101
|
Gupta S, Vanderbilt CM, Cotzia P, Arias-Stella JA, Chang JC, Zehir A, Benayed R, Nafa K, Razavi P, Hyman DM, Baselga J, Berger MF, Ladanyi M, Arcila ME, Ross DS. Next-Generation Sequencing-Based Assessment of JAK2, PD-L1, and PD-L2 Copy Number Alterations at 9p24.1 in Breast Cancer: Potential Implications for Clinical Management. J Mol Diagn 2019; 21:307-317. [PMID: 30576871 PMCID: PMC6432425 DOI: 10.1016/j.jmoldx.2018.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/28/2018] [Accepted: 10/24/2018] [Indexed: 01/01/2023] Open
Abstract
Genomic amplification at 9p24.1, including the loci for JAK2, PD-L1, and PD-L2, has recently been described as a mechanism of resistance in postchemotherapy, triple-negative breast cancer. This genomic signature holds significant promise as a prognostic biomarker and has implications for targeted therapy with JAK2 inhibitors, as well as with immunotherapy. To guide future screening strategies, the frequency of these alterations was determined. A total of 5399 cases were included in the study. This encompassed 2890 institutional cases tested by the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets assay and 2509 cases from The Cancer Genome Atlas (TCGA). The combined incidence of 9p24.1 amplifications in both the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets and TCGA cohorts was 1.0% (56/5399 cases) and showed a >10-fold higher incidence in triple-negative breast cancer (triple-negative: 5.1%; non-triple-negative: 0.5%). Tumor mutation burden and stromal tumor infiltrating lymphocytes, parameters used to assess response to immunotherapy, were not significantly higher for these cases. The significance of genomic losses at 9p24.1 is unclear, and further studies are needed. Herein, we studied the spectrum of copy number alterations in breast cancer cases within our institutional clinical sequencing cohort and those profiled by TCGA to determine the frequency of genomic alterations that may predict response or resistance to JAK2 inhibitors and/or immunotherapy.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paolo Cotzia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Khedouja Nafa
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pedram Razavi
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David M Hyman
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - José Baselga
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
102
|
Bang YJ, Van Cutsem E, Fuchs CS, Ohtsu A, Tabernero J, Ilson DH, Hyung WJ, Strong VE, Goetze TO, Yoshikawa T, Tang LH, Hwang PMT, Webb N, Adelberg D, Shitara K. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol 2019; 15:943-952. [PMID: 30777447 DOI: 10.2217/fon-2018-0581] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Surgical resection is the only curative treatment option for gastric cancer. Despite widespread adoption of multimodality perioperative treatment strategies, 5-year overall survival rates remain low. In patients with advanced gastric or gastroesophageal junction adenocarcinoma, pembrolizumab has demonstrated promising efficacy and manageable safety as monotherapy in previously treated patients and as first-line therapy in combination with cisplatin and 5-fluorouracil. Combining chemotherapy with pembrolizumab in the neoadjuvant/adjuvant setting may benefit patients with locally advanced, resectable disease. AIM To describe the design and rationale for the global, multicenter, randomized, double-blind, Phase III KEYNOTE-585 study to evaluate the efficacy and safety of pembrolizumab plus chemotherapy compared with placebo plus chemotherapy as neoadjuvant/adjuvant treatment for localized gastric or gastroesophageal junction adenocarcinoma. ClinicalTrials.gov : NCT03221426.
Collapse
Affiliation(s)
- Yung-Jue Bang
- Seoul National University College of Medicine, 101 Daehak-ro, Ihwa-Dong, Seoul, Republic of Korea
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven & KU Leuven, 49 Herestraat, Leuven 3001, Belgium
| | - Charles S Fuchs
- Yale Cancer Center and Smilow Cancer Hospital, 333 Cedar Street, New Haven, CT 06510, USA
| | - Atsushi Ohtsu
- National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Josep Tabernero
- Vall d'Hebron University Hospital & Institute of Oncology (VHIO), Vall d'Hebron 119-129, 08035 Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David H Ilson
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street, BAIC 1031, New York, NY 10065, USA
| | - Woo Jin Hyung
- Yonsei Cancer Hospital, Yonsei University Health System, 50-1 Yonsei-Ro Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Vivian E Strong
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Thorsten Oliver Goetze
- Institute of Clinical Cancer Research, UCT University Cancer Center Frankfurt, Steinbacher Hohl 2-26, Frankfurt 60488, Germany
| | - Takaki Yoshikawa
- National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Laura H Tang
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | | | - Nancy Webb
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - David Adelberg
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Kohei Shitara
- National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| |
Collapse
|
103
|
Cecchini M, Rubin EH, Blumenthal GM, Ayalew K, Burris HA, Russell-Einhorn M, Dillon H, Lyerly HK, Reaman GH, Boerner S, LoRusso PM. Challenges with Novel Clinical Trial Designs: Master Protocols. Clin Cancer Res 2019; 25:2049-2057. [PMID: 30696689 DOI: 10.1158/1078-0432.ccr-18-3544] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/07/2019] [Accepted: 01/25/2019] [Indexed: 11/16/2022]
Abstract
The 2018 Accelerating Anticancer Agent Development (AAADV) Workshop assembled a panel of experts for an in-depth discussion session to present "Challenges with Novel Clinical Trial Designs." This panel offered assessments of the challenges faced by industry, the FDA, investigators, institutional review boards, and patients. The panel focused on master protocols, which include umbrella trials, platform trials, and basket trials. Umbrella trials and platform trials share many commonalities, whereas basket trials are more distinct. Umbrella and platform trials are generally designed with multiple arms where patients of the same histology or other unifying characteristics are enrolled into different arms and multiple investigational agents are evaluated in a single protocol. In contrast, basket studies generally enroll patients with different tumor types based on the presence of a specific mutation or biomarker regardless of histology; these trials may include expansion cohorts. These novel designs offer the promise of expedited drug assessment and approval, but they also place new challenges on all the stakeholders involved in the drug development process. Only by identifying the challenges of these complex, innovative clinical trial designs and highlighting challenges from each perspective can we begin to address these challenges. The 2018 AAADV Workshop convened a panel of experts from relevant disciplines to highlight the challenges that are created by master protocols, and, where appropriate, offer strategies to address these challenges.
Collapse
Affiliation(s)
| | | | | | - Kassa Ayalew
- U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Howard A Burris
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | | | - Hildy Dillon
- Cancer Support Community, Washington, District of Columbia
| | | | | | | | | |
Collapse
|
104
|
Lyons TG, Traina TA. Emerging Novel Therapeutics in Triple-Negative Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:377-399. [PMID: 31456195 DOI: 10.1007/978-3-030-20301-6_20] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mortality from breast cancer has steadily decreased due in part to early detection and advances in therapy. The treatment options for breast cancer vary considerably depending on the histological subtype. There are a number of very effective targeted therapies available for estrogen receptor-positive disease and for human epidermal growth factor receptor 2-positive disease. However, triple-negative breast cancer is a particularly aggressive subtype. This subtype represents an unmet need for improved therapies. TNBC is a heterogenous subtype of breast cancer that is beginning to be refined by its molecular characteristics and clinical response to a targeted therapeutic approach. Here we review the recent advances in the treatment of TNBC with emphasis on the many emerging novel targeted therapies.
Collapse
Affiliation(s)
- Tomas G Lyons
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Tiffany A Traina
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weil Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
105
|
Levinson K, Dorigo O, Rubin K, Moore K. Immunotherapy in Gynecologic Cancers: What We Know Now and Where We Are Headed. Am Soc Clin Oncol Educ Book 2019; 39:e126-e140. [PMID: 31099679 DOI: 10.1200/edbk_237967] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Immunotherapy, mainly in the form of immune checkpoint inhibitors (ICIs), has been transformative in both solid tumor and hematologic malignancies. Patients with previously terminal illnesses have experienced profound responses of great durability with these agents, fueling excitement among patients and providers regarding their use. Unfortunately, the gains seen in some solid tumors have not been replicated in a large percentage of patients with gynecologic cancer. This review focuses on the clinical benefits seen to date, toxicities and management when using ICIs, ways to improve prediction of who should receive immunotherapy, and a discussion of next-generation immunotherapy with cellular therapeutics and how these might relate to gynecologic cancers.
