451
|
Solinas C, Aiello M, Rozali E, Lambertini M, Willard-Gallo K, Migliori E. Programmed cell death-ligand 2: A neglected but important target in the immune response to cancer? Transl Oncol 2020; 13:100811. [PMID: 32622310 PMCID: PMC7332529 DOI: 10.1016/j.tranon.2020.100811] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Programmed cell death-ligand 2 (PD-L2) is one of the two ligands of the programmed cell death-1 (PD-1) receptor, an inhibitory protein mainly expressed on activated immune cells that is targeted in the clinic, with successful and remarkable results. The PD-1/PD-Ls axis was shown to be one of the most relevant immunosuppressive pathways in the immune microenvironment, and blocking this interaction gave rise to an impressive clinical benefit in a broad variety of solid and hematological malignancies. Although PD-L2 has been historically considered a minor ligand, it binds to PD-1 with a two- to six-fold higher affinity as compared to PD-L1. PD-L2 can be expressed by immune, stromal, or tumor cells. The aims of this narrative review are to summarize PD-L2 biology in the physiological responses of the immune system and its role, expression, and clinical significance in cancer.
Collapse
Affiliation(s)
- Cinzia Solinas
- Azienda USL Valle d'Aosta, Regional Hospital of Valle d'Aosta, Aosta, Italy
| | - Marco Aiello
- Medical Oncology Unit, A.O.U. Policlinico San Marco, Catania, Italy
| | - Esdy Rozali
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Matteo Lambertini
- IRCCS Ospedale Policlinico San Martino and University of Genova, Genova, Italy
| | | | - Edoardo Migliori
- Columbia University Medical Center, Columbia Center for Translational Immunology, New York, NY, USA.
| |
Collapse
|
452
|
Keup C, Kimmig R, Kasimir-Bauer S. Liquid Biopsies to Evaluate Immunogenicity of Gynecological/Breast Tumors: On the Way to Blood-Based Biomarkers for Immunotherapies. Breast Care (Basel) 2020; 15:470-480. [PMID: 33223990 PMCID: PMC7650128 DOI: 10.1159/000510509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite the assumption of breast cancer (BC) as a cold, non-immunogenic tumor, immune checkpoint inhibitor (ICI) therapy is favorable for a subgroup of patients. Immunohistochemical assessment of the programmed cell death ligand 1 (PD-L1) is the only approved companion diagnostic for anti-PD-L1 therapy in metastatic triple-negative BC; however, challenges regarding the standardization of PD-L1 scoring in tumor tissue still remain. Consequently, to select patients most likely to respond to ICI, blood-based biomarkers are urgently needed. SUMMARY AND KEY MESSAGES Liquid biopsy, comprising circulating immune cells, circulating tumor cells and extracellular vesicles, as well as their surface proteins, is of high potential, and these analytes were already shown to be molecular correlates or regulators of the evasion from antitumoral immune reaction. Liquid biopsy, also enabling the evaluation of tumor mutational burden (TMB), microsatellite instability, and the T-cell receptor repertoire, allows serial sampling for monitoring purposes and reflects intra-tumoral heterogeneity which qualifies as marker for immunogenicity. Only a very few studies have already elucidated the potential of these analytes as biomarkers under ICI therapy. Nonetheless, the topic is of growing interest and has high relevance for the future. However, for clinical implementation, these multifarious analytes first need to pass robust standardization and validation procedures.
Collapse
Affiliation(s)
| | | | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| |
Collapse
|
453
|
Radosa JC, Stotz L, Müller C, Kaya AC, Solomayer EF, Radosa MP. Clinical Data on Immunotherapy in Breast Cancer. Breast Care (Basel) 2020; 15:450-469. [PMID: 33223989 PMCID: PMC7650095 DOI: 10.1159/000511788] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Breast cancer has traditionally been considered to have a low immunogenic potential compared to other tumor entities. SUMMARY The most extensively studied immunotherapeutic agents for breast cancer to date are immune checkpoint inhibitors, with the results of the IMpassion130 trial leading to the approval of atezolizumab plus nab-paclitaxel for first-line treatment of programmed cell death ligand 1-positive, metastatic, triple-negative breast cancer, and studies in earlier stages have yielded promising results. Other immunotherapeutic options being assessed in phases 2 and 3 trials include vaccine-based therapies and treatment with anti-human epidermal growth factor receptor 2 (H-directed immune-linked antibodies) and substances evaluated in early clinical trials as cellular therapies (adoptive cell therapy and chimeric antigen receptor T cells). KEY MESSAGES Immunotherapy is an emerging modality for the treatment of breast cancer, as evidenced by the plethora of preclinical and clinical concepts and ongoing trials. Early studies established the role of immunotherapeutic agents in the metastatic setting. Ongoing studies will expand our knowledge about the timing of administration, best partners for combination therapy, and predictive biomarkers to guide immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Julia Caroline Radosa
- Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, Germany
| | - Lisa Stotz
- Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, Germany
| | - Carolin Müller
- Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, Germany
| | - Askin Canguel Kaya
- Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, Germany
| | - Erich-Franz Solomayer
- Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, Germany
| | - Marc Philipp Radosa
- Department of Gynecology and Obstetrics, Klinikum Bremen-Nord, Bremen, Germany
| |
Collapse
|
454
|
Purwanto I, Heriyanto DS, Ghozali A, Widodo I, Dwiprahasto I, Aryandono T, Haryana SM. Overexpression of Programmed Death-Ligand 1 Receptor mRNA as an Independent Negative Prognostic Factor for Triple Negative Breast Cancer. World J Oncol 2020; 11:216-222. [PMID: 33117465 PMCID: PMC7575275 DOI: 10.14740/wjon1302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/23/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) (represents roughly 25% of all breast cancers in Yogyakarta) still has the worst survival compared to other breast cancer subtypes. Results from recent studies have shown that inhibition of programmed death-ligand 1 receptor (PD-L1) in TNBC patients is associated with better prognosis. Currently, data on PD-L1 expression and its prognostic value in Indonesian TNBC patients are still relatively unknown. This study aimed to investigate the expression of PD-L1 in Indonesian TNBC patients as preliminary proof to support PD-L1 inhibitor as a possible treatment option near in the future. METHODS We retrospectively included stage I-III TNBC patients diagnosed between 2014 and 2017 in Dr. Sardjito Hospital, Yogyakarta, Indonesia. Clinical variables were collected from medical record. Paraffin blocks of biopsy specimen were retrieved to examine mRNA level of PD-L1. RESULTS We included 48 subjects with mean age of 51.09 years and mean body mass index (BMI) of 24.58. The 3-year overall survival (OS) was 58.3%. Overexpression of PD-L1 mRNA in TNBC patients is associated with worse prognosis (P < 0.01). There were no statistically significant associations between PD-L1 mRNA expression and any of the clinicopathologic variables examined. CONCLUSIONS In summary, PD-L1 mRNA overexpression is associated with worse survival in Indonesian TNBC patients, independent of other established risk factors. PD-L1 mRNA is expressed in all of our samples, presenting as a feasible alternative or complementary method in deciding which patient might benefit from receiving PD-L1 inhibitor.
Collapse
Affiliation(s)
- Ibnu Purwanto
- Hematology and Medical Oncology Division, Department of Internal Medicine, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Ahmad Ghozali
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Irianiwati Widodo
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Iwan Dwiprahasto
- Department of Clinical Pharmacology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Teguh Aryandono
- Department of Surgical Oncology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Sofia Mubarika Haryana
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| |
Collapse
|
455
|
Fares J, Kanojia D, Rashidi A, Ulasov I, Lesniak MS. Landscape of combination therapy trials in breast cancer brain metastasis. Int J Cancer 2020; 147:1939-1952. [PMID: 32086955 PMCID: PMC7423704 DOI: 10.1002/ijc.32937] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/05/2020] [Accepted: 02/18/2020] [Indexed: 12/30/2022]
Abstract
Combination therapy has become a cornerstone in cancer treatment to potentiate therapeutic effectiveness and overcome drug resistance and metastasis. In this work, we explore combination trials in breast cancer brain metastasis (BCBM), highlighting deficiencies in trial design and underlining promising combination strategies. On October 31, 2019, we examined ClinicalTrials.gov for interventional and therapeutic clinical trials involving combination therapy for BCBM, without limiting for date or location. Information on trial characteristics was collected. Combination therapies used in trials were analyzed and explored in line with evidence from the medical literature. Sixty-five combination therapy trials were selected (n = 65), constituting less than 0.7% of all breast cancer trials. Most trials (62%) combined ≥2 chemotherapeutic agents. Chemotherapy with radiation was main-stay in 23% of trials. Trastuzumab was mostly used in combination (31%), followed by lapatinib (20%) and capecitabine (15%). Common strategies involved combining tyrosine kinase inhibitors with thymidylate synthase inhibitors (6 trials), dual HER-dimerization inhibitors (3 trials), microtubule inhibitors and tyrosine kinase inhibitors (3 trials), and HER-dimerization inhibitors and tyrosine kinase inhibitors (3 trials). The combination of tucatinib and capecitabine yielded the highest objective response rate (83%) in early phase trials. The triple combination of trastuzumab, tucatinib and capecitabine lowered the risk of disease progression or death by 52% in patients with HER2-positive BCBM. Combining therapeutic agents based on biological mechanisms is necessary to increase the effectiveness of available anti-cancer regimens. Significant survival benefit has yet to be achieved in future combination therapy trials. Enhancing drug delivery through blood-brain barrier permeable agents may potentiate the overall therapeutic outcomes.
Collapse
Affiliation(s)
- Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- High Impact Cancer Research program, Harvard Medical School, Boston, MA, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
456
|
Emens LA, Esteva FJ, Beresford M, Saura C, De Laurentiis M, Kim SB, Im SA, Wang Y, Salgado R, Mani A, Shah J, Lambertini C, Liu H, de Haas SL, Patre M, Loi S. Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase 2, multicentre, randomised, double-blind trial. Lancet Oncol 2020; 21:1283-1295. [PMID: 33002436 DOI: 10.1016/s1470-2045(20)30465-4] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND HER2-positive metastatic breast cancer is incurable and new treatments are needed. Addition of atezolizumab to trastuzumab emtansine might potentiate anticancer immunity and enhance the HER2-targeted cytotoxic activity of trastuzumab emtansine. We aimed to test this combination in HER2-positive advanced breast cancer that had progressed after previous treatment with trastuzumab and a taxane. METHODS The KATE2 study is a randomised, double-blind, placebo-controlled, phase 2 study at 68 centres from nine countries across Asia, Australia, North America, and western Europe. Eligible patients were adults (aged ≥18 years) with an Eastern Cooperative Oncology Group performance status of 0 or 1 and centrally confirmed, measurable, HER2-positive advanced breast cancer previously treated with trastuzumab and a taxane. Patients were randomly assigned (2:1) either trastuzumab emtansine (3·6 mg/kg of bodyweight) plus atezolizumab (1200 mg) or trastuzumab emtansine plus placebo; all study drugs were administered by intravenous infusion every 3 weeks. Randomisation was done via an interactive voice and web response system using a permuted block scheme (block size of six) and was stratified by PD-L1 status, world region, and liver metastases. Patients, investigators, and study team members were masked to treatment allocation. The primary endpoint was investigator-assessed progression-free survival in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, NCT02924883, and the study has been completed. FINDINGS Between Sept 26, 2016, and Aug 7, 2017, 330 patients were screened for the study, of whom 202 were randomly allocated either atezolizumab (n=133) or placebo (n=69). At the recommendation of the independent data monitoring committee, treatment assignment was unmasked on Dec 11, 2017, due to futility and the numerically higher frequency of adverse events among patients assigned atezolizumab. This date was set as the clinical cutoff for the primary analysis. Median follow-up was 8·5 months (IQR 6·1-11·5) for patients assigned atezolizumab and 8·4 months (5·3-11·1) for those assigned placebo. Median progression-free survival was 8·2 months (95% CI 5·8-10·7) for patients assigned atezolizumab versus 6·8 months (4·0-11·1) for those assigned placebo (stratified hazard ratio 0·82, 95% CI 0·55-1·23; p=0·33). The most common grade 3 or worse adverse events were thrombocytopenia (17 [13%] among 132 patients who received atezolizumab vs three [4%] among 68 who received placebo), increased aspartate aminotransferase (11 [8%] vs two [3%]), anaemia (seven [5%] vs 0), neutropenia (six [5%] vs three [4%]), and increased alanine aminotransferase (six [5%] vs two [3%]). Serious adverse events occurred in 43 (33%) of 132 patients who received atezolizumab and 13 (19%) of 68 patients who received placebo. One patient who received atezolizumab died due to a treatment-related adverse event (haemophagocytic syndrome). INTERPRETATION Addition of atezolizumab to trastuzumab emtansine did not show a clinically meaningful improvement in progression-free survival and was associated with more adverse events. Further study of trastuzumab emtansine plus atezolizumab is warranted in a subpopulation of patients with PD-L1-positive, HER2-positive advanced breast cancer. FUNDING F Hoffman-La Roche.
