1
|
Izadi A, Naimi A, Amjadi E, Beheshtiparvar D, Soltan M. The Prevalence of PD-L1 Expression in Triple-Negative Breast Cancer Patients and Its Correlation with Survival Rates and Other Prognostic Factors: A Survival Analysis. Adv Biomed Res 2024; 13:86. [PMID: 39512412 PMCID: PMC11542687 DOI: 10.4103/abr.abr_2_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 11/15/2024] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a leading cause of cancer-related mortality among women, with a poor prognosis. The programmed cell death 1 (PD-1) pathway has emerged as a potential immunotherapy target. This study aimed to assess PD-L1 expression in TNBC patients and its relationship with prognostic variables. Materials and Methods This cross-sectional study included 107 TNBC patients recruited between 2016 and 2020. Patient age, tumor grade, and Ki67 expression were obtained from pathology reports. Immunohistochemistry was utilized to determine PD-L1 status, and 2-year survival data were collected through telephone follow-up. Results PD-L1 expression frequency in TNBC patients was 76.6%. Grade 3 was the most common cancer grade, significantly more prevalent in the PD-L1 positive group (P = 0.01). High Ki67 expression (≥14%) was observed in 89% of patients, significantly higher in the PD-L1 positive group (P = 0.003). The 2-year survival rates for the PD-L1 positive and negative groups were 84.1% and 92%, respectively, with no significant difference between the groups (P = 0.512). Conclusion This study investigated PD-L1 expression prevalence in TNBC patients and its correlation with prognostic variables. PD-L1 expression was associated with higher tumor grade and elevated Ki67 expression, indicating a potential role in tumor aggressiveness. However, despite these associations, PD-L1 expression did not significantly impact the 2-year survival rate in TNBC patients. These results emphasize the complexity of the immune microenvironment in TNBC and the necessity for further research to elucidate the precise role of PD-L1 in disease progression and patient outcomes.
Collapse
Affiliation(s)
- Arefeh Izadi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azar Naimi
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Amjadi
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Maryam Soltan
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
2
|
Huang X, Anderson SA, Siegal GP, Wei S, Liu S, Yang J, Roisin P, Pickens JT, Huo L, Sahin AA, Granada CP, Chen S. Comparison of PD-L1 (22C3) Expression in Paired Primary and Metastatic Breast Carcinoma. Clin Breast Cancer 2024; 24:e310-e318. [PMID: 38492995 DOI: 10.1016/j.clbc.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/18/2024] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
INTRODUCTION PD-L1 immunohistochemistry (IHC) is being used as a predictive marker of the benefit derived from immunotherapy in several cancer types, including breast cancer. However, the insight gleaned of the prognostic and predictive value of PD-L1 status and its correlation with molecular characteristics during breast cancer progression remains limited. METHODS We performed an PD-L1 (22C3) assay in pre-treatment primary and metastatic tumor sections from 33 patients with breast carcinoma, matched for post neoadjuvant chemotherapy (p-NACT). PD-L1 expression was evaluated using 3 scoring methods: immune cell (IC) and tumor cell (TC) with a 1% as the cutoff value, and combined positive scores (CPS) with a 1 as the cutoff value. Twenty-two samples from 11 patients had successful fluorescence in situ hybridization (FISH)-based molecular data available for analysis. RESULTS In the 33 pre-treatment primary tumors, PD-L1 IC, TC, and CPS showed positive correlation with stromal tumor infiltrate lymphocytes (sTIL), histological grade 3, and triple negative breast carcinoma (TNBC). In the matched metastatic tumors, only PD-L1 IC showed a positive correlation with sTIL. The primary tumors showed a higher PD-L1 expression than the matched metastatic tumors by IC and CPS. Negative to positive conversion by CPS was identified in the metastatic tumors from lung, pleura and liver. p-NACT tumors also showed a trend of lower PD-L1 expression compared to the pre-treatment tumors. Six patients had matched samples for molecular and PD-L1 comparison, and none of them showed consistent gene alterations or PD-L1 expression among the primary, p-NACT and metastatic tumors. CONCLUSION Our study showed a decrease in PD-L1 expression and disconnected molecular features during breast cancer progression. Repeating PD-L1 IHC testing could be considered in some specific metastatic sites if primary tumors were negative. Further studies are needed to identify other predictive factors for immune checkpoint inhibitor (ICI) therapy in patients with breast carcinoma.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL.