Collapse
Affiliation(s)
- Kimberly Levinson
- 1 The Kelly Gynecologic Oncology Service, Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | - Kathleen Moore
- 4 Stephenson Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
106
|
Pelekanou V, Villarroel-Espindola F, Schalper KA, Pusztai L, Rimm DL. CD68, CD163, and matrix metalloproteinase 9 (MMP-9) co-localization in breast tumor microenvironment predicts survival differently in ER-positive and -negative cancers. Breast Cancer Res 2018; 20:154. [PMID: 30558648 PMCID: PMC6298021 DOI: 10.1186/s13058-018-1076-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Abstract
Background The role of tumor-associated macrophages (TAMs) in the cancer immune landscape and their potential as treatment targets or modulators of response to treatment are gaining increasing interest. TAMs display high molecular and functional complexity. Therefore their objective assessment as breast cancer biomarkers is critical. The aims of this study were to objectively determine the in situ expression and significance of TAM biomarkers (CD68, CD163, and MMP-9) in breast cancer and to identify subclasses of patients who could benefit from TAM-targeting therapies. Methods We measured CD68, CD163, and MMP-9 protein expression in formalin-fixed paraffin-embedded tissues of breast carcinomas represented in tissue microarray format using multiplexed quantitative immunofluorescence (QIF) in two independent Yale cohorts: cohort A—n = 398, estrogen receptor–positive (ER+) and ER− cases—and the triple-negative breast cancer (TNBC)-only cohort B (n = 160). Associations between macrophage markers, ER status, and survival were assessed. Protein expression measured by QIF was compared with mRNA expression data from the METABRIC study. Results All three macrophage markers were co-expressed, displaying higher expression in ER− cancers. High pan-macrophage marker CD68 correlated with poorer overall survival (OS) only in ER− cases of cohort A (P = 0.02). High expression of CD163 protein in TAMs was associated with improved OS in ER− cases (cohort A, P = 0.03 and TNBC cohort B, P = 0.04, respectively) but not in ER+ cancers. MMP-9 protein was not individually associated with OS. High expression of MMP-9 in the CD68+/CD163+ TAMs was associated with worse OS in ER+ tumors (P <0.001) but not in ER− cancers. In the METABRIC dataset, mRNA levels followed the co-expression pattern observed in QIF but did not always show the same trend regarding OS. Conclusions Macrophage activity markers correlate with survival differently in ER+ and ER− cancers. The association between high co-expression and co-localization of MMP-9/CD163/CD68 and poor survival in ER+ cancers suggests that these cancers may be candidates for macrophage-targeted therapies. Electronic supplementary material The online version of this article (10.1186/s13058-018-1076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vasiliki Pelekanou
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA. .,Sanofi US Services Inc., Bridgewater Township, USA.
| | - Franz Villarroel-Espindola
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| | - Lajos Pusztai
- Department of Medical Oncology, Yale School of Medicine, 330 Cedar Street, New Haven, 06520, CT, USA
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| |
Collapse
|
107
|
Altan M, Kidwell KM, Pelekanou V, Carvajal-Hausdorf DE, Schalper KA, Toki MI, Thomas DG, Sabel MS, Hayes DF, Rimm DL. Association of B7-H4, PD-L1, and tumor infiltrating lymphocytes with outcomes in breast cancer. NPJ Breast Cancer 2018; 4:40. [PMID: 30564631 PMCID: PMC6288133 DOI: 10.1038/s41523-018-0095-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023] Open
Abstract
B7-H4 (VTCN1) is a member of the CD28/B7 family of immune co-inhibitory molecules. The relationship of tumor and stromal B7-H4 protein expression with PD-L1, tumor infiltrating lymphocytes (TILs) and its association with clinico-pathological variables are not well defined. Herein, we explore the expression level of B7-H4 protein in breast cancer and evaluate its association with TILs, levels of PD-L1 expression, and clinico-pathological characteristics in two independent populations. In this study, we used multiplexed automated quantitative immunofluorescence (QIF) to measure the levels of B7-H4 and PD-L1 protein and determined TILs through pathologist assessment of H&E-stained preparations in over a thousand breast cancer cases from two institutions represented in tissue microarray format. Associations between the marker levels, major clinico-pathological variables, and survival were analyzed. We detected B7-H4 protein was highly expressed in both breast cancer and stromal cells. Its expression was independent of breast cancer intrinsic subtypes. PD-L1 expression was higher in triple negative breast cancers. Neither B7-H4 nor PD-L1 were associated with survival in breast cancer. Our study shows there is a mutually exclusive pattern of B7-H4 with both tumor PD-L1 expression and TILs in all breast cancers, independent of breast cancer intrinsic subtype. This exclusive pattern suggests that some breast tumors may preferentially use one B7-related immune evasion mechanism/pathway. This could explain the clinical benefit that is seen only in a fraction of patients with immune checkpoint inhibitors directed exclusively towards PD-L1 in breast cancer.
Collapse
Affiliation(s)
- Mehmet Altan
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Kelley M. Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI USA
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | | | - Daniel E. Carvajal-Hausdorf
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
- Anatomic Pathology, Clinica Alemana-Facultad de Medicina Universidad de Desarrollo, Vitacura, Santiago Chile
| | - Kurt A. Schalper
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Maria I. Toki
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Dafydd G. Thomas
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Michael S. Sabel
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Daniel F. Hayes
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - David L. Rimm
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
108
|
Fremd C, Jaeger D, Schneeweiss A. Targeted and immuno-biology driven treatment strategies for triple-negative breast cancer: current knowledge and future perspectives. Expert Rev Anticancer Ther 2018; 19:29-42. [PMID: 30351981 DOI: 10.1080/14737140.2019.1537785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Accounting for about 15% of breast cancer patients, triple-negative breast cancer (TNBC) is responsible for 25% of disease related deaths, more frequent distant spread and visceral metastasis. However, improving survival in TNBC failed and primary resistance, immunological ignorance and tumor heterogeneity limit clinical activity of novel therapies. In view of recent molecular, genetic and immunologic insights, this review aims to describe the current status of immunological and targeted treatments from a hypothesis driven perspective. Areas covered: Recent preclinical studies and ongoing clinical trials for immune directed and targeted treatments of TNBC are summarized, including immune-checkpoint blockade, resistance mechanisms, inhibition of poly (ADP-ribose) polymerase (PARP), combinatorial strategies as well as preclinical, hypothesis generating studies. Expert commentary: Sustained responses have been observed with immune-checkpoint blockade and PARP inhibitors demonstrated remarkable efficacy in germline BRCA mutated TNBC. In order to generate clinical success of many other, to date ineffective, targeted and immune therapies, the integration of multidimensional, large amounts of data, will be essential and likely accelerate treatment progress of TNBC.
Collapse
Affiliation(s)
- Carlo Fremd
- a National Center for Tumor Diseases, Department of Medical Oncology , University of Heidelberg , Heidelberg , Germany
| | - Dirk Jaeger
- a National Center for Tumor Diseases, Department of Medical Oncology , University of Heidelberg , Heidelberg , Germany
| | - Andreas Schneeweiss
- a National Center for Tumor Diseases, Department of Medical Oncology , University of Heidelberg , Heidelberg , Germany
| |
Collapse
|
109
|
Abstract
Early-stage non-small cell lung cancer is a potentially curable disease, but with relapse rates exceeding 50% with standard treatments, this is a patient population in critical need of therapy innovation. Immunotherapy with immune checkpoint blockade has revolutionized the treatment strategy for advanced lung cancer. However, the role of this therapy in earlier-stage disease is largely unknown. The study of immunotherapy in earlier-stage disease has many advantages, including assessment of pathologic response and incorporation of translational scientific analyses to evaluate antitumor immune responses. Multiple clinical trials are currently under way, with promising early results.
Collapse
Affiliation(s)
- Samuel Rosner
- Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA; , ,
| | - Joshua E Reuss
- Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA; , ,
| | - Patrick M Forde
- Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA; , ,
| |
Collapse
|
110
|
Escrivá-de-Romaní S, Arumí M, Zamora E, Bellet M. Neoadjuvant Model as a Platform for Research in Breast Cancer and Novel Targets under Development in this Field. Breast Care (Basel) 2018; 13:251-262. [PMID: 30319327 DOI: 10.1159/000492122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
For decades, the neoadjuvant setting has provided a useful scenario for research in breast cancer. Historically, neoadjuvant clinical trials, either hormone therapy-based or chemotherapy-based, have tried to recapitulate the results of their counterpart adjuvant studies, but with smaller patient numbers, more rapid outcomes (clinical response and/or pathologic complete response (pCR)), together with additional biologic information. As for neoadjuvant chemotherapy trials, the increase in pCR rates has been recently accepted as an appropriate surrogate marker to accelerate drug approval in high-risk breast cancer patients. In this setting, with the exception of luminal A tumors, pCR has been associated with improved long-term outcomes, particularly when the analysis is based on specific trials for each breast cancer subtype. For luminal tumors receiving neoadjuvant endocrine therapy, Ki67 at 2-4 weeks and the preoperative endocrine prognostic index score are the most accepted intermediate markers of efficacy, which will be validated in ongoing larger trials. In this review, we describe the different neoadjuvant designs: from the classical randomized trials in which treatment is delivered for 6 or more months to short non-therapeutic presurgical studies lasting just 2 or 3 weeks. We also review the main neoadjuvant trials, either ongoing or completed, for luminal, triple-negative, and HER2-positive breast cancer. The translational effort and research of biomarkers conducted in these studies will be particularly addressed.