Collapse
Affiliation(s)
- Leisha A Emens
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Francisco J Esteva
- Perlmutter Cancer Center at New York University Langone Health, New York, NY, USA
| | | | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Michelino De Laurentiis
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale Tumori Fondazione Pascale, Naples, Italy
| | - Sung-Bae Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Roberto Salgado
- Department of Pathology, Gasthuis Zusters Antwerpen-Ziekenhuis Netwerk Antwerpen Hospitals, Antwerp, Belgium
| | | | | | | | | | | | | | - Sherene Loi
- Division of Research, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.
| |
Collapse
|
457
|
Shi J, Liu F, Song Y. Progress: Targeted Therapy, Immunotherapy, and New Chemotherapy Strategies in Advanced Triple-Negative Breast Cancer. Cancer Manag Res 2020; 12:9375-9387. [PMID: 33061626 PMCID: PMC7533235 DOI: 10.2147/cmar.s272685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, accounting for approximately 15% of cases, and is defined by the lack of expression of hormone receptors (estrogen and progesterone receptors) and lack of amplification or overexpression of human epidermal growth receptor 2 (HER2). Due to the lack of targets of hormone receptors and HER2, treatment of TNBC or advanced TNBC relies on conventional chemotherapeutic agents, but their efficacy and prognosis are poor. In patients with advanced TNBC, poorer outcomes are observed. Recently, with the launch of clinical trials and advancements in molecular studies, targeted therapy for signaling transduction pathways, immunotherapy for immune checkpoints, and new chemotherapy strategies have provided feasible or potential therapeutic options for advanced TNBC. This review aimed to summarize recent progress in targeted therapy, immunotherapy, and chemotherapy for advanced TNBC.
Collapse
Affiliation(s)
- Jinhong Shi
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Feiqi Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yanqiu Song
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
458
|
Schardt J. [The use of immune checkpoint inhibitors in routine oncology]. Z Rheumatol 2020; 79:809-817. [PMID: 32936368 PMCID: PMC7653782 DOI: 10.1007/s00393-020-00876-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2020] [Indexed: 11/12/2022]
Abstract
Hintergrund Die Einführung von Immuncheckpoint-Inhibitoren (ICI) hat die Behandlungskonzepte der Onkologie für eine Vielzahl von unterschiedlichen Krebsarten maßgeblich verändert. Dabei werden in der klinischen Routine v. a. humanisierte Antikörper gegen Immuncheckpoints wie „cytotoxic T‑lymphocyte associated protein 4“ (CTLA-4) oder „programmed cell death 1/programmed cell death ligand 1“ (PD1/PD-L1) eingesetzt. Fragestellung Übersicht zur Therapielandschaft mit Immuncheckpoint-Inhibitoren bei mehrheitlich soliden Tumoren in der Onkologie. Material und Methoden Darstellung und Diskussion aktueller Studienresultate, Einbezug aktueller Behandlungsempfehlungen und Zulassungsindikationen. Ergebnisse Sieben verschiedene Immuncheckpoint-Inhibitoren werden in der Onkologie therapeutisch eingesetzt: ein Anti-CTLA-4-Antikörper, 3 Anti-PD1-Antikörper und 3 Anti-PD-L1-Antiköper. FDA-Zulassung auf dem US-Markt für 17 verschiedene Tumorentitäten und einer agnostischen Indikation (Tumoren mit defizienter Mismatch-repair-Maschinerie/hohe Mikrosatelliteninstabilität). Langzeitremissionen sind in ca. zwei Drittel der Patienten mit Tumoransprechen möglich. Schlussfolgerungen Nutzen der Immuncheckpoint-Inhibitoren nur für einen Teil der behandelten Patienten. Primäre und sekundäre Resistenzmechanismen erst in Anfängen verstanden. Kombinationstherapien der Immuncheckpoint-Inhibitoren mit z. B. Chemotherapie, neuen Immuncheckpoint-Inhibitoren (z. B. Anti-LAG3-Antikörper) oder gezielten Therapien (z. B. CDK4/6, PARP-Inhibitoren) zur Verbesserung der Wirksamkeit werden in klinischen Studien untersucht. Verlässliche, prädiktive Marker sind dringend erforderlich.
Collapse
Affiliation(s)
- Julian Schardt
- Universitätsklinik für Medizinische Onkologie, Inselspital, Freiburgstr. 41G, 3010, Bern, Schweiz.
| |
Collapse
|
459
|
Beyond Chemotherapies: Recent Strategies in Breast Cancer Treatment. Cancers (Basel) 2020; 12:cancers12092634. [PMID: 32947780 PMCID: PMC7565588 DOI: 10.3390/cancers12092634] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022] Open
Abstract
In 2018, about 2.1 million women have been diagnosed with breast cancer worldwide. Treatments include-among others-surgery, chemotherapy, radiotherapy, or endocrine therapy. The current policy of care tends rather at therapeutic de-escalation, and systemic treatment such as chemotherapies alone are not systematically considered as the best option anymore. With recent advances in the understanding of cancer biology, and as a complement to anatomic staging, some biological factors (assessed notably via gene-expression signatures) are taken into account to evaluate the benefit of a chemotherapy regimen. The first aim of this review will be to summarize when chemotherapies can be avoided or used only combined with other treatments. The second aim will focus on molecules that can be used instead of chemotherapeutic drugs or used in combination with chemotherapeutic drugs to improve treatment outcomes. These therapeutic molecules have emerged from the collaboration between fundamental and clinical research, and include molecules, such as tyrosine kinase inhibitors, CDK4/6 inhibitors, and monoclonal antibodies (such as anti-PD-L1). In the fight against cancer, new tools aiding decision making are of the utmost importance: gene-expression signatures have proven to be valuable in the clinic, notably, to know when chemotherapies can be avoided. When substitution treatments are also available, a big step can be made toward personalized medicine for the patient's benefit.
Collapse
|
460
|
Cerbelli B, Scagnoli S, Mezi S, De Luca A, Pisegna S, Amabile MI, Roberto M, Fortunato L, Costarelli L, Pernazza A, Strigari L, Della Rocca C, Marchetti P, d’Amati G, Botticelli A. Tissue Immune Profile: A Tool to Predict Response to Neoadjuvant Therapy in Triple Negative Breast Cancer. Cancers (Basel) 2020; 12:E2648. [PMID: 32947953 PMCID: PMC7565153 DOI: 10.3390/cancers12092648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/06/2020] [Accepted: 09/11/2020] [Indexed: 12/31/2022] Open
Abstract
Pathological complete response (pCR) after neoadjuvant chemotherapy (NACT) can predict better survival outcomes in patients with early triple negative breast cancer (TNBC). Tumor infiltrating lymphocytes (TILs), Programmed Death-Ligand 1 (PD-L1), and Cluster of Differentiation 73 (CD73) are immune-related biomarkers that can be evaluated in the tumor microenvironment. We investigated if the contemporary expression of these biomarkers combined in a tissue immune profile (TIP) can predict pCR better than single biomarkers in TNBC. Tumor infiltrating lymphocytes (TILs), CD73 expression by cancer cells (CC), and PD-L1 expression by immune cells (IC) were evaluated on pre-NACT biopsies. We defined TIP positive (TIP+) as the simultaneous presence of TILS ≥ 50%, PD-L1 ≥ 1%, and CD73 ≤ 40%. To consider the effects of all significant variables on the pCR, multivariate analysis was performed. Akaike information criterion (AIC) and Bayesian information criterion (BIC) were used for model selection. We retrospectively analyzed 60 biopsies from patients with TNBC who received standard NACT. Pathological complete response was achieved in 23 patients (38.0%). Twelve (20.0%) cases resulted to be TIP+. The pCR rate was significantly different between TIP+ (91.7%) and TIP- (25.0%) (p < 0.0001). Using a multivariate analysis, TIP was confirmed as an independent predictive factor of pCR (OR 49.7 (6.30-392.4), p < 0.0001). Finally, we compared the efficacy of TIP versus each single biomarker in predicting pCR by AIC and BIC. The combined immune profile is more accurate in predicting pCR (AIC 68.3; BIC 74.5) as compared to single biomarkers. The association between TIP+ and pCR can be proposed as a novel link between immune background and response to chemotherapy in TNBC, highlighting the need to consider an immunological patients' profile rather than single biomarkers.
Collapse
Affiliation(s)
- Bruna Cerbelli
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (B.C.); (S.M.); (S.P.); (G.d.)
| | - Simone Scagnoli
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Silvia Mezi
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (B.C.); (S.M.); (S.P.); (G.d.)
| | - Alessandro De Luca
- Department of Surgical Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.D.L.); (M.I.A.)
| | - Simona Pisegna
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (B.C.); (S.M.); (S.P.); (G.d.)
| | - Maria Ida Amabile
- Department of Surgical Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.D.L.); (M.I.A.)
| | - Michela Roberto
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00187 Rome, Italy; (M.R.); (P.M.); (A.B.)
| | - Lucio Fortunato
- Azienda Ospedaliera San Giovanni-Addolorata, 00184 Rome, Italy; (L.F.); (L.C.)
| | - Leopoldo Costarelli
- Azienda Ospedaliera San Giovanni-Addolorata, 00184 Rome, Italy; (L.F.); (L.C.)
| | - Angelina Pernazza
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00161 Rome, Italy; (A.P.); (C.D.R.)
| | - Lidia Strigari
- Medical Physics Unit, “S. Orsola-Malpighi” Hospital, 40138 Bologna, Italy;
| | - Carlo Della Rocca
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00161 Rome, Italy; (A.P.); (C.D.R.)
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00187 Rome, Italy; (M.R.); (P.M.); (A.B.)
| | - Giulia d’Amati
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (B.C.); (S.M.); (S.P.); (G.d.)
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00187 Rome, Italy; (M.R.); (P.M.); (A.B.)
| |
Collapse
|
461
|
Xu T, Wang Z, Dong M, Wu D, Liao S, Li X. Chloride intracellular channel protein 2: prognostic marker and correlation with PD-1/PD-L1 in breast cancer. Aging (Albany NY) 2020; 12:17305-17327. [PMID: 32915772 PMCID: PMC7521498 DOI: 10.18632/aging.103712] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
Immune checkpoint inhibition has emerged as an effective treatment for multiple solid tumors, including advanced-stage breast cancer (BC). During the past decade, the US Food and Drug Administration has approved a number of agents for immune checkpoint blockade (ICB). However, the limited data on monotherapy anti-tumor activity in BC underscores the need for robust predictive biomarker development. Here, we used weighted gene coexpression network analysis of genes differentially expressed between BC and normal tissue to identify genes coexpressed with programmed death-1 (PD-1) and its ligand (PD-L1). Tumor Immune Estimation Resource and Gene Expression Profiling Interaction Analysis were used to assess the relationship between gene expression and the abundance of tumor-infiltrating lymphocytes (TILs). We found that chloride intracellular channel protein 2 (CLIC2) was not only coexpressed with PD-1 and PD-L1, but its increased expression was associated with a favorable prognosis and enrichment of multiple TIL types, particularly CD8+ T cells. These results suggest that CLIC2 is a potentially useful biomarker for identifying BC patients who could benefit from ICB.
