the biological background and clinical development of immune checkpoint inhibitors in colorectal cancer (CRC). Early trial results on PD1 and PD-L1 blockade appear promising, especially in CRC patients with microsatellite instability (MSI). Clinical trials are ongoing to confirm these preliminary results, evaluate combination strategies and identify biomarkers to predict which patients are most likely to benefit from, or show resistance to, the effects of checkpoint inhibition.
Collapse
|
1068
|
Denkert C, Liedtke C, Tutt A, von Minckwitz G. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet 2017; 389:2430-2442. [PMID: 27939063 DOI: 10.1016/s0140-6736(16)32454-0] [Citation(s) in RCA: 612] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer is a heterogeneous disease and specific therapies have not been available for a long time. Therefore, conventional chemotherapy is still considered the clinical state of the art. Different subgroups of triple-negative breast cancer have been identified on the basis of protein expression, mRNA signatures, and genomic alterations. Important elements of triple-negative breast cancer biology include high proliferative activity, an increased immunological infiltrate, a basal-like and a mesenchymal phenotype, and deficiency in homologous recombination, which is in part associated with loss of BRCA1 or BRCA2 function. A minority of triple-negative tumours express luminal markers, such as androgen receptors, and have a lower proliferative activity. These biological subgroups are overlapping and currently cannot be combined into a unified model of triple-negative breast cancer biology. Nevertheless, the molecular analysis of this disease has identified potential options for targeted therapeutic intervention. This has led to promising clinical strategies, including modified chemotherapy approaches targeting the DNA damage response, angiogenesis inhibitors, immune checkpoint inhibitors, or even anti-androgens, all of which are being evaluated in phase 1-3 clinical studies. This Series paper focuses on the most relevant clinical questions, summarises the results of recent clinical trials, and gives an overview of ongoing studies and trial concepts that will lead to a more refined therapy for this tumour type.
Collapse
Affiliation(s)
- Carsten Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Germany.
| | | | - Andrew Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research and Breast Cancer Now Research Unit, King's College, London, UK
| | | |
Collapse
|
1069
|
Bettaieb A, Paul C, Plenchette S, Shan J, Chouchane L, Ghiringhelli F. Precision medicine in breast cancer: reality or utopia? J Transl Med 2017. [PMID: 28623955 PMCID: PMC5474301 DOI: 10.1186/s12967-017-1239-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Many cancers, including breast cancer, have demonstrated prognosis and support advantages thanks to the discovery of targeted therapies. The advent of these new approaches marked the rise of precision medicine, which leads to improve the diagnosis, prognosis and treatment of cancer. Precision medicine takes into account the molecular and biological specificities of the patient and their tumors that will influence the treatment determined by physicians. This new era of medicine is accessible through molecular genetics platforms, the development of high-speed sequencers and means of analysis of these data. Despite the spectacular results in the treatment of cancers including breast cancer, described in this review, not all patients however can benefit from this new strategy. This seems to be related to the many genetic mutations, which may be different from one patient to another or within the same patient. It comes to give new impetus to the research—both from a technological and biological point of view—to make the hope of precision medicine accessible to all.
Collapse
Affiliation(s)
- Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France. .,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France. .,Immunology and Immunotherapy of Cancer Laboratory, EA7269, Université de Bourgogne, EPHE 7 Bd Jeanne d'Arc, 21079, Dijon, France.
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France
| | - Stéphanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France
| | - Jingxuan Shan
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Lotfi Chouchane
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - 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 1231 Inserm-Université de Bourgogne Franche Comté, UFR des Sciences de Santé, 21000, Dijon, France.,Université de Bourgogne, 21000, Dijon, France
| |
Collapse
|
1070
|
Abstract
Despite great advances in early detection, as well as surgical resection of breast tumours, breast cancer remains the deadliest cancer for women worldwide. Moreover, its incidence is without pair, accounting for twice as many new cancer cases as the second most prevalent cancer, colorectal carcinoma. There is therefore a strong need for new therapeutic approaches to breast cancers. Immunotherapies are novel treatment modalities which aim to use immune mediators to attack cancerous cells. Recent clinical results show that these may not only mediate tumour regressions but also cures in some cases. In this review, we discuss the relevance of the immune system in the development of new carcinomas, as well as its importance in mediating cancer regression. We also dissect the known different approaches to harness the immune system to attack breast tumours. Namely, therapies using the passive transfer of either tumour-specific antibodies or cytotoxic cells have been researched and in some cases are already standard of care. Additionally, therapeutic vaccines and immune checkpoint blockade have recently demonstrated great therapeutic efficacy and have generated great excitement for the development of new treatments. Immunotherapies have the potential to generate tumour specific responses, as well as long-lasting remissions, which is why studying those approaches is crucial for the future of cancer medicine.
Collapse
Affiliation(s)
- Gwennaëlle C Monnot
- Department of Fundamental Oncology, Ludwig Cancer Research Center at University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Pedro Romero
- Department of Fundamental Oncology, Ludwig Cancer Research Center at University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| |
Collapse
|
1071
|
Nathan MR, Schmid P. The emerging world of breast cancer immunotherapy. Breast 2017; 37:200-206. [PMID: 28583398 DOI: 10.1016/j.breast.2017.05.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 05/24/2017] [Accepted: 05/25/2017] [Indexed: 01/21/2023] Open
Abstract
Over the last few years, the developments around cancer immunotherapy (CIT) have led to a paradigm shift in the treatment of many different cancers, in particular melanoma, renal, bladder and lung cancers with a remarkable impact on response rate and, most importantly, overall survival. Breast cancer is most commonly considered to be a 'non-inflamed' cancer and so this shift has been less marked within its treatment. However, some subsets of breast cancer, most notably triple negative breast cancer, are deemed to be more 'inflamed' and therefore may prove to be an appropriate cohort for CIT. This review looks back at the theory of the cancer immunity cycle and mechanism of action behind immune checkpoint inhibitors and goes on to explore their role within the various subtypes of breast cancer. It looks at the first trials performed using CIT monotherapy which demonstrated that breast cancer could respond to CIT with a small population reaping considerable benefit. It then examines the continuing body of work being undertaken to explore CIT in combination with chemotherapy to try to increase the proportion of patients who might reap the considerable rewards on offer.
