1
|
Stroes CI, Meijer SL, Creemers GJ, Hooijer GKJ, Mohammad NH, Los M, Slingerland M, Hospers GAP, Cats A, Beerepoot LV, Bijlsma MF, van Laarhoven HWM. Tumor immune microenvironmental characteristics in Human Epidermal Growth Factor-2 (HER2) positive esophageal adenocarcinoma: A comparative analysis and biomarker study. Transl Oncol 2024; 49:102079. [PMID: 39151279 PMCID: PMC11375138 DOI: 10.1016/j.tranon.2024.102079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/11/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND HER2 targeting in esophageal adenocarcinoma (EAC) has shown potential, but often fails to show durable response. Given the contributions of the tumor immune microenvironment (TIME) to therapeutic responses, we aimed to chart the TIME characteristics of HER2 positive tumors. METHODS 84 biopsies were taken from the TRAP cohort (neoadjuvant chemoradiotherapy (nCRT) according to CROSS with trastuzumab and pertuzumab; n = 40; HER2+n = 40) and a control cohort with nCRT only (n = 44; HER2- n = 40, HER2+n = 4) before treatment. Biopsies were analysed using targeted gene expression analysis (Nanostring immune-oncology panel, 750 genes). Differential gene expression was assessed between HER2 positive (n = 44) vs. negative biopsies (n = 40), and non-responders (n = 17) vs. responders (n = 23) to anti-HER2 treatment. Statistical significance was determined as p-value <0.05, adjusted for multiple testing correction. RESULTS 83 biopsies were eligible for analyses following quality control (TRAP cohort n = 40; control cohort n = 43); there were no significant differences in clinical characteristics between the TRAP vs. control the cohort or HER2 positive vs. HER2 negative biopsies. HER2 expression was found to associate with epithelial markers (EPCAM p < 0.001; E-cadherin p < 0.001). Moreover, HER2 expression was associated with a lower expression of immune cell infiltration, such as NK-cells (p < 0.001) and CD8 T-cells (p < 0.001), but also lower expression of immune exhaustion markers (PDCD1LG2, CTLA4; p < 0.001). In non-responders to anti-HER2 treatment, baseline biopsies showed increased expression of immune exhaustion markers, as well as hypoxia and VEGF signalling. DISCUSSION HER2 expression was associated with epithelial tumor characteristics. The HER2 positive TIME showed reduced immune cell infiltration but also lower expression of inhibitory signals associated with immune exhaustion, questioning the mechanism behind potential clinical benefit of co-administration of anti-HER2 agents and checkpoint inhibitors. As limited response was associated with increased VEGF signalling, studies could investigate potential synergism of targeting VEGF and HER2.
Collapse
Affiliation(s)
- Charlotte I Stroes
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands.
| | - Sybren L Meijer
- Amsterdam UMC, Location University of Amsterdam, Department of Pathology, Amsterdam, the Netherlands
| | - Geert-Jan Creemers
- Catharina Hospital, Department of Medical Oncology, Eindhoven, the Netherlands
| | - Gerrit K J Hooijer
- Amsterdam UMC, Location University of Amsterdam, Department of Pathology, Amsterdam, the Netherlands
| | - Nadia Haj Mohammad
- Universtiy Medical Center Utrecht, Department of Medical Oncology, Utrecht University, Utrecht, the Netherlands
| | - Maartje Los
- Sint Antonius Hospital, Department of Medical Oncology, Nieuwegein, the Netherlands
| | - Marije Slingerland
- Leiden University Medical Center, Department of Medical Oncology, Leiden, the Netherlands
| | - Geke A P Hospers
- Universtiy Medical Center Groningen, Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Annemieke Cats
- Netherlands Cancer Institute, Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | - Laurens V Beerepoot
- Elisabeth-TweeSteden Hospital, Department of Medical Oncology, Tilburg, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Cai A, Chen Y, Wang LS, Cusick JK, Shi Y. Depicting Biomarkers for HER2-Inhibitor Resistance: Implication for Therapy in HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:2635. [PMID: 39123362 PMCID: PMC11311605 DOI: 10.3390/cancers16152635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is highly expressed in a variety of cancers, including breast, lung, gastric, and pancreatic cancers. Its amplification is linked to poor clinical outcomes. At the genetic level, HER2 is encoded by the ERBB2 gene (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), which is frequently mutated or amplified in cancers, thus spurring extensive research into HER2 modulation and inhibition as viable anti-cancer strategies. An impressive body of FDA-approved drugs, including anti-HER2 monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and HER2-tyrosine kinase inhibitors (TKIs), have demonstrated success in enhancing overall survival (OS) and disease progression-free survival (PFS). Yet, drug resistance remains a persistent challenge and raises the risks of metastatic potential and tumor relapse. Research into alternative therapeutic options for HER2+ breast cancer therefore proves critical for adapting to this ever-evolving landscape. This review highlights current HER2-targeted therapies, discusses predictive biomarkers for drug resistance, and introduces promising emergent therapies-especially combination therapies-that are aimed at overcoming drug resistance in the context of HER2+ breast cancer.
Collapse
Affiliation(s)
- Alvan Cai
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Lily S. Wang
- University of California, Berkeley, CA 94720, USA;
| | - John K. Cusick
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
3
|
Biswas M. Understanding tissue-resident macrophages unlocks the potential for novel combinatorial strategies in breast cancer. Front Immunol 2024; 15:1375528. [PMID: 39104525 PMCID: PMC11298421 DOI: 10.3389/fimmu.2024.1375528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/14/2024] [Indexed: 08/07/2024] Open
Abstract
Tissue-resident macrophages (TRMs) are an integral part of the innate immune system, but their biology is not well understood in the context of cancer. Distinctive resident macrophage populations are identified in different organs in mice using fate mapping studies. They develop from the yolk sac and self-maintain themselves lifelong in specific tissular niches. Similarly, breast-resident macrophages are part of the mammary gland microenvironment. They reside in the breast adipose tissue stroma and close to the ductal epithelium and help in morphogenesis. In breast cancer, TRMs may promote disease progression and metastasis; however, precise mechanisms have not been elucidated. TRMs interact intimately with recruited macrophages, cytotoxic T cells, and other immune cells along with cancer cells, deciding further immunosuppressive or cytotoxic pathways. Moreover, triple-negative breast cancer (TNBC), which is generally associated with poor outcomes, can harbor specific TRM phenotypes. The influence of TRMs on adipose tissue stroma of the mammary gland also contributes to tumor progression. The complex crosstalk between TRMs with T cells, stroma, and breast cancer cells can establish a cascade of downstream events, understanding which can offer new insight for drug discovery and upcoming treatment choices. This review aims to acknowledge the previous research done in this regard while exploring existing research gaps and the future therapeutic potential of TRMs as a combination or single agent in breast cancer.
Collapse
Affiliation(s)
- Manjusha Biswas
- Department of Molecular Biomedicine, Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| |
Collapse
|
4
|
Sanad AM, Ibrahim WS, Ezzo IM, Sabry RM. Is Programmed Death-Ligand 1 of Prognostic Significance in Triple-Negative Female Mammary Carcinoma? J Microsc Ultrastruct 2024; 12:6-13. [PMID: 38633572 PMCID: PMC11019586 DOI: 10.4103/jmau.jmau_77_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction The most widespread female malignancy is breast cancer (BC), considerable percentage of patients with triple-negative BC (TNBC) experience rapid progression, recurrence, and metastasis. BC has not historically been treated as an immunogenic cancer. Nonetheless, several researchers have started to concentrate on immunotherapy. Aim The aim of the study is to investigate the immunohistochemical (IHC) expression of programmed death-ligand 1 (PD-L1) by stromal tumor-infiltrating lymphocytes (TILs) and tumor cells (TC) in female (TNBC) and to correlate with pathological features of such tumors, particularly those determine biologic behavior, such as the grade and stage the overall survival. Methodology This is a retrospective study which includes 49 paraffin-embedded tumor tissue sections which were collected from breast surgery specimens either radical or conservative of female patients with TNBC. The samples were analyzed immunohistochemically for PD-L1 expression. Results There were statistically significant relations among TC PD-L1 expression and TILs PD-L1 expression as well as relations among TILs PD-L1 expression with histologic grade, stromal TILs, and Ki-67 were statistically significant. Correlations between TC PD-L1 expression and N stage, histologic grade, and anatomic stage were statistically significant. Improved survival was detected within TILs PD-L1-positive cases; however, the correlation between the overall survival and PD-L1 expression in both TCs and stromal TIL was not statistically significant. Conclusion PD-L1 expressed in tumors with poor prognostic features such as the high grade, advanced T stage, and high Ki-67 index, TILs PD-L1-positive cases experienced improved survival supporting its prognostic significance. However, the correlation with overall survival was not statistically significant.
Collapse
Affiliation(s)
- Amal Mostafa Sanad
- Department of Pathology, Faculty of Medicine, Tanta Cancer Center, Tanta University, Tanta, Egypt
| | - Wael Shawky Ibrahim
- Department of Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Iman Mohamed Ezzo
- Department of Pathology, Tanta Cancer Center, Tanta University, Tanta, Egypt
| | - Rania Mohamed Sabry
- Department of Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
5
|
Mandelkow T, Bady E, Lurati MCJ, Raedler JB, Müller JH, Huang Z, Vettorazzi E, Lennartz M, Clauditz TS, Lebok P, Steinhilper L, Woelber L, Sauter G, Berkes E, Bühler S, Paluchowski P, Heilenkötter U, Müller V, Schmalfeldt B, von der Assen A, Jacobsen F, Krech T, Krech RH, Simon R, Bernreuther C, Steurer S, Burandt E, Blessin NC. Automated Prognosis Marker Assessment in Breast Cancers Using BLEACH&STAIN Multiplexed Immunohistochemistry. Biomedicines 2023; 11:3175. [PMID: 38137396 PMCID: PMC10741079 DOI: 10.3390/biomedicines11123175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/12/2023] [Accepted: 11/18/2023] [Indexed: 12/24/2023] Open
Abstract
Prognostic markers in routine clinical management of breast cancer are often assessed using RNA-based multi-gene panels that depend on fluctuating tumor purity. Multiplex fluorescence immunohistochemistry (mfIHC) holds the potential for an improved risk assessment. To enable automated prognosis marker detection (i.e., progesterone receptor [PR], estrogen receptor [ER], androgen receptor [AR], GATA3, TROP2, HER2, PD-L1, Ki67, TOP2A), a framework for automated breast cancer identification was developed and validated involving thirteen different artificial intelligence analysis steps and an algorithm for cell distance analysis using 11+1-marker-BLEACH&STAIN-mfIHC staining in 1404 invasive breast cancers of no special type (NST). The framework for automated breast cancer detection discriminated normal glands from malignant glands with an accuracy of 98.4%. This approach identified that five (PR, ER, AR, GATA3, PD-L1) of nine biomarkers were associated with prolonged overall survival (p ≤ 0.0095 each) and two of these (PR, AR) were found to be independent risk factors in multivariate analysis (p ≤ 0.0151 each). The combined assessment of PR-ER-AR-GATA3-PD-L1 as a five-marker prognosis score showed strong prognostic relevance (p < 0.0001) and was an independent risk factor in multivariate analysis (p = 0.0034). Automated breast cancer detection in combination with an artificial intelligence-based analysis of mfIHC enables a rapid and reliable analysis of multiple prognostic parameters. The strict limitation of the analysis to malignant cells excludes the impact of fluctuating tumor purity on assay precision.