Collapse
Affiliation(s)
- Tao Xu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China,Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China
| | - Zhi Wang
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China
| | - Menglu Dong
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China
| | - Di Wu
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China
| | - Shujie Liao
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of HUST, Wuhan 430030, Hubei, People’s Republic of China
| |
Collapse
|
462
|
Sirait-Fischer E, Olesch C, Fink AF, Berkefeld M, Huard A, Schmid T, Takeda K, Brüne B, Weigert A. Immune Checkpoint Blockade Improves Chemotherapy in the PyMT Mammary Carcinoma Mouse Model. Front Oncol 2020; 10:1771. [PMID: 33014872 PMCID: PMC7513675 DOI: 10.3389/fonc.2020.01771] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the success of immune checkpoint blockade in cancer, the number of patients that benefit from this revolutionary treatment option remains low. Therefore, efforts are being undertaken to sensitize tumors for immune checkpoint blockade, which includes combining immune checkpoint blocking agents such as anti-PD-1 antibodies with standard of care treatments. Here we report that a combination of chemotherapy (doxorubicin) and immune checkpoint blockade (anti-PD-1 antibodies) induces superior tumor control compared to chemotherapy and immune checkpoint blockade alone in the murine autochthonous polyoma middle T oncogene-driven (PyMT) mammary tumor model. Using whole transcriptome analysis, we identified a set of genes that were upregulated specifically upon chemoimmunotherapy. This gene signature and, more specifically, a condensed four-gene signature predicted favorable survival of human mammary carcinoma patients in the METABRIC cohort. Moreover, PyMT tumors treated with chemoimmunotherapy contained higher levels of cytotoxic lymphocytes, particularly natural killer cells (NK cells). Gene set enrichment analysis and bead-based ELISA measurements revealed increased IL-27 production and signaling in PyMT tumors upon chemoimmunotherapy. Moreover, IL-27 signaling improved NK cell cytotoxicity against PyMT cells in vitro. Taken together, our data support recent clinical observations indicating a benefit of chemoimmunotherapy compared to monotherapy in breast cancer and suggest potential underlying mechanisms.
Collapse
Affiliation(s)
- Evelyn Sirait-Fischer
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Catherine Olesch
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Annika F Fink
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Matthias Berkefeld
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Arnaud Huard
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Tobias Schmid
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Kazuhiko Takeda
- Research Center of Oncology, ONO Pharmaceutical Co., LTD, Osaka, Japan
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.,Branch for Translational Medicine and Pharmacology TMP of the Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| |
Collapse
|
463
|
Marinelli D, Mazzotta M, Pizzuti L, Krasniqi E, Gamucci T, Natoli C, Grassadonia A, Tinari N, Tomao S, Sperduti I, Sanguineti G, Botticelli A, Fabbri A, Botti C, Ciliberto G, Barba M, Vici P. Neoadjuvant Immune-Checkpoint Blockade in Triple-Negative Breast Cancer: Current Evidence and Literature-Based Meta-Analysis of Randomized Trials. Cancers (Basel) 2020; 12:cancers12092497. [PMID: 32899209 PMCID: PMC7565914 DOI: 10.3390/cancers12092497] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Breast cancer is a heterogeneous disease, which encompasses several subgroups of entities widely varying by clinical-pathological features. Triple negative breast cancer is characterized by a particularly aggressive biological behavior. The administration of chemotherapy has long represented the most efficacious weapon in combating triple negative breast cancer in both its initial and late phase of development. A pivot point has been recently reached throughout the approval of the immunotherapic agent atezolizumab in combination with chemotherapy as first-line treatment for programmed-death ligand 1-positive, unresectable locally advanced, or metastatic triple-negative breast cancer. Results from the registrative trial, IMpassion 130, have increasingly fueled the flourishing of studies of immune-checkpoint inhibitors in the early stage of triple negative breast cancer development. We critically interpret results from the most recent literature in light of relevant issues of methodological nature and also present a quantitative summary of data from the inherent trials. Future directions are also highlighted. Abstract Chemotherapy based on the sequential use of anthracyclines and taxanes has long represented the most efficacious approach in the management of early-stage, triple-negative breast cancer, whose aggressive behavior is widely renowned. This standard chemotherapy backbone was subsequently enriched by the use of carboplatin, based on its association with increased pathologic complete response and efficacy in the metastatic setting. Following the results from the IMpassion130 trial, the recent approval of the immunotherapic agent atezolizumab in combination with chemotherapy as first-line treatment for programmed-death ligand 1-positive, unresectable locally advanced, or metastatic triple-negative breast cancer increasingly fueled the flourishing of trials of immune-checkpoint inhibitors in the early setting. In this work, we review the most recent inherent literature in light of key methodological issues and provide a quantitative summary of the results from phase II–III randomized trials of immunotherapic agents combined with chemotherapy in the setting of interest. Hints regarding future directions are also discussed.
Collapse
Affiliation(s)
- Daniele Marinelli
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy;
| | - Marco Mazzotta
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
- Correspondence: (L.P.); (M.B.); Tel.: +39-06-52665698 (L.P.); +39-06-52665419 (M.B.)
| | - Eriseld Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| | - Teresa Gamucci
- Medical Oncology, Sandro Pertini Hospital, 00157 Rome, Italy;
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Antonino Grassadonia
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I, ‘Sapienza’ University of Rome, 00161 Rome, Italy;
| | - Isabella Sperduti
- Biostatistics Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | | | - Agnese Fabbri
- Medical Oncology Unit, Belcolle Hospital, 01100 Viterbo, Italy;
| | - Claudio Botti
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
- Correspondence: (L.P.); (M.B.); Tel.: +39-06-52665698 (L.P.); +39-06-52665419 (M.B.)
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| |
Collapse
|
464
|
Ho AY, Wright JL, Blitzblau RC, Mutter RW, Duda DG, Norton L, Bardia A, Spring L, Isakoff SJ, Chen JH, Grassberger C, Bellon JR, Beriwal S, Khan AJ, Speers C, Dunn SA, Thompson A, Santa-Maria CA, Krop IE, Mittendorf E, King TA, Gupta GP. Optimizing Radiation Therapy to Boost Systemic Immune Responses in Breast Cancer: A Critical Review for Breast Radiation Oncologists. Int J Radiat Oncol Biol Phys 2020; 108:227-241. [PMID: 32417409 PMCID: PMC7646202 DOI: 10.1016/j.ijrobp.2020.05.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Immunotherapy using immune checkpoint blockade has revolutionized the treatment of many types of cancer. Radiation therapy (RT)-particularly when delivered at high doses using newer techniques-may be capable of generating systemic antitumor effects when combined with immunotherapy in breast cancer. These systemic effects might be due to the local immune-priming effects of RT resulting in the expansion and circulation of effector immune cells to distant sites. Although this concept merits further exploration, several challenges need to be overcome. One is an understanding of how the heterogeneity of breast cancers may relate to tumor immunogenicity. Another concerns the need to develop knowledge and expertise in delivery, sequencing, and timing of RT with immunotherapy. Clinical trials addressing these issues are under way. We here review and discuss the particular opportunities and issues regarding this topic, including the design of informative clinical and translational studies.
Collapse
Affiliation(s)
- Alice Y Ho
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts.
| | - Jean L Wright
- Department of Radiation Oncology, Johns Hopkins Cancer Center, Brooklandville, Maryland
| | - Rachel C Blitzblau
- Department of Radiation Oncology, Duke Cancer Center, Durham, North Carolina
| | - Robert W Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Dan G Duda
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Larry Norton
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aditya Bardia
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Laura Spring
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Steven J Isakoff
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan H Chen
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Clemens Grassberger
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer R Bellon
- Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Sushil Beriwal
- Department of Radiation Oncology, University of Pittsburgh Cancer Center, Pittsburgh, Pennslyvania
| | - Atif J Khan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Samantha A Dunn
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Alastair Thompson
- Department of Surgical Oncology, Baylor College of Medicine Medical Center, Houston, Texas
| | - Cesar A Santa-Maria
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ian E Krop
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Elizabeth Mittendorf
- Department of Surgical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Tari A King
- Department of Surgical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Gaorav P Gupta
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
465
|
Wang T, Shen Y, Luyten S, Yang Y, Jiang X. Tissue-resident memory CD8+ T cells in cancer immunology and immunotherapy. Pharmacol Res 2020; 159:104876. [DOI: 10.1016/j.phrs.2020.104876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
|
466
|
Affiliation(s)
- Rachna Malani
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, NY 10065, USA
| |
Collapse
|
467
|
Sheybani ND, Witter AR, Thim EA, Yagita H, Bullock TNJ, Price RJ. Combination of thermally ablative focused ultrasound with gemcitabine controls breast cancer via adaptive immunity. J Immunother Cancer 2020; 8:jitc-2020-001008. [PMID: 32819975 PMCID: PMC7443308 DOI: 10.1136/jitc-2020-001008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 12/05/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) remains recalcitrant to most targeted therapy approaches. However, recent clinical studies suggest that inducing tumor damage can render TNBC responsive to immunotherapy. We therefore tested a strategy for immune sensitization of murine TNBC (4T1 tumors) through combination of focused ultrasound (FUS) thermal ablation and a chemotherapy, gemcitabine (GEM), known to attenuate myeloid-derived suppressor cells (MDSCs). Methods We applied a sparse-scan thermally ablative FUS regimen at the tumor site in combination with systemically administered GEM. We used flow cytometry analysis to investigate the roles of monotherapy and combinatorial therapy in mediating local and systemic immunity. We also tested this combination in Rag1−/− mice or T cell-depleted wild-type mice to determine the essentiality of adaptive immunity. Further, we layered Programmed cell death protein 1 (PD-1) blockade onto this combination to evaluate its impact on tumor outgrowth and survival. Results The immune-modulatory effect of FUS monotherapy was insufficient to promote a robust T cell response against 4T1, consistent with the dominant MDSC-driven immunosuppression evident in this model. The combination of FUS+GEM significantly constrained primary TNBC tumor outgrowth and extended overall survival of mice. Tumor control correlated with increased circulating antigen-experienced T cells and was entirely dependent on T cell-mediated immunity. The ability of FUS+GEM to control primary tumor outgrowth was moderately enhanced by either neoadjuvant or adjuvant treatment with anti-PD-1. Conclusion Thermally ablative FUS in combination with GEM restricts primary tumor outgrowth, improves survival and enhances immunogenicity in a murine metastatic TNBC model. This treatment strategy promises a novel option for potentiating the role of FUS in immunotherapy of metastatic TNBC and is worthy of future clinical evaluation. Trial registration numbers NCT03237572 and NCT04116320.
Collapse
Affiliation(s)
- Natasha D Sheybani
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | | | - Eric A Thim
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Hideo Yagita
- Department of Immunology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | | | - Richard J Price
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Radiology & Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
468
|
Prognostication of a 13-immune-related-gene signature in patients with early triple-negative breast cancer. Breast Cancer Res Treat 2020; 184:325-334. [PMID: 32812178 DOI: 10.1007/s10549-020-05874-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/10/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE We investigated the expression profiles of immune genes in patients with triple-negative breast cancer (TNBC) to identify the prognostic value of immune genes and their clinical implications. METHODS NanoString nCounter Analysis of 770 immune-related genes was used to measure immune gene expression in patients with TNBC who underwent curative surgery followed by adjuvant chemotherapy at Samsung Medical Center between 2000 and 2004. Statistical analyses were conducted to identify the associations between gene expression and distant recurrence-free survival (DRFS). RESULTS Of 1189 patients who underwent curative BC surgery, 200 TNBC patients were included and stage was the only clinical factor predictive of DRFS. In terms of immune genes, 155 of 770 genes were associated with DRFS (p < 0.01). Further multivariate analysis revealed that 13 genes, CD1B, CD53, CT45A1, GTF3C1, IL11RA, IL1RN, LRRN3, MAPK1, NEFL, PRKCE, PTPRC, SPACA3 and TNFSF11, were associated with patient prognosis (p < 0.05). The prognostic value of stage and expression levels of 13 immune genes was analyzed and the area under the receiver operating characteristic curve (AUC) was 0.923. Based on the AUC, we divided patients into three genetic risk groups and DRFS rate was significantly different according to genetic risk groups, even in the same stage (p < 0.001). CONCLUSIONS In this study, a 13-gene expression profile in combination with stage precisely predicted distant recurrence of early TNBC. Therefore, this 13-immune-gene signature could help predict TNBC prognosis and provide guidance for treatment as well as the opportunity to develop new targets for immunotherapy in TNBC patients.