Collapse
|
1072
|
Rubovszky G, Horváth Z. Recent Advances in the Neoadjuvant Treatment of Breast Cancer. J Breast Cancer 2017; 20:119-131. [PMID: 28690648 PMCID: PMC5500395 DOI: 10.4048/jbc.2017.20.2.119] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/22/2017] [Indexed: 12/13/2022] Open
Abstract
In the last few decades, neoadjuvant therapy for breast cancer has gained considerable therapeutic importance. Despite extensive clinical investigations, it has not yet been clarified whether neoadjuvant therapy would result in improved survival in comparison with the standard adjuvant setting in any subgroups of patients with breast cancer. Chemotherapy is especially effective in the treatment of endocrine insensitive tumors, and such ther-apeutic benefit can be assumed for patients with triple-negative, or hormone receptor-negative and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, dose escalation, modification of the therapeutic regimens according to early tumor response, as well as the optimal sequence of administration are still matters of debate. There is a current debate between clinical experts regarding the concomitant and sequential administration of carboplatin and capecitabine, respectively, as part of the standard neoadjuvant treatment, as well as the use of bevacizumab, as part of the preoperative treatment. In case of HER2 positive tumors, an anti-HER2 agent can be administered as part of the preoperative treatment, and according to preliminary clinical data, dual HER2 blockade can also be reasonable. Further, chemotherapy-free regimens can be justified in highly endocrine sensitive tumors, while immune modulating agents may also gain particular importance in the case of certain subtypes of breast cancer. Several small-molecule targeted therapies are under clinical investigation and are expected to provide new neoadjuvant treatment options. However, novel, more predictive biomarkers are required for further evaluation of the neoadjuvant therapies, as well as the effect of novel targeted agents intended to be incorporated into neoadjuvant therapy.
Collapse
Affiliation(s)
- Gábor Rubovszky
- Department of Medical Oncology and Clinical Pharmacology “B”, National Institute of Oncology, Budapest, Hungary
| | - Zsolt Horváth
- Faculty of Medicine, Institute of Oncology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
1073
|
Vonderheide RH, Domchek SM, Clark AS. Immunotherapy for Breast Cancer: What Are We Missing? Clin Cancer Res 2017; 23:2640-2646. [PMID: 28572258 PMCID: PMC5480967 DOI: 10.1158/1078-0432.ccr-16-2569] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 02/15/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
The recent demonstration of modest single-agent activity of programmed death-ligand 1 (PD-L1) and programmed death receptor-1 (PD-1) antibodies in patients with breast cancer has generated hope that breast cancer can be made amenable to immunotherapy. Depending on the subtype of breast cancer, it is now clear in both primary and metastatic disease that the extent of tumor-infiltrating T cells is not only prognostic for survival but predictive of response to nonimmune, standard therapies. Despite these findings, immune cytolytic activity in spontaneous breast tumors, the burden of nonsynonymous tumor mutations, and the predicted load of neoepitopes-factors linked to response to checkpoint blockade in other malignancies-are all relatively modest in breast cancer compared with melanoma or lung cancer. Thus, in breast cancer, combinations of immune agents with nonredundant mechanisms of action are high-priority strategies. For most breast cancers that exhibit relatively modest T-cell infiltration, major challenges include immune suppression in the tumor microenvironment as well as failed or suboptimal T-cell priming. Agents that trigger de novo T-cell responses may be critical for the successful development of cancer immunotherapy and immune prevention in breast cancer. Success may also require reaching beyond nonsynonymous mutations as the T-cell epitopes to target, especially as numerous unmutated proteins were validated as breast cancer-associated antigens in the pre-checkpoint era. A deeper understanding of the immunobiology of breast cancer will be critical for immunotherapy to become broadly relevant in this disease. Clin Cancer Res; 23(11); 2640-6. ©2017 AACRSee all articles in this CCR Focus section, "Breast Cancer Research: From Base Pairs to Populations."
Collapse
Affiliation(s)
- Robert H Vonderheide
- Abramson Cancer Center, Hematology-Oncology Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Susan M Domchek
- Abramson Cancer Center, Hematology-Oncology Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy S Clark
- Abramson Cancer Center, Hematology-Oncology Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
1074
|
Kourie HR, Awada G, Awada A. The second wave of immune checkpoint inhibitor tsunami: advance, challenges and perspectives. Immunotherapy 2017; 9:647-657. [DOI: 10.2217/imt-2017-0029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
After the first wave of the tsunami of immune checkpoint inhibitors, 2016 was marked by the second wave, revealed by numerous US FDA approvals, publications and abstracts in relation with these drugs in different cancers and settings. First, we reported all new indications of anti-CTLA4, anti-programmed cell death protein 1 and anti-PDL1 approved by the FDA, the positive clinical trials published and the abstracts reported at important scientific meetings during 2016. Then, we highlighted the updates on debatable issues related to checkpoint inhibitors, since the first wave published in a previous issue. We focused on the predictive biomarkers, combination therapies, tumor response patterns and efficacy in particular settings and the side effect management. Finally, the impact of checkpoint inhibitors development on the care management of cancer centers will be discussed.
Collapse
Affiliation(s)
- Hampig Raphael Kourie
- Department of Oncology, Faculty of medicine, Saint Joseph University, Beirut, Lebanon
- Medical Oncology clinic, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Gil Awada
- Internal medicine, Vrije Universiteit, Brussels, Belgium
| | - Ahmad Awada
- Medical Oncology clinic, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
1075
|
Muraro MG, Muenst S, Mele V, Quagliata L, Iezzi G, Tzankov A, Weber WP, Spagnoli GC, Soysal SD. Ex-vivo assessment of drug response on breast cancer primary tissue with preserved microenvironments. Oncoimmunology 2017; 6:e1331798. [PMID: 28811974 DOI: 10.1080/2162402x.2017.1331798] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022] Open
Abstract
Interaction between cancerous, non-transformed cells, and non-cellular components within the tumor microenvironment plays a key role in response to treatment. However, short-term culture or xenotransplantation of cancer specimens in immunodeficient animals results in dramatic modifications of the tumor microenvironment, thus preventing reliable assessment of compounds or biologicals of potential therapeutic relevance. We used a perfusion-based bioreactor developed for tissue engineering purposes to successfully maintain the tumor microenvironment of freshly excised breast cancer tissue obtained from 27 breast cancer patients and used this platform to test the therapeutic effect of antiestrogens as well as checkpoint-inhibitors on the cancer cells. Viability and functions of tumor and immune cells could be maintained for over 2 weeks in perfused bioreactors. Next generation sequencing authenticated cultured tissue specimens as closely matching the original clinical samples. Anti-estrogen treatment of cultured estrogen receptor positive breast cancer tissue as well as administration of pertuzumab to a Her2 positive breast cancer both had an anti-proliferative effect. Treatment with anti-programmed-death-Ligand (PD-L)-1 and anti-cytotoxic T lymphocyte-associated protein (CTLA)-4 antibodies lead to immune activation, evidenced by increased lymphocyte proliferation, increased expression of IFNγ, and decreased expression of IL10, accompanied by a massive cancer cell death in ex vivo triple negative breast cancer specimens. In the era of personalized medicine, the ex vivo culture of breast cancer tissue represents a promising approach for the pre-clinical evaluation of conventional and immune-mediated treatments and provides a platform for testing of innovative treatments.