Collapse
Affiliation(s)
- Tim Mandelkow
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Elena Bady
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Magalie C. J. Lurati
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jonas B. Raedler
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- College of Arts and Sciences, Boston University, Boston, MA 02215, USA
| | - Jan H. Müller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Zhihao Huang
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eik Vettorazzi
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Till S. Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Institute of Pathology, Clinical Center Osnabrück, 49076 Osnabrück, Germany
| | - Lisa Steinhilper
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Linn Woelber
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Enikö Berkes
- Department of Gynecology, Albertinen Clinic Schnelsen, 22457 Hamburg, Germany
| | - Simon Bühler
- Department of Gynecology, Amalie Sieveking Clinic, 22359 Hamburg, Germany
| | - Peter Paluchowski
- Department of Gynecology, Regio Clinic Pinneberg, 25421 Pinneberg, Germany
| | - Uwe Heilenkötter
- Department of Gynecology, Clinical Centre Itzehoe, 25524 Itzehoe, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Till Krech
- Institute of Pathology, Clinical Center Osnabrück, 49076 Osnabrück, Germany
| | - Rainer H. Krech
- Institute of Pathology, Clinical Center Osnabrück, 49076 Osnabrück, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Niclas C. Blessin
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
6
|
Swaminathan H, Saravanamurali K, Yadav SA. Extensive review on breast cancer its etiology, progression, prognostic markers, and treatment. Med Oncol 2023; 40:238. [PMID: 37442848 DOI: 10.1007/s12032-023-02111-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
As the most frequent and vulnerable malignancy among women, breast cancer universally manifests a formidable healthcare challenge. From a biological and molecular perspective, it is a heterogenous disease and is stratified based on the etiological factors driving breast carcinogenesis. Notably, genetic predispositions and epigenetic impacts often constitute the heterogeneity of this disease. Typically, breast cancer is classified intrinsically into histological subtypes in clinical landscapes. These stratifications empower physicians to tailor precise treatments among the spectrum of breast cancer therapeutics. In this pursuit, numerous prognostic algorithms are extensively characterized, drastically changing how breast cancer is portrayed. Therefore, it is a basic requisite to comprehend the multidisciplinary rationales of breast cancer to assist the evolution of novel therapeutic strategies. This review aims at highlighting the molecular and genetic grounds of cancer additionally with therapeutic and phytotherapeutic context. Substantially, it also renders researchers with an insight into the breast cancer cell lines as a model paradigm for breast cancer research interventions.
Collapse
Affiliation(s)
- Harshini Swaminathan
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - K Saravanamurali
- Virus Research and Diagnostics Laboratory, Department of Microbiology, Coimbatore Medical College, Coimbatore, India
| | - Sangilimuthu Alagar Yadav
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| |
Collapse
|
7
|
Pan L, Han J, Lin M. Targeting breast cancer stem cells directly to treat refractory breast cancer. Front Oncol 2023; 13:981247. [PMID: 37251931 PMCID: PMC10213424 DOI: 10.3389/fonc.2023.981247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/02/2023] [Indexed: 05/31/2023] Open
Abstract
For patients with refractory breast cancer (BC), integrative immunotherapies are emerging as a critical component of treatment. However, many patients remain unresponsive to treatment or relapse after a period. Different cells and mediators in the tumor microenvironment (TME) play important roles in the progression of BC, and cancer stem cells (CSCs) are deemed the main cause of relapse. Their characteristics depend on their interactions with their microenvironment as well as on the inducing factors and elements in this environment. Strategies to modulate the immune system in the TME of BC that are aimed at reversing the suppressive networks within it and eradicating residual CSCs are, thus, essential for improving the current therapeutic efficacy of BC. This review focuses on the development of immunoresistance in BCs and discusses the strategies that can modulate the immune system and target breast CSCs directly to treat BC including immunotherapy with immune checkpoint blockades.
Collapse
Affiliation(s)
- Liping Pan
- Wuhan Center for Clinical Laboratory, Wuhan, China
| | - Juan Han
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Lin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Mondal SK, Bhattacharya S, Sarkar AP, Saha R. Expression of programmed death ligand-1 (PD-L1) and tumor infiltrating lymphocytes (TILs) in breast carcinoma and their clinical significance. J Cancer Res Ther 2023; 19:S81-S86. [PMID: 37147987 DOI: 10.4103/jcrt.jcrt_2013_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Background and Aim Carcinoma of the breast is the second most common cause of cancer death in women. Expression of programmed death ligand-1 (PD-L1) in cancer cells plays an important role in tailored therapy. This can be evaluated by immunohistochemistry using a monoclonal PD-L1 antibody in formalin-fixed and paraffin-embedded (FFPE) specimens. Our aim was to evaluate the expression of PD-L1 and tumor infiltrating lymphocytes (TILs) in invasive carcinoma of breast and their clinicopathological correlation. Materials and Methods Immunohistochemical staining for PD-L1 and TILs was done in paraffin-embedded tissues of histologically diagnosed 50 cases of breast carcinoma. Statistical analysis was done using Statistical Package for the Social Sciences (SPSS) 22 software. Results Out of these 50 cases, PD-L1 and TIL expression were seen in 16 (32%) cases and 18 (36%) cases, respectively. PD-L1 positivity was seen in 33.33% cases of grade 1 breast carcinoma, 13.79% of cases of grade 2 breast carcinoma, and in 75% case of grade 3 breast carcinoma. TILs showed positivity in 6.9% cases of grade 1 breast carcinoma, 13.79% of cases of grade 2 breast carcinoma, and in 100% cases of grade 3 breast carcinoma. Proportion of patients having PD-L1 expression was higher in grade 3 carcinoma than in grade 1 or 2. The differences were statistically significant (Chi-square value = 13.417, degree of freedom = 1, P < 0.05). The Chi-square value for TILs was 28.07, degree of freedom was 1, and P value was <0.05, which was statistically significant. Conclusion Both PD-L1 and TILs showed maximum positivity in grade 3 breast carcinoma.
Collapse
Affiliation(s)
- Santosh Kumar Mondal
- Department of Pathology, All India Institute of Medical Science, Kalyani, West Bengal, India
| | | | - Aditya Prasad Sarkar
- Community Medicine, Bankura Sammilani Medical College, Bankura, West Bengal, India
| | - Rama Saha
- Department of Pathology, All India Institute of Medical Science, Kalyani, West Bengal, India
| |
Collapse
|
9
|
Mercogliano MF, Bruni S, Mauro FL, Schillaci R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15071987. [PMID: 37046648 PMCID: PMC10093019 DOI: 10.3390/cancers15071987] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of death. HER2 overexpression is found in approximately 20% of breast cancers and is associated with a poor prognosis and a shorter overall survival. Tratuzumab, a monoclonal antibody directed against the HER2 receptor, is the standard of care treatment. However, a third of the patients do not respond to therapy. Given the high rate of resistance, other HER2-targeted strategies have been developed, including monoclonal antibodies such as pertuzumab and margetuximab, trastuzumab-based antibody drug conjugates such as trastuzumab-emtansine (T-DM1) and trastuzumab-deruxtecan (T-DXd), and tyrosine kinase inhibitors like lapatinib and tucatinib, among others. Moreover, T-DXd has proven to be of use in the HER2-low subtype, which suggests that other HER2-targeted therapies could be successful in this recently defined new breast cancer subclassification. When patients progress to multiple strategies, there are several HER2-targeted therapies available; however, treatment options are limited, and the potential combination with other drugs, immune checkpoint inhibitors, CAR-T cells, CAR-NK, CAR-M, and vaccines is an interesting and appealing field that is still in development. In this review, we will discuss the highlights and pitfalls of the different HER2-targeted therapies and potential combinations to overcome metastatic disease and resistance to therapy.
Collapse
|
10
|
Duro-Sánchez S, Alonso MR, Arribas J. Immunotherapies against HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15041069. [PMID: 36831412 PMCID: PMC9954045 DOI: 10.3390/cancers15041069] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths among women worldwide. HER2-positive breast cancer, which represents 15-20% of all cases, is characterized by the overexpression of the HER2 receptor. Despite the variety of treatments available for HER2-positive breast cancer, both targeted and untargeted, many patients do not respond to therapy and relapse and eventually metastasize, with a poor prognosis. Immunotherapeutic approaches aim to enhance the antitumor immune response to prevent tumor relapse and metastasis. Several immunotherapies have been approved for solid tumors, but their utility for HER2-positive breast cancer has yet to be confirmed. In this review, we examine the different immunotherapeutic strategies being tested in HER2-positive breast cancer, from long-studied cancer vaccines to immune checkpoint blockade, which targets immune checkpoints in both T cells and tumor cells, as well as the promising adoptive cell therapy in various forms. We discuss how some of these new approaches may contribute to the prevention of tumor progression and be used after standard-of-care therapies for resistant HER2-positive breast tumors, highlighting the benefits and drawbacks of each. We conclude that immunotherapy holds great promise for the treatment of HER2-positive tumors, with the potential to completely eradicate tumor cells and prevent the progression of the disease.
Collapse
Affiliation(s)
- Santiago Duro-Sánchez
- Preclinical & Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Campus de la UAB, 08193 Bellaterra, Spain
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Macarena Román Alonso
- Preclinical & Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Campus de la UAB, 08193 Bellaterra, Spain
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Joaquín Arribas
- Preclinical & Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Campus de la UAB, 08193 Bellaterra, Spain
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|
11
|
Durak Ö, Bozkurt KK, Çiriş İM, Kocer M, Eroğlu HE. Programmed cell death 1 and programmed cell death ligand 1 expression in invasive breast carcinoma using CAL10 and NAT105 immunostaining. Biotech Histochem 2023; 98:147-154. [PMID: 36281760 DOI: 10.1080/10520295.2022.2137586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increased incidence of breast cancer has stimulated development of new diagnostic and therapeutic methods. The programmed cell death 1 (PD1) pathway and its inhibitors are promising avenues for investigation. PD1 includes PD ligands 1 (PDL1) and 2 (PDL2). We investigated the expression of PD1 and PDL1 in invasive breast carcinomas using immunohistochemical staining. We used 171 invasive breast carcinoma specimens from which tissue microarray blocks were created. Immunohistochemical staining of PD1 using NAT105, and PDL1 using CAL10 was performed on tissue microarray sections. NAT105 and CAL10 are useful clones for detecting expression of PD1 and PDL1. PD1 and PDL1 immunostaining was significantly stronger in carcinomas with basal-like phenotype compared to other molecular breast cancer types. PD1 and PDL1 expression also was associated with a high histologic grade and a high Ki-67 index. PD1 expression also was associated with lymphovascular invasion and axillary metastasis. PD1 and PDL1 expression is associated with aggressive tumor behavior and a basal-like phenotype in breast cancer. We suggest that inhibition of the PD1/PDL1 pathway, particularly in triple negative breast carcinomas with basal-like phenotype, might be useful for targeted immunotherapy.