Collapse
Affiliation(s)
- Santiago Escrivá-de-Romaní
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Miriam Arumí
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Esther Zamora
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Meritxell Bellet
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
111
|
Noor ZS, Master A. Updates on Targeted Therapy for Triple-Negative Breast Cancer (TNBC). CURRENT BREAST CANCER REPORTS 2018. [DOI: 10.1007/s12609-018-0291-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
112
|
Yoon HK, Kim TH, Park S, Jung H, Quan X, Park SJ, Han J, Lee A. Effect of anthracycline and taxane on the expression of programmed cell death ligand-1 and galectin-9 in triple-negative breast cancer. Pathol Res Pract 2018; 214:1626-1631. [DOI: 10.1016/j.prp.2018.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/29/2018] [Accepted: 08/08/2018] [Indexed: 01/22/2023]
|
113
|
Constantinidou A, Alifieris C, Trafalis DT. Targeting Programmed Cell Death -1 (PD-1) and Ligand (PD-L1): A new era in cancer active immunotherapy. Pharmacol Ther 2018; 194:84-106. [PMID: 30268773 DOI: 10.1016/j.pharmthera.2018.09.008] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Improved understanding of the immune system and its role in cancer development and progression has led to impressive advances in the field of cancer immunotherapy over the last decade. Whilst the field is rapidly evolving and the list of drugs receiving regulatory approval for the treatment of various cancers is fast growing, the group of PD1- PDL-1 inhibitors is establishing a leading role amongst immunomodulatory agents. PD1- PDL-1 inhibitors act against pathways involved in adaptive immune suppression resulting in immune checkpoint blockade. Within the last four years two PD-1 and three PD-L1 inhibitors have been utilized in clinical practice against a variety of malignancies. Focus was initially placed on targeting cancers considered immunogenic such as melanoma, renal and lung cancers but subsequently the application expanded to include amongst others Hodgkin Lymphoma, urothelial as well as head and neck cancer. This article provides a comprehensive review of the early and late phase trials that led to the regulatory approval of all five PD1- PDL-1 inhibitors in the corresponding cancer types. It presents available data on the combinations of PD1- PDL-1 inhibitors with other therapies (immunotherapy, targeted therapy and chemotherapy), the toxicity profile of the PD1- PDL-1 inhibitors and ongoing trials testing the efficacy of these agents in cancer types beyond those that have been addressed already. Finally, current and future challenges in the application of PD-1 and PD-L1 inhibitors are discussed with emphasis on the role of predictive biomarkers.
Collapse
Affiliation(s)
| | - Constantinos Alifieris
- Laboratory of Pharmacology, Clinical Pharmacology and Therapeutic Oncology Unit, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Clinical Pharmacology and Therapeutic Oncology Unit, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
114
|
Rosato RR, Dávila-González D, Choi DS, Qian W, Chen W, Kozielski AJ, Wong H, Dave B, Chang JC. Evaluation of anti-PD-1-based therapy against triple-negative breast cancer patient-derived xenograft tumors engrafted in humanized mouse models. Breast Cancer Res 2018; 20:108. [PMID: 30185216 PMCID: PMC6125882 DOI: 10.1186/s13058-018-1037-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022] Open
Abstract
Background Breast cancer has been considered not highly immunogenic, and few patients benefit from current immunotherapies. However, new strategies are aimed at changing this paradigm. In the present study, we examined the in vivo activity of a humanized anti-programmed cell death protein 1 (anti-PD-1) antibody against triple-negative breast cancer (TNBC) patient-derived xenograft (PDX) tumor models. Methods To circumvent some of the limitations posed by the lack of appropriate animal models in preclinical studies of immunotherapies, partially human leukocyte antigen-matched TNBC PDX tumor lines from our collection, as well as human melanoma cell lines, were engrafted in humanized nonobese diabetic/severe combined immunodeficiency IL2Rγnull (hNSG) mice obtained by intravenous injection of CD34+ hematopoietic stem cells into nonlethally irradiated 3–4-week-old mice. After both PDXs and melanoma cell xenografts reached ~ 150–200 mm3, animals were treated with humanized anti-PD-1 antibody or anti-CTLA-4 and evaluated for tumor growth, survival, and potential mechanism of action. Results Human CD45+, CD20+, CD3+, CD8+, CD56+, CD68+, and CD33+ cells were readily identified in blood, spleen, and bone marrow collected from hNSG, as well as human cytokines in blood and engrafted tumors. Engraftment of TNBC PDXs in hNSG was high (~ 85%), although they grew at a slightly slower pace and conserved their ability to generate lung metastasis. Human CD45+ cells were detectable in hNSG-harbored PDXs, and consistent with clinical observations, anti-PD-1 antibody therapy resulted in both a significant reduction in tumor growth and increased survival in some of the hNSG PDX tumor lines, whereas no such effects were observed in the corresponding non-hNSG models. Conclusions This study provides evidence associated with anti-PD-1 immunotherapy against TNBC tumors supporting the use of TNBC PDXs in humanized mice as a model to overcome some of the technical difficulties associated with the preclinical investigation of immune-based therapies. Electronic supplementary material The online version of this article (10.1186/s13058-018-1037-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Roberto R Rosato
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA.
| | | | - Dong Soon Choi
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Wei Qian
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Wen Chen
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Anthony J Kozielski
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Helen Wong
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Bhuvanesh Dave
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Jenny C Chang
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| |
Collapse
|
115
|
Zerdes I, Matikas A, Bergh J, Rassidakis GZ, Foukakis T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations. Oncogene 2018; 37:4639-4661. [PMID: 29765155 PMCID: PMC6107481 DOI: 10.1038/s41388-018-0303-3] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/27/2018] [Accepted: 04/13/2018] [Indexed: 02/06/2023]
Abstract
The programmed death protein 1 (PD-1) and its ligand (PD-L1) represent a well-characterized immune checkpoint in cancer, effectively targeted by monoclonal antibodies that are approved for routine clinical use. The regulation of PD-L1 expression is complex, varies between different tumor types and occurs at the genetic, transcriptional and post-transcriptional levels. Copy number alterations of PD-L1 locus have been reported with varying frequency in several tumor types. At the transcriptional level, a number of transcriptional factors seem to regulate PD-L1 expression including HIF-1, STAT3, NF-κΒ, and AP-1. Activation of common oncogenic pathways such as JAK/STAT, RAS/ERK, or PI3K/AKT/MTOR, as well as treatment with cytotoxic agents have also been shown to affect tumoral PD-L1 expression. Correlative studies of clinical trials with PD-1/PD-L1 inhibitors have so far shown markedly discordant results regarding the value of PD-L1 expression as a marker of response to treatment. As the indications for immune checkpoint inhibition broaden, understanding the regulation of PD-L1 in cancer will be of utmost importance for defining its role as predictive marker but also for optimizing strategies for cancer immunotherapy. Here, we review the current knowledge of PD-L1 regulation, and its use as biomarker and as therapeutic target in cancer.
Collapse
Affiliation(s)
- Ioannis Zerdes
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - George Z Rassidakis
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden.
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
116
|
Everett AS, De Los Santos JF, Boggs DH. The Evolving Role of Postmastectomy Radiation Therapy. Surg Clin North Am 2018; 98:801-817. [DOI: 10.1016/j.suc.2018.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
117
|
Moore-Smith L, Forero-Torres A, Stringer-Reasor E. Future Developments in Neoadjuvant Therapy for Triple-Negative Breast Cancer. Surg Clin North Am 2018; 98:773-785. [PMID: 30005773 DOI: 10.1016/j.suc.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Breast cancer is the 2nd leading cause of cancer-related death in women in the United States. In general, advances in targeted treatment for breast cancer have improved over the last twenty years, except in the triple-negative breast cancer (TNBC) subtype. TNBC is an aggressive breast cancer subtype with limited treatment options as compared to hormone positive breast cancers. Recently, genomic profiling of TNBC shows promise in aiding clinicians to develop personalized targeted agents. Prioritizing novel molecular-based therapies in the neoadjuvant setting may help investigators understand mechanisms of resistance and ultimately improve patient outcomes in TNBC.