Collapse
Affiliation(s)
- Manuele G Muraro
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Simone Muenst
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Valentina Mele
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Giandomenica Iezzi
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Walter P Weber
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Giulio C Spagnoli
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Savas D Soysal
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.,Department of Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
1076
|
Yam C, Mani SA, Moulder SL. Targeting the Molecular Subtypes of Triple Negative Breast Cancer: Understanding the Diversity to Progress the Field. Oncologist 2017; 22:1086-1093. [PMID: 28559413 DOI: 10.1634/theoncologist.2017-0095] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/12/2017] [Indexed: 12/17/2022] Open
Abstract
Triple negative breast cancers (TNBCs) represent 10%-20% of primary breast cancers, and despite having greater initial sensitivity to cytotoxic chemotherapy, patients with TNBCs have higher rates of distant metastasis and a poorer prognosis compared with patients with hormone receptor positive and/or human epidermal growth factor receptor 2 positive disease. TNBC has historically been treated as a single disease entity in targeted therapy trials, but advances in gene expression profiling and other molecular diagnostic techniques over the last decade have revealed considerable biologic heterogeneity within TNBCs, including subgroups with distinct, targetable aberrations. Such molecular heterogeneity explains, in part, the disappointing performance of targeted therapeutics in unselected TNBC. Here we discuss the history of gene expression profiling in breast cancer and its application in partitioning TNBCs into subtypes that may lead to more consistent therapeutic successes in this heterogeneous disease. IMPLICATIONS FOR PRACTICE Triple negative breast cancers (TNBCs) have historically been regarded as a single entity in clinical trial design. Over the last decade, molecular characterization has revealed much heterogeneity in TNBCs, explaining in part the lackluster performance of targeted therapeutics in TNBCs as a group. In this article, we review the history of the molecular classification of breast cancer based on gene expression profiling and discuss its role in TNBCs.
Collapse
Affiliation(s)
- Clinton Yam
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stacy L Moulder
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
1077
|
Hu ZI, Ho AY, McArthur HL. Combined Radiation Therapy and Immune Checkpoint Blockade Therapy for Breast Cancer. Int J Radiat Oncol Biol Phys 2017; 99:153-164. [PMID: 28816141 DOI: 10.1016/j.ijrobp.2017.05.029] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/02/2017] [Accepted: 05/18/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE Treatment with checkpoint inhibitors has shown durable responses in a number of solid tumors, including melanoma, lung, and renal cell carcinoma. However, most breast cancers are resistant to monotherapy with checkpoint inhibitors. Radiation therapy (RT) has been shown to have a number of immunostimulatory effects, including priming the immune system, recruiting immune cells to the tumor environment, and altering the immunosuppressive effects of the tumor microenvironment. RT therefore represents a promising adjuvant therapy to checkpoint blockade in breast cancer. METHODS AND MATERIALS We review the data from the checkpoint blockade studies on breast cancer reported to date, the mechanisms by which RT potentiates immune responses, the preclinical and clinical data of checkpoint blockade and RT combinations, and the landscape of current clinical trials of RT and immune checkpoint inhibitor combinations in breast cancer. RESULTS Clinical trials with checkpoint blockade therapy have demonstrated response rates of up to 19% in breast cancer, and many of the responses are durable. Preclinical data indicate that RT combined with checkpoint inhibition synergizes not only to enhance antitumor efficacy but also to induce responses outside of the radiation field. Thus multiple clinical trials are currently investigating the combination of checkpoint inhibition with RT. CONCLUSIONS The use of combination strategies that incorporate chemotherapy and/or local strategies such as RT may be needed to augment responses to immune therapy in breast cancer. Preclinical and clinical results show that RT in combination with checkpoint blockade may be a promising therapeutic option in breast cancer.
Collapse
Affiliation(s)
- Zishuo I Hu
- Icahn School of Medicine, Mount Sinai Health System, New York, New York.
| | - Alice Y Ho
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Heather L McArthur
- Breast Oncology Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
1078
|
Gonçalves A, Monneur A, Viens P, Bertucci F. The use of systemic therapies to prevent progression of inflammatory breast cancer: which targeted therapies to add on cytotoxic combinations? Expert Rev Anticancer Ther 2017; 17:593-606. [PMID: 28506194 DOI: 10.1080/14737140.2017.1330655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Inflammatory breast cancer is a rare but frequently fatal disease, essentially because of its high ability to develop distant metastases. Even though the prognosis of IBC was significantly improved by multimodal management, including the systematic use of cytotoxic-based induction, the prognosis remains largely dismal. Areas covered: This review presents the main achievements in the systemic treatment of IBC during the past 30 years. It focuses more specifically on recent results obtained with targeted therapies, including anti-HER2 and anti-angiogenic agents. Novel approaches under investigation are presented. Expert commentary: Current management of IBC is subtype-specific and the largest benefit has been achieved in HER2-positive disease. The identification of breakthrough therapeutic advances is eagerly awaited and will require the development of IBC-specific clinical trials. Future clinical investigations should not only aim to increase the pathological response rate but also to eradicate distant metastases, which ultimately lead to patient death.