Collapse
Affiliation(s)
- Özlem Durak
- Department of Pathology, Kastamonu State Hospital, Kastamonu, Turkey
| | | | | | - Murat Kocer
- Department of Medical Oncology, Antalya Education and Research Hospital, Health Sciences University, Antalya, Turkey
| | | |
Collapse
|
12
|
Aisa A, Weng S, Li X, Zhang D, Yuan Y. Immune Checkpoint Inhibitors combined with HER-2 targeted therapy in HER-2 positive Gastroesophageal cancer. Crit Rev Oncol Hematol 2022; 180:103864. [DOI: 10.1016/j.critrevonc.2022.103864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
|
13
|
Lee DY, Im E, Yoon D, Lee YS, Kim GS, Kim D, Kim SH. Pivotal role of PD-1/PD-L1 immune checkpoints in immune escape and cancer progression: Their interplay with platelets and FOXP3+Tregs related molecules, clinical implications and combinational potential with phytochemicals. Semin Cancer Biol 2022; 86:1033-1057. [PMID: 33301862 DOI: 10.1016/j.semcancer.2020.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 01/27/2023]
Abstract
Immune checkpoint proteins including programmed cell death protein 1 (PD-1), its ligand PD-L1 and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are involved in proliferation, angiogenesis, metastasis, chemoresistance via immune escape and immune tolerance by disturbing cytotoxic T cell activation. Though many clinical trials have been completed in several cancers by using immune checkpoint inhibitors alone or in combination with other agents to date, recently multi-target therapy is considered more attractive than monotherapy, since immune checkpoint proteins work with other components such as surrounding blood vessels, dendritic cells, fibroblasts, macrophages, platelets and extracellular matrix within tumor microenvironment. Thus, in the current review, we look back on research history of immune checkpoint proteins and discuss their associations with platelets or tumor cell induced platelet aggregation (TCIPA) and FOXP3+ regulatory T cells (Tregs) related molecules involved in immune evasion and tumor progression, clinical implications of completed trial results and signaling networks by phytochemicals for combination therapy with immune checkpoint inhibitors and suggest future research perspectives.
Collapse
Affiliation(s)
- Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Eunji Im
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dahye Yoon
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Geum-Soog Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Donghwi Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
14
|
Chow CY, Lie EF, Wu CH, Chow LW. Clinical implication of genetic composition and molecular mechanism on treatment strategies of HER2-positive breast cancers. Front Oncol 2022; 12:964824. [PMID: 36387174 PMCID: PMC9659858 DOI: 10.3389/fonc.2022.964824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/14/2022] [Indexed: 12/01/2022] Open
Abstract
The current clinical management model of HER2-positive breast cancers is commonly based on guidelines, which in turn are based on the design and outcome of clinical trials. While this model is useful to most practicing clinicians, the treatment outcome of individual patient is not certain at the start of treatment. As the understanding of the translational research of carcinogenesis and the related changes in cancer genetics and tumor microenvironment during treatment is critical in the selection of right choice of treatment to maximize the successful clinical outcome for the patient, this review article intends to discuss the latest developments in the genetic and molecular mechanisms of cancer progression and treatment resistance, and how they influence the planning of the treatment strategies of HER2-positive breast cancers.
Collapse
Affiliation(s)
- Christopher Y.C. Chow
- UNIMED Medical Institute, Hong Kong, Hong Kong SAR, China
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | | | - Cheng-Hsun Wu
- Department of Anatomy, China Medical University, Taichung, Taiwan
| | - Louis W.C. Chow
- UNIMED Medical Institute, Hong Kong, Hong Kong SAR, China
- Organisation for Oncology and Translational Research, Hong Kong, Hong Kong SAR, China
- *Correspondence: Louis W.C. Chow,
| |
Collapse
|
15
|
Elliott MJ, Wilson B, Cescon DW. Current Treatment and Future Trends of Immunotherapy in Breast Cancer. Curr Cancer Drug Targets 2022; 22:667-677. [PMID: 35301950 DOI: 10.2174/1568009622666220317091723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022]
Abstract
Immunotherapy continues to redefine the solid tumor treatment landscape with inhibitors of the PD-L1/PD-1 immune checkpoint having the most widespread impact. As the most common cancer diagnosed worldwide, there is significant interest in the development of immunotherapy for the treatment of breast cancer in both the early and metastatic settings. Recently reported results of several clinical trials have identified potential roles for immunotherapy agents alone or in combination with standard treatment for early and metastatic disease. While trials to date have been promising, immunotherapy thus far has been shown to benefit only a select group of patients with breast cancer, defined by tumor subtype, PD-L1 expression, and line of therapy. With over 250 trials ongoing, emerging data will enable the further refinement of breast cancer immunotherapy strategies. The integration of multiple putative biomarkers and consideration of dynamic markers of early response or resistance may inform optimal patient selection for immunotherapy investigation and integration into clinical practice. This review will summarize the current evidence for immune-checkpoint blockade (ICB) in the treatment of early and metastatic breast cancer and highlight current and potential future biomarkers of therapeutic response.
Collapse
Affiliation(s)
- Mitchell J Elliott
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Brooke Wilson
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
- University of New South Wales, Kensington, New South Wales, Australia
| | - David W Cescon
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Wilson GM, Dinh P, Pathmanathan N, Graham JD. Ductal Carcinoma in Situ: Molecular Changes Accompanying Disease Progression. J Mammary Gland Biol Neoplasia 2022; 27:101-131. [PMID: 35567670 PMCID: PMC9135892 DOI: 10.1007/s10911-022-09517-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/13/2022] [Indexed: 10/26/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive ductal carcinoma (IDC), whereby if left untreated, approximately 12% of patients develop invasive disease. The current standard of care is surgical removal of the lesion, to prevent potential progression, and radiotherapy to reduce risk of recurrence. There is substantial overtreatment of DCIS patients, considering not all DCIS lesions progress to invasive disease. Hence, there is a critical imperative to better predict which DCIS lesions are destined for poor outcome and which are not, allowing for tailored treatment. Active surveillance is currently being trialed as an alternative management practice, but this approach relies on accurately identifying cases that are at low risk of progression to invasive disease. Two DCIS-specific genomic profiling assays that attempt to distinguish low and high-risk patients have emerged, but imperfections in risk stratification coupled with a high price tag warrant the continued search for more robust and accessible prognostic biomarkers. This search has largely turned researchers toward the tumor microenvironment. Recent evidence suggests that a spectrum of cell types within the DCIS microenvironment are genetically and phenotypically altered compared to normal tissue and play critical roles in disease progression. Uncovering the molecular mechanisms contributing to DCIS progression has provided optimism for the search for well-validated prognostic biomarkers that can accurately predict the risk for a patient developing IDC. The discovery of such markers would modernize DCIS management and allow tailored treatment plans. This review will summarize the current literature regarding DCIS diagnosis, treatment, and pathology.
Collapse
Affiliation(s)
- Gemma M Wilson
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Phuong Dinh
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Nirmala Pathmanathan
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - J Dinny Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
17
|
Nicotine treatment regulates PD-L1 and PD-L2 expression via inhibition of Akt pathway in HER2-type breast cancer cells. PLoS One 2022; 17:e0260838. [PMID: 35085258 PMCID: PMC8794171 DOI: 10.1371/journal.pone.0260838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
The immune checkpoint molecules such as PD-L1 and PD-L2 have a substantial contribution to cancer immunotherapy including breast cancer. Microarray expression profiling identified several molecular subtypes, namely luminal-type (with a good-prognosis), HER2-type (with an intermediate-prognosis), and triple-negative breast cancer (TNBC)-type (with a poor-prognosis). We found that PD-L1 and PD-L2 mRNA expressions were highly expressed in TNBC-type cell lines (HCC1937, MDA-MB-231), moderately expressed in HER2-type cell line (SK-BR-3), and poorly expressed in luminal-type cell lines (MDA-MB-361, MCF7). The PD-L1 and PD-L2 expression in SK-BR-3 cells, but not those in HCC1937 and MDA-MB-231 cells, decreased by nicotine stimulation in a dose-dependent manner. In addition, nicotine treatment decreased the phosphorylation of Akt in SK-BR-3 cells, but not in other cell lines. These results show that nicotine regulates the expression of immune checkpoint molecules, PD-L1 and PD-L2, via inhibition of Akt phosphorylation. This findings may provide the new therapeutic strategies for the treatment of breast cancer.
Collapse
|
18
|
Botticelli A, Pomati G, Marchetti P. Target therapy in cancer treatment. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
19
|
Liu X, Mai J, Meng C, Spiegel AJ, Wei W, Shen H. Antitumor Immunity from Abdominal Flap-Embedded Therapeutic Cancer Vaccine. Int J Nanomedicine 2022; 17:203-212. [PMID: 35046655 PMCID: PMC8760982 DOI: 10.2147/ijn.s341394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/26/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Xiaoling Liu
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, People’s Republic of China
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Chaoyang Meng
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Aldona J Spiegel
- Institute for Reconstructive Surgery, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX, USA
| | - Wei Wei
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, People’s Republic of China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
- Correspondence: Haifa Shen Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USATel +1 713-441-7321 Email
| |
Collapse
|
20
|
Lee S, Kim JY, Lee SJ, Kwon SW, Jung HJ, Jung SJ, Kim KB, Choi KU, Lee CH, Huh GY, Kim A. Lymphocyte-activating gene-3 expression is associated with tumor-infiltrating lymphocyte levels in HER2-positive breast cancers. Medicine (Baltimore) 2021; 100:e28057. [PMID: 34918659 PMCID: PMC8678028 DOI: 10.1097/md.0000000000028057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
Lymphocyte-activating gene-3 (LAG-3, CD223) is the third inhibitory receptor targeted for immunotherapy. Several clinical trials investigating the use of interventions targeting LAG-3 are underway. The exact signaling mechanism downstream of LAG-3 is largely unknown, especially in breast cancer. The prognostic significance of tumor-infiltrating lymphocytes (TILs) in breast cancer has been previously determined.Among 167 human epidermal growth factor receptor 2 (HER2)-positive breast cancer patients, 90 and 78 patients were positive and negative for the hormone receptor, respectively. LAG-3 mRNA and protein expression levels in TILs were evaluated by quantitative real-time polymerase chain reaction and immunohistochemistry, respectively, among 12 and 167 HER2-positive breast cancer samples, respectively.High expression of LAG-3 in TILs was significantly correlated with high levels of TILs (P = .003) and an abundance of tertiary lymphoid structures around invasive components (P = .014). In addition, high expression of LAG3 was significantly associated with positivity for programmed death-ligand 1 (PD-L1) in tumor cells, a high immunostaining score of PD-L1 in TILs, and a high total immunostaining score for PD-L1 in tumor cells and TILs (all, P < .001). High expression levels of LAG-3 mRNA were associated with high levels of TILs (P = .091).LAG-3 protein expression was not a prognostic factor in HER2-positive breast cancers, and LAG-3 expression in TILs was significantly associated with the levels of TILs in HER2-positive breast cancer, although it was not a prognostic factor.