Collapse
Affiliation(s)
- Lakisha Moore-Smith
- Department of Medicine, Brookwood Baptist Health - Princeton, 833 Princeton Avenue, POB III Suite 200, Birmingham, AL 35211-1311, USA
| | - Andres Forero-Torres
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2517, Birmingham, AL 35294-3300, USA
| | - Erica Stringer-Reasor
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2501, Birmingham, AL 35294-3300, USA.
| |
Collapse
|
118
|
Bartsch R, Bergen E, Galid A. Current concepts and future directions in neoadjuvant chemotherapy of breast cancer. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2018; 11:199-203. [PMID: 30220926 PMCID: PMC6132793 DOI: 10.1007/s12254-018-0421-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/10/2018] [Indexed: 12/21/2022]
Abstract
Preoperative administration of chemotherapy is a widespread treatment approach in early stage breast cancer whenever chemotherapy is indicated in principle. In addition, neoadjuvant treatment is today regarded as the preferred way of delivering systemic therapy in triple-negative and HER2-positive breast cancer. While preoperative chemotherapy allows for disease downstaging and increases breast conservation rates, achieving pathologic complete remission (pCR) is usually regarded as the most pertinent aim as pCR predicts for improved long-term outcome in high-risk breast cancer subtypes. A multitude of clinical trials therefore have focused on strategies to increase pCR rates. This short review summarizes outcomes of selected studies investigating the addition of further chemotherapeutic drugs or biologically targeted agents to standard regimens and provides an overview of novel strategies currently under clinical evaluation.
Collapse
Affiliation(s)
- Rupert Bartsch
- 1Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Elisabeth Bergen
- 1Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Arik Galid
- 2Department of Gynaecology, Hanusch Hospital, Vienna, Austria
| |
Collapse
|
119
|
Wein L, Luen SJ, Savas P, Salgado R, Loi S. Checkpoint blockade in the treatment of breast cancer: current status and future directions. Br J Cancer 2018; 119:4-11. [PMID: 29808015 PMCID: PMC6035268 DOI: 10.1038/s41416-018-0126-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 04/03/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022] Open
Abstract
There is now accumulating evidence that the host immune system plays an important role in influencing response to treatment and prognosis in breast cancer. Immunotherapy with immune checkpoint inhibitors is a promising and rapidly growing field of interest in many solid tumours, including breast cancer. Trials to date have largely focused on metastatic triple-negative disease, a genomically unstable subtype of breast cancer that is believed to be the most immunogenic and following the development of treatment resistance, has limited treatment options and a particularly poor prognosis. Both checkpoint inhibitor monotherapy and combinations with chemotherapy are being investigated. In this review, we discuss the current evidence for PD-1/PD-L1 blockade in metastatic triple-negative breast cancer (TNBC), HER2+ breast cancer and ER+ disease, as well as the emerging evidence for use in the early-stage (neoadjuvant) setting. We also propose potential ways of improving responses to checkpoint blockade in breast cancer.
Collapse
Affiliation(s)
- Lironne Wein
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Stephen J Luen
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Peter Savas
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Roberto Salgado
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Pathology/GZA, Antwerp, Belgium
| | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
120
|
de la Cruz-Merino L, Palazón-Carrión N, Henao-Carrasco F, Nogales-Fernández E, Álamo-de la Gala M, Vallejo-Benítez A, Chiesa M, Sánchez-Margalet V. New horizons in breast cancer: the promise of immunotherapy. Clin Transl Oncol 2018; 21:117-125. [PMID: 29916188 DOI: 10.1007/s12094-018-1907-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022]
Abstract
Immunology and immunotherapy of cancer is an expanding field in oncology, with recent great achievements obtained through the new successful approaches implemented to circumvent immune evasion, which is undoubtedly considered a novel hallmark of cancer. Translational research in this topic has revealed targets that can be modulated in the clinical setting with new compounds and strategies. Like most of the tumors, breast cancer is considered a complex and heterogeneous disease in which host immune responses have been also recently demonstrated of critical relevance. T infiltrating lymphocyte measurement is suggested as a powerful new tool necessary to predict early breast cancer evolution, especially for the her2-positive and triple-negative subtypes. Other biomarkers in tissue and peripheral blood are under intense scrutiny to ascertain their eventual role as prognostic and/or predictive factors. This background has fueled the interest in developing clinical research strategies to test activity of modern immunotherapy in breast cancer, which constitutes the main focus of this review.
Collapse
Affiliation(s)
- L de la Cruz-Merino
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain.
| | - N Palazón-Carrión
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain
| | - F Henao-Carrasco
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain
| | - E Nogales-Fernández
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain
| | - M Álamo-de la Gala
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain
| | - A Vallejo-Benítez
- Pathology Department, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain
| | - M Chiesa
- Spanish Breast Cancer Research Group (GEICAM), 28703, Madrid, Spain
| | - V Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Hospital Universitario Virgen Macarena, Avda Dr Fedriani s/n, 41009, Seville, Spain
| | | |
Collapse
|
121
|
Abstract
An effective antitumor immune response requires interaction between cells of the adaptive and innate immune system. Three key elements are required: generation of activated tumor-directed T cells, infiltration of activated T cells into the tumor microenvironment, and killing of tumor cells by activated T cells. Tumor immune evasion can occur as a result of the disruption of each of these three key T cell activities, resulting in three distinct cancer-immune phenotypes. The immune inflamed phenotype, characterized by the presence of a robust tumor immune infiltrate, suggests impaired activated T cell killing of tumor cells related to the presence of inhibitory factors. Programmed death receptor-1 (PD-1) is an inhibitory transmembrane protein expressed on T cells, B cells, and NK cells. The interaction between PD-1 and its ligands (PD-L1/L2) functions as an immune checkpoint against unrestrained cytotoxic T effector cell activity-it promotes peripheral T effector cell exhaustion and conversion of T effector cells to immunosuppressive T regulatory (Treg) cells. Immune checkpoint inhibitors, which block the PD-1/PD-L1 axis and reactivate cytotoxic T effector cell function, are actively being investigated for the treatment of breast cancer.
Collapse
Affiliation(s)
- April Swoboda
- Section of Hematology/Oncology, 5841 S. Maryland Ave, MC 2115, Chicago, IL, 60616, USA
| | - Rita Nanda
- Section of Hematology/Oncology, 5841 S. Maryland Ave, MC 2115, Chicago, IL, 60616, USA.
| |
Collapse
|
122
|
Groheux D. Now Is the Time to Use 18F-FDG PET/CT to Optimize Neoadjuvant Treatment in Triple-Negative Breast Cancer! J Nucl Med 2018; 59:863-864. [DOI: 10.2967/jnumed.118.210922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/12/2018] [Indexed: 11/16/2022] Open
|
123
|
Makhoul I, Atiq M, Alwbari A, Kieber-Emmons T. Breast Cancer Immunotherapy: An Update. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2018; 12:1178223418774802. [PMID: 29899661 PMCID: PMC5985550 DOI: 10.1177/1178223418774802] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 04/08/2018] [Indexed: 12/22/2022]
Abstract
The immune system plays a major role in cancer surveillance. Harnessing its power to treat many cancers is now a reality that has led to cures in hopeless situations where no other solutions were available from traditional anticancer drugs. These spectacular achievements rekindled the oncology community's interest in extending the benefits to all cancers including breast cancer. The first section of this article reviews the biological foundations of the immune response to different subtypes of breast cancer and the ways cancer may overcome the immune attack leading to cancer disease. The second section is dedicated to the actual immune treatments including breast cancer vaccines, checkpoint inhibitors, monoclonal antibodies, and the "unconventional" immune role of chemotherapy.
Collapse
Affiliation(s)
- Issam Makhoul
- Divisions of Hematology and Medical Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mohammad Atiq
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ahmed Alwbari
- Divisions of Hematology and Medical Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Thomas Kieber-Emmons
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
124
|
Telli ML, Stover DG, Loi S, Aparicio S, Carey LA, Domchek SM, Newman L, Sledge GW, Winer EP. Homologous recombination deficiency and host anti-tumor immunity in triple-negative breast cancer. Breast Cancer Res Treat 2018; 171:21-31. [PMID: 29736741 DOI: 10.1007/s10549-018-4807-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is associated with worse outcomes relative to other breast cancer subtypes. Chemotherapy remains the standard-of-care systemic therapy for patients with localized or metastatic disease, with few biomarkers to guide benefit. METHODS We will discuss recent advances in our understanding of two key biological processes in TNBC, homologous recombination (HR) DNA repair deficiency and host anti-tumor immunity, and their intersection. RESULTS Recent advances in our understanding of homologous recombination (HR) deficiency, including FDA approval of PARP inhibitor olaparib for BRCA1 or BRCA2 mutation carriers, and host anti-tumor immunity in TNBC offer potential for new and biomarker-driven approaches to treat TNBC. Assays interrogating HR DNA repair capacity may guide treatment with agents inducing or targeting DNA damage repair. Tumor infiltrating lymphocytes (TILs) are associated with improved prognosis in TNBC and recent efforts to characterize infiltrating immune cell subsets and activate host anti-tumor immunity offer promise, yet challenges remain particularly in tumors lacking pre-existing immune infiltrates. Advances in these fields provide potential biomarkers to stratify patients with TNBC and guide therapy: induction of DNA damage in HR-deficient tumors and activation of existing or recruitment of host anti-tumor immune cells. Importantly, these advances provide an opportunity to guide use of existing therapies and development of novel therapies for TNBC. Efforts to combine therapies that exploit HR deficiency to enhance the activity of immune-directed therapies offer promise. CONCLUSIONS HR deficiency remains an important biomarker target and potentially effective adjunct to enhance immunogenicity of 'immune cold' TNBCs.