Collapse
Affiliation(s)
- Anthony Gonçalves
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Audrey Monneur
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Patrice Viens
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - François Bertucci
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| |
Collapse
|
1079
|
Siniard RC, Harada S. Immunogenomics: using genomics to personalize cancer immunotherapy. Virchows Arch 2017; 471:209-219. [DOI: 10.1007/s00428-017-2140-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 01/06/2023]
|
1080
|
Yardley DA, Arrowsmith ER, Daniel BR, Eakle J, Brufsky A, Drosick DR, Kudrik F, Bosserman LD, Keaton MR, Goble SA, Bubis JA, Priego VM, Pendergrass K, Manalo Y, Bury M, Gravenor DS, Rodriguez GI, Inhorn RC, Young RR, Harwin WN, Silver C, Hainsworth JD, Burris HA. TITAN: phase III study of doxorubicin/cyclophosphamide followed by ixabepilone or paclitaxel in early-stage triple-negative breast cancer. Breast Cancer Res Treat 2017; 164:649-658. [PMID: 28508185 DOI: 10.1007/s10549-017-4285-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/05/2017] [Indexed: 11/26/2022]
Abstract
PURPOSE Ixabepilone is a microtubule stabilizer with activity in taxane-refractory metastatic breast cancer and low susceptibility to taxane-resistance mechanisms including multidrug-resistant phenotypes and high β-III tubulin expression. Since these resistance mechanisms are common in triple-negative breast cancer (TNBC), ixabepilone may have particular advantages in this patient population. This study evaluated the substitution of ixabepilone for paclitaxel following doxorubicin/cyclophosphamide (AC) in the adjuvant treatment of early-stage TNBC. METHODS Patients with operable TNBC were eligible following definitive breast surgery. Patients were randomized (1:1) to receive four cycles of AC followed by either four cycles (12 weeks) of ixabepilone or 12 weekly doses of paclitaxel. RESULTS 614 patients were randomized: 306 to AC/ixabepilone and 308 to AC/paclitaxel. At a median follow-up of 48 months, 59 patients had relapsed (AC/ixabepilone, 29; AC/paclitaxel, 30). The median time from diagnosis to relapse was 20.8 months. The 5-year disease-free survival (DFS) rates of the two groups were similar [HR 0.92; ixabepilone 87.1% (95% CI 82.6-90.5) vs. paclitaxel 84.7% (95% CI 79.7-88.6)]. The estimated 5-year overall survival (OS) rates were also similar [HR 1.1; ixabepilone 89.7% (95% CI 85.5-92.7) vs. paclitaxel 89.6% (95% CI 85.0-92.9)]. Peripheral neuropathy was the most common grade 3/4 event. Dose reductions and treatment discontinuations occurred more frequently during paclitaxel treatment. CONCLUSIONS Treatment with AC/ixabepilone provided similar DFS and OS in patients with operable TNBC when compared to treatment with AC/paclitaxel. The two regimens had similar toxicity, although treatment discontinuation, dose modifications, and overall peripheral neuropathy were more frequent with AC/paclitaxel. TRIAL REGISTRATION Clinical Trials.gov Identifier, NCT00789581.
Collapse
Affiliation(s)
- Denise A Yardley
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA.
- Tennessee Oncology, Nashville, TN, USA.
| | - Edward R Arrowsmith
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Tennessee Oncology, Chattanooga, TN, USA
| | - Brooke R Daniel
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Tennessee Oncology, Chattanooga, TN, USA
| | - Janice Eakle
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Florida Cancer Specialists, Fort Meyers, FL, USA
| | - Adam Brufsky
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - David R Drosick
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Oncology Hematology Care, Cincinnati, OH, USA
| | - Fred Kudrik
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- South Carolina Oncology Associates, Columbia, SC, USA
| | | | | | | | - Jeffrey A Bubis
- ICON/Cancer Specialists of North Florida, Jacksonville, FL, USA
| | | | | | | | - Martin Bury
- Cancer Research Consortium of West Michigan, Grand Rapids, MI, USA
| | | | | | | | - Robyn R Young
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- The Center for Cancer and Blood Disorders, Ft Worth, TX, USA
| | - William N Harwin
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Florida Cancer Specialists, Fort Meyers, FL, USA
| | - Caryn Silver
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Florida Cancer Specialists, Fort Meyers, FL, USA
| | - John D Hainsworth
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Tennessee Oncology, Nashville, TN, USA
| | - Howard A Burris
- Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, TN, 37203, USA
- Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
1081
|
Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, Saba NF, Weiss J, Wirth L, Sukari A, Kang H, Gibson MK, Massarelli E, Powell S, Meister A, Shu X, Cheng JD, Haddad R. Pembrolizumab for Platinum- and Cetuximab-Refractory Head and Neck Cancer: Results From a Single-Arm, Phase II Study. J Clin Oncol 2017; 35:1542-1549. [PMID: 28328302 PMCID: PMC5946724 DOI: 10.1200/jco.2016.70.1524] [Citation(s) in RCA: 510] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose There are no approved treatments for recurrent/metastatic head and neck squamous cell carcinoma refractory to platinum and cetuximab. In the single-arm, phase II KEYNOTE-055 study, we evaluated pembrolizumab, an anti-programmed death 1 receptor antibody, in this platinum- and cetuximab-pretreated population with poor prognosis. Methods Eligibility stipulated disease progression within 6 months of platinum and cetuximab treatment. Patients received pembrolizumab 200 mg every 3 weeks. Imaging was performed every 6 to 9 weeks. Primary end points: overall response rate (Response Evaluation Criteria in Solid Tumors v1.1, central review) and safety. Efficacy was assessed in all dosed patients and in subgroups on the basis of programmed death ligand 1 (PD-L1) expression and human papillomavirus (HPV) status. Results Among 171 patients treated, 75% received two or more prior lines of therapy for metastatic disease, 82% were PD-L1 positive, and 22% were HPV positive. At the time of analysis, 109 patients (64%) experienced a treatment-related adverse event; 26 patients (15%) experienced a grade ≥ 3 event. Seven patients (4%) discontinued treatment, and one died of treatment-related adverse events. Overall response rate was 16% (95% CI, 11% to 23%), with a median duration of response of 8 months (range, 2+ to 12+ months); 75% of responses were ongoing at the time of analysis. Response rates were similar in all HPV and PD-L1 subgroups. Median progression-free survival was 2.1 months, and median overall survival was 8 months. Conclusion Pembrolizumab exhibited clinically meaningful antitumor activity and an acceptable safety profile in recurrent/metastatic head and neck squamous cell carcinoma previously treated with platinum and cetuximab.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- B7-H1 Antigen/analysis
- Carcinoma, Squamous Cell/chemistry
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/secondary
- Carcinoma, Squamous Cell/virology
- Cetuximab/therapeutic use
- Disease Progression
- Disease-Free Survival
- Female
- Head and Neck Neoplasms/chemistry
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/pathology
- Head and Neck Neoplasms/virology
- Humans
- Male
- Middle Aged
- Neoplasm Recurrence, Local/drug therapy
- Papillomavirus Infections/complications
- Platinum Compounds/therapeutic use
- Response Evaluation Criteria in Solid Tumors
- Retreatment
- Survival Rate
Collapse
Affiliation(s)
- Joshua Bauml
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Tanguy Y. Seiwert
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - David G. Pfister
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Francis Worden
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Stephen V. Liu
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Jill Gilbert
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Nabil F. Saba
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Jared Weiss