Collapse
|
469
|
Chao X, Liu L, Sun P, Yang X, Li M, Luo R, Huang Y, He J, Yun J. Immune parameters associated with survival in metaplastic breast cancer. Breast Cancer Res 2020; 22:92. [PMID: 32811533 PMCID: PMC7437173 DOI: 10.1186/s13058-020-01330-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/11/2020] [Indexed: 12/31/2022] Open
Abstract
Background Metaplastic breast carcinoma (MBC) is a rare histological type of breast cancer, which commonly shows resistance to standard therapies and is associated with poor prognosis. The immune microenvironment in MBC and its significance has not been well established due to its low incurrence rate and complex components. We aimed to investigate the diversity of immune parameters including subsets of TILs and PDL1/PD1 expression in MBC, as well as its correlation with prognosis. Methods A total of 60 patients diagnosed with MBC from January 2006 to December 2017 were included in our study. The percentage (%) and quantification (per mm2) of TILs and presence of tertiary lymphoid structures (TLS) were evaluated by hematoxylin and eosin staining (HE). The quantification of CD4+, CD8+ TILs (per mm2), and PD-1/PDL1 expression were evaluated through immunohistochemistry and analyzed in relation to clinicopathological characteristics. A ≥ 1% membranous or cytoplasmatic expression of PD1 and PDL1 was considered a positive expression. Results We found squamous cell carcinoma MBC (33/60, 55%) exhibiting most TILs of all the MBC subtypes (p = 0.043). Thirty-three of 60 (50%) of the patients had coexisting invasive ductal carcinoma of no special type (IDC-NST), and the average percentage of TILs in MBC components was lower compared with NST components (p < 0.001). Thirty (50%) patients exhibited positive (≥ 1%) PDL1 expression in their tumor cells, while 36 (60%) had positive (≥ 1%) PDL1 expression in their TILs. Twenty-seven (45%) of all the patients had positive (≥ 1%) PD1 expression in their tumor cells and 33 (55%) had PD1-positive (≥ 1%) stromal TILs. More CD8+ TILs were associated with positive PDL1 expression of tumor cells as well as positive PD1 expression in stromal cells. Greater number of stromal TILS (> 300/mm2, 20%), CD4+ TILs (> 250/mm2), and CD8+ TILs (> 70/mm2) in MBC were found associated with longer disease-free survival. Positive expression of PDL1 in tumor cells (≥ 1%) and PD1 in stromal cells (≥ 1%) were also associated with longer survival. Conclusions The immune characteristics differ in various subtypes as well as components of MBC. Immune parameters are key predictive factors of MBC and provide the clinical significance of applying immune checkpoint therapies in patients with MBC.
Collapse
Affiliation(s)
- Xue Chao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Lili Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Peng Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Xia Yang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Mei Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Rongzhen Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Yuhua Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Jiehua He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China
| | - Jingping Yun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China. .,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
470
|
Kern R, Correa SC, Scandolara TB, Carla da Silva J, Pires BR, Panis C. Current advances in the diagnosis and personalized treatment of breast cancer: lessons from tumor biology. Per Med 2020; 17:399-420. [PMID: 32804054 DOI: 10.2217/pme-2020-0070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Breast cancer treatment has advanced enormously in the last decade. Most of this is due to advances reached in the knowledge regarding tumor biology, mainly in the field of diagnosis and treatment. This review brings information about how the genomics-based information contributed to advances in breast cancer diagnosis and prognosis perspective, as well as presents how tumor biology discoveries fostered the main therapeutic approaches available to treat such patients, based on a personalized point of view.
Collapse
Affiliation(s)
- Rodrigo Kern
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Post-Graduation Program in Health-Applied Sciences, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil
| | - Stephany Christiane Correa
- Center for Bone Marrow Transplantation, Laboratory of Stem Cells, National Cancer Institute (INCA), Rio de Janeiro 20230-130, RJ, Brazil
| | - Thalita Basso Scandolara
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil
| | - Janaína Carla da Silva
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Post-Graduation Program in Health-Applied Sciences, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil
| | - Bruno Ricardo Pires
- Instituto Nacional de Câncer José Alencar Gomes da Silva, Rio de Janeiro 20230-130, RJ, Brazil.,Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Post-Graduation Program in Health-Applied Sciences, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil
| |
Collapse
|
471
|
Zou Y, Zou X, Zheng S, Tang H, Zhang L, Liu P, Xie X. Efficacy and predictive factors of immune checkpoint inhibitors in metastatic breast cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2020; 12:1758835920940928. [PMID: 32874208 PMCID: PMC7436841 DOI: 10.1177/1758835920940928] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/12/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown encouraging treatment efficacy for metastatic breast cancer in several clinical trials. However, response only occurred in a small population. Evidence predicting response and survival of patients with metastatic breast cancer following ICI treatment with existing biomarkers has not been well summarized. This review aimed to summarize the efficacy and predictive factors of immune checkpoint therapy in metastatic breast cancer, which is critical for clinical practice. METHODS PubMed, Embase, Cochrane Library, Web of Science, www.clinicaltrials.gov, and meeting abstracts were comprehensively searched to identify clinical trials. The outcomes were objective response rate (ORR), treatment-related adverse events (trAEs), immune-related adverse events (irAEs), progression-free survival (PFS), and overall survival (OS). RESULTS In this review, 27 studies with 1746 patients were included for quantitative synthesis. The pooled ORR was 19% [95% confidence interval (CI) = 12-27%]. Programmed death-ligand 1 (PD-L1)-positive patients had a higher response rate [odds ratio (OR) = 1.44, p = 0.01]. First-line immunotherapy had a better ORR than second-line immunotherapy (OR = 2.00, p = 0.02). Tumor-infiltrating lymphocytes (TILs) ⩾5% (OR = 2.53, p = 0.002) and high infiltrated CD8+ T-cell level (OR = 4.33, p = 0.006) were ideal predictors of immune checkpoint therapy response. Liver metastasis indicated poor response (OR = 0.19, p = 0.009). However, the difference was non-significant in ORR based on age, performance status score, lymph node metastasis, and lactate dehydrogenase (LDH) level. In addition, the PD-L1-positive subgroup had a better 1-year PFS (OR = 1.55, p = 0.04) and 2-year OS (OR = 2.28, p = 0.02) following ICI treatment. The pooled incidence during ICI therapy of grade 3-4 trAEs was 25% (95% CI = 16-34%), whereas for grade 3-4 irAEs it was 15% (95% CI = 11-19%). CONCLUSIONS Metastatic breast cancer had modest response to ICI therapy. PD-L1-positive, first-line immunotherapy, non-liver metastasis, and high TIL and CD8+ T-cell infiltrating levels could predict better response to ICI treatment. Patients with PD-L1-positive tumor could gain more survival benefits from immune checkpoint therapy.
Collapse
Affiliation(s)
- Yutian Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Xuxiazi Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Shaoquan Zheng
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Hailin Tang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Lijuan Zhang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Peng Liu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 East Dongfeng Road, Guangzhou 510060, People’s Republic of China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 East Dongfeng Road, Guangzhou 510060, People’s Republic of China
| |
Collapse
|
472
|
Mo H, Xu B. Progress in systemic therapy for triple-negative breast cancer. Front Med 2020; 15:1-10. [PMID: 32789731 DOI: 10.1007/s11684-020-0741-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/28/2019] [Indexed: 12/18/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a heterogeneous genetic profile. Chemotherapy exhibits substantial activity in a small subset of these patients. Drug resistance is inevitable. Major progress has been made in the genetic analysis of TNBC to identify novel targets and increase the precision of therapeutic intervention. Such progress has translated into major advances in treatment strategies, including modified chemotherapy approaches, immune checkpoint inhibitors, and targeted therapeutic drugs. All of these strategies have been evaluated in clinical trials. Nevertheless, patient selection remains a considerable challenge in clinical practice.
Collapse
Affiliation(s)
- Hongnan Mo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
473
|
Wu SY, Wang H, Shao ZM, Jiang YZ. Triple-negative breast cancer: new treatment strategies in the era of precision medicine. SCIENCE CHINA-LIFE SCIENCES 2020; 64:372-388. [PMID: 32803712 DOI: 10.1007/s11427-020-1714-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancer (TNBC) remains the most aggressive cluster of all breast cancers, which is due to its rapid progression, high probabilities of early recurrence, and distant metastasis resistant to standard treatment. Following the advances in cancer genomics and transcriptomics that can illustrate the comprehensive profiling of this heterogeneous disease, it is now possible to identify different subclasses of TNBC according to both intrinsic signals and extrinsic microenvironment, which have a huge influence on predicting response to established therapies and picking up novel therapeutic targets for each cluster. In this review, we summarize basic characteristics and critical subtyping systems of TNBC, and particularly discuss newly found prospective targets and relevant medications, which were proved promising in clinical trials, thus shedding light on the future development of precision treatment strategies.
Collapse
Affiliation(s)
- Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hai Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
474
|
Gu Y, Zhang H, Liu Z, Xia Y, Liang B, Liang L. Different patterns of treatment-related adverse events of programmed cell death-1 and its ligand-1 inhibitors in different cancer types: A meta-analysis and systemic review of clinical trials. Asia Pac J Clin Oncol 2020; 16:e160-e178. [PMID: 32779383 DOI: 10.1111/ajco.13385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 05/13/2020] [Indexed: 12/17/2022]
Abstract
Programmed cell death receptor-1 and its ligand-1 (PD-1/PD-L1) inhibitors have been applied to many cancers, but the difference of treatment-related adverse events (AEs) across cancer types remains unknown. We performed a meta-analysis and systemic review to compare the incidences of commonly reported all-grade AEs across cancer types and found that the most frequent AEs were fatigue, rash/pruritus, loss of appetite/nausea and diarrhea. However, each cancer type also had its higher incidences of AEs involving a relevant system, such as melanoma with epidermal AEs (rash, diarrhea and enterocolitis), lung cancer with dyspnea and pneumonitis, digestive system cancers with amylase and lipase elevation; and renal cell and urothelial cancer with kidney injury (creatinine elevation and proteinuria). However, the incidence of hepatitis did not follow the pattern to show a difference. We did another comparison between PD-1 and PD-L1 inhibitors in lung cancer and urothelial cancer respectively, and found that the risk of most AEs did not differ much, except for more hypothyroidism in PD-1 inhibitors, and more kidney injury in PD-L1 inhibitors. Besides possible immunological mechanisms for treatment-related AEs, the influence of previous radiotherapy and the clinical characteristics of the diseases themselves should also be considered and is worth further investigation. With the result of this meta-analysis, clinicians could estimate the risk of certain AE in certain cancer type, to make treatment options and to customize monitor strategies.
Collapse
Affiliation(s)
- Yangchun Gu
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, P.R. China
| | - Hua Zhang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, P.R. China
| | - Zexiang Liu
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, P.R. China
| | - Yifan Xia
- Institute of Medical Technology, Health Science Center, Peking University, Beijing, P.R. China
| | - Baosheng Liang
- Department of Biostatistics, School of Public Health, Peking University, Beijing, P.R. China
| | - Li Liang
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, P.R. China
| |
Collapse
|
475
|
Pascual T, Cejalvo JM, Oliveira M, Vidal M, Vega E, Ganau S, Julve A, Zamora E, Miranda I, Delgado A, Bermejo B, la Cruz-Merino LD, Juan M, Ferrero-Cafiero JM, Canes J, Gonzalez X, Villagrasa P, Prat A. SOLTI-1503 PROMETEO TRIAL: combination of talimogene laherparepvec with atezolizumab in early breast cancer. Future Oncol 2020; 16:1801-1813. [PMID: 32633563 DOI: 10.2217/fon-2020-0246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
New treatment strategies such as immune checkpoint inhibitors and oncolytic viruses are opening new possibilities in cancer therapy. Preliminary results in melanoma and other tumors showed that the combination of talimogene laherparepvec with an anti-PD-1/PD-L1 or anti-CTLA4 has greater efficacy than either therapy alone, without additional safety concerns beyond those expected for each agent. The presence of residual cancer after neoadjuvant chemotherapy in early breast cancer patients is an unmet medical need. SOLTI-1503 PROMETEO is a window of opportunity trial, which evaluates the combination of talimogene laherparepvec in combination with atezolizumab in women with operable HER2-negative breast cancer who present residual disease after neoadjuvant chemotherapy. The primary end point is the rate of residual cancer burden 0/1. Clinical Trial Registration: NCT03802604.