Collapse
Affiliation(s)
- M L Telli
- Stanford University School of Medicine, Stanford, CA, USA
| | - D G Stover
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA. .,Ohio State University Comprehensive Cancer Center, Biomedical Research Tower, Room 512, 460 West 12th Ave, Columbus, OH, 43210, USA.
| | - S Loi
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - S Aparicio
- University of British Columbia, Vancouver, BC, Canada
| | - L A Carey
- University of North Carolina, Chapel Hill, NC, USA
| | - S M Domchek
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - L Newman
- Henry Ford Hospital, Detroit, MI, USA
| | - G W Sledge
- Stanford University School of Medicine, Stanford, CA, USA
| | - E P Winer
- Dana Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
125
|
Park JH, Ahn JH, Kim SB. How shall we treat early triple-negative breast cancer (TNBC): from the current standard to upcoming immuno-molecular strategies. ESMO Open 2018; 3:e000357. [PMID: 29765774 PMCID: PMC5950702 DOI: 10.1136/esmoopen-2018-000357] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2018] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a long-lasting orphan disease in terms of little therapeutic progress during the past several decades and still the standard of care remains chemotherapy. Experimental discovery of molecular signatures including the ‘BRCAness’ highlighted the innate heterogeneity of TNBC, generating the diversity of TNBC phenotypes. As it contributes to enhancing genomic instability, it has widened the therapeutic spectrum of TNBC. In particular, unusual sensitivity to DNA damaging agents was denoted in patients with BRCA deficiency, suggesting therapeutic benefit from platinum and poly(ADP-ribose) polymerase inhibitors. However, regardless of enriched chemosensitivity and immunogenicity, majority of patients with TNBC still suffer from dismal clinical outcomes including early relapse and metastatic spread. Therefore, efforts into more precise and personalised treatment are critical at this point. Accordingly, the advance of multiomics has revealed novel actionable targets including PI3K-Akt-mTOR and epidermal growth factor receptor signalling pathways, which might actively participate in modulating the chemosensitivity and immune system. Also, TNBC has long been considered a potential protagonist of immunotherapy in breast cancer, supported by abundant tumour-infiltrating lymphocytes and heterogeneous tumour microenvironment. Despite that, earlier studies showed somewhat unsatisfactory results of monotherapy with immune-checkpoint inhibitors, consistently durable responses in responders were noteworthy. Based on these results, further combinatorial trials either with other chemotherapy or targeted agents are underway. Incorporating immune-molecular targets into combination as well as refining the standard chemotherapy might be the key to unlock the future of TNBC. In this review, we share the current and upcoming treatment options of TNBC in the framework of scientific and clinical data, especially focusing on early stage of TNBC.
Collapse
Affiliation(s)
- Ji Hyun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea; Department of Hemato-Oncology, Konkuk Medical Center, University of Konkuk College of Medicine, Gwangjin-gu, Seoul, Korea
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea.
| |
Collapse
|
126
|
Abstract
PURPOSE OF REVIEW The treatment landscape for many cancers has dramatically changed with the development of checkpoint inhibitors. This article will review the literature concerning the use of checkpoint inhibitors in breast cancer. RECENT FINDINGS The histological subtype of BC with the strongest signal of efficacy has been triple-negative breast cancer (TNBC). Early trials of single-agent checkpoint inhibitors did not demonstrate a uniformly positive signal. Clinical studies suggest response rates between 5 and 10% in pretreated patients and roughly 20-25% for untreated advanced TNBC. However, in the small subset of patients who do respond, the response is often durable. More encouraging results have been reported with their use in combination with chemotherapy in the neoadjuvant setting. Larger phase III studies are underway to confirm these earlier findings. An immune-directed therapeutic approach for the management of BC is underway, and it is likely that combination therapy will be required to achieve a level of efficacy worthy of use in the BC treatment paradigm. These agents are not without both economic and clinical toxicity; therefore, it is imperative that we identify patients most likely to benefit from these therapies through well-designed biologically plausible clinical studies and by evaluating novel combinatorial approaches with informative biomarker driven correlative studies.
Collapse
|
127
|
Abstract
PURPOSE OF REVIEW This review summarizes current immunotherapies in breast cancer, with an emphasis on immune checkpoint inhibitors and vaccines. RECENT FINDINGS Combination immunotherapy with checkpoint inhibitors and cytotoxic therapies have shown promising results. Active clinical trials are ongoing in both early stage and metastatic settings for triple negative, HER2+, and hormone-positive breast cancer patients. SUMMARY Ongoing challenges remain in defining biomarkers that predict response to immunotherapy, determining the optimal combination immunotherapies, and enhancing the immunogenicity of breast cancer subtypes.
Collapse
Affiliation(s)
- Zishuo I Hu
- 1Mount Sinai Health System, Icahn School of Medicine, New York, NY USA
| | - Heather L McArthur
- 2Department of Medicine, Breast Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, 1S35, Los Angeles, CA 90048 USA
| |
Collapse
|
128
|
Abstract
OPINION STATEMENT Triple-negative breast cancer (TNBC) accounts for 15% of all breast cancers and is associated with poor long-term outcomes compared to other breast cancer subtypes. Currently, chemotherapy remains the main modality of treatment for early-stage TNBC, as there is no approved targeted therapy for this subtype. The biologic heterogeneity of TNBC has hindered the development and evaluation of novel agents, but recent advancements in subclassifying TNBC have paved the way for further investigation of more effective systemic therapies, including cytotoxic and targeted agents. TNBC is enriched for germline BRCA mutation and for somatic deficiencies in homologous recombination DNA repair, the so-called "BRCAness" phenotype. Together, germline BRCA mutations and BRCAness are promising biomarkers of susceptibility to DNA-damaging therapy. Various investigational approaches are consequently being investigated in early-stage TNBC, including immune checkpoint inhibitors, platinum compounds, PI3K pathway inhibitors, and androgen receptor inhibitors. Due to the biological diversity found within TNBC, patient selection based on molecular biomarkers could aid the design of early-phase clinical trials, ultimately accelerating the clinical application of effective new agents. TNBC is an aggressive breast cancer subtype, for which multiple targeted approaches will likely be required for patient outcomes to be substantially improved.
Collapse
|
129
|
Heymach J, Krilov L, Alberg A, Baxter N, Chang SM, Corcoran RB, Dale W, DeMichele A, Magid Diefenbach CS, Dreicer R, Epstein AS, Gillison ML, Graham DL, Jones J, Ko AH, Lopez AM, Maki RG, Rodriguez-Galindo C, Schilsky RL, Sznol M, Westin SN, Burstein H. Clinical Cancer Advances 2018: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology. J Clin Oncol 2018; 36:1020-1044. [PMID: 29380678 DOI: 10.1200/jco.2017.77.0446] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A MESSAGE FROM ASCO'S PRESIDENT I remember when ASCO first conceived of publishing an annual report on the most transformative research occurring in cancer care. Thirteen reports later, the progress we have chronicled is remarkable, and this year is no different. The research featured in ASCO's Clinical Cancer Advances 2018 report underscores the impressive gains in our understanding of cancer and in our ability to tailor treatments to tumors' genetic makeup. The ASCO 2018 Advance of the Year, adoptive cell immunotherapy, allows clinicians to genetically reprogram patients' own immune cells to find and attack cancer cells throughout the body. Chimeric antigen receptor (CAR) T-cell therapy-a type of adoptive cell immunotherapy-has led to remarkable results in young patients with acute lymphoblastic leukemia (ALL) and in adults with lymphoma and multiple myeloma. Researchers are also exploring this approach in other types of cancer. This advance would not be possible without robust federal investment in cancer research. The first clinical trial of CAR T-cell therapy in children with ALL was funded, in part, by grants from the National Cancer Institute (NCI), and researchers at the NCI Center for Cancer Research were the first to report on possible CAR T-cell therapy for multiple myeloma. These discoveries follow decades of prior research on immunology and cancer biology, much of which was supported by federal dollars. In fact, many advances that are highlighted in the 2018 Clinical Cancer Advances report were made possible thanks to our nation's support for biomedical research. Funding from the US National Institutes of Health and the NCI helps researchers pursue critical patient care questions and addresses vital, unmet needs that private industry has little incentive to take on. Federally supported cancer research generates the biomedical innovations that fuel the development and availability of new and improved treatments for patients. We need sustained federal research investment to accelerate the discovery of the next generation of cancer treatments. Another major trend in this year's report is progress in precision medicine approaches to treat cancer. Although precision medicine offers promise to people with cancer and their families, that promise is only as good as our ability to make these treatments available to all patients. My presidential theme, "Delivering Discoveries: Expanding the Reach of Precision Medicine," focuses on tackling this formidable challenge so that new targeted therapies are accessible to anyone who faces a cancer diagnosis. By improving access to high-quality care, harnessing big data on patient outcomes from across the globe, and pursuing innovative clinical trials, I am optimistic that we will speed the delivery of these most promising treatments to more patients. Sincerely, Bruce E. Johnson, FASCO ASCO President, 2017 to 2018.