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Lori Wirth
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Ammar Sukari
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Hyunseok Kang
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Michael K. Gibson
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Erminia Massarelli
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Steven Powell
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Amy Meister
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Xinxin Shu
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Jonathan D. Cheng
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| | - Robert Haddad
- Joshua Bauml, University of Pennsylvania, Philadelphia, PA; Tanguy Y. Seiwert, University of Chicago, Chicago, IL; David G. Pfister, Memorial Sloan Kettering Cancer Center, New York, NY; Francis Worden, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ammar Sukari, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Stephen V. Liu, Georgetown University Hospital, Washington, DC; Jill Gilbert, Vanderbilt University School of Medicine, Nashville, TN; Nabil F. Saba, Winship Cancer Institute/Emory University, Atlanta, GA; Jared Weiss, Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC; Lori Wirth, Massachusetts General Hospital; Robert Haddad, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Hyunseok Kang, Johns Hopkins University School of Medicine, Baltimore, MD; Michael K. Gibson, University Hospitals Cleveland Medical Center, Cleveland, OH; Erminia Massarelli, The University of Texas MD Anderson Cancer Center, Houston, TX; Steven Powell, Sanford Health, Sioux Falls, SD; and Amy Meister, Xinxin Shu, and Jonathan D. Cheng, Merck & Co., Inc., Kenilworth, NJ
| |
Collapse
|
1082
|
Elevated T cell activation score is associated with improved survival of breast cancer. Breast Cancer Res Treat 2017; 164:689-696. [PMID: 28488141 DOI: 10.1007/s10549-017-4281-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 05/04/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE Immune checkpoints cytotoxic T lymphocyte antigen 4 (CTLA4) and programmed cell death 1 receptor (PD-1) negatively regulate CD8+ T cell functions, impeding the capacity of effector T cells to kill tumors. Here, we study the prognostic significance of CTLA4, PD-1 and T cell activation status in breast cancer. METHODS Using a publicly accessed RNA-seq dataset including 1087 breast cancer patients, we performed Kaplan-Meier survival curves and multivariate Cox regression models to evaluate the associations of CTLA4, PD-1, and weighted T cell activation score with patients' overall survival. RESULTS Survival analyses showed that high CTLA4 but low PD-1 expression was associated with a poor overall survival, and that high T cell activation score was associated with an improved survival. The median survival was 216.6 months (95% CI 114.1-244.9) for the T activation group, 127.0 months (95% CI 112.3-212.1) for the intermediate, and 120.5 months (95% CI 93.8 to ∞) for the exhaustion (Log-rank p = 0.084). This association was verified in multivariate Cox regression analysis. The hazard ratios were 0.81 (95% CI 0.56-1.19) for the intermediate group, and 0.48 (95% CI 0.26-0.86) for the activation group, respectively, in comparison to the exhaustion group (p value for trend = 0.016). CONCLUSIONS T cell activation score has significantly positive relationship with patients' overall survival, and may serve as a marker of personalized immunotherapy in breast cancer patients. Cocktail rather than single immune checkpoint blockade may yield more benefit for breast cancer patients.
Collapse
|
1083
|
Moreno Ayala MA, Gottardo MF, Asad AS, Zuccato C, Nicola A, Seilicovich A, Candolfi M. Immunotherapy for the treatment of breast cancer. Expert Opin Biol Ther 2017; 17:797-812. [PMID: 28446053 DOI: 10.1080/14712598.2017.1324566] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Breast cancer is the most common cancer as well as the first cause of death by cancer in women worldwide. Although routine treatment improves the outcome of early stage breast cancer patients, there is no effective therapy for the disseminated disease. Immunotherapy has emerged as a powerful therapeutic strategy for the treatment of many cancers. Although traditionally conceived as a non-immunogenic tumor, breast cancer is now considered a potential target for immunotherapy. Areas covered: In this review, the authors discuss different immunotherapeutic strategies that are currently being tested for the treatment of breast cancer: These strategies include: (i) blockade of immunological checkpoints, (ii) antitumor vaccines, (iii) regulatory T cell blockade, (iv) adoptive T cell transfer therapy, (iv) adoptive immunotherapy with monoclonal antibodies, and (v) combination of immunotherapy with chemotherapy. Expert opinion: A growing body of evidence indicates that immunotherapeutic strategies can benefit a larger cohort of breast cancer patients than hitherto anticipated. Since breast tumors entail multiple mechanisms to impair antitumor immunity, the immunological characterization of individual tumors and the selection of suitable combinations of chemotherapeutic and immunotherapeutic approaches are required to achieve significant clinical benefit in these patients.
Collapse
Affiliation(s)
- Mariela A Moreno Ayala
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Maria Florencia Gottardo
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Antonela S Asad
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Camila Zuccato
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Alejandro Nicola
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Adriana Seilicovich
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Marianela Candolfi
- a Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
1084
|
Kitano A, Ono M, Yoshida M, Noguchi E, Shimomura A, Shimoi T, Kodaira M, Yunokawa M, Yonemori K, Shimizu C, Kinoshita T, Fujiwara Y, Tsuda H, Tamura K. Tumour-infiltrating lymphocytes are correlated with higher expression levels of PD-1 and PD-L1 in early breast cancer. ESMO Open 2017; 2:e000150. [PMID: 28761741 PMCID: PMC5519809 DOI: 10.1136/esmoopen-2016-000150] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 12/31/2022] Open
Abstract
Background The presence of tumour-infiltrating lymphocytes (TILs) is a favourable prognostic factor in patients with early breast cancer. Programmed cell death-1 (PD-1) and its ligand PD-L1 are associated with a variety of adverse features. The purpose of this study was to clarify the relationships between TILs, PD-1 and PD-L1 as well as their prognostic implications in early breast cancer. Methods We investigated 180 patients with breast cancer who received neoadjuvant chemotherapy and underwent subsequent surgery for stage II–III invasive breast carcinoma between 1999 and 2007. TIL expression was classified as low or high using a previously reported scoring model. PD-1 and PD-L1 expression levels were determined by immunohistochemistry. The correlation between PD-1 expression in TILs and PD-L1 expression in cancer cells was also investigated. Results Higher tumour grade was significantly correlated with PD-L1 expression in tumours (p<0.0001). PD-1 and PD-L1 expression levels were associated with tumour subtype and were highest in triple-negative tumours (p<0.0001). Furthermore, expression of each of PD-1 and PD-L1 was significantly correlated with higher TIL expression and pathological complete response (pCR) (p<0.0001). PD-L1 expression in cancer cells was significantly correlated with PD-1 expression in TILs (p=0.03). The correlations between pCR and expression of each of PD-L1 and PD-1 were not significant. Conclusion Expression of PD-L1 and PD-1 in early breast cancer is associated with higher TIL scores and pCR; conversely, expression of these proteins correlates with poor prognostic clinicopathological factors such as tumour grade and subtype. TILs, PD-1 and PD-L1 can potentially predict the response to treatment.