Collapse
Affiliation(s)
- Tomas Pascual
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Juan M Cejalvo
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Breast Cancer Biology Research Group, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Mafalda Oliveira
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Vidal
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Estela Vega
- Medical Oncology Department, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Sergi Ganau
- Radiology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Ana Julve
- Radiology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Esther Zamora
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ignacio Miranda
- Radiology Department, Breast Imaging Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ana Delgado
- Radiology Department, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Breast Cancer Biology Research Group, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Luis de la Cruz-Merino
- Medical Oncology Department, Hospital Universitario Virgen Macarena. Sevilla, Spain
- Medicine Department, Universidad de Sevilla, Sevilla, Spain
| | - Manel Juan
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Biomedicine Department, University of Barcelona, Barcelona, Spain
- Immunogenetics of the Autoinflammatory Response, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Jordi Canes
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Xavier Gonzalez
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department Hospital Universitari General de Catalunya, Sant Cugat del Vallès, Spain
| | | | - Aleix Prat
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| |
Collapse
|
476
|
Yamashita D, Shimada K, Kohno K, Kogure Y, Kataoka K, Takahara T, Suzuki Y, Satou A, Sakakibara A, Nakamura S, Asano N, Kato S. PD-L1 expression on tumor or stromal cells of nodal cytotoxic T-cell lymphoma: A clinicopathological study of 50 cases. Pathol Int 2020; 70:513-522. [PMID: 32424876 PMCID: PMC7496983 DOI: 10.1111/pin.12950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/12/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
Inhibitors of programmed cell-death 1 (PD-1) and programmed cell-death ligand 1 (PD-L1) have revolutionized cancer therapy. Nodal cytotoxic T-cell lymphoma (CTL) is characterized by a poorer prognosis compared to nodal non-CTLs. Here we investigated PD-L1 expression in 50 nodal CTL patients, with and without EBV association (25 of each). We identified seven patients (14%) with neoplastic PD-L1 (nPD-L1) expression on tumor cells, including three males and four females, with a median age of 66 years. One of the seven cases was TCRαβ type, three were TCRγδ type and three were TCR-silent type. Six of the seven cases exhibited a lethal clinical course despite multi-agent chemotherapy, of whom four patients died within one year of diagnosis. Morphological findings were uniform, with six cases showing centroblastoid appearance. Among nPD-L1+ cases, two of three examined had structural variations of PD-L1 disrupting 3'-UTR region. Notably, all of the TCRγδ-type nodal CTL cases showed nPD-L1 or miPD-L1 positivity (3 and 10 cases, respectively). TCRγδ-type cases comprised 42% of nPD-L1+ cases (P = 0.043 vs. PD-L1- ), and 35% of miPD-L1+ cases (P = 0.037 vs. PD-L1- ). The results indicate that PD-L1+ nodal CTL cases, especially of the TCRγδ type, are potential candidates for anti-PD-1/PD-L1 therapies.
Collapse
MESH Headings
- Adult
- Aged
- B7-H1 Antigen/metabolism
- Biomarkers, Tumor/metabolism
- Epstein-Barr Virus Infections/diagnosis
- Epstein-Barr Virus Infections/pathology
- Female
- Humans
- Lymphoma, T-Cell/diagnosis
- Lymphoma, T-Cell/metabolism
- Lymphoma, T-Cell/pathology
- Lymphoma, T-Cell, Peripheral/diagnosis
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Male
- Middle Aged
- Prognosis
Collapse
Affiliation(s)
- Daisuke Yamashita
- Department of Pathology and Laboratory MedicineNagoya University HospitalAichiJapan
- Department of PathologyKobe City Hospital Organization Kobe City Medical Center General HospitalHyōgoJapan
| | - Kazuyuki Shimada
- Department of Hematology and OncologyNagoya University Graduate School of MedicineAichiJapan
| | - Kei Kohno
- Department of Pathology and Laboratory MedicineNagoya University HospitalAichiJapan
| | - Yasunori Kogure
- Division of Molecular OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Keisuke Kataoka
- Division of Molecular OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Taishi Takahara
- Department of Surgical PathologyAichi Medical University HospitalAichiJapan
| | - Yuka Suzuki
- Department of Pathology and Laboratory MedicineNagoya University HospitalAichiJapan
| | - Akira Satou
- Department of Surgical PathologyAichi Medical University HospitalAichiJapan
| | - Ayako Sakakibara
- Department of Pathology and Laboratory MedicineNagoya University HospitalAichiJapan
| | - Shigeo Nakamura
- Department of Pathology and Laboratory MedicineNagoya University HospitalAichiJapan
| | - Naoko Asano
- Department of Molecular DiagnosticsNagano Prefectural Suzaka HospitalNaganoJapan
| | - Seiichi Kato
- Department of Pathology and Laboratory MedicineNagoya University HospitalAichiJapan
- Department of Pathology and Molecular DiagnosticsAichi Cancer Center HospitalAichiJapan
| |
Collapse
|
477
|
Affiliation(s)
- Kylie M Quinn
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.,Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
478
|
Jiang YZ, Liu Y, Xiao Y, Hu X, Jiang L, Zuo WJ, Ma D, Ding J, Zhu X, Zou J, Verschraegen C, Stover DG, Kaklamani V, Wang ZH, Shao ZM. Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: the FUTURE trial. Cell Res 2020; 31:178-186. [PMID: 32719455 PMCID: PMC8027015 DOI: 10.1038/s41422-020-0375-9] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/30/2020] [Indexed: 01/20/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous disease, and molecular subtyping may result in improved diagnostic precision and targeted therapies. Our previous study classified TNBCs into four subtypes with putative therapeutic targets. Here, we conducted the FUTURE trial (ClinicalTrials.gov identifier: NCT03805399), a phase Ib/II subtyping-based and genomic biomarker-guided umbrella trial, to evaluate the efficacy of these targets. Patients with refractory metastatic TNBC were enrolled and stratified by TNBC subtypes and genomic biomarkers, and assigned to one of these seven arms: (A) pyrotinib with capecitabine, (B) androgen receptor inhibitor with CDK4/6 inhibitor, (C) anti PD-1 with nab-paclitaxel, (D) PARP inhibitor included, (E) and (F) anti-VEGFR included, or (G) mTOR inhibitor with nab-paclitaxel. The primary end point was the objective response rate (ORR). We enrolled 69 refractory metastatic TNBC patients with a median of three previous lines of therapy (range, 1–8). Objective response was achieved in 20 (29.0%, 95% confidence interval (CI): 18.7%–41.2%) of the 69 intention-to-treat (ITT) patients. Our results showed that immunotherapy (arm C), in particular, achieved the highest ORR (52.6%, 95% CI: 28.9%–75.6%) in the ITT population. Arm E demonstrated favorable ORR (26.1%, 95% CI: 10.2%–48.4% in the ITT population) but with more high grade (≥ 3) adverse events. Somatic mutations of TOP2A and CD8 immunohistochemical score may have the potential to predict immunotherapy response in the immunomodulatory subtype of TNBC. In conclusion, the phase Ib/II FUTURE trial suggested a new concept for TNBC treatment, demonstrating the clinical benefit of subtyping-based targeted therapy for refractory metastatic TNBC.
Collapse
Affiliation(s)
- Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yin Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi Xiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Hu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lin Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wen-Jia Zuo
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiahan Ding
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoyu Zhu
- Jiangsu Hengrui Medicine Co Ltd, Lianyungang, Jiangsu, 222002, China
| | - Jianjun Zou
- Jiangsu Hengrui Medicine Co Ltd, Lianyungang, Jiangsu, 222002, China
| | - Claire Verschraegen
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Daniel G Stover
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Virginia Kaklamani
- Division of Hematology/Oncology, University of Texas Health Science Center San Antonio, San Antonio, TX, 78284, USA
| | - Zhong-Hua Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
479
|
Terranova-Barberio M, Pawlowska N, Dhawan M, Moasser M, Chien AJ, Melisko ME, Rugo H, Rahimi R, Deal T, Daud A, Rosenblum MD, Thomas S, Munster PN. Exhausted T cell signature predicts immunotherapy response in ER-positive breast cancer. Nat Commun 2020; 11:3584. [PMID: 32681091 PMCID: PMC7367885 DOI: 10.1038/s41467-020-17414-y] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/24/2020] [Indexed: 01/05/2023] Open
Abstract
Responses to immunotherapy are uncommon in estrogen receptor (ER)-positive breast cancer and to date, lack predictive markers. This randomized phase II study defines safety and response rate of epigenetic priming in ER-positive breast cancer patients treated with checkpoint inhibitors as primary endpoints. Secondary and exploratory endpoints included PD-L1 modulation and T-cell immune-signatures. 34 patients received vorinostat, tamoxifen and pembrolizumab with no excessive toxicity after progression on a median of five prior metastatic regimens. Objective response was 4% and clinical benefit rate (CR + PR + SD > 6 m) was 19%. T-cell exhaustion (CD8+ PD-1+/CTLA-4+) and treatment-induced depletion of regulatory T-cells (CD4+ Foxp3+/CTLA-4+) was seen in tumor or blood in 5/5 patients with clinical benefit, but only in one non-responder. Tumor lymphocyte infiltration was 0.17%. Only two non-responders had PD-L1 expression >1%. This data defines a novel immune signature in PD-L1-negative ER-positive breast cancer patients who are more likely to benefit from immune-checkpoint and histone deacetylase inhibition (NCT02395627).
Collapse
Affiliation(s)
| | - Nela Pawlowska
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Mallika Dhawan
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Mark Moasser
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Amy J Chien
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Michelle E Melisko
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Hope Rugo
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Roshun Rahimi
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Travis Deal
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Adil Daud
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | | | - Scott Thomas
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA
| | - Pamela N Munster
- Division of Hematology and Oncology, University of California, San Francisco, CA, USA.
| |
Collapse
|
480
|
Zhao B, Zhao H, Zhao J. Efficacy of PD-1/PD-L1 blockade monotherapy in clinical trials. Ther Adv Med Oncol 2020; 12:1758835920937612. [PMID: 32728392 PMCID: PMC7366397 DOI: 10.1177/1758835920937612] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 06/05/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Inhibitors targeting programmed cell death 1 (PD-1) and programmed
death-ligand 1 (PD-L1) have unprecedented effects in cancer treatment.
However, the objective response rates (ORRs), progression-free survival
(PFS), and overall survival (OS) of PD-1/PD-L1 blockade monotherapy have not
been systematically evaluated. Methods: We searched Embase, PubMed, and Cochrane database from inception to July 2019
for prospective clinical trials on single-agent PD-1/PD-L1 antibodies
(avelumab, atezolizumab, durvalumab, cemiplimab, pembrolizumab, and
nivolumab) with information regarding ORR, PFS, and OS. Results: Totally, 28,304 patients from 160 perspective trials were included. Overall,
4747 responses occurred in 22,165 patients treated with PD-1/PD-L1
monotherapy [ORR, 20.21%; 95% confidence interval (CI), 18.34–22.15%].
Compared with conventional therapy, PD-1/PD-L1 blockade immunotherapy was
associated with more tumor responses (odds ratio, 1.98; 95% CI, 1.52–2.57)
and better OS [hazard ratio (HR), 0.75; 95% CI, 0.67–0.83]. The ORRs varied
significantly across cancer types and PD-L1 expression status. Line of
treatment, clinical phase and drug target also impacted the response rates
in some tumors. A total of 2313 of 9494 PD-L1 positive patients (ORR,
24.39%; 95% CI, 22.29–26.54%) and 456 of 4215 PD-L1 negative patients (ORR,
10.34%; 95% CI, 8.67–12.14%) achieved responses. For PD-L1 negative
patients, the ORR (odds ratio, 0.92; 95% CI, 0.70–1.20) and PFS (HR, 1.15;
95% CI, 0.87–1.51) associated with immunotherapy and conventional treatment
were similar. However, PD-1/PD-L1 blockade monotherapy decreased the risk of
death in both PD-L1 positive (HR, 0.66; 95% CI, 0.60–0.72) and PD-L1
negative (HR, 0.86; 95% CI, 0.74–0.99) patients compared with conventional
therapy. Conclusion: The efficacies associated with PD-1/PD-L1 monotherapy vary significantly
across cancer types and PD-L1 expression. This comprehensive summary of
clinical benefit from immunotherapy in cancer patients provides an important
guide for clinicians.