Collapse
Affiliation(s)
- Seokwon Lee
- Department of Surgery, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Jee Yeon Kim
- Department of Pathology, Yangsan Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - So Jeong Lee
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Soon Wook Kwon
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Ho Jin Jung
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Se Jin Jung
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Kyung Bin Kim
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Kyung Un Choi
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Chang Hun Lee
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Gi Yeong Huh
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| | - Ahrong Kim
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, Busan, Republic of Korea
| |
Collapse
|
21
|
Matusz-Fisher A, Tan AR. Combination of HER2-targeted agents with immune checkpoint inhibitors in the treatment of HER2-positive breast cancer. Expert Opin Biol Ther 2021; 22:385-395. [PMID: 34806498 DOI: 10.1080/14712598.2021.1981284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Human epidermal growth factor receptor 2 (HER2)-positive breast cancers account for approximately 15 to 20% of breast cancer diagnoses. Historically, HER2-positive breast cancers had been associated with poorer prognosis. The addition of HER2-targeted agents to treatment regimens has significantly improved outcomes for patients with HER2-positive breast cancer. Despite this, relapses continue to occur in about 20% of patients. Newer therapeutic strategies are needed. The role of immunotherapy in the treatment of HER2-positive breast cancer is currently under clinical investigation. AREAS COVERED This article will focus on the clinical trial data evaluating immune checkpoint inhibitors, including pembrolizumab, atezolizumab, avelumab, durvalumab, and nivolumab in the treatment of HER2-positive breast cancer. EXPERT OPINION The incorporation of immunotherapy in the treatment of HER2-positive breast cancer is a reasonable strategy. Clinical trials of checkpoint inhibitors with HER2-targeted agents show clinical activity in HER2-positive breast cancer tumors that are programmed cell death-ligand 1 (PD-L1) positive and also when used as an earlier line of therapy in the metastatic setting. Treatment of HER2-positive breast cancer with immunotherapy and HER2-targeted agents warrants continued clinical investigation.
Collapse
Affiliation(s)
- Ashley Matusz-Fisher
- Hematology/Oncology Fellow, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Antoinette R Tan
- Chief of Breast Medical Oncology, Co-Director, Phase I Program, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| |
Collapse
|
22
|
Watase C, Shiino S, Shimoi T, Noguchi E, Kaneda T, Yamamoto Y, Yonemori K, Takayama S, Suto A. Breast Cancer Brain Metastasis-Overview of Disease State, Treatment Options and Future Perspectives. Cancers (Basel) 2021; 13:cancers13051078. [PMID: 33802424 PMCID: PMC7959316 DOI: 10.3390/cancers13051078] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary In this review, we present the latest information on the pathophysiology, diagnosis, and local and systemic treatment of brain metastases from breast cancer, with a focus on recent publications. Improving the local treatment and subtype-specific systemic therapies through advancements in basic and translational research will contribute to better clinical outcomes for patients with breast cancer brain metastasis. Abstract Breast cancer is the second most common origin of brain metastasis after lung cancer. Brain metastasis in breast cancer is commonly found in patients with advanced course disease and has a poor prognosis because the blood–brain barrier is thought to be a major obstacle to the delivery of many drugs in the central nervous system. Therefore, local treatments including surgery, stereotactic radiation therapy, and whole-brain radiation therapy are currently considered the gold standard treatments. Meanwhile, new targeted therapies based on subtype have recently been developed. Some drugs can exceed the blood–brain barrier and enter the central nervous system. New technology for early detection and personalized medicine for metastasis are warranted. In this review, we summarize the historical overview of treatment with a focus on local treatment, the latest drug treatment strategies, and future perspectives using novel therapeutic agents for breast cancer patients with brain metastasis, including ongoing clinical trials.
Collapse
Affiliation(s)
- Chikashi Watase
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan; (C.W.); (S.S.); (S.T.)
| | - Sho Shiino
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan; (C.W.); (S.S.); (S.T.)
| | - Tatsunori Shimoi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan; (T.S.); (E.N.); (K.Y.)
| | - Emi Noguchi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan; (T.S.); (E.N.); (K.Y.)
| | - Tomoya Kaneda
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Yusuke Yamamoto
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan;
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan; (T.S.); (E.N.); (K.Y.)
| | - Shin Takayama
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan; (C.W.); (S.S.); (S.T.)
| | - Akihiko Suto
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan; (C.W.); (S.S.); (S.T.)
- Correspondence: ; Tel.: +81-3-3542-2511; Fax: +81-3-3545-3567
| |
Collapse
|
23
|
Zhang JQ, Plitas G. Immunotherapeutic strategies in breast cancer: A clinical update. J Surg Oncol 2021; 123:710-717. [PMID: 33155281 PMCID: PMC7889634 DOI: 10.1002/jso.26287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022]
Abstract
Immunotherapy has been incorporated into the standard of care for a wide range of malignancies. The study of tumor-infiltrating lymphocytes has emphasized the importance of the host antitumor immune response in the natural history of breast cancer. Recent clinical trials have used immunotherapeutic approaches to augment this response and improve outcomes for patients with breast cancer. Here, we review several current clinical trial data that indicate checkpoint blockade may mediate clinically significant responses.
Collapse
Affiliation(s)
- Jennifer Q. Zhang
- Department of Surgery, Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - George Plitas
- Department of Surgery, Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Ludwig Center at Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
24
|
Kuznetsova OA, Zavalishina LE, Andreeva YY, Vinogradov MI, Shomova MV, Frank GA. [Immunohistochemical study of MSI markers in breast cancer]. Arkh Patol 2021; 83:12-17. [PMID: 33512122 DOI: 10.17116/patol20218301112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate the expression of microsatellite instability (MSI) markers, which is detected by an immunohistochemical technique, and to compare the expression with the PD-L1 status in luminal B, HER2-negative and triple-negative breast cancer. MATERIAL AND METHODS The investigation included tumors from 40 patients with triple-negative and luminal B, HER2-negative subtypes. Immunohistochemical study was performed using Ventana antibodies: anti-MLH1 (clone M1), anti-MSH2 (clone G219-1129), anti-PMS2 (clone A16-4), and anti-MSH6 (clone SP93). MSI was assessed according to the standard criteria. RESULTS The PD-L1-positive status was present in 14 (35%) of the 40 patients. Moreover, MSI-H was detected in only 1 (2.50%) case. The two-year survival rate was 87.5%; it should be noted that the median survival rate was not reached either in the study sample or in the groups divided according to PD-L1 and MSI statuses. The overall survival rate for patients with MSI was 75% (3/4). CONCLUSION The first comparative study of the expression of PD-L1 and immunohistochemical MSI markers, which has been conducted on a small sample, fails to draw unambiguous conclusions, but shows the need to investigate this phenomenon on large samples and by using genetic methods.
Collapse
Affiliation(s)
- O A Kuznetsova
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia, Moscow, Russia
| | - L E Zavalishina
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia, Moscow, Russia
| | - Yu Yu Andreeva
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia, Moscow, Russia
| | | | - M V Shomova
- Regional Clinical Oncology Dispensary, Ryazan, Russia
| | - G A Frank
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
25
|
Tonse R, Rubens M, Appel H, Tom MC, Hall MD, Odia Y, McDermott MW, Ahluwalia MS, Mehta MP, Kotecha R. Systematic review and meta-analysis of PD-L1 expression discordance between primary tumor and lung cancer brain metastasis. Neurooncol Adv 2021; 3:vdab166. [PMID: 34988451 PMCID: PMC8704382 DOI: 10.1093/noajnl/vdab166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Novel immunotherapeutic strategies targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis are often administered when metastatic tumors show PD-L1 positivity, even in the setting of lung cancer brain metastasis (LCBM). However, biological differences exist between primary tumors and metastatic sites. The objective of this study was to analyze rates of PD-L1 receptor discordance between primary tumors and LCBM. Methods A systematic review of studies of biopsied or resected LCBM evaluating PD-L1 discordance published in the Medline database was performed using PRISMA guidelines. Weighted random effects models were used to calculate pooled estimates. Results Six full-text articles (n = 230 patients) with a median of 32 patients in each study (range: 24–73) reported PD-L1 receptor expression analyses of both primary lung tumors and brain metastases and met inclusion criteria. The pooled estimate for tumor cell (TC) PD-L1 receptor discordance between primary tumors and LCBM was 19% (95% confidence interval [CI]: 10–27%). For PD-L1 receptor expression in tumor-infiltrating lymphocytes (TIL), the weighted pooled estimate for discordance was 21% (95% CI: 8–44%). For primary versus LCBM, the positive rates by expression levels of <1%, 1–50%, and >50% were 52% (95% CI: 30–73%) versus 56% (95% CI: 34–76%), 30% (95% CI: 22–40%) versus 20% (95% CI: 10–35%), and 15% (95% CI: 6–36%) versus 22% (95% CI: 15–31%) (P = .425), respectively. Conclusions PD-L1 discordance occurs in ~20% of LCBM, with the greatest discordance in the 1–50% expression category. Although controversial, confirming discordance might be important for selection of immune checkpoint inhibitor therapy and in the analysis of patterns of failure after treatment.