Collapse
Affiliation(s)
- Atsuko Kitano
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Makiko Ono
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.,Department of Medical Oncology, Cancer Institute Hospital, Tokyo, Japan
| | - Masayuki Yoshida
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Emi Noguchi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsunori Shimoi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Makoto Kodaira
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Mayu Yunokawa
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Chikako Shimizu
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takayuki Kinoshita
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
1085
|
Costa RL, Gradishar WJ. Triple-Negative Breast Cancer: Current Practice and Future Directions. J Oncol Pract 2017; 13:301-303. [DOI: 10.1200/jop.2017.023333] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Ricardo L.B. Costa
- Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - William J. Gradishar
- Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| |
Collapse
|
1086
|
Abstract
HER2 and CDK4/6 are undoubted two most important biological targets for breast cancer. Anti-HER2 treatments enhance objective response and progression-free survival/disease-free survival as well as overall survival. Three CDK4/6 inhibitors consistently improve objective response and progression-free survival; however, overall survival data are waited. Optimization of chemotherapy and endocrine strategies remains an unmet need. Check point inhibitor-based immunotherapy combined with chemotherapy is a promising field, especially for triple-negative breast cancer.
Collapse
Affiliation(s)
- Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Wei Huang
- Roche Product Development in Asia Pacific.5F, Tower C, Parkview Green, No.9, Dongdaqiao Road, Chaoyang District, Beijing, 100020 People’s Republic of China
| | - Minhao Fan
- Hutchison MediPharma Limited, Building 4 917 Halei Road Zhangjiang Hi-Tech Park, Shanghai, 201203 China
| |
Collapse
|
1087
|
Osada T, Morse MA, Hobeika A, Diniz MA, Gwin WR, Hartman Z, Wei J, Guo H, Yang XY, Liu CX, Kaneko K, Broadwater G, Lyerly HK. Vaccination targeting human HER3 alters the phenotype of infiltrating T cells and responses to immune checkpoint inhibition. Oncoimmunology 2017; 6:e1315495. [PMID: 28680745 DOI: 10.1080/2162402x.2017.1315495] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023] Open
Abstract
Expression of human epidermal growth factor family member 3 (HER3), a critical heterodimerization partner with EGFR and HER2, promotes more aggressive biology in breast and other epithelial malignancies. As such, inhibiting HER3 could have broad applicability to the treatment of EGFR- and HER2-driven tumors. Although lack of a functional kinase domain limits the use of receptor tyrosine kinase inhibitors, HER3 contains antigenic targets for T cells and antibodies. Using novel human HER3 transgenic mouse models of breast cancer, we demonstrate that immunization with recombinant adenoviral vectors encoding full length human HER3 (Ad-HER3-FL) induces HER3-specific T cells and antibodies, alters the T cell infiltrate in tumors, and influences responses to immune checkpoint inhibitions. Both preventative and therapeutic Ad-HER3-FL immunization delayed tumor growth but were associated with both intratumoral PD-1 expressing CD8+ T cells and regulatory CD4+ T cell infiltrates. Immune checkpoint inhibition with either anti-PD-1 or anti-PD-L1 antibodies increased intratumoral CD8+ T cell infiltration and eliminated tumor following preventive vaccination with Ad-HER3-FL vaccine. The combination of dual PD-1/PD-L1 and CTLA4 blockade slowed the growth of tumor in response to Ad-HER3-FL in the therapeutic model. We conclude that HER3-targeting vaccines activate HER3-specific T cells and induce anti-HER3 specific antibodies, which alters the intratumoral T cell infiltrate and responses to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Marcio A Diniz
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - William R Gwin
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| | - Zachary Hartman
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Junping Wei
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Hongtao Guo
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Kensuke Kaneko
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Gloria Broadwater
- Duke University, Division of Biostatistics Duke Cancer Institute, Durham, NC, USA
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
1088
|
Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci 2017; 24:26. [PMID: 28376884 PMCID: PMC5381059 DOI: 10.1186/s12929-017-0329-9] [Citation(s) in RCA: 476] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has recently emerged as the fourth pillar of cancer treatment, joining surgery, radiation, and chemotherapy. While early immunotherapies focused on accelerating T-cell activity, current immune-checkpoint inhibitors take the brakes off the anti-tumor immune responses. Successful clinical trials with PD-1 monoclonal antibodies and other immune-checkpoint inhibitors have opened new avenues in cancer immunology. However, the failure of a large subset of cancer patients to respond to these new immunotherapies has led to intensified research on combination therapies and predictive biomarkers. Here we summarize the development of PD-1-blockade immunotherapy and current issues in its clinical use.
Collapse
Affiliation(s)
- Yoshiko Iwai
- Department of Molecular Biology, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu-shi, Fukuoka, 807-8555, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
1089
|
Law AMK, Lim E, Ormandy CJ, Gallego-Ortega D. The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy. Endocr Relat Cancer 2017; 24:R123-R144. [PMID: 28193698 PMCID: PMC5425956 DOI: 10.1530/erc-16-0404] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
A cancer cell-centric view has long dominated the field of cancer biology. Research efforts have focussed on aberrant cancer cell signalling pathways and on changes to cancer cell DNA. Mounting evidence demonstrates that many cancer-associated cell types within the tumour stroma co-evolve and support tumour growth and development, greatly modifying cancer cell behaviour, facilitating invasion and metastasis and controlling dormancy and sensitivity to drug therapy. Thus, these stromal cells represent potential targets for cancer therapy. Among these cell types, immune cells have emerged as a promising target for therapy. The adaptive and the innate immune system play an important role in normal mammary development and breast cancer. The number of infiltrating adaptive immune system cells with tumour-rejecting capacity, primarily, T lymphocytes, is lower in breast cancer compared with other cancer types, but infiltration occurs in a large proportion of cases. There is strong evidence demonstrating the importance of the immunosuppressive role of the innate immune system during breast cancer progression. A consideration of components of both the innate and the adaptive immune system is essential for the design and development of immunotherapies in breast cancer. In this review, we focus on the importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Andrew M K Law
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Elgene Lim
- Connie Johnson Breast Cancer Research LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Christopher J Ormandy
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - David Gallego-Ortega
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| |
Collapse
|
1090
|
Parvani JG, Jackson MW. Silencing the roadblocks to effective triple-negative breast cancer treatments by siRNA nanoparticles. Endocr Relat Cancer 2017; 24:R81-R97. [PMID: 28148541 PMCID: PMC5471497 DOI: 10.1530/erc-16-0482] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/01/2017] [Indexed: 12/12/2022]
Abstract
Over the past decade, RNA interference (RNAi) has been ubiquitously utilized to study biological function in vitro; however, limitations were associated with its utility in vivo More recently, small interfering RNA (siRNA) nanoparticles with improved biocompatibility have gained prevalence as a potential therapeutic option for the treatment of various diseases. The adaptability of siRNA nanoparticles enables the delivery of virtually any siRNA, which is especially advantageous for therapeutic applications in heterogeneous diseases that lack unifying molecular features, such as triple-negative breast cancer (TNBC). TNBC is an aggressive subtype of breast cancer that is stratified by the lack of estrogen receptor/progesterone receptor expression and HER2 amplification. There are currently no FDA-approved targeted therapies for the treatment of TNBCs, making cytotoxic chemotherapy the only treatment option available to these patients. In this review, we outline the current status of siRNA nanoparticles in clinical trials for cancer treatment and discuss the promising preclinical approaches that have utilized siRNA nanoparticles for TNBC treatment. Next, we address TNBC subtype-specific therapeutic interventions and highlight where and how siRNA nanoparticles fit into these strategies. Lastly, we point out ongoing challenges in the field of siRNA nanoparticle research that, if addressed, would significantly improve the efficacy of siRNA nanoparticles as a therapeutic option for cancer treatment.