Collapse
Affiliation(s)
- John Heymach
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Lada Krilov
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Anthony Alberg
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Nancy Baxter
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Susan Marina Chang
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Ryan B Corcoran
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - William Dale
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Angela DeMichele
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Catherine S Magid Diefenbach
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Robert Dreicer
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Andrew S Epstein
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Maura L Gillison
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - David L Graham
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Joshua Jones
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Andrew H Ko
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Ana Maria Lopez
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Robert G Maki
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Carlos Rodriguez-Galindo
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Richard L Schilsky
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Mario Sznol
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Shannon Neville Westin
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Harold Burstein
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| |
Collapse
|
130
|
Kwa MJ, Adams S. Checkpoint inhibitors in triple-negative breast cancer (TNBC): Where to go from here. Cancer 2018; 124:2086-2103. [DOI: 10.1002/cncr.31272] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/22/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Maryann J. Kwa
- Laura and Issac Perlmutter Cancer Center; NYU Langone Medical Center; New York New York
| | - Sylvia Adams
- Laura and Issac Perlmutter Cancer Center; NYU Langone Medical Center; New York New York
| |
Collapse
|
131
|
Emens LA. Breast Cancer Immunotherapy: Facts and Hopes. Clin Cancer Res 2018; 24:511-520. [PMID: 28801472 PMCID: PMC5796849 DOI: 10.1158/1078-0432.ccr-16-3001] [Citation(s) in RCA: 518] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/07/2017] [Accepted: 08/08/2017] [Indexed: 12/21/2022]
Abstract
Immunotherapy is revolutionizing the management of multiple solid tumors, and early data have revealed the clinical activity of programmed cell death-1/programmed death ligand-1 (PD-1/PD-L1) antagonists in small numbers of patients with metastatic breast cancer. Clinical activity appears more likely if the tumor is triple negative, PD-L1+, and/or harbors higher levels of tumor-infiltrating leukocytes. Responses to atezolizumab and pembrolizumab appear to be durable in metastatic triple-negative breast cancer (TNBC), suggesting that these agents may transform the lives of responding patients. Current clinical efforts are focused on developing immunotherapy combinations that convert nonresponders to responders, deepen those responses that do occur, and surmount acquired resistance to immunotherapy. Identifying biomarkers that can predict the potential for response to single-agent immunotherapy, identify the best immunotherapy combinations for a particular patient, and guide salvage immunotherapy in patients with progressive disease are high priorities for clinical development. Smart clinical trials testing rational immunotherapy combinations that include robust biomarker evaluations will accelerate clinical progress, moving us closer to effective immunotherapy for almost all patients with breast cancer. Clin Cancer Res; 24(3); 511-20. ©2017 AACR.
Collapse
Affiliation(s)
- Leisha A Emens
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
132
|
Taube JM, Galon J, Sholl LM, Rodig SJ, Cottrell TR, Giraldo NA, Baras AS, Patel SS, Anders RA, Rimm DL, Cimino-Mathews A. Implications of the tumor immune microenvironment for staging and therapeutics. Mod Pathol 2018; 31:214-234. [PMID: 29192647 PMCID: PMC6132263 DOI: 10.1038/modpathol.2017.156] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/06/2017] [Accepted: 09/24/2017] [Indexed: 02/06/2023]
Abstract
Characterizing the tumor immune microenvironment enables the identification of new prognostic and predictive biomarkers, the development of novel therapeutic targets and strategies, and the possibility to guide first-line treatment algorithms. Although the driving elements within the tumor microenvironment of individual primary organ sites differ, many of the salient features remain the same. The presence of a robust antitumor milieu characterized by an abundance of CD8+ cytotoxic T-cells, Th1 helper cells, and associated cytokines often indicates a degree of tumor containment by the immune system and can even lead to tumor elimination. Some of these features have been combined into an 'Immunoscore', which has been shown to complement the prognostic ability of the current TNM staging for early stage colorectal carcinomas. Features of the immune microenvironment are also potential therapeutic targets, and immune checkpoint inhibitors targeting the PD-1/PD-L1 axis are especially promising. FDA-approved indications for anti-PD-1/PD-L1 are rapidly expanding across numerous tumor types and, in certain cases, are accompanied by companion or complimentary PD-L1 immunohistochemical diagnostics. Pathologists have direct visual access to tumor tissue and in-depth knowledge of the histological variations between and within tumor types and thus are poised to drive forward our understanding of the tumor microenvironment. This review summarizes the key components of the tumor microenvironment, presents an overview of and the challenges with PD-L1 antibodies and assays, and addresses newer candidate biomarkers, such as CD8+ cell density and mutational load. Characteristics of the local immune contexture and current pathology-related practices for specific tumor types are also addressed. In the future, characterization of the host antitumor immune response using multiplexed and multimodality biomarkers may help predict which patients will respond to immune-based therapies.
Collapse
Affiliation(s)
- Janis M Taube
- Department of Dermatology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD
- Department of Pathology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, Centre de Recherche des Cordeliers, Paris, France
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tricia R Cottrell
- Department of Pathology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
| | - Nicolas A Giraldo
- Department of Dermatology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD
- Department of Pathology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
| | - Alexander S Baras
- Department of Pathology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
| | - Sanjay S Patel
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Robert A Anders
- Department of Pathology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Ashley Cimino-Mathews
- Department of Pathology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University SOM and Bloomberg-Kimmel Institute for Immunotherapy, Baltimore, MD, USA
| |
Collapse
|
133
|
Denkert C, Wienert S, Klauschen F. Analyzing the Immunological Landscape of a Tumor—Heterogeneity of Immune Infiltrates in Breast Cancer as a New Prognostic Indicator. J Natl Cancer Inst 2018. [DOI: 10.1093/jnci/djx188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Carsten Denkert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Charité, Berlin, Germany
| | - Stephan Wienert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frederick Klauschen
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Charité, Berlin, Germany
| |
Collapse
|
134
|
Voorwerk L, Kat M, Kok M. Towards predictive biomarkers for immunotherapy response in breast cancer patients. BREAST CANCER MANAGEMENT 2018. [DOI: 10.2217/bmt-2017-0014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immunotherapy using anti-PD(L)1 has revolutionized treatment for various tumor types. Early data have shown durable responses in a small subgroup of breast cancer patients. So far, the response rates appear higher for breast tumors that are triple negative, PDL1-positive and/or harbor high levels of immune cells. Both comprehensive analyses of the breast tumor microenvironment and exploiting research on biomarkers in other cancer types, such as melanoma and lung cancer, may contribute to the discovery of accurate biomarkers to select breast cancer patients for immunotherapy. Here we summarize key features of the breast tumor microenvironment as well as putative predictive biomarkers established in other tumor types. Insights from both fields can guide future studies to enable personalized breast cancer immunotherapy.
Collapse
Affiliation(s)
- Leonie Voorwerk
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute Amsterdam, Amsterdam, The Netherlands
| | - Marije Kat
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute Amsterdam, Amsterdam, The Netherlands
| | - Marleen Kok
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
135
|
Checkpoint inhibitors in breast cancer – Current status. Cancer Treat Rev 2018; 63:122-134. [DOI: 10.1016/j.ctrv.2017.12.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
|
136
|
Nixon MJ, Balko JM. Biomarkers for assessing the effectiveness of immunotherapy in breast cancer. Biomark Med 2018; 12:97-100. [PMID: 29338306 DOI: 10.2217/bmm-2017-0408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mellissa J Nixon
- Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232-6307, USA
| | - Justin M Balko
- Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232-6307, USA
| |
Collapse
|
137
|
Ye JC, Formenti SC. Integration of radiation and immunotherapy in breast cancer - Treatment implications. Breast 2018; 38:66-74. [PMID: 29253718 DOI: 10.1016/j.breast.2017.12.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/15/2017] [Accepted: 12/04/2017] [Indexed: 01/18/2023] Open
Abstract
Radiation therapy (RT) has been successfully used in the treatment of breast cancer (BC) for over a century. While historically thought to be immunosuppressive, new data have shown that RT can work together with the immune system to eliminate cancer. It can cause immunogenic cell death and facilitate tumor neoantigen presentation and cross-priming of tumor-specific T-lymphocytes, turning irradiated tumor into an in-situ vaccine. Unfortunately, due to various immune escape mechanism put in place by the tumor, RT alone rarely results in a systemic response of metastatic disease sites (known as the abscopal effect). Immunotherapy, a series of agents designed to stimulate the immune system in order to generate tumor-specific immune response, is showing promise in treatment of various cancers, including BC, and can be an ideal complement to RT in stimulating a systemic immune response to reject the tumor cells. This review discusses the mechanisms in which RT can trigger an immune response for tumor rejection, and provide emerging preclinical and clinical data of combination immunoradiotherapy, and its potential in treating BC.