| | - Sarah A Anderson
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL
| | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL
| | - Shi Wei
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Shanrun Liu
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL
| | - Jingyun Yang
- Department of Neurological Sciences, RUSH University, Chicago, IL
| | | | - J Taylor Pickens
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL
| | - Lei Huo
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aysegul A Sahin
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carlos Prieto Granada
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Shuojun Chen
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
3
|
Han EK, Woo JW, Suh KJ, Kim SH, Kim JH, Park SY. PD-L1 (SP142) Expression in Primary and Recurrent/Metastatic Triple-Negative Breast Cancers and Its Clinicopathological Significance. Cancer Res Treat 2024; 56:557-566. [PMID: 38097920 PMCID: PMC11016636 DOI: 10.4143/crt.2023.1025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/11/2023] [Indexed: 04/13/2024] Open
Abstract
PURPOSE The programmed death-ligand 1 (PD-L1) SP142 assay identifies patients with triple-negative breast cancer (TNBC) who are most likely to respond to the anti-PD-L1 agent atezolizumab. We aimed to compare PD-L1 (SP142) expression between primary and recurrent/metastatic TNBCs and elucidate the clinicopathological features associated with its expression. MATERIALS AND METHODS Primary and recurrent/metastatic TNBCs tested with PD-L1 (SP142) were collected, and clinicopathological information of these cases was obtained through a review of slides and medical records. RESULTS PD-L1 (SP142) positivity was observed in 50.9% (144/283) of primary tumors and 37.8% (31/82) of recurrent/metastatic TNBCs with a significant difference. Recurrent or metastatic sites were associated with PD-L1 positivity, with high PD-L1 positivity in the lung, breast, and soft tissues, and low positivity in the bone, skin, liver, and brain. When comparing PD-L1 expression between primary and matched recurrent/metastatic TNBCs using 55 paired samples, 20 cases (36.4%) showed discordance; 10 cases revealed positive conversion, and another 10 cases revealed negative conversion during metastatic progression. In primary TNBCs, PD-L1 expression was associated with a higher histologic grade, lower T category, pushing border, and higher tumor-infiltrating lymphocyte infiltration. In survival analyses, PD-L1 positivity, especially high positivity, was found to be associated with favorable prognosis of patients. CONCLUSION PD-L1 (SP142) expression was lower in recurrent/metastatic TNBCs, and substantial cases showed discordance in its expression between primary and recurrent/metastatic sites, suggesting that multiple sites may need to be tested for PD-L1 (SP142) when considering atezolizumab therapy. PD-L1 (SP142)-positive TNBCs seems to be associated with favorable clinical outcomes.
Collapse
Affiliation(s)
- Eun Kyung Han
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Ji Won Woo
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Koung Jin Suh
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Se Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jee Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
4
|
Cerbelli B, Cirillo A, Pomati G, Pernazza A, Ascione A, Pisegna S, Pisano A, Leopizzi M, Pignataro MG, Costarelli L, Mulè A, Vecchione A, Catalano P, Coppola L, Perrone G, Perracchio L, Anemona L, Mastracchio A, Nardi S, Reitano R, Massari A, Grillo LR, Liberati F, Della Rocca C, Marchetti P, Botticelli A, D'Amati G. PD-L1 testing in metastatic triple negative breast cancer: Results of an Italian survey. TUMORI JOURNAL 2024; 110:44-48. [PMID: 37726962 DOI: 10.1177/03008916231196781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
BACKGROUND Immunotherapy has revolutionized the approach to metastatic triple-negative breast cancers. Atezolizumab was approved for patients with metastatic triple-negative breast cancers whose tumors express PD-L1, determined by SP 142 assay. To assess the availability and practice of SP142 test we administered a survey to all the 15 pathology departments of the Lazio Region during a six-month period. METHODS The survey comprised 12 questions regarding the availability of SP142 in the pathology departments, the percentage of positive tests, the difficulties of pathologists in cases close to cut-off value and the tested samples. RESULTS The SP142 assay was available in only eight centers. In case of positive result, most centers (5/8, 62.5%) reported values of PD-L1 expression ranging from > 1 to ⩽ 5%, with values close to the cut-off point (⩾ 1% or < 1%) being the greatest challenge.Most of the centers (6/8, 75%) tested material from both their own and other hospitals. In most centers, the evaluations were performed either on primary tumors or metastasis, in particular lymph nodes (5/8, 62.5%), followed by lung (3/8, 37.5%) and liver (1/8, 12.5%) metastasis. CONCLUSION Our results raise some important issues concerning the evaluation of PD-L1 in the "real-life" setting, providing strategies for its implementation.