Collapse
Affiliation(s)
- Jenny G Parvani
- Department of Biomedical EngineeringCase Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer CenterCase Western Reserve University, Cleveland, Ohio, USA
| | - Mark W Jackson
- Case Comprehensive Cancer CenterCase Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
1091
|
Ocana A, Pandiella A. Targeting oncogenic vulnerabilities in triple negative breast cancer: biological bases and ongoing clinical studies. Oncotarget 2017; 8:22218-22234. [PMID: 28108739 PMCID: PMC5400659 DOI: 10.18632/oncotarget.14731] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/11/2017] [Indexed: 12/15/2022] Open
Abstract
Triple negative breast cancer (TNBC) is still an incurable disease despite the great scientific effort performed during the last years. The huge heterogeneity of this disease has motivated the evaluation of a great number of therapies against different molecular alterations. In this article, we review the biological bases of this entity and how the known molecular evidence supports the current preclinical and clinical development of new therapies. Special attention will be given to ongoing clinical studies and potential options for future drug combinations.
Collapse
Affiliation(s)
- Alberto Ocana
- Unidad de Investigación Traslacional, Hospital Universitario de Albacete, Universidad de Castilla La Mancha, Albacete, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer and CIBERONC. CSIC-Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
1092
|
Abstract
Immune escape of malignant cells is an important hallmark of cancer, necessary for tumor formation and progression. Accordingly, in recent years, therapies that enhance the immune system have had remarkable success in treating a myriad of malignancies. Particularly successful has been immune checkpoint blockade (ICB), which is a therapy that targets T-cell inhibitory receptors, or immune checkpoints. Despite these encouraging clinical results, most patients do not respond to such agents. Therefore, determining methods to better target and enhance the therapeutic efficacy of ICB is of paramount importance. One appealing approach is to use standard anticancer therapies, such as radiation, chemotherapy, and targeted biologics, to favorably modulate the immune system and enhance the anticancer immune response. For example, although radiation therapy has classically been thought of as a local therapy, there is significant potential for combining radiation therapy with ICB to both optimize local control and to treat metastatic disease. This concept is supported by numerous preclinical studies and clinical case reports and has since led to many early and ongoing clinical trials. However, it is still unclear how to optimally combine radiation and ICB to maximize the therapeutic effect. In this review, we highlight relevant preclinical and clinical studies in the field of radiation and ICB and discuss optimal strategies for combination therapies moving forward.
Collapse
Affiliation(s)
- Jacob E Shabason
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Andy J Minn
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
1093
|
Luen S, Virassamy B, Savas P, Salgado R, Loi S. The genomic landscape of breast cancer and its interaction with host immunity. Breast 2017; 29:241-50. [PMID: 27481651 DOI: 10.1016/j.breast.2016.07.015] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 07/08/2016] [Indexed: 12/15/2022] Open
Abstract
Molecular profiling of thousands of primary breast cancers has uncovered remarkable genomic diversity between breast cancer subtypes, and even within subtypes. Only a few driver genes are recurrently altered at high frequency highlighting great challenges for precision medicine. Considerable evidence also confirms the role of host immunosurveillance in influencing response to therapy and prognosis in HER2+ and triple negative breast cancer. The role of immunosurveillance in ER + disease remains unclear. Advances in both these fields have lead to intensified interest in the interaction between genomic landscapes and host anti-tumour immune responses in breast cancer. In this review, we discuss the potential genomic determinants of host anti-tumour immunity - mutational load, driver alterations, mutational processes and neoantigens - and their relationship with immunity in breast cancer. Significant differences exist in both the genomic and immune characteristics amongst breast cancer subtypes. While ER + disease appears to be less immunogenic than HER2+ and triple negative breast cancer, it displays the greatest degree of heterogeneity. Mutational and neoantigen load appears to incompletely explains immune responses in breast cancer. Driver alterations do not appear to increase immunogenicity. Instead, they could contribute to immune-evasion or an immunosuppressive microenvironment, and therefore represent potential therapeutic targets. Finally, we also discuss the tailoring of immunotherapeutic strategies by genomic alterations, with possible multimodal combination approaches to maximise clinical benefits.
Collapse
Affiliation(s)
- Stephen Luen
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Balaji Virassamy
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter Savas
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Sherene Loi
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
1094
|
Castrellon AB, Pidhorecky I, Valero V, Raez LE. The Role of Carboplatin in the Neoadjuvant Chemotherapy Treatment of Triple Negative Breast Cancer. Oncol Rev 2017; 11:324. [PMID: 28382189 PMCID: PMC5365000 DOI: 10.4081/oncol.2017.324] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast (TNBC) cancer constitutes a heterogeneous group of disease with histologic and molecular differences. Complete pathologic response to neoadjuvant chemotherapy (NACT) in TNBC is associated with improved outcomes. Efforts have been made in identifying drug combinations that will increase the response rate to preoperative chemotherapy. In this review we present recent studies that have incorporated carboplatin (Cb) in the NACT of TNBC. We discuss the homologous recombination deficiency score and the somatic or germline mutation for BRCA as potential biomarkers for future selection of patients that could benefit from the addition of Cb to NACT.