Collapse
Affiliation(s)
- Raees Tonse
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Muni Rubens
- Office of Clinical Research, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Haley Appel
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Martin C Tom
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Matthew D Hall
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Yazmin Odia
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Division of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Michael W McDermott
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Department of Neurosurgery, Miami Neuroscience Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Manmeet S Ahluwalia
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| |
Collapse
|
26
|
Chatzopoulos K, Collins AR, Sotiriou S, Keeney MG, Visscher DW, Rivera M, Schembri-Wismayer DJ, Lewis JE, Greipp PT, Sukov WR, Chintakuntlawar AV, Price KA, Garcia JJ. Increased ERBB2 Gene Copy Numbers Reveal a Subset of Salivary Duct Carcinomas with High Densities of Tumor Infiltrating Lymphocytes and PD-L1 Expression. Head Neck Pathol 2020; 14:951-965. [PMID: 32350809 PMCID: PMC7669929 DOI: 10.1007/s12105-020-01163-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/22/2020] [Indexed: 02/08/2023]
Abstract
Salivary duct carcinoma (SDC) commonly expresses androgen receptor (AR) and HER2, giving rise to treatment implications. SDC may also express programmed-death-ligand-1 (PD-L1), a predictive marker of response to checkpoint inhibitors. PD-L1 can be associated with genomic instability and high density of tumor infiltrating lymphocytes (TILs). Evaluation of HER2 immunohistochemistry (IHC) in SDC is not standardized, and relationships between ERBB2 copy numbers, PD-L1 expression and TILs in SDC are unknown. We evaluated 32 SDCs for HER2, AR and PD-L1 expression (IHC), ERBB2 status (FISH) and TILs (slide review). HER2 was scored with three different systems (breast, gastric, proposed salivary gland). PD-L1 was evaluated with the combined positive score. Most patients were older men, presenting at advanced clinical stage with nodal or distant metastases. During follow-up (mean 5 years, range 6 months to 21 years), 25 of the 32 patients (78%) died of SDC. We propose a HER2 IHC scoring system which accurately predicts underlying ERBB2 amplification or increased copy numbers in SDC. Most tumors had increased ERBB2 copy numbers (19/32 amplification, 6/32 aneusomy), a finding associated with higher TIL densities (p = 0.045) and PD-L1 expression (p = 0.025). Patients with TILs ≥ 40% had better prognoses (Log-Rank p = 0.013), with TILs being favorable prognosticators in univariate analysis (Hazard ratio: 0.18, p = 0.024). A subset of SDCs with increased ERBB2 copy numbers have higher TILs and PD-L1 expression. TILs ≥ 40% are associated with better prognosis.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- B7-H1 Antigen/biosynthesis
- Biomarkers, Tumor/analysis
- Carcinoma, Ductal/genetics
- Carcinoma, Ductal/immunology
- Carcinoma, Ductal/pathology
- DNA Copy Number Variations
- Female
- Gene Amplification
- Genes, erbB-2
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Salivary Gland Neoplasms/genetics
- Salivary Gland Neoplasms/immunology
- Salivary Gland Neoplasms/pathology
Collapse
Affiliation(s)
| | | | - Sotiris Sotiriou
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Michael G. Keeney
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Daniel W. Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Michael Rivera
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | | | - Jean E. Lewis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Patricia T. Greipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - William R. Sukov
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | | | | | - Joaquin J. Garcia
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| |
Collapse
|
27
|
Evangelou Z, Papoudou-Bai A, Karpathiou G, Kourea H, Kamina S, Goussia A, Harissis H, Peschos D, Batistatou A. PD-L1 Expression and Tumor-infiltrating Lymphocytes in Breast Cancer: Clinicopathological Analysis in Women Younger than 40 Years Old. In Vivo 2020; 34:639-647. [PMID: 32111764 DOI: 10.21873/invivo.11818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/17/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIM To evaluate the association between programmed cell death ligand 1 (PD-L1) expression on both tumor cells (TC) and inflammatory cells (IC), tumor infiltrating lymphocytes (TILs), CD3+ and CD8+ lymphocytes and other clinicopathological parameters in primary infiltrative breast cancer (IBC) of young women, a population shown to have a worse prognosis. MATERIALS AND METHODS A retrospective study was performed collecting data from patients younger than 40 years old. Forty-five young women with IBC were included. Whole tissue sections were used to evaluate all parameters. RESULTS Twenty percent (20%) of cases showed PD-L1 expression by tumor cells (PDL1TC) and 44.4% showed PD-L1 expression by immune cells (PDL1IC). Furthermore, 28.88% revealed high stromal TILs. PDL1TC and PDL1IC expression were significantly associated with tumor diameter and expression of estrogen (ER) and progesterone (PR) receptors and Ki67. PDL1TC expression was also associated with grade. High TILs were associated with tumor diameter, ER and Ki67 expression. PDL1TC, PDL1IC expression and TILs were associated with the density of CD3+ and CD8+ lymphocytes. CONCLUSION Our results are similar to those of other age groups, as reported in the literature.
Collapse
Affiliation(s)
- Zoi Evangelou
- Department of Pathology, University Hospital of Ioannina, Ioannina, Greece
| | | | - Georgia Karpathiou
- Department of Pathology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Helen Kourea
- Department of Pathology, University Hospital of Patras, Patras, Greece
| | - Sevasti Kamina
- Department of Pathology, University Hospital of Ioannina, Ioannina, Greece
| | - Anna Goussia
- Department of Pathology, University Hospital of Ioannina, Ioannina, Greece
| | - Haralambos Harissis
- Department of Surgery, Breast Unit, University Hospital of Ioannina, Ioannina, Greece
| | - Dimitrios Peschos
- Department of Physiology, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Anna Batistatou
- Department of Pathology, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
28
|
Zimmer AS, Van Swearingen AED, Anders CK. HER2‐positive
breast cancer brain metastasis: A new and exciting landscape. Cancer Rep (Hoboken) 2020; 5:e1274. [PMID: 32881421 PMCID: PMC9124511 DOI: 10.1002/cnr2.1274] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
Background Brain metastases (BrM) incidence is 25% to 50% in women with advanced human epidermal growth factor receptor 2 (HER2)‐positive breast cancer. Radiation and surgery are currently the main local treatment approaches for central nervous system (CNS) metastases. Systemic anti‐HER2 therapy following a diagnosis of BrM improves outcomes. Previous preclinical data has helped elucidate HER2 brain trophism, the blood‐brain/blood‐tumor barrier(s), and the brain tumor microenvironment, all of which can lead to development of novel therapeutic options. Recent findings Several anti‐HER2 agents are currently available and reviewed here, some of which have recently shown promising effects in BrM patients, specifically. New strategies driven by and focusing on brain metastasis‐specific genomics, immunotherapy, and preventive strategies have shown promising results and are under development. Conclusions The field of HER2+ breast cancer, particularly for BrM, continues to evolve as new therapeutic strategies show promising results in recent clinical trials. Increasing inclusion of patients with BrM in clinical studies, and a focus on assessing their outcomes both intracranially and extracranially, is changing the landscape for patients with HER2+ CNS metastases by demonstrating the ability of newer agents to improve outcomes.
Collapse
Affiliation(s)
| | | | - Carey K. Anders
- Duke Center for Brain and Spine MetastasisDuke Cancer Institute Durham North Carolina USA
| |
Collapse
|
29
|
Choong GM, Cullen GD, O'Sullivan CC. Evolving standards of care and new challenges in the management of HER2-positive breast cancer. CA Cancer J Clin 2020; 70:355-374. [PMID: 32813307 DOI: 10.3322/caac.21634] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/29/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
The management of human epidermal growth factor receptor (HER2)-positive breast cancer (BC) has rapidly evolved over the last 20 years. Major advances have led to US Food and Drug Administration approval of 7 HER2-targeted therapies for the treatment of early-stage and/or advanced-stage disease. Although oncologic outcomes continue to improve, most patients with advanced HER2-positive BC ultimately die of their disease because of primary or acquired resistance to therapy, and patients with HER2-positive early BC who have residual invasive disease after preoperative systemic therapy are at a higher risk of distant recurrence and death. The concept of treatment de-escalation and escalation is increasingly important to optimally tailor therapy for patients with HER2-positive BC and is a major focus of the current review. Research efforts in this regard are discussed as well as updates regarding the evolving standard of care in the (neo)adjuvant and metastatic settings, including the use of novel combination therapies. The authors also briefly discuss ongoing challenges in the management of HER2-positive BC (eg, intrinsic vs acquired drug resistance, the identification of predictive biomarkers, the integration of imaging techniques to guide clinical practice), and the treatment of HER2-positive brain metastases. Research aimed at superseding these challenges will be imperative to ensure continued progress in the management of HER2-positive BC going forward.
Collapse
Affiliation(s)
- Grace M Choong
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Grace D Cullen
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
30
|
Clinicopathological correlation of PD-L1 and TET1 expression with tumor-infiltrating lymphocytes in non-small cell lung cancer. Pathol Res Pract 2020; 216:153188. [PMID: 32919305 DOI: 10.1016/j.prp.2020.153188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 11/22/2022]
Abstract
The immunohistochemical analysis of PD-L1 expression is still important in cancer immunotherapy. PD-L1 expression is affected by various tumor microenvironmental factors including tumor infiltrating lymphocytes (TILs) and DNA methylation biomarkers. Given the complex communication between tumor cells and immune cells, we analyzed the expression of PD-L1 and TET1 with TILs in human NSCLC and the correlation with various clinicopathological characteristics and patient prognosis. A total of 96 cases of NSCLC were enrolled in this study. Using tissue microarray, we performed immunohistochemical staining to analyze PD-L1 and TET1 expression. Image-Pro Plus was used as an automated imaging analysis software program to analyze the density of CD3+, CD4+ and CD8 + TILs. PD-L1 expression was positively correlated with the density of CD3+, CD4+ and CD8 + TILs (p = 0.038, p = 0.020, and p = 0.009, respectively); however, no significant relationship existed between TET1 expression and any TILs. The survival analysis revealed that a high PD-L1 expression was associated with favorable prognosis for OS (p = 0.049) and DFS (p = 0.029) in advanced-stage II-IV patients, but not in early stage I. Density of CD8+ TILs was an independent and favorable prognostic factor for DFS (p = 0.008) and OS (p = 0.002) in early-stage I patients. However, high TET-1 expression was associated with poor prognosis for OS (p = 0.029) in total NSCLC patients. These findings suggest the correlation and favorable prognostic impact of PD-L1 and TILs in NSCLC. In addition, DNA demethylase TET1 has oncogenic effects, showing association with poor prognosis.