Collapse
Affiliation(s)
- Jason C Ye
- USC Keck School of Medicine, Los Angeles, CA, USA
| | | |
Collapse
|
138
|
Vidula N, Rugo HS. Emerging data on improving response to hormone therapy: the role of novel targeted agents. Expert Rev Anticancer Ther 2017; 18:3-18. [PMID: 29192520 DOI: 10.1080/14737140.2018.1412259] [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: 12/19/2022]
Abstract
INTRODUCTION Hormone receptor positive (HR+) breast cancer represents the most common subtype of breast cancer. Metastatic HR+ breast cancer may develop resistance to standard hormone therapies, arising from genomic alterations in the estrogen receptor and/or upregulation of other signal transduction pathways. Areas covered: In this review, we discuss hormone resistance and strategies to overcome it, from the pre-clinical and clinical perspectives. This review includes a discussion of inhibition of the PI3K/AKT/mTOR, CDK 4/6, histone deacetylation, fibroblast growth factor receptor, and immune pathways, based on review of relevant literature. Expert commentary: Several emerging novel therapies to improve the response to hormone therapy are approved or are in development. The most promising agents at present are inhibitors of CDK 4/6 and mTOR, which have already been incorporated into treatment in the advanced stage setting and are under study for early stage disease.
Collapse
Affiliation(s)
- Neelima Vidula
- a Division of Hematology and Oncology , Massachusetts General Hospital , Boston , MA , USA
| | - Hope S Rugo
- b Division of Hematology and Oncology , University of California San Francisco, Helen Diller Family Comprehensive Cancer Center , San Francisco , CA , USA
| |
Collapse
|
139
|
Swoboda A, Nanda R. Immunotherapy Approaches to Breast Cancer. CURRENT BREAST CANCER REPORTS 2017. [DOI: 10.1007/s12609-017-0252-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
140
|
Solinas C, Gombos A, Latifyan S, Piccart-Gebhart M, Kok M, Buisseret L. Targeting immune checkpoints in breast cancer: an update of early results. ESMO Open 2017; 2:e000255. [PMID: 29177095 PMCID: PMC5687552 DOI: 10.1136/esmoopen-2017-000255] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 08/21/2017] [Indexed: 12/16/2022] Open
Abstract
The immune tumour microenvironment has been shown to play a crucial role in the development and progression of cancer. Expression of gene signatures, reflecting immune activation, and the presence of tumour-infiltrating lymphocytes were associated with favourable outcomes in HER2-positive and triple-negative breast cancer. Recently, immunotherapy with immune checkpoint blockade induced long-lasting responses and improved survival in hard-to-treat malignancies (ie, melanoma and non-small cell lung cancer) and are changing treatment paradigms in a variety of neoplastic diseases. Immune checkpoint blockade has been evaluated in breast cancer, particularly in the triple-negative subtype, with promising results observed in monotherapy or in combination with chemotherapy in the metastatic and neoadjuvant settings. However, identification of patients who are most likely to benefit from immune checkpoint blockade remains challenging, with many patients not responding to treatments and a significant financial cost. The combination of immune checkpoint blockade with conventional cancer treatments such as chemotherapy, radiotherapy, targeted therapies or with other immunotherapies is a promising strategy to potentiate its efficacy in breast cancer although further research is required to effectively identify who will respond to these immunotherapies. In this review we report the most recent results that emerged from trials testing immune checkpoint blockade and potential predictive biomarkers and emphasise the new strategies that are under clinical development in breast cancer.
Collapse
Affiliation(s)
- Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrea Gombos
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sofiya Latifyan
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Martine Piccart-Gebhart
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Marleen Kok
- Department of Medical Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Laurence Buisseret
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
141
|
Wang C, Kar S, Lai X, Cai W, Arfuso F, Sethi G, Lobie PE, Goh BC, Lim LHK, Hartman M, Chan CW, Lee SC, Tan SH, Kumar AP. Triple negative breast cancer in Asia: An insider's view. Cancer Treat Rev 2017; 62:29-38. [PMID: 29154023 DOI: 10.1016/j.ctrv.2017.10.014] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/29/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
Abstract
While tremendous improvement has been made for the treatment of breast cancers, the treatment of triple negative breast cancer (TNBC) still remains a challenge due to its aggressive characteristics and limited treatment options. Most of the studies on TNBC were conducted in Western population and TNBC is reported to be more frequent in the African women. This review encapsulates the studies conducted on TNBC patients in Asian population and elucidates the similarities and differences between these two regions. The current treatment of TNBC includes surgery, radiotherapy and chemotherapy. In addition to the current chemotherapies, which mainly include cytotoxic agents, such as taxanes and anthracyclines, many clinical trials are investigating the potential use of other chemotherapy drugs, targeted therapeutics and combinational therapies to treat TNBC. Moreover, this review also integrates the studies involving novel markers, which will help us to dissect the pathologic process of TNBC and in turn facilitate the development of better treatment strategies to combat TNBC.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shreya Kar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xianning Lai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wanpei Cai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Peter E Lobie
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School at Shenzhen, Shenzhen, China
| | - Boon C Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore
| | - Lina H K Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; NUS Immunology Program, National University of Singapore, Singapore
| | - Mikael Hartman
- Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; Department of Surgery, National University Cancer Institute, National University Health System, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Ching W Chan
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore
| | - Soo C Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore
| | - Sing H Tan
- Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore; OncoCare Cancer Centre, Gleneagles Medical Centre, Singapore.
| | - Alan P Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
142
|
Cohen IJ, Blasberg R. Impact of the Tumor Microenvironment on Tumor-Infiltrating Lymphocytes: Focus on Breast Cancer. Breast Cancer (Auckl) 2017; 11:1178223417731565. [PMID: 28979132 PMCID: PMC5617083 DOI: 10.1177/1178223417731565] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022] Open
Abstract
Immunotherapy is revolutionizing cancer care across disciplines. The original success of immune checkpoint blockade in melanoma has already been translated to Food and Drug Administration-approved therapies in a number of other cancers, and a large number of clinical trials are underway in many other disease types, including breast cancer. Here, we review the basic requirements for a successful antitumor immune response, with a focus on the metabolic and physical barriers encountered by lymphocytes entering breast tumors. We also review recent clinical trials of immunotherapy in breast cancer and provide a number of interesting questions that will need to be answered for successful breast cancer immunotherapy.