Collapse
Affiliation(s)
| | - Ihor Pidhorecky
- Surgical Oncology Services & Pancreaticobiliary Center, Memorial Cancer Institute, Memorial Healthcare System, Hollywood, FL
| | - Vicente Valero
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luis Estuardo Raez
- Hematology/Oncology and Medical Director of Memorial Cancer Institute, Memorial Healthcare System, Hollywood, FL, USA
| |
Collapse
|
1095
|
Bhattacharya R, Banerjee K, Mukherjee N, Sen M, Mukhopadhyay A. From molecular insight to therapeutic strategy: The holistic approach for treating triple negative breast cancer. Pathol Res Pract 2017; 213:177-182. [DOI: 10.1016/j.prp.2017.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/02/2017] [Accepted: 01/02/2017] [Indexed: 02/07/2023]
|
1096
|
Immune checkpoint inhibitors and targeted therapies for metastatic melanoma: A network meta-analysis. Cancer Treat Rev 2017; 54:34-42. [DOI: 10.1016/j.ctrv.2017.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/14/2017] [Accepted: 01/16/2017] [Indexed: 01/22/2023]
|
1097
|
Hubalek M, Czech T, Müller H. Biological Subtypes of Triple-Negative Breast Cancer. Breast Care (Basel) 2017; 12:8-14. [PMID: 28611535 DOI: 10.1159/000455820] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancers (TNBCs) are defined as tumors that are negative for estrogen, progesterone and HER-2 receptor. At a percentage of 10-20% TNBCs represent a minority in all breast cancers. However, because of the poor prognosis this particular subtype, triple negative disease accounts for a disproportionate number of metastatic cases and breast cancer deaths. Identification of its subtypes is essential for understanding the biological characteristics and clinical behavior of TNBC, as well as for developing personalized treatments. This review will focus on the great progress that has been made in the past few years on identifying new targets in TNBC subtypes and a variety of new treatment options that are on the verge of routine clinical application.
Collapse
Affiliation(s)
- Michael Hubalek
- Department of Gynecology and Obstetrics, Brustzentrum Schwaz, Bezirkskrankenhaus Schwaz, Schwaz, Austria
| | - Theresa Czech
- Dept. of Gynecology and Obstretrics, Brustgesundheitzentrum Tirol, Medical University Innsbruck, Innsbruck, Austria
| | - Hannes Müller
- Dept. of Gynecology and Obstretrics, Brustgesundheitzentrum Tirol, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
1098
|
MT4-MMP and EGFR expression levels are key biomarkers for breast cancer patient response to chemotherapy and erlotinib. Br J Cancer 2017; 116:742-751. [PMID: 28196064 PMCID: PMC5355928 DOI: 10.1038/bjc.2017.23] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022] Open
Abstract
Background: Triple-negative breast cancers (TNBC) are heterogeneous cancers with poor prognosis. We aimed to determine the clinical relevance of membrane type-4 matrix metalloproteinase (MT4-MMP), a membrane type matrix metalloproteinase that interacts with epidermal growth factor receptor (EGFR) overexpressed in >50% of TNBC. Methods: We conducted a retrospective immunohistochemical analysis on human TNBC samples (n=81) and validated our findings in in vitro and in vivo assays. Results: Membrane type-4 matrix metalloproteinase and EGFR are produced in 72.5% of TNBC samples, whereas those proteins are faintly produced by healthy tissues. Unexpectedly, tumour relapse after chemotherapy was reduced in samples highly positive for MT4-MMP. Mechanistically, this is ascribed to a higher sensitivity of MT4-MMP-producing cells to alkylating or intercalating chemotherapeutic agents, as assessed in vitro. In sharp contrast, MT4-MMP expression did not affect tumour cell sensitivity to paclitaxel that interferes with protease trafficking. Importantly, MT4-MMP expression sensitised cancer cells to erlotinib, a tyrosine kinase EGFR inhibitor. In a pre-clinical model, the growth of MT4-MMP overexpressing xenografts, but not of control ones, was reduced by epirubicin or erlotinib. The combination of suboptimal drug doses blocked drastically the growth of MT4-MMP-producing tumours. Conclusions: We demonstrate that MT4-MMP defines a sub-population of TNBC sensitive to a combination of DNA-targeting chemotherapeutic agents and anti-EGFR drugs.
Collapse
|
1099
|
Kumar V, Chaudhary N, Garg M, Floudas CS, Soni P, Chandra AB. Current Diagnosis and Management of Immune Related Adverse Events (irAEs) Induced by Immune Checkpoint Inhibitor Therapy. Front Pharmacol 2017; 8:49. [PMID: 28228726 PMCID: PMC5296331 DOI: 10.3389/fphar.2017.00049] [Citation(s) in RCA: 426] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/23/2017] [Indexed: 12/18/2022] Open
Abstract
The indications of immune checkpoint inhibitors (ICIs) are set to rise further with the approval of newer agent like atezolimumab for use in patients with advanced stage urothelial carcinoma. More frequent use of ICIs has improved our understanding of their unique side effects, which are known as immune-related adverse events (irAEs). The spectrum of irAEs has expanded beyond more common manifestations such as dermatological, gastrointestinal and endocrine effects to rarer presentations involving nervous, hematopoietic and urinary systems. There are new safety data accumulating on ICIs in patients with previously diagnosed autoimmune conditions. It is challenging for clinicians to continuously update their working knowledge to diagnose and manage these events successfully. If diagnosed timely, the majority of events are completely reversible, and temporary immunosuppression with glucocorticoids, infliximab or other agents is warranted only in the most severe grade illnesses. The same principles of management will possibly apply as newer anti- cytotoxic T lymphocytes-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1/PD-L1) antibodies are introduced. The current focus of research is for prophylaxis and for biomarkers to predict the onset of these toxicities. In this review we summarize the irAEs of ICIs and emphasize their growing spectrum and their management algorithms, to update oncology practitioners.
Collapse
Affiliation(s)
- Vivek Kumar
- Department of Medicine, Maimonides Medical Center Brooklyn, NY, USA
| | - Neha Chaudhary
- Department of Pediatrics, Maimonides Medical Center Brooklyn, NY, USA
| | - Mohit Garg
- Department of Medicine, Maimonides Medical Center Brooklyn, NY, USA
| | | | - Parita Soni
- Department of Medicine, Maimonides Medical Center Brooklyn, NY, USA
| | | |
Collapse
|
1100
|
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
|