Collapse
|
31
|
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: 91] [Impact Index Per Article: 18.2] [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
|
32
|
Wilkes JG, Czerniecki BJ, Costa RLB. Treatment from within: Ductal Carcinoma as an Opportunity to Harness the Immune System. CURRENT BREAST CANCER REPORTS 2020. [DOI: 10.1007/s12609-020-00356-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
33
|
Serum PD-1/PD-L1 Levels, Tumor Expression and PD-L1 Somatic Mutations in HER2-Positive and Triple Negative Normal-Like Feline Mammary Carcinoma Subtypes. Cancers (Basel) 2020; 12:cancers12061386. [PMID: 32481540 PMCID: PMC7352561 DOI: 10.3390/cancers12061386] [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: 04/16/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment has gained great relevance due to its ability to regulate distinct checkpoints mediators, orchestrating tumor progression. Serum programmed cell death protein-1 (PD-1) and programmed death ligand-1 (PD-L1) levels were compared with healthy controls and with serum cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and tumor necrosis factor-alpha (TNF-α) levels in order to understand the role of PD-1/PD-L1 axis in cats with mammary carcinoma. PD-1 and PD-L1 expression was evaluated in tumor-infiltrating lymphocytes (TILs) and cancer cells, as the presence of somatic mutations. Results showed that serum PD-1 and PD-L1 levels were significantly higher in cats with HER2-positive (p = 0.017; p = 0.032) and triple negative (TN) normal-like mammary carcinomas (p = 0.004; p = 0.015), showing a strong positive correlation between serum CTLA-4 and TNF-α levels. In tumors, PD-L1 expression in cancer cells was significantly higher in HER2-positive samples than in TN normal-like tumors (p = 0.010), as the percentage of PD-L1-positive TILs (p = 0.037). PD-L1 gene sequencing identified two heterozygous mutations in exon 4 (A245T; V252M) and one in exon 5 (T267S). In summary, results support the use of spontaneous feline mammary carcinoma as a model for human breast cancer and suggest that the development of monoclonal antibodies may be a therapeutic strategy.
Collapse
|
34
|
Zerdes I, Sifakis EG, Matikas A, Chrétien S, Tobin NP, Hartman J, Rassidakis GZ, Bergh J, Foukakis T. Programmed death-ligand 1 gene expression is a prognostic marker in early breast cancer and provides additional prognostic value to 21-gene and 70-gene signatures in estrogen receptor-positive disease. Mol Oncol 2020; 14:951-963. [PMID: 32115850 PMCID: PMC7191187 DOI: 10.1002/1878-0261.12654] [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: 12/09/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
Gene and protein expression of programmed death‐ligand 1 (PD‐L1) are prognostic in early breast cancer (BC), but their prognostic information is inconsistent at least in some biological subgroups. The validated prognostic gene signatures (GS) in BC are mainly based on proliferation and estrogen receptor (ER)‐related genes. Here, we aimed to explore the prognostic capacity of PD‐L1 expression at the protein vs mRNA levels and to investigate the prognostic information that PD‐L1 can potentially add to routinely used GS. Gene expression data were derived from two early BC cohorts (cohort 1: 562 patients; cohort 2: 1081 patients). Tissue microarrays from cohort 1 were immunohistochemically (IHC) stained for PD‐L1 using the SP263 clone. GS scores (21‐gene, 70‐gene) were calculated, and likelihood‐ratio (LR) tests and concordance indices were used to evaluate the additional prognostic information for each signature. The immune cell composition was also evaluated using the CIBERSORT in silico tool. PD‐L1 gene and protein expressions were independently associated with better prognosis. In ER+/HER2− patients, PD‐L1 gene expression provided significant additional prognostic information beyond that of both 21‐GS [LR‐Δχ2 = 15.289 and LR‐Δχ2 = 8.812, P < 0.01 for distant metastasis‐free interval (DMFI) in cohorts 1 and 2, respectively] and 70‐GS score alone (LR‐Δχ2 = 18.198 and LR‐Δχ2 = 8.467, P < 0.01 for DMFI in cohorts 1 and 2, respectively). PD‐L1 expression was correlated with IHC‐determined CD3+ cells (r = 0.41, P < 0.001) and with CD8+ (r = 0.62, P < 0.001) and CD4+ memory activated (r = 0.66, P < 0.001) but not with memory resting (r = −0.063, P = 0.14) or regulatory (r = −0.12, P < 0.01) T cells in silico. PD‐L1 gene expression represents a promising favorable prognostic marker and can provide additional prognostic value to 21‐ and 70‐gene scores in ER+/HER2− BC.
Collapse
Affiliation(s)
- Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Alexios Matikas
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Sebastian Chrétien
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Nicholas P Tobin
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - George Z Rassidakis
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Costa RLB, Czerniecki BJ. Clinical development of immunotherapies for HER2 + breast cancer: a review of HER2-directed monoclonal antibodies and beyond. NPJ Breast Cancer 2020; 6:10. [PMID: 32195333 PMCID: PMC7067811 DOI: 10.1038/s41523-020-0153-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer accounts for ~25% of breast cancer cases. Monoclonal antibodies (mAbs) against HER2 have led to unparalleled clinical benefit for a subset of patients with HER2+ breast cancer. In this narrative review, we summarize advances in the understanding of immune system interactions, examine clinical developments, and suggest rationales for future investigation of immunotherapies for HER2+ breast cancer. Complex interactions have been found between different branches of the immune system, HER2+ breast cancer, and targeted treatments (approved and under investigation). A new wave of immunotherapies, such as novel HER2-directed mAbs, antibody drug conjugates, vaccines, and adoptive T-cell therapies, are being studied in a broad population of patients with HER2-expressing tumors. The development of immunotherapies for HER2+ breast cancer represents an evolving field that should take into account interactions between different components of the immune system.
Collapse
Affiliation(s)
- Ricardo L B Costa
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Brian J Czerniecki
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| |
Collapse
|
36
|
Padmanabhan R, Kheraldine HS, Meskin N, Vranic S, Al Moustafa AE. Crosstalk between HER2 and PD-1/PD-L1 in Breast Cancer: From Clinical Applications to Mathematical Models. Cancers (Basel) 2020; 12:E636. [PMID: 32164163 PMCID: PMC7139939 DOI: 10.3390/cancers12030636] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is one of the major causes of mortality in women worldwide. The most aggressive breast cancer subtypes are human epidermal growth factor receptor-positive (HER2+) and triple-negative breast cancers. Therapies targeting HER2 receptors have significantly improved HER2+ breast cancer patient outcomes. However, several recent studies have pointed out the deficiency of existing treatment protocols in combatting disease relapse and improving response rates to treatment. Overriding the inherent actions of the immune system to detect and annihilate cancer via the immune checkpoint pathways is one of the important hallmarks of cancer. Thus, restoration of these pathways by various means of immunomodulation has shown beneficial effects in the management of various types of cancers, including breast. We herein review the recent progress in the management of HER2+ breast cancer via HER2-targeted therapies, and its association with the programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) axis. In order to link research in the areas of medicine and mathematics and point out specific opportunities for providing efficient theoretical analysis related to HER2+ breast cancer management, we also review mathematical models pertaining to the dynamics of HER2+ breast cancer and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Regina Padmanabhan
- Department of Electrical Engineering, Qatar University, 2713 Doha, Qatar;
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
| | - Hadeel Shafeeq Kheraldine
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
- College of Pharmacy, QU Health, Qatar University, 2713 Doha, Qatar
| | - Nader Meskin
- Department of Electrical Engineering, Qatar University, 2713 Doha, Qatar;
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
| | - Ala-Eddin Al Moustafa
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
| |
Collapse
|
37
|
Role of innate and adaptive immunity in the efficacy of anti-HER2 monoclonal antibodies for HER2-positive breast cancer. Crit Rev Oncol Hematol 2020; 149:102927. [PMID: 32172224 DOI: 10.1016/j.critrevonc.2020.102927] [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: 05/24/2019] [Revised: 02/06/2020] [Accepted: 03/02/2020] [Indexed: 01/09/2023] Open
Abstract
Anti-HER2 monoclonal antibodies (mAbs) such as trastuzumab are effective for all stages of HER2-positive breast cancer (BC). However, intrinsic or acquired resistance to these drugs may occur in a significant number of patients (pts) and, except for HER2 status, no validated predictive factors of response/resistance have been identified to date. This lack is in part due to the not yet fully elucidated mechanism of action of mAbs in vivo. Increasing evidence suggests a significant contribution of both innate and adaptive immunity to the antitumor effects of mAbs. The aim of this review was to describe the role of innate and adaptive immunity in the efficacy of anti-HER2 mAbs and to report known and novel strategies to be used for optimizing immune effects of anti-HER2 therapies for HER2-positive BC.
Collapse
|
38
|
Arab A, Yazdian-Robati R, Behravan J. HER2-Positive Breast Cancer Immunotherapy: A Focus on Vaccine Development. Arch Immunol Ther Exp (Warsz) 2020; 68:2. [PMID: 31915932 PMCID: PMC7223380 DOI: 10.1007/s00005-019-00566-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023]
Abstract
Clinical progress in the field of HER2-positive breast cancer therapy has been dramatically improved by understanding of the immune regulatory mechanisms of tumor microenvironment. Passive immunotherapy utilizing recombinant monoclonal antibodies (mAbs), particularly trastuzumab and pertuzumab has proved to be an effective strategy in HER2-positive breast cancer treatment. However, resistance to mAb therapy and relapse of disease are still considered important challenges in clinical practice. There are increasing reports on the induction of cellular and humoral immune responses in HER2-positive breast cancer patients. More recently, increasing efforts are focused on using HER2-derived peptide vaccines for active immunotherapy. Here, we discuss the development of various HER2-derived vaccines tested in animal models and human clinical trials. Different formulations and strategies to improve immunogenicity of the antigens in animal studies are also discussed. Furthermore, other immunotherapeutic approaches to HER2 breast cancer including, CTLA-4 inhibitors, immune checkpoint inhibitors, anti PD-1/PD-L1 antibodies are presented.
Collapse
Affiliation(s)
- Atefeh Arab
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Rezvan Yazdian-Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Javad Behravan
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,School of Pharmacy, University of Waterloo, Waterloo, ON, Canada. .,Theraphage Inc., Kitchener, ON, Canada.
| |
Collapse
|
39
|
Verdura S, Cuyàs E, Cortada E, Brunet J, Lopez-Bonet E, Martin-Castillo B, Bosch-Barrera J, Encinar JA, Menendez JA. Resveratrol targets PD-L1 glycosylation and dimerization to enhance antitumor T-cell immunity. Aging (Albany NY) 2020; 12:8-34. [PMID: 31901900 PMCID: PMC6977679 DOI: 10.18632/aging.102646] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/23/2019] [Indexed: 12/24/2022]
Abstract
New strategies to block the immune evasion activity of programmed death ligand-1 (PD-L1) are urgently needed. When exploring the PD-L1-targeted effects of mechanistically diverse metabolism-targeting drugs, exposure to the dietary polyphenol resveratrol (RSV) revealed its differential capacity to generate a distinct PD-L1 electrophoretic migration pattern. Using biochemical assays, computer-aided docking/molecular dynamics simulations, and fluorescence microscopy, we found that RSV can operate as a direct inhibitor of glyco-PD-L1-processing enzymes (α-glucosidase/α-mannosidase) that modulate N-linked glycan decoration of PD-L1, thereby promoting the endoplasmic reticulum retention of a mannose-rich, abnormally glycosylated form of PD-L1. RSV was also predicted to interact with the inner surface of PD-L1 involved in the interaction with PD-1, almost perfectly occupying the target space of the small compound BMS-202 that binds to and induces dimerization of PD-L1. The ability of RSV to directly target PD-L1 interferes with its stability and trafficking, ultimately impeding its targeting to the cancer cell plasma membrane. Impedance-based real-time cell analysis (xCELLigence) showed that cytotoxic T-lymphocyte activity was notably exacerbated when cancer cells were previously exposed to RSV. This unforeseen immunomodulating mechanism of RSV might illuminate new approaches to restore T-cell function by targeting the PD-1/PD-L1 immunologic checkpoint with natural polyphenols.