Collapse
Affiliation(s)
- Bruna Cerbelli
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University of Roma, Lazio, Italy
| | - Alessio Cirillo
- Department of Experimental Medicine, Sapienza University of Rome, Lazio, Italy
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| | - Giulia Pomati
- Department of Molecular Medicine, Sapienza University of Rome, Lazio, Italy
| | - Angelina Pernazza
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University of Roma, Lazio, Italy
| | - Andrea Ascione
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| | - Simona Pisegna
- Department of Experimental Medicine, Sapienza University of Rome, Lazio, Italy
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| | - Annalinda Pisano
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University of Roma, Lazio, Italy
| | - Maria Gemma Pignataro
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| | | | - Antonino Mulè
- Breast Unit Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, Pathology Unit, Sant'Andrea Hospital, Sapienza University of Rome, Lazio, Italy
| | - Piera Catalano
- Department of Pathology, San Giovanni Calibita Fatebenefratelli Hospital, Rome, Lazio, Italy
| | - Luigi Coppola
- Unit of Anatomy, Pathological Histology and Diagnostic Cytology, Department of Diagnostic and Pharma-Ceutical Services, Sandro Pertini Hospital, Rome, Italy
| | - Giuseppe Perrone
- Campus Bio-Medico University Hospital Foundation of Rome, Rome, Lazio, Italy
| | - Letizia Perracchio
- Pathology Department, IRCCS Regina Elena National Cancer Institute, Rome, Emilia-Romagna, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, Pathology Unit, Tor Vergata University, Rome, Italy
| | | | - Stefano Nardi
- Department of Pathology, S. Maria Goretti Hospital, Latina, Italy
| | - Renato Reitano
- Department of Pathology, Spaziani Hospital, Frosinone, Italy
| | - Annalisa Massari
- Department of Pathology, Belcolle Hospital, Rome, Viterbo, Italy
| | | | - Fabrizio Liberati
- Department of Anatomic Pathology and Histology, San Camillo De Lellis Hospital, Rieti, Italy
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University of Roma, Lazio, Italy
| | | | - Andrea Botticelli
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| | - Giulia D'Amati
- Department of Radiological, Oncological and Anatomo-pathological Science, Sapienza University of Roma, Lazio, Italy
| |
Collapse
|
5
|
Miyakoshi J, Yazaki S, Shimoi T, Onishi M, Saito A, Kita S, Yamamoto K, Kojima Y, Sumiyoshi-Okuma H, Nishikawa T, Sudo K, Noguchi E, Murata T, Shiino S, Takayama S, Suto A, Fujiwara Y, Yoshida M, Yonemori K. Discordance in PD-L1 expression using 22C3 and SP142 assays between primary and metastatic triple-negative breast cancer. Virchows Arch 2023; 483:855-863. [PMID: 37668667 DOI: 10.1007/s00428-023-03634-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023]
Abstract
AIMS SP142 and 22C3 assays are approved companion diagnostic assays for anti-PD-1/PD-L1 therapy selection in metastatic triple-negative breast cancer (TNBC). The discordance in PD-L1 status between primary and metastatic tumors in the same patient has been poorly characterized. Here, we examined the concordance of PD-L1 status between the two assays and between primary tumors and metastases for each assay. METHODS We retrospectively evaluated tumor samples from 160 patients with TNBC, including 45 patients with paired primary and metastatic tumors. PD-L1 status was assessed using SP142 and 22C3 assays, to determine the immune cell (IC) score, tumor cell (TC) score (SP142 and 22C3), and combined proportion score (CPS: 22C3). RESULTS The concordance of PD-L1 positivity at diagnostic cutoffs for SP142 (IC ≥ 1) and 22C3 (CPS ≥ 10) was substantial (κ = 0.80) in primary tumors and moderate (κ = 0.60) in metastatic tumors. In comparison, between primary and metastatic tumors, the concordance with 22C3 was moderate (κ = 0.50), whereas that with SP142 was poor (κ = -0.03). Among patients who were PD-L1 negative for both assays in primary tumors, 7/30 (23.3%) were PD-L1 positive for both or either 22C3 or SP142 in the metastatic tumors. CONCLUSIONS The inter-assay concordance of PD-L1 positivity at diagnostic cutoffs was substantial in primary tumors and moderate in metastatic tumors. Discordance between PD-L1 status in primary and metastatic tumors was frequently observed, especially with SP142. Some patients with a PD-L1-negative status in primary tumors may still be candidates for immunotherapy, depending on the PD-L1 status in their metastatic tumors.