Collapse
Affiliation(s)
- Ivan J Cohen
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Ronald Blasberg
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
143
|
Schlotter CM, Tietze L, Vogt U, Heinsen CV, Hahn A. Ki67 and lymphocytes in the pretherapeutic core biopsy of primary invasive breast cancer: positive markers of therapy response prediction and superior survival. Horm Mol Biol Clin Investig 2017; 32:/j/hmbci.ahead-of-print/hmbci-2017-0022/hmbci-2017-0022.xml. [PMID: 28937963 DOI: 10.1515/hmbci-2017-0022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/22/2017] [Indexed: 11/15/2022]
Abstract
Background Core needle biopsy plays a crucial role as diagnostic tool for BC. Both Ki67 and likely tumor-infiltrating lymphocytes (TILs) in the near future are determining the kind of systemic therapy. The role of TILs in BC is still an issue for clinical research, albeit preliminary results of neoadjuvant and adjuvant clinical studies already now highlight the crucial impact of TILs on therapy response and survival. Methods Evaluation of related publications (pubmed) and meeting abstracts (ASCO, SABCS). Results The monoclonal antibody Ki67 recognizing a nuclear antigene in proliferating cells is a positive marker of therapy response and superior survival. Endocrine responsive tumors of low proliferation (Ki67 < 14%/11%) respond to tamoxifen, in contrast postmenopausal tumors with higher proliferation respond better to aromatase-inhibitors. Pathological complete response (pCR)-rates increase in tumors with higher proliferation (Ki67 > 19%) vs. tumors with lower proliferation after neoadjuvant chemotherapy (NAC). pCR-rates of up to 60% can be seen in TNBC and HR-, HER2+BC, lower pCR-rates, however, in HR+, HER2- BC. Increased stromal TILs are found in 30% of TNBC and in 19% of HR-, HER2+BC. The percentage of TILs is a significant independent parameter for pCR after NAC. Lymphocyte-predominant BC (LPBC) respond with higher pCR-rates than non-LPBC or tumors without any TILs. Increased TILs in TN and HR-, HER2+ subtypes predict benefit from addition of carboplatin to NAC. TILs are also associated with improved DFS and OS among patients with TNBC and HR-, HER2+ BC. Conversly and interestingly increased TILs in patients with HR+, HER2-(luminal) BC are associated with a 10% higher risk of death per 10% increase of TILs. Interactions between immune system and cancer are complex. The cancer-immunity cycle characterizes these interactions. BC subtypes with higher number of mutations such as TNBC and HR-, HER2+BC are considered to provide a raising number of tumor-associated antigens, thereby capable to build up a higher endogenous immune response. TILs may serve as surrogate marker of both an existing endogenous immune response and the probability to respond to cancer immune therapies. As cancer co-opt immune checkpoint-pathways as a major mechanism of immune resistance, in particular, against cytotoxic T-cells, blockades of checkpoint-pathways by antibodies are one of the goals of the current cancer immunotherapy studies. Therapy studies with antigene-based strategies (vaccines) and antibodies against the immune checkpoints PD-1 and CTLA-4 and their inhibitory pathways in order to enhance cytotoxic T-cell activities against cancer cells with or without chemotherapy are underway. Conclusions It can be suggested that the use of multigene expression testing will increase in order to select more clearly primary HR+, HER2- BC patients with intermediate recurrence risk who likely may benefit from chemotherapy. Furthermore Ki67 and the multigene expression test Oncotype DX can act as dynamic markers to avoid cytostatic overtreatment and endocrine undertreatment. A data-derived optimal Ki67 cut point for pCR and DFS as well as OS is currently not feasible. The integration of stromal TILs into the immunohisto-pathological report after their evaluation has been standardized is likely helpful to determine patients who profit by additional carboplatin chemotherapy. Oncologists need an enlarged information about the tumor-microenvironment in future. The preliminary results of current BC immunotherapy studies are encouraging.
Collapse
Affiliation(s)
- Claus M Schlotter
- Department of Obstetrics and Gynecology, Klinikum-Mittelbaden, Breast Center, D-76532 Baden-Baden, Balger-Str. 50, Germany, Phone: +4917620123472
| | - Lothar Tietze
- Institute of Pathology, Ortenau Klinikum Lahr-Ettenheim, Lahr, Germany
| | - Ulf Vogt
- European Laboratory Association, Ibbenbueren, Germany
| | - Carlos Villena Heinsen
- Department of Obstetrics and Gynecology, Luzerner Kantonsspital Sursee, Sursee, Switzerland
| | - Antje Hahn
- Department of Obstetrics and Gynecology, Breast Center, Klinikum-Mittelbaden, Baden-Baden, Germany
| |
Collapse
|
144
|
Saraiva DP, Guadalupe Cabral M, Jacinto A, Braga S. How many diseases is triple negative breast cancer: the protagonism of the immune microenvironment. ESMO Open 2017; 2:e000208. [PMID: 29018573 PMCID: PMC5604720 DOI: 10.1136/esmoopen-2017-000208] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/22/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a type of breast cancer (BC) that does not express the oestrogen and the progesterone receptors and the human epidermal growth factor receptor type 2 (HER2). Since there are no positive markers to reliably classify TNBC, these tumours are not yet treated with targeted therapies. Perhaps for this reason they are the most aggressive form of breast carcinomas. However, the clinical observation that these patients do not carry a uniformly dismal prognosis, coupled with data coming from pathology and epidemiology, suggests that this negative definition is not capturing a single clinical entity, but several. We critically evaluate this evidence in this paper, reviewing clinical and epidemiological data and new studies that aim to subclassify TNBC. Moreover, evidence on the role of tumour infiltrating lymphocytes (TILs) on TNBC progression, response to chemotherapy and patient outcome have been published. The heterogeneity, observed even at TILs level, highlights the idea that TNBC is much more than a single disease with a unique treatment. The exploration of the immune environment present at the tumour site could indeed help in answering the question 'How many diseases is TNBC' and will help to define prognosis and eventually develop new therapies, by stimulating the immune effector cells or by inhibiting immunological repressor molecules. In this review, we focus on the prospect of the patient's diverse immune signatures within the tumour as potential biomarkers and how they could be modulated to fight the disease.
Collapse
Affiliation(s)
- Diana P Saraiva
- CEDOC, Nova Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| | - M Guadalupe Cabral
- CEDOC, Nova Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| | - António Jacinto
- CEDOC, Nova Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| | - Sofia Braga
- CEDOC, Nova Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto CUF de Oncologia, Lisbon, Portugal
| |
Collapse
|
145
|
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: 81] [Impact Index Per Article: 11.6] [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
|
146
|
Harshman L, Drake C, Haas N, Manola J, Puligandla M, Signoretti S, Cella D, Gupta R, Bhatt R, Van Allen E, Lara P, Choueiri T, Kapoor A, Heng D, Shuch B, Jewett M, George D, Michaelson D, Carducci M, McDermott D, Allaf M. Transforming the Perioperative Treatment Paradigm in Non-Metastatic RCC-A Possible Path Forward. KIDNEY CANCER 2017; 1:31-40. [PMID: 30334002 PMCID: PMC6179104 DOI: 10.3233/kca-170010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In 2017, there is no adjuvant systemic therapy proven to increase overall survival in non-metastatic renal cell carcinoma (RCC). The anti-PD-1 antibody nivolumab improves overall survival in metastatic treatment refractory RCC and is generally tolerable. Mouse solid tumor models have revealed a benefit with a short course of neoadjuvant PD-1 blockade compared to adjuvant therapy. Two ongoing phase 2 studies of perioperative nivolumab in RCC patients have shown preliminary feasibility and safety with no surgical delays or complications. The recently opened PROSPER RCC trial (A Phase 3 RandOmized Study Comparing PERioperative Nivolumab vs. Observation in Patients with Localized Renal Cell Carcinoma Undergoing Nephrectomy; EA8143) will examine if the addition of perioperative nivolumab to radical or partial nephrectomy can improve clinical outcomes in patients with high risk localized and locally advanced RCC. With the goal of increasing cure and recurrence-free survival (RFS) rates in non-metastatic RCC, we are executing a three-pronged, multidisciplinary approach of presurgical priming with nivolumab followed by resection and adjuvant PD-1 blockade. We plan to enroll 766 patients with clinical stage ≥T2 or node positive M0 RCC of any histology in this global, randomized, unblinded, phase 3 National Clinical Trials Network study. The investigational arm will receive two doses of nivolumab 240 mg IV prior to surgery followed by adjuvant nivolumab for 9 months. The control arm will undergo the current standard of care: surgical resection followed by observation. Patients are stratified by clinical T stage, node positivity, and histology. The trial is powered to detect a 14.4% absolute benefit in the primary endpoint of RFS from the ASSURE historical control of 55.8% to 70.2% at 5 years (HR = 0.70). The study is also powered to detect a significant overall survival benefit (HR 0.67). Key safety, feasibility, and quality of life endpoints are incorporated. PROSPER RCC exemplifies team science with a host of planned correlative work to investigate the impact of the baseline immune milieu and changes after neoadjuvant priming on clinical outcomes.
Collapse
Affiliation(s)
- L.C. Harshman
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - C.G. Drake
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - N.B. Haas
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - J. Manola
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - M. Puligandla
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - S. Signoretti
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - D. Cella
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - R.T. Gupta
- Departments of Radiology and Surgery and The Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - R. Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - E. Van Allen
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - P. Lara
- University of California Davis School of Medicine, Sacramento, CA, USA
| | - T.K. Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - A. Kapoor
- Division of Urology, McMaster University, Hamilton, ON, Canada
| | - D.Y.C. Heng
- Tom Baker Cancer Center, Calgary, AB, Canada
| | - B. Shuch
- Division of Urology, Yale Cancer Institute, New Haven, CT, USA
| | - M. Jewett
- Departments of Surgery(Urology) and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada
| | - D. George
- Duke University Departments of Medicine, Surgery, and Pharmacology and Cancer Biology, Division of Medical Oncology, The Duke Cancer Institute, Durham, NC, USA
| | - D. Michaelson
- Genitourinary Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - M.A. Carducci
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - D. McDermott
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - M. Allaf
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|