Collapse
Affiliation(s)
- Sara Verdura
- Program against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Elisabet Cuyàs
- Program against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Eric Cortada
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain.,Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, Girona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Joan Brunet
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain.,Department of Medical Sciences, Medical School University of Girona, Girona, Spain.,Hereditary Cancer Programme, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain.,Hereditary Cancer Programme, Catalan Institute of Oncology (ICO), Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Eugeni Lopez-Bonet
- Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona, Spain
| | | | - Joaquim Bosch-Barrera
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain.,Medical Oncology, Catalan Institute of Oncology, Girona, Spain.,Department of Medical Sciences, Medical School University of Girona, Girona, Spain
| | - José Antonio Encinar
- Institute of Research, Development and Innovation in Biotechnology of Elche (IDiBE) and Molecular and Cell Biology Institute (IBMC), Miguel Hernández University (UMH), Elche, Spain
| | - Javier A Menendez
- Program against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| |
Collapse
|
40
|
Vranic S, Cyprian FS, Gatalica Z, Palazzo J. PD-L1 status in breast cancer: Current view and perspectives. Semin Cancer Biol 2019; 72:146-154. [PMID: 31883913 DOI: 10.1016/j.semcancer.2019.12.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022]
Abstract
Breast cancer was traditionally not considered a particularly immunogenic tumor. However, recent developments have shown that some aggressive triple-negative breast cancers are immunogenic, exhibit a resistance to chemotherapy and have a poor prognosis. These cancers have been shown to express molecules identified as targets for immunotherapy. Despite the advances, the challenges are many, and include identifying the patients that may benefit from immunotherapy. The best methods to analyze these samples and to evaluate immunogenicity are also major challenges. Therefore, the most accurate and reliable assessment of immune cells as potential targets is one of the most important aims in the current research in breast immunotherapy. In the present review, we briefly discuss the mechanisms of the regulation of checkpoint inhibitors (PD-1/PD-L1) in breast cancer and explore the predictive aspects in the PD-L1 testing.
Collapse
Affiliation(s)
- Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | | | | |
Collapse
|
41
|
Al-Dughaishi M, Shalaby A, Al-Ribkhi K, Boudaka A, Boulassel MR, Saleh J. The Value of Programmed Death Ligand 1 Expression in Cancer Patients Treated with Neoadjuvant Chemotherapy. Sultan Qaboos Univ Med J 2019; 19:e277-e283. [PMID: 31897310 PMCID: PMC6930029 DOI: 10.18295/squmj.2019.19.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/01/2019] [Accepted: 09/30/2019] [Indexed: 01/26/2023] Open
Abstract
Programmed death ligand 1 (PD-L1) is an inhibitory molecule expressed by cancer cells to supress T-cell activity and escape anti-tumour immunity. The role of PD-L1 in cancer has been studied extensively as it is considered an important immune checkpoint against immune over-activation through its interaction with Programmed death receptor 1 (PD-1) expressed on activated lymphocytes. PD-L1 expression was found to be enhanced by chemotherapy through different proliferation pathways. However, the predictive and prognostic value for PD-L1 expression in cancer patients treated with neoadjuvant chemotherapy (NAC) is not yet established. This review focused on the potential effects of chemotherapy on PD-L1 expression and the role of PD-L1 as a prognostic and predictive marker in NAC-treated cancer patients. In addition, the potential use of this marker in clinical practice is discussed.
Collapse
Affiliation(s)
| | - Asem Shalaby
- Department of Pathology, Sultan Qaboos University, Muscat, Oman
| | - Khawla Al-Ribkhi
- Department of Biochemistry, Sultan Qaboos University, Muscat, Oman
| | - Ammar Boudaka
- Department of Physiology, Sultan Qaboos University, Muscat, Oman
| | | | - Jumana Saleh
- Department of Biochemistry, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
42
|
Miglietta F, Griguolo G, Guarneri V, Dieci MV. Programmed Cell Death Ligand 1 in Breast Cancer: Technical Aspects, Prognostic Implications, and Predictive Value. Oncologist 2019; 24:e1055-e1069. [PMID: 31444294 PMCID: PMC6853089 DOI: 10.1634/theoncologist.2019-0197] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/15/2019] [Indexed: 12/22/2022] Open
Abstract
In the light of recent advances in the immunotherapy field for breast cancer (BC) treatment, especially in the triple-negative subtype, the identification of reliable biomarkers capable of improving patient selection is paramount, because only a portion of patients seem to derive benefit from this appealing treatment strategy. In this context, the role of programmed cell death ligand 1 (PD-L1) as a potential prognostic and/or predictive biomarker has been intensively explored, with controversial results. The aim of the present review is to collect available evidence on the biological relevance and clinical utility of PD-L1 expression in BC, with particular emphasis on technical aspects, prognostic implications, and predictive value of this promising biomarker. IMPLICATIONS FOR PRACTICE: In the light of the promising results coming from trials of immune checkpoint inhibitors for breast cancer treatment, the potential predictive and/or prognostic role of programmed cell death ligand 1 (PD-L1) in breast cancer has gained increasing interest. This review provides clinicians with an overview of the available clinical evidence regarding PD-L1 as a biomarker in breast cancer, focusing on both data with a possible direct impact on clinic and methodological pitfalls that need to be addressed in order to optimize PD-L1 implementation as a clinically useful tool for breast cancer management.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| |
Collapse
|
43
|
Viale G, Morganti S, Ferraro E, Zagami P, Marra A, Curigliano G. What therapies are on the horizon for HER2 positive breast cancer? Expert Rev Anticancer Ther 2019; 19:811-822. [PMID: 31448640 DOI: 10.1080/14737140.2019.1660164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Despite dramatic improvements in survival achieved with currently available anti-HER2 agents, HER2-positive metastatic breast cancer remains an almost invariably deadly disease, with primary or acquired resistance to HER2-directed agents developing during treatment. Many efforts are focused on identifying new agents that may more effectively inhibit HER2 signaling and on possible combination strategies. Areas covered: This review summarizes the landscape of drugs under development for HER2-positive metastatic breast cancer, as antibody-drug conjugates, monoclonal anti-HER2 antibodies, bispecific antibodies, or novel tyrosine kinase inhibitors. Moreover, available data for possible combination of anti-HER2 drugs and different agents, as immunotherapy, PI3K/mTOR inhibitors, CDK4/6 inhibitors currently under evaluation are reviewed. These strategies may overcome mechanisms of resistance and further improve patient outcomes. Expert opinion: Identification of valuable predictive biomarkers is needed to better inform choice of treatment sequence for the individual patient and limit the financial toxicity of these agents.
Collapse
Affiliation(s)
- Giulia Viale
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Emanuela Ferraro
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Paola Zagami
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Antonio Marra
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| |
Collapse
|
44
|
Mittal D, Vijayan D, Neijssen J, Kreijtz J, Habraken MMJM, Van Eenennaam H, Van Elsas A, Smyth MJ. Blockade of ErbB2 and PD-L1 using a bispecific antibody to improve targeted anti-ErbB2 therapy. Oncoimmunology 2019; 8:e1648171. [PMID: 31646095 DOI: 10.1080/2162402x.2019.1648171] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/14/2019] [Accepted: 07/23/2019] [Indexed: 01/04/2023] Open
Abstract
A significant proportion of human epidermal growth factor receptor 2 (Her2/ErbB2)-positive metastatic breast cancer patients are refractory to Her2-targeted trastuzumab-like therapy. Some of this resistance has been attributed to the upregulation of immune checkpoints such as programmed cell death-1 (PD-1) and its ligand, PD-L1 in Her2-positive breast cancer patients. Therefore, therapies targeting both the PD-1/PD-L1 interaction and oncogenic Her2 signaling are of significant clinical interest. Here, we constructed a mouse bispecific antibody targeting PD-L1 and rat Her2 (referred to as BsPD-L1xrErbB2) aiming to redirect the anti-PD-L1 response toward Her2-expressing tumor cells. BsPD-L1xrErbB2 demonstrated additive binding to interferon (IFN)-γ treated Her2+ TUBO tumor cells, but it did not affect the proliferation of tumor cells in-vitro. BsPD-L1xrErbB2 also blocked the PD-1/PD-L1 interaction. This bispecific antibody was constructed with a mouse IgG2a Fc backbone and interacted with Fcγ receptors and resulted in complement deposition (C3). ADCC and complement action could be potential mechanisms of action of this molecule. BsPD-L1xrErbB2 successfully reduced TUBO tumor growth and increased tumor rejection rate compared to the monovalent anti-PD-L1, monovalent anti-ErbB2 or the combination of anti-PD-L1 and anti-ErbB2 monotherapies. The enhanced anti-tumor effect of BsPD-L1xrErbB2 was dependent on CD8+ T lymphocytes and IFN-γ, as depletion of CD8+ T lymphocytes and neutralization of IFN-γ completely abolished the antitumor activity of the bispecific antibody. Consistently, BsPD-L1xrErbB2 treatment also increased the frequency of intratumor CD8+ T lymphocytes. Taken together, our data support a bispecific antibody approach to enhance the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade in Her2-positive metastatic breast cancers.
Collapse
Affiliation(s)
- Deepak Mittal
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
| | - Dipti Vijayan
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
| | | | | | | | | | | | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
| |
Collapse
|
45
|
Criscitiello C, Curigliano G. Treatment in real-life patients with HER2-positive metastatic breast cancer: What we learn from the KAMILLA trial? Eur J Cancer 2019; 117:1-4. [DOI: 10.1016/j.ejca.2019.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
|
46
|
Huang W, Ran R, Shao B, Li H. Prognostic and clinicopathological value of PD-L1 expression in primary breast cancer: a meta-analysis. Breast Cancer Res Treat 2019; 178:17-33. [PMID: 31359214 DOI: 10.1007/s10549-019-05371-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/19/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE To evaluate the association between PD-L1 expression (PD-L1+) and clinicopathological characteristics and effect on prognosis in primary breast cancer (PBC). METHODS A systematic search of the PubMed, Web of Science, and Embase databases was conducted in November 2018. Studies detecting PD-L1 using immunohistochemistry, and concerning its prognostic or clinicopathological significance in PBC were included. The HR with 95% CI for survival, and the events for clinicopathological features were pooled. RESULTS Forty-seven studies were included, with a total of 14,367 PBC patients. PD-L1+ tumor cells (TCs) were associated with ductal carcinomas, large tumor size, histological Grade 3 tumors, high Ki-67, ER and PR negative, and triple-negative breast cancer; and also, related to high tumor-infiltrating lymphocytes (TILs) and PD-1 expression. PD-L1+ TCs were significantly associated with shorter disease-free survival (DFS, HR = 1.43, 95% CI 1.21-1.70, P < 0.0001) and overall survival (OS, HR = 1.58, 95% CI 1.14-2.20, P = 0.006). And the HRs of PD-L1+ TCs on DFS and OS were higher (1.48 and 1.70, respectively) and homogeneous when using whole tissue section, compared with tumor microarrays. However, PD-L1+ TILs related to better DFS (HR = 0.45, 95% CI 0.28-0.73, P = 0.001) and OS (HR = 0.41, 95% CI 0.27-0.63, P < 0.0001). CONCLUSION PD-L1 expression on TCs associates with high-risk clinicopathological parameters and poor prognosis in PBC patients, while PD-L1+ TILs may relate to a better survival. Comprehensive assessment of TCs and TILs is required when evaluating the clinical relevance of PD-L1 expression in future studies.