Collapse
Affiliation(s)
- Bin Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Rd, Wenzhou, 325035, China
| | - Hong Zhao
- The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Jiaxin Zhao
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
481
|
Zheng S, Zou Y, Xie X, Liang JY, Yang A, Yu K, Wang J, Tang H, Xie X. Development and validation of a stromal immune phenotype classifier for predicting immune activity and prognosis in triple-negative breast cancer. Int J Cancer 2020; 147:542-553. [PMID: 32285442 DOI: 10.1002/ijc.33009] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/25/2020] [Accepted: 03/24/2020] [Indexed: 12/20/2022]
Abstract
Our study aims to construct a prognosis-related immune phenotype classifier for predicting clinical prognosis and immune activity in triple-negative breast cancer (TNBC). A total of 237 patients with TNBC from Sun Yat-sen University Cancer Center (SYSUCC) and 533 patients with TNBC from public datasets were included in our study. A stromal immune quantified index was generated with a LASSO Cox regression model based on five prognosis-related immune cells evaluated by CIBERSORT or IHC and was used to determine immune phenotypes. Immune features were evaluated in the samples before chemotherapy. A total of 119 patients in the SYSUCC training cohort were classified into immune Phenotypes A and B according to the density of stromal CD4+ T cells, γδ T cells, monocytes, M1 macrophages and M2 macrophages. Phenotype A predicted better survival than Phenotype B, and the classification was further validated in the testing cohort of 118 patients and the validation cohort of 533 patients. In the combined cohort, significant differences were found in Phenotype A compared to Phenotype B for the 5-year overall survival (83.5% vs 65.8%, respectively, P < .01) and the 5-year disease-free survival (87.3% vs 76.0%, respectively, P < .01). In Phenotype A, immune-related pathways were significantly enriched, and a higher level of immune checkpoint molecules, including PD-L1, PD-1 and CTLA-4, could be observed. The immune phenotype classification was an independent prognostic indicator for TNBC and might serve as a potential predictor for immune activity within the tumor microenvironment.
Collapse
Affiliation(s)
- Shaoquan Zheng
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yutian Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xinhua Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jie-Ying Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Anli Yang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kai Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hailin Tang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
482
|
Shi Y. Clinical Translation of Nanomedicine and Biomaterials for Cancer Immunotherapy: Progress and Perspectives. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900215] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yang Shi
- Department of Nanomedicine and Theranostics Institute for Experimental Molecular Imaging Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering Faculty of Medicine RWTH Aachen University Aachen 52074 Germany
| |
Collapse
|
483
|
Guo L, Wei R, Lin Y, Kwok HF. Clinical and Recent Patents Applications of PD-1/PD-L1 Targeting Immunotherapy in Cancer Treatment-Current Progress, Strategy, and Future Perspective. Front Immunol 2020; 11:1508. [PMID: 32733486 PMCID: PMC7358377 DOI: 10.3389/fimmu.2020.01508] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Targeting PD-L1 and PD-1 interactions is a relatively new therapeutic strategy used to treat cancer. Inhibitors of PD-1/PD-L1 include peptides, small molecule chemical compounds, and antibodies. Several approved antibodies targeting PD-1 or PD-L1 have been patented with good curative effect in various cancer types in clinical practices. While the current antibody therapy is facing development bottleneck, some companies have tried to develop PD-L1 companion tests to select patients with better diagnosis potential. Meanwhile, many companies have recently synthesized small molecule inhibitors of PD-1/PD-L1 interactions and focused on searching for novel biomarker to predict the efficacy of anti-PD-1/PD-L1 drugs. This review summarized clinical studies and patent applications related to PD-1/PD-L1 targeted therapy and also discussed progress in inhibitors of PD-1/PD-L1.
Collapse
Affiliation(s)
- Libin Guo
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, China
| | - Ran Wei
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, China
| | - Yao Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, China
| |
Collapse
|
484
|
Wang YN, Lee HH, Hsu JL, Yu D, Hung MC. The impact of PD-L1 N-linked glycosylation on cancer therapy and clinical diagnosis. J Biomed Sci 2020; 27:77. [PMID: 32620165 PMCID: PMC7333976 DOI: 10.1186/s12929-020-00670-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022] Open
Abstract
N-linked glycosylation is one of the most abundant posttranslational modifications of membrane-bound proteins in eukaryotes and affects a number of biological activities, including protein biosynthesis, protein stability, intracellular trafficking, subcellular localization, and ligand-receptor interaction. Accumulating evidence indicates that cell membrane immune checkpoint proteins, such as programmed death-ligand 1 (PD-L1), are glycosylated with heavy N-linked glycan moieties in human cancers. N-linked glycosylation of PD-L1 maintains its protein stability and interaction with its cognate receptor, programmed cell death protein 1 (PD-1), and this in turn promotes evasion of T-cell immunity. Studies have suggested targeting PD-L1 glycosylation as a therapeutic option by rational combination of cancer immunotherapies. Interestingly, structural hindrance by N-glycan on PD-L1 in fixed samples impedes its recognition by PD-L1 diagnostic antibodies. Notably, the removal of N-linked glycosylation enhances PD-L1 detection in a variety of bioassays and more accurately predicts the therapeutic efficacy of PD-1/PD-L1 inhibitors, suggesting an important clinical implication of PD-L1 N-linked glycosylation. A detailed understanding of the regulatory mechanisms, cellular functions, and diagnostic limits underlying PD-L1 N-linked glycosylation could shed new light on the clinical development of immune checkpoint inhibitors for cancer treatment and deepen our knowledge of biomarkers to identify patients who would benefit the most from immunotherapy. In this review, we highlight the effects of protein glycosylation on cancer immunotherapy using N-linked glycosylation of PD-L1 as an example. In addition, we consider the potential impacts of PD-L1 N-linked glycosylation on clinical diagnosis. The notion of utilizing the deglycosylated form of PD-L1 as a predictive biomarker to guide anti-PD-1/PD-L1 immunotherapy is also discussed.
Collapse
Affiliation(s)
- Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, 91 Hsueh-Shih Rd, North District, Taichung, 404, Taiwan. .,Department of Biotechnology, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
485
|
Gaynor N, Crown J, Collins DM. Immune checkpoint inhibitors: Key trials and an emerging role in breast cancer. Semin Cancer Biol 2020; 79:44-57. [PMID: 32623044 DOI: 10.1016/j.semcancer.2020.06.016] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/15/2022]
Abstract
This review focuses on immune checkpoint inhibitors - immunomodulatory agents that aim to relieve tumour-mediated immune-cell suppression. Immune checkpoint proteins can be expressed on the tumour-cell or immune-cell populations. Immune checkpoint proteins dampen the immune response by inactivating immune cells capable of tumour destruction. Blockade of immune checkpoints has shown impressive results in a range of solid cancers, particularly melanoma and non-small cell lung cancer. The potential benefit of this class of drugs is widespread across most cancer types and an unprecedented number of clinical studies are underway to examine the benefit of these agents. The aims of this review are to: provide an overview of the key early immune checkpoint inhibitor trials involving drugs targeting programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in multiple disease types; provide an overview of emerging therapies aimed at these targets; and provide a detailed exploration of the status of immune checkpoint inhibitors in breast cancer.
Collapse
Affiliation(s)
- Nicola Gaynor
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland; Department of Medical Oncology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.
| | - Denis M Collins
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
486
|
Vici P, Pizzuti L, Krasniqi E, Botticelli A, Ciliberto G, Barba M. Risk of SARS-CoV-2 infection and disease in metastatic triple-negative breast cancer patients treated with immune checkpoint inhibitors. Immunotherapy 2020; 12:675-679. [PMID: 32489118 PMCID: PMC7273903 DOI: 10.2217/imt-2020-0142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/22/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Eriseld Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Botticelli
- Department of Clinical & Molecular Oncology, University of Rome “Sapienza”, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
487
|
Clinical Development of PD-1/PD-L1 Inhibitors in Breast Cancer: Still a Long Way to Go. Curr Treat Options Oncol 2020; 21:59. [PMID: 32556894 DOI: 10.1007/s11864-020-00756-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Currently, only patients with metastatic triple-negative breast cancer whose tumors are PD-L1 positive are eligible for receiving immunotherapy. Other studies have explored new combinations with PD-1/PD-L1 inhibitors in different disease settings and populations. Data from neoadjuvant trials testing the addition of PD-1/PD-L1 inhibitors to standard treatment are promising and have led to increases in pathologic complete response rates; however, data on survival outcomes are still immature. There is still much work needed to optimize benefits of immunotherapy in breast cancer and correlative studies in patients treated with immunotherapy are urgently needed to inform the best strategies for further development.
Collapse
|
488
|
Loibl S, Untch M, Burchardi N, Huober J, Sinn BV, Blohmer JU, Grischke EM, Furlanetto J, Tesch H, Hanusch C, Engels K, Rezai M, Jackisch C, Schmitt WD, von Minckwitz G, Thomalla J, Kümmel S, Rautenberg B, Fasching PA, Weber K, Rhiem K, Denkert C, Schneeweiss A. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol 2020; 30:1279-1288. [PMID: 31095287 DOI: 10.1093/annonc/mdz158] [Citation(s) in RCA: 461] [Impact Index Per Article: 92.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Combining immune-checkpoint inhibitors with chemotherapy yielded an increased response rates in patients with metastatic triple-negative breast cancer (TNBC). Therefore, we evaluated the addition of durvalumab to standard neoadjuvant chemotherapy (NACT) in primary TNBC. PATIENTS AND METHODS GeparNuevo is a randomised phase II double-blind placebo-controlled study randomising patients with TNBC to durvalumab or placebo given every 4 weeks in addition to nab-paclitaxel followed by standard EC. In the window-phase durvalumab/placebo alone was given 2 weeks before start of nab-paclitaxel. Randomisation was stratified by stromal tumour-infiltrating lymphocyte (sTILs). Patients with primary cT1b-cT4a-d disease, centrally confirmed TNBC and sTILs were included. Primary objective was pathological complete response (pCR) (ypT0 ypN0). RESULTS A total of 174 patients were randomised, 117 participated in the window-phase. Median age was 49.5 years (range 23-76); 47 patients (27%) were younger than 40 years; 113 (65%) had stage ≥IIA disease, 25 (14%) high sTILs, 138 of 158 (87%) were PD-L1-positive. pCR rate with durvalumab was 53.4% (95% CI 42.5% to 61.4%) versus placebo 44.2% (95% CI 33.5% to 55.3%; unadjusted continuity corrected χ2P = 0.287), corresponding to OR = 1.45 (95% CI 0.80-2.63, unadjusted Wald P = 0.224). Durvalumab effect was seen only in the window cohort (pCR 61.0% versus 41.4%, OR = 2.22, 95% CI 1.06-4.64, P = 0.035; interaction P = 0.048). In both arms, significantly increased pCR (P < 0.01) were observed with higher sTILs. There was a trend for increased pCR rates in PD-L1-positive tumours, which was significant for PD-L1-tumour cell in durvalumab (P = 0.045) and for PD-L1-immune cell in placebo arm (P = 0.040). The most common immune-related adverse events were thyroid dysfunction any grade in 47%. CONCLUSIONS Our results suggest that the addition of durvalumab to anthracycline-/taxane-based NACT increases pCR rate particularly in patients treated with durvalumab alone before start of chemotherapy. TRIAL REGISTRATION ClinicalTrials.gov number: NCT02685059.