Collapse
Affiliation(s)
- Jun Miyakoshi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shu Yazaki
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Mai Onishi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Ayumi Saito
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shosuke Kita
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kasumi Yamamoto
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Hitomi Sumiyoshi-Okuma
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Emi Noguchi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takeshi Murata
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Sho Shiino
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shin Takayama
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Akihiko Suto
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Masayuki Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| |
Collapse
|
6
|
Suzuki K, Ohe R, Kabasawa T, Kitaoka T, Kawai M, Motoi F, Futakuchi M. Histological spatial analysis on the induction of PD-L1 + macrophages by CD8 + T cells at the marginal microenvironment of triple-negative breast cancer. Breast Cancer 2023; 30:1094-1104. [PMID: 37792212 PMCID: PMC10587303 DOI: 10.1007/s12282-023-01507-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/24/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) plays important roles in the evasion of antitumor immunity. Because we observed the localization of PD-L1-positive (PD-L1+) cells in the marginal region of triple-negative breast cancer (TNBC) specimens, we hypothesized that the marginal microenvironment of TNBC would involve the induction of PD-L1+ cells. METHODS One hundred and one TNBC surgical specimens were examined. We performed immunohistochemical (IHC) studies of PD-L1, CD68, CD8, and pan-cytokeratin in these specimens. We analyzed the localization of IHC-positive cells and the distance between these cells by histological spatial analysis. RESULTS In 30.7% of TNBC specimens, PD-L1+ cells were located in the marginal region. Approximately three PD-L1+ cells accumulated around a single TNBC cell. Most PD-L1+ cells were located within 50 μm of TNBC cells. PD-L1+ cells were indicated to interact with TNBC cells in the marginal region. PD-L1+CD68+ cells were located in the marginal region, while CD68+ macrophages (MΦs) were observed either in the marginal region or the core region. PD-L1 expression in MΦs was induced in the marginal region. The colocalization of CD8+ T cells in the marginal region indicates that PD-L1 expression in MΦs would be induced by interaction with CD8+ T cells. Because CD8+ T cells are positive for CCL2, CCL2 may induce PD-L1 expression in MΦs. CONCLUSION At the marginal microenvironment of TNBC, PD-L1 expression would be induced in MΦs by interaction with CD8+ T cells through CCL2. The interaction between PD-L1+ MΦs and TNBC cells would facilitate the growth of TNBC under antitumor immunity. These interactions would be potential targets for restoring antitumor immunity and suppressing TNBC progression.
Collapse
Affiliation(s)
- Kazushi Suzuki
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan.
| | - Rintaro Ohe
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Takanobu Kabasawa
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Takumi Kitaoka
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Masaaki Kawai
- Department of Surgery 1, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Fuyuhiko Motoi
- Department of Surgery 1, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Mitsuru Futakuchi
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| |
Collapse
|
7
|
Tierno D, Grassi G, Scomersi S, Bortul M, Generali D, Zanconati F, Scaggiante B. Next-Generation Sequencing and Triple-Negative Breast Cancer: Insights and Applications. Int J Mol Sci 2023; 24:9