Collapse
Affiliation(s)
- Wenfa Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Ran Ran
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Bin Shao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
47
|
Lee DW, Ryu HS, Jin MS, Lee KH, Suh KJ, Youk J, Kim JY, Min A, Lee HB, Moon HG, Kim TY, Han SW, Oh DY, Han W, Park IA, Noh DY, Im SA. Immune recurrence score using 7 immunoregulatory protein expressions can predict recurrence in stage I-III breast cancer patients. Br J Cancer 2019; 121:230-236. [PMID: 31292534 PMCID: PMC6738097 DOI: 10.1038/s41416-019-0511-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Immune cells in the tumour microenvironment play an essential role in tumorigenesis. This study aimed to evaluate the immunoregulatory protein expression of breast cancer and reveal their prognostic role. METHODS Expression of 10 immune markers (PD-1/PD-L1/PD-L2/IDO/TIM-3/OX40/OX40L/B7-H2/ B7-H3/B7-H4) with known/possible clinical relevance was identified in stromal tumour-infiltrating lymphocytes or tumour tissue of stage I-III breast cancer patients. RESULTS A total of 392 patients, including 271(69.1%) luminal A, 36(9.2%) luminal B, 32(8.2%) HER2-positive and 53(13.5%) triple negative disease, were included. Expression of PD-1 and PD-L1 was higher in HER2-positive and triple negative disease. By contrast, expression of TIM-3, OX40 and OX40L were higher in luminal disease. We devised an immune recurrence score (IRS) using seven markers with prognostic value (B7-H2/B7-H3/B7-H4/OX40/OX40L/PD-L1/PD-L2). Patients were classified as high-risk (7.9%), intermediate-risk (67.6%), or low-risk (24.5%). In the multivariate analysis, IRS low-risk (adjusted HR 0.14, p = 0.001) and intermediate-risk (adjusted HR 0.32, p = 0.002) had significantly lower risk of recurrence compared with high-risk. The prognostic role of IRS was maintained in both luminal A and non-luminal A patients. CONCLUSIONS This study identified immunoregulatory protein expression of breast cancer patients using 10 immune markers. In addition, we devised an IRS which may predict recurrence in stage I-III breast cancer patients.
Collapse
Affiliation(s)
- Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Min-Sun Jin
- Department of Pathology, Bucheon St. Mary's Hospital, Bucheon, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.
| | - Koung Jin Suh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jeonghwan Youk
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jung Youn Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Han-Byoel Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyeong-Gon Moon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Wonshik Han
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Dong-Young Noh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
48
|
Matikas A, Zerdes I, Lövrot J, Richard F, Sotiriou C, Bergh J, Valachis A, Foukakis T. Prognostic Implications of PD-L1 Expression in Breast Cancer: Systematic Review and Meta-analysis of Immunohistochemistry and Pooled Analysis of Transcriptomic Data. Clin Cancer Res 2019; 25:5717-5726. [PMID: 31227501 DOI: 10.1158/1078-0432.ccr-19-1131] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/15/2019] [Accepted: 06/19/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Conflicting data have been reported on the prognostic value of PD-L1 protein and gene expression in breast cancer.Experimental Design: Medline, Embase, Cochrane Library, and Web of Science Core Collection were searched, and data were extracted independently by two researchers. Outcomes included pooled PD-L1 protein positivity in tumor cells, immune cells, or both, per subtype and per antibody used, and its prognostic value for disease-free and overall survival. A pooled gene expression analysis of 39 publicly available transcriptomic datasets was also performed. RESULTS Of the initial 4,184 entries, 38 retrospective studies fulfilled the predefined inclusion criteria. The overall pooled PD-L1 protein positivity rate was 24% (95% CI, 15%-33%) in tumor cells and 33% (95% CI, 14%- 56%) in immune cells. PD-L1 protein expression in tumor cells was prognostic for shorter overall survival (HR, 1.63; 95% CI, 1.07-2.46; P = 0.02); there was significant heterogeneity (I2 = 80%, P heterogeneity < 0.001). In addition, higher PD-L1 gene expression predicted better survival in multivariate analysis in the entire population (HR, 0.82; 95% CI, 0.74-0.90; P < 0.001 for OS) and in basal-like tumors (HR, 0.64; 95% CI, 0.52-0.80; P < 0.001 for OS; P interaction 0.005). CONCLUSIONS The largest to our knowledge meta-analysis on the subject informs on PD-L1 protein positivity rates and its prognostic value in breast cancer. Standardization is needed prior to routine implementation. PD-L1 gene expression is a promising prognostic factor, especially in basal-like breast cancer. Discrepant prognostic information might be related to PD-L1 gene expression in the stroma.
Collapse
Affiliation(s)
- Alexios Matikas
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden. .,Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Zerdes
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - John Lövrot
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Jonas Bergh
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Theodoros Foukakis
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Barroso-Sousa R, Barry WT, Guo H, Dillon D, Tan YB, Fuhrman K, Osmani W, Getz A, Baltay M, Dang C, Yardley D, Moy B, Marcom PK, Mittendorf EA, Krop IE, Winer EP, Tolaney SM. The immune profile of small HER2-positive breast cancers: a secondary analysis from the APT trial. Ann Oncol 2019; 30:575-581. [PMID: 30753274 PMCID: PMC8033534 DOI: 10.1093/annonc/mdz047] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Previous data suggest that the immune microenvironment plays a critical role in human epidermal growth factor receptor 2 (HER2) -positive breast cancer; however, there is little known about the immune profiles of small HER2-positive tumors. In this study, we aimed to characterize the immune microenvironment of small HER2-positive breast cancers included in the Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer (APT) trial and to correlate the immune markers with pathological and molecular tumor characteristics. PATIENTS AND METHODS The APT trial was a multicenter, single-arm, phase II study of paclitaxel and trastuzumab in patients with node-negative HER2-positive breast cancer. The study included 406 patients with HER2-positive, node-negative breast cancer, measuring up to 3 cm. Exploratory analysis of tumor infiltrating lymphocytes (TIL), programmed death-ligand 1 (PD-L1) expression (by immunohistochemistry), and immune gene signatures using data generated by nCounter PanCancer Pathways Panel (NanoString Technologies, Seattle, WA), and their association with pathological and molecular characteristics was carried out. RESULTS Of the 406 patients, 328 (81%) had at least one immune assay carried out: 284 cases were evaluated for TIL, 266 for PD-L1, and 213 for immune gene signatures. High TIL (≥60%) were seen with greater frequency in hormone-receptor (HR) negative, histological grades 2 and 3, as well in HER2-enriched and basal-like tumors. Lower stromal PD-L1 (≤1%) expression was seen with greater frequency in HR-positive, histological grade 1, and in luminal tumors. Both TIL and stromal PD-L1 were positively correlated with 10 immune cell signatures, including Th1 and B cell signatures. Luminal B tumors were negatively correlated with those signatures. Significant correlation was seen among these immune markers; however, the magnitude of correlation did not indicate a monotonic relationship between them. CONCLUSION Immune profiles of small HER2-positive breast cancers differ according to HR status, histological grade, and molecular subtype. Further work is needed to explore the implication of these findings on disease outcome. CLINICAL TRIAL REGISTRATION clinicaltrials.gov identifier: NCT00542451.
Collapse
Affiliation(s)
| | - W T Barry
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston
| | - H Guo
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston
| | - D Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - Y B Tan
- Department of Pathology, Brigham and Women's Hospital, Boston
| | | | | | - A Getz
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - M Baltay
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - C Dang
- Breast Cancer Medicine Service, Department of Medicine, Solid Tumor Division, Memorial Sloan Kettering Cancer Center, New York; Department of Medicine, Weill Cornell Medical Center, New York
| | | | - B Moy
- Department of Hematology-Oncology, Massachusetts General Hospital, Boston
| | - P K Marcom
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Durham
| | - E A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston; Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| | | | | | | |
Collapse
|
50
|
Pernas S, Tolaney SM. HER2-positive breast cancer: new therapeutic frontiers and overcoming resistance. Ther Adv Med Oncol 2019; 11:1758835919833519. [PMID: 30911337 PMCID: PMC6425535 DOI: 10.1177/1758835919833519] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/23/2019] [Indexed: 12/29/2022] Open
Abstract
The introduction of anti-HER2 therapies to the treatment of patients with HER2-positive breast cancer has led to dramatic improvements in survival in both early and advanced settings. Despite this breakthrough, nearly all patients with metastatic HER2-positive breast cancer eventually progress on anti-HER2 therapy due to de novo or acquired resistance. A better understanding not only of the underlying mechanisms of HER2 therapy resistance but of tumor heterogeneity as well as the host and tumor microenvironment is essential for the development of new strategies to further improve patient outcomes. One strategy has focused on inhibiting the HER2 signaling pathway more effectively with dual-blockade approaches and developing improved anti-HER2 therapies like antibody-drug conjugates, new anti-HER2 antibodies, bispecific antibodies, or novel tyrosine kinase inhibitors that might replace or be used in addition to some of the current anti-HER2 treatments. Combinations of anti-HER2 therapy with other agents like immune checkpoint inhibitors, CDK4/6 inhibitors, and PI3K/AKT/mTOR inhibitors are also being extensively evaluated in clinical trials. These add-on strategies of combining optimized targeted therapies could potentially improve outcomes for patients with HER2-positive breast cancer but may also allow de-escalation of treatment in some patients, potentially sparing some from unnecessary treatments, and their related toxicities and costs.
Collapse
Affiliation(s)
- Sonia Pernas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medical Oncology-Breast Cancer Unit, Institut Catala d’Oncologia (ICO)-H.U. Bellvitge-IDIBELL, Barcelona, Spain
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|