Collapse
Affiliation(s)
- S Loibl
- German Breast Group, Neu-Isenburg; Oncological Practice Bethanien, Cancer Center Frankfurt Northeast, Frankfurt am Main.
| | - M Untch
- HELIOS Klinikum Berlin-Buch, Berlin
| | | | - J Huober
- Brustzentrum, Universitätsfrauenklinik Ulm, Ulm
| | - B V Sinn
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin; Berlin Institute of Health (BIH), Berlin
| | - J-U Blohmer
- Gynäkologie mit Brustzentrum, Charité-Universitätsmedizin Berlin, Berlin
| | | | | | - H Tesch
- Oncological Practice Bethanien, Cancer Center Frankfurt Northeast, Frankfurt am Main
| | - C Hanusch
- Rotkreuzklinikum München Frauenklinik, München
| | - K Engels
- Zentrum für Pathologie, Zytologie und Molekularpathologie Neuss, Neuss
| | - M Rezai
- Medical Center, Luisenkrankenhaus Düsseldorf, Düsseldorf
| | - C Jackisch
- Brustzentrum, Sana-Klinikum Offenbach, Offenbach
| | - W D Schmitt
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin
| | | | - J Thomalla
- Praxisklinik für Hämatologie und Onkologie Koblenz, Koblenz
| | - S Kümmel
- Breast Unit, Kliniken Essen-Mitte, Essen
| | - B Rautenberg
- Klinik für Frauenheilkunde, Universitätsklinikum Freiburg, Freiburg
| | - P A Fasching
- Brustzentrum, Universitätsklinikum Erlangen, Erlangen
| | - K Weber
- German Breast Group, Neu-Isenburg
| | - K Rhiem
- Center for Hereditary Breast and Ovarian Cancer, University Hospital Cologne, Cologne
| | - C Denkert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin
| | - A Schneeweiss
- National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
489
|
|
490
|
Pilipow K, Darwich A, Losurdo A. T-cell-based breast cancer immunotherapy. Semin Cancer Biol 2020; 72:90-101. [PMID: 32492452 DOI: 10.1016/j.semcancer.2020.05.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/22/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy has witnessed a new renaissance with the advent of immune checkpoint inhibitors, which reactivate T cells and foster endogenous anti-tumor responses. The excellent results of immunotherapy in the field of melanoma, renal cancer, lung cancer, and other cancer types that have traditionally been known to be immunogenic, rekindled the interest of the oncology community in extending the benefits to all cancers including breast cancer (BC). In this review, we highlight the current state of using T cells as both markers for clinical practice and therapeutic options for BC.
Collapse
Affiliation(s)
- Karolina Pilipow
- Laboratory of Translational Immunology, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy
| | - Abbass Darwich
- Laboratory of Mucosal Immunology and Microbiota, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy; Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, MI, Italy
| | - Agnese Losurdo
- Laboratory of Translational Immunology, Italy; Medical Oncology and Hematology Unit, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy.
| |
Collapse
|
491
|
Malhotra MK, Emens LA. The evolving management of metastatic triple negative breast cancer. Semin Oncol 2020; 47:229-237. [PMID: 32563561 DOI: 10.1053/j.seminoncol.2020.05.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Advanced triple negative breast cancer (TNBC) is an incurable disease classified by its lack of expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2. Due to its lack of therapeutic targets, it has historically been treated with single agent chemotherapy, with combination cytotoxic therapy typically reserved for patients with high disease burdens, symptomatic disease, and/or impending visceral crisis. Recent molecular analyses have revealed that this clinical group of TNBCs is in fact quite biologically heterogeneous, with multiple TNBC subtypes defined by distinct biology and clinical behavior. Building on this biology, 2 targeted strategies are now approved for selected patients with advanced TNBC: the poly (ADP-ribose) polymerase inhibitors for advanced TNBC with a germline mutation in BRCA1/2, and the combination of the programmed death ligand 1-specific antibody atezolizumab with nab-paclitaxel for advanced TNBC that expresses programmed death ligand 1 on immune cells within the tumor. These targeted agents tend to be associated with a more favorable side effect profile and longer disease control than standard chemotherapy. A number of other targeted therapies have shown promise in early clinical trials, and several are now in definitive phase 3 testing for advanced TNBC. These include the antiapoptotic kinase inhibitors ipatisertib and capivasertib, and the antibody-drug conjugate sacituzumab govitecan-hziy. Approved biomarker-driven treatment options for this disease are thus likely to expand in the near-term. Here we review current treatment options and emerging targeted therapies for advanced TNBC. For patients who do not meet criteria for approved targeted therapies, participation in clinical trials evaluating precision medicines with candidate predictive biomarkers in advanced TNBC should be encouraged.
Collapse
Affiliation(s)
- Monica K Malhotra
- University of Pittsburgh Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Leisha A Emens
- University of Pittsburgh Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA.
| |
Collapse
|
492
|
Pérez-García J, Soberino J, Racca F, Gion M, Stradella A, Cortés J. Atezolizumab in the treatment of metastatic triple-negative breast cancer. Expert Opin Biol Ther 2020; 20:981-989. [PMID: 32450725 DOI: 10.1080/14712598.2020.1769063] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) accounts for approximately 10%-15% of all diagnosed breast cancers and is associated with an aggressive natural history and poor clinical outcomes. Immunotherapy using immune checkpoint inhibitors has emerged as an effective therapeutic option for TNBC. The results of the IMpassion130 trial have recently led to the approval of the combination of atezolizumab and nab-paclitaxel in the first-line treatment of patients with unresectable locally advanced or metastatic, PD-L1-positive TNBC. AREAS COVERED This article summarizes the clinical development and ongoing research on atezolizumab in the treatment of metastatic TNBC. Results of atezolizumab monotherapy trials and data from combination studies with chemotherapy in the advanced setting are reviewed, with special focus on the design, methods, and key findings of the IMpassion130 trial. EXPERT OPINION The approval of atezolizumab plus nab-paclitaxel represents an important advance in the treatment of metastatic TNBC. This combination has a favorable risk-benefit profile and is associated with clinically meaningful outcomes. However, further research is needed to identify better predictive biomarkers of response as well as novel immunotherapeutic strategies with atezolizumab and other anticancer drugs.
Collapse
Affiliation(s)
- José Pérez-García
- IOB Institute of Oncology, Quironsalud Group , Madrid & Barcelona, Spain.,Medica Scientia Innovation Research (MedSIR) , Barcelona, Spain
| | - Jesús Soberino
- IOB Institute of Oncology, Quironsalud Group , Madrid & Barcelona, Spain
| | - Fabricio Racca
- IOB Institute of Oncology, Quironsalud Group , Madrid & Barcelona, Spain
| | - María Gion
- IOB Institute of Oncology, Quironsalud Group , Madrid & Barcelona, Spain.,Hospital Universitario Ramón y Cajal , Madrid, Spain
| | - Agostina Stradella
- IOB Institute of Oncology, Quironsalud Group , Madrid & Barcelona, Spain.,Institut Català d'Oncologia, Hospitalet de Llobregat , Barcelona, Spain
| | - Javier Cortés
- IOB Institute of Oncology, Quironsalud Group , Madrid & Barcelona, Spain.,Medica Scientia Innovation Research (MedSIR) , Barcelona, Spain.,Vall d´Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| |
Collapse
|
493
|
Sugie T, Sato E, Miyashita M, Yamaguchi R, Sakatani T, Kozuka Y, Moritani S, Suzuki E, Kakimi K, Mikami Y, Moriya T. Multispectral quantitative immunohistochemical analysis of tumor-infiltrating lymphocytes in relation to programmed death-ligand 1 expression in triple-negative breast cancer. Breast Cancer 2020; 27:519-526. [PMID: 32447649 DOI: 10.1007/s12282-020-01110-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/13/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) expression on immune cells (ICs) is a predictive marker for PD-L1 checkpoint blockade in patients with triple-negative breast cancer (TNBC). However, the level of PD-L1 expression and the percentage of cells that are PD-L1+ are continuous variables not dichotomous variables for tumor-infiltrating lymphocytes (TILs) and other cells. METHODS Multiplexed immunohistochemistry was applied to 31 archived surgical specimens from untreated TNBC patients. TIL levels were visually scored, and CD8+ T cells and PD-L1+ ICs were quantified using an automated multispectral imaging system. PD-L1 expression was assessed within a multiplexed context (CD8 combined spectral composite). RESULTS The mean value of stromal TILs (i.e., the percentage of the stromal area with a dese mononuclear infiltrate) was 20%. The frequency of patients with PD-L1-positive tumor cells (TC) and ICs was 38.7% and 32.2%, respectively, with a significant association between them. TIL levels were correlated with CD8+ T cell infiltration in the stroma (Spearman r = 0.795, p < 0.0001). PD-L1 expression on IC was significantly associated with TIL levels (Spearman r = 0.790, p < 0.001) and infiltration of CD8+ T cells (Spearman r = 0.683, p < 0.0001). CONCLUSIONS The level of PD-L1 on IC was correlated with the level of PD-L1 on TC as well as TIL levels and infiltration of CD8+ T cells. These results suggest that high PD-L1 on IC may reflect T cell-inflamed tumors with the amount of TILs present, including the CD8+ T cells required for anti-tumor responses.
Collapse
Affiliation(s)
- Tomoharu Sugie
- Breast Surgery, Kansai Medical University Hospital, Hirakata, Japan.
| | - Eiichi Sato
- Department of Pathology (Medical Research Center), Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Rin Yamaguchi
- Department of Pathology and Clinical Medicine, Kurume University Medical Center, Kurume, Japan
| | - Takashi Sakatani
- Department of Diagnostic Pathology, Nippon Medical School Hospital, Tokyo, Japan
| | - Yuji Kozuka
- Department of Pathology, Mie University Hospital, Tsu, Japan
| | - Suzuko Moritani
- Department of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Eiji Suzuki
- Breast Surgery, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Kazuhiro Kakimi
- Cancer Immunology Data Multi-level Integration Unit, Medical Science Innovation, Hub Program, RIKEN, Tokyo, Japan
| | - Yoshiki Mikami
- Department of Diagnostic Pathology, Kumamoto University Hospital, Kumamoto, Japan
| | - Takuya Moriya
- Department of Pathology, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
494
|
Cao L, Niu Y. Triple negative breast cancer: special histological types and emerging therapeutic methods. Cancer Biol Med 2020; 17:293-306. [PMID: 32587770 PMCID: PMC7309458 DOI: 10.20892/j.issn.2095-3941.2019.0465] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/05/2020] [Indexed: 12/23/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a complex and malignant breast cancer subtype that lacks expression of the estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2), thereby making therapeutic targeting difficult. TNBC is generally considered to have high malignancy and poor prognosis. However, patients diagnosed with certain rare histomorphologic subtypes of TNBC have better prognosis than those diagnosed with typical triple negative breast cancer. In addition, with the discovery and development of novel treatment targets such as the androgen receptor (AR), PI3K/AKT/mTOR and AMPK signaling pathways, as well as emerging immunotherapies, the therapeutic options for TNBC are increasing. In this paper, we review the literature on various histological types of TNBC and focus on newly developed therapeutic strategies that target and potentially affect molecular pathways or emerging oncogenes, thus providing a basis for future tailored therapies focused on the mutational aspects of TNBC.
Collapse
Affiliation(s)
- Lu Cao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yun Niu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
495
|
Hudeček J, Voorwerk L, van Seijen M, Nederlof I, de Maaker M, van den Berg J, van de Vijver KK, Sikorska K, Adams S, Demaria S, Viale G, Nielsen TO, Badve SS, Michiels S, Symmans WF, Sotiriou C, Rimm DL, Hewitt SM, Denkert C, Loibl S, Loi S, Bartlett JMS, Pruneri G, Dillon DA, Cheang MCU, Tutt A, Hall JA, Kos Z, Salgado R, Kok M, Horlings HM. Application of a risk-management framework for integration of stromal tumor-infiltrating lymphocytes in clinical trials. NPJ Breast Cancer 2020; 6:15. [PMID: 32436923 PMCID: PMC7217941 DOI: 10.1038/s41523-020-0155-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 02/18/2020] [Indexed: 02/08/2023] Open
Abstract
Stromal tumor-infiltrating lymphocytes (sTILs) are a potential predictive biomarker for immunotherapy response in metastatic triple-negative breast cancer (TNBC). To incorporate sTILs into clinical trials and diagnostics, reliable assessment is essential. In this review, we propose a new concept, namely the implementation of a risk-management framework that enables the use of sTILs as a stratification factor in clinical trials. We present the design of a biomarker risk-mitigation workflow that can be applied to any biomarker incorporation in clinical trials. We demonstrate the implementation of this concept using sTILs as an integral biomarker in a single-center phase II immunotherapy trial for metastatic TNBC (TONIC trial, NCT02499367), using this workflow to mitigate risks of suboptimal inclusion of sTILs in this specific trial. In this review, we demonstrate that a web-based scoring platform can mitigate potential risk factors when including sTILs in clinical trials, and we argue that this framework can be applied for any future biomarker-driven clinical trial setting.
Collapse
Affiliation(s)
- Jan Hudeček
- Department of Research IT, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leonie Voorwerk
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maartje van Seijen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Iris Nederlof
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michiel de Maaker
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jose van den Berg
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Karolina Sikorska
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sylvia Adams
- Department of Medicine, Perlmutter Cancer Center, New York University School of Medicine, New York, NY USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY USA
| | - Giuseppe Viale
- International Breast Cancer Study Group Central Pathology Office, Department of Pathology and Laboratory Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy
- University of Milan, Milan, Italy
| | - Torsten O. Nielsen
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, BC Canada
| | - Sunil S. Badve
- Department of Pathology and Laboratory Medicine, Indiana University Simon Cancer Center, Indianapolis, IN USA
| | - Stefan Michiels
- Service de Biostatistique et d’Epidémiologie, Gustave Roussy, CESP, Université-Paris Sud, Université Paris-Saclay, Villejuif, France
- CESP, Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
| | | | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, U-CRC, Université Libre de Bruxelles, Brussels, Belgium
| | - David L. Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Stephen M. Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD USA
| | - Carsten Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Pathology, Philipps-University Marburg, Marburg, Germany
| | | | - Sherene Loi
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC Australia
| | - John M. S. Bartlett
- Ontario Institute for Cancer Research, Toronto, ON Canada
- IGMM, Edinburgh, UK
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, IRCCS Fondazion - Instituto Nazionale Tumori, Milan, Italy
- School of Medicine, University of Milan, Milan, Italy
| | - Deborah A. Dillon
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Maggie C. U. Cheang
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, Surrey, UK
| | - Andrew Tutt
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - Zuzana Kos
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON Canada
| | - Roberto Salgado
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC Australia
- Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
| | - Marleen Kok
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hugo M. Horlings
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
496
|
Botticelli A, Cirillo A, Scagnoli S, Cerbelli B, Strigari L, Cortellini A, Pizzuti L, Vici P, De Galitiis F, Di Pietro FR, Cerbelli E, Ghidini M, D’Amati G, Della Rocca C, Mezi S, Gelibter A, Giusti R, Cortesi E, Ascierto PA, Nuti M, Marchetti P. The Agnostic Role of Site of Metastasis in Predicting Outcomes in Cancer Patients Treated with Immunotherapy. Vaccines (Basel) 2020; 8:vaccines8020203. [PMID: 32353934 PMCID: PMC7349154 DOI: 10.3390/vaccines8020203] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors have revolutionized treatment and outcome of melanoma and many other solid malignancies including non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC). Unfortunately, only a minority of patients have a long-term benefit, while the remaining demonstrate primary or acquired resistance. Recently, it has been demonstrated that the prevalence of programmed death-ligand 1 (PD-L1) and tumor-infiltrating lymphocytes (TILs) varies based on the anatomical site of metastases. In particular, liver seems to have more immunosuppressive microenvironment while both the presence of lymph nodal disease and lung metastases seem to have the highest prevalence of PD-L1 and TILs. The aim of the present study is to investigate the possible role of site of metastases as a predictive factor for response or resistance to immunotherapy in several types of cancer. In this multicenter retrospective study, we enrolled patients with metastatic NSCLC, melanoma, RCC, urothelial, merkel carcinoma, and colon cancer who received immunotherapy from April 2015 to August 2019. Major clinicopathological parameters were retrieved and correlated with patients’ survival outcomes in order to assess their prognostic value and build a useful tool to assist in the decision-making process. A total of 291 patients were included in this study. One hundred eighty-seven (64%) patients were male and 104 (36%) female. The tumor histology was squamous NSCLC in 56 (19%) patients, non-squamous NSCLC in 99 (34%) patients, melanoma in 101 (35%) patients, RCC in 28 (10%) patients, and other tumors in the remaining 7 (2%) patients. The number of metastatic sites was 1 in 103 patients (35%), 2 in 104 patients (36%) and 3 in 84 patients (29%). Out of 183 valuable patients, the entity of response was complete response (CR), partial response (PR), stable disease (SD), and progression disease (PD) in 15, 53, 31, and 79 patients, respectively. Using an univariate analysis (UVA), tumor burden (p = 0.0004), the presence of liver (p = 0.0009), bone (p = 0.0016), brain metastases (p < 0.0001), the other metastatic sites (p = 0.0375), the number of metastatic sites (p = 0.0039), the histology (p = 0.0034), the upfront use of immunotherapy (p = 0.0032), and Eastern Cooperative Oncology Group (ECOG) Perfomance status (PS) ≥ 1 (p < 0.0001) were significantly associated with poor overall survival (OS). Using a multivariate analysis (MVA) the presence of liver (p = 0.0105) and brain (p = 0.0026) metastases, the NSCLC diagnosis (p < 0.0001) and the ECOG PS (p < 0.0001) resulted as significant prognostic factors of survival. Regarding the progression free survival (PFS), using a UVA of the tumor burden (p = 0.0004), bone (p = 0.0098) and brain (p = 0.0038) metastases, the presence of other metastatic sites (p = 0.0063), the number of metastatic sites (p = 0.0007), the histology (p = 0.0007), the use of immunotherapy as first line (p = 0.0031), and the ECOG PS ≥ 1 (p ≤ 0.0001) were associated with a lower PFS rate. Using an MVA, the presence of brain (p = 0.0088) and liver metastases (p = 0.024) and the ECOG PS (p < 0.0001) resulted as predictors of poor PFS. Our study suggests that the site of metastases could have a role as prognostic and predictive factor in patients treated with immunotherapy. Indeed, regardless of the histology, the presence of liver and brain metastases was associated with a shorter PFS and OS, but these results must be confirmed in further studies. In this context, a deep characterization of microenvironment could be crucial to prepare patients through novel strategies with combination or sequential immunotherapy in order to improve treatment response.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and molecular oncology, University of Rome “Sapienza”, 00185 Rome, Italy; (A.B.); (P.M.)
| | - Alessio Cirillo
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Simone Scagnoli
- Department of Medical and Surgical Sciences and Translational Medicine, University of Rome “Sapienza”, 00185 Rome, Italy
- Correspondence: ; Tel.: +39-3280306525
| | - Bruna Cerbelli
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Lidia Strigari
- Medical Physics Unit, “S. Orsola-Malpighi” Hospital, 40138 Bologna, Italy;
| | - Alessio Cortellini
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 671000 L’Aquila, Italy; (A.C.); (L.P.)
| | - Laura Pizzuti
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 671000 L’Aquila, Italy; (A.C.); (L.P.)
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | | | | | - Edoardo Cerbelli
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Michele Ghidini
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy;
| | - Giulia D’Amati
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, 00185 Roma, Italy;
| | - Silvia Mezi
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Alain Gelibter
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Raffaele Giusti
- Sant’Andrea Hospital, Department of clinical and molecular medicine, Sapienza, University of Rome, 00153 Roma, Italy;
| | - Enrico Cortesi
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Via Mariano Semmola snc, 80131 Naples, Italy;
| | - Marianna Nuti
- Department of Experimental Medicine, University Sapienza, 00185 Rome, Italy;
| | - Paolo Marchetti
- Department of Clinical and molecular oncology, University of Rome “Sapienza”, 00185 Rome, Italy; (A.B.); (P.M.)
| |
Collapse
|
497
|
Manjili MH. The premise of personalized immunotherapy for cancer dormancy. Oncogene 2020; 39:4323-4330. [PMID: 32322001 PMCID: PMC7260096 DOI: 10.1038/s41388-020-1295-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022]
Abstract
Progress in cancer therapies has resulted in improved survival of patients with early stage breast cancer. However, mortality remains high in patients with distant recurrence of the disease after initially successful treatment of early stage breast cancer. To this end, tumor recurrences have been attributed to the presence of dormant tumor cells in breast cancer patients and cancer survivors. Current clinical practice guidelines recommend a “wait and watch” approach for tumor recurrence. This is because of our limited understanding of tumor dormancy. Dormant tumor cells are quiescent, and thus, do not respond to chemotherapies or radiation therapies, and they are not operable. Therefore, immunotherapy is the only option for the treatment of tumor dormancy. However, gaps in our knowledge as to dormancy-specific antigens prevent a relapse preventing vaccine design. Here, we provide a critical review of cancer immunotherapy, and discuss empirical evidence related to naturally-occurring tumor dormancy and treatment-induced tumor dormancy at the site of primary tumor and in distant organs before and after cancer therapies. Finally, we suggest that personalized vaccines targeting dormancy-associated neoantigens, which can be given to patients with early stage disease after the completion of neoadjuvant therapies and tumor resection as well as to cancer survivors could eliminate relapse causing dormant cells and offer a cure for cancer.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, VCU School of Medicine, VCU Institute of Molecular Medicine, Massey Cancer Center, Richmond, VA, USA.
| |
Collapse
|
498
|
Monaco SE, Dacic S, Seigh L, Hartman DJ, Xing J, Pantanowitz L. Quantitative image analysis for CD8 score in lung small biopsies and cytology cell-blocks. Cytopathology 2020; 31:393-401. [PMID: 32065467 DOI: 10.1111/cyt.12812] [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] [Received: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Immunotherapy has shown promising results in non-small cell lung cancer (NSCLC), for which tumour-infiltrating cytotoxic (CD8+) T cells play a critical role. We investigated the utility of image analysis (IA) to quantify CD8+ T cells in a series of matched small biopsies and resections of NSCLC. METHODS CD8 immunohistochemistry was performed on cell-blocks (CB), core needle biopsies (CNB) and corresponding resections from primary NSCLCs. Slides were digitised using an Aperio AT2 scanner (Leica) and annotated by whole slide image (WSI) or fields of view occupied by tissue spots (TS). Quantitative IA was performed with a customised Aperio algorithm (Leica). CD8 scores (number of T cells with 1-3+ staining/total area) were then compared. RESULTS Forty-four cases with CB or CNB material and a corresponding resection were analysed. Average CD8 score was determined in CB (7.67 WSI, 77.67 TS) and/or CNB (47.35 WSI, 325.67 TS), and corresponding resections (190.35 WSI, 336.58 TS). CD8 score concordance was highest (78.6%) for CNBs using WSI annotation. Overall, small biopsies (CB or CNB) correlated with the resection in 71.4% cases using WSI and 63.3% cases using TS annotation. IA performed better for low CD8 scores. CONCLUSIONS These findings show that CD8 density in NSCLC can be quantified by IA in small biopsies and cell blocks, achieving the best concordance using WSI scores. Discrepancies were attributed to values near the cut-off and background detection of staining. These data warrant future studies with more cases and follow-up data to further investigate the clinical utility of IA for CD8 analysis in NSCLC.
Collapse
Affiliation(s)
- Sara E Monaco
- Department of Pathology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Lindsey Seigh
- Department of Pathology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Douglas J Hartman
- Department of Pathology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Juan Xing
- Department of Pathology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Liron Pantanowitz
- Department of Pathology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| |
Collapse
|
499
|
Zhao J, Huang J. Breast cancer immunology and immunotherapy: targeting the programmed cell death protein-1/programmed cell death protein ligand-1. Chin Med J (Engl) 2020; 133:853-862. [PMID: 32106121 PMCID: PMC7147660 DOI: 10.1097/cm9.0000000000000710] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 12/28/2022] Open
Abstract
Historically, breast cancer has been regarded as an immunogenic "cold" tumor. However, the discovery of immune checkpoint inhibitors has made immunotherapy becoming an emerging new treatment modality for breast cancer. This review discusses the immune system, immune features of breast cancer, and the programmed cell death protein-1/programmed cell death protein ligand-1 (PD-1/PD-L1) inhibitors used in the treatment of breast cancer. High T lymphocyte infiltration and mutation burden were observed in triple-negative breast cancer and human epidermal growth factor receptor 2 positive breast cancer. Increasing breast cancer immunogenicity and modulating the tumor microenvironment has been reported to improve the therapeutic efficacy of immunotherapy. Recent clinical trials involving PD-1/PD-L1 inhibitors monotherapy in breast cancer has revealed little efficacy, which highlights the need to develop combinations of PD-1/PD-L1 inhibitors with chemotherapy, molecularly targeted therapies, and other immunotherapies to maximize the clinical efficacy. Collectively, the immunotherapy might be a promising therapeutic strategy for breast cancer and several clinical trials are still on-going.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
500
|
Hypoxia-sensitive supramolecular nanogels for the cytosolic delivery of ribonuclease A as a breast cancer therapeutic. J Control Release 2020; 320:83-95. [DOI: 10.1016/j.jconrel.2020.01.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/02/2020] [Accepted: 01/09/2020] [Indexed: 01/08/2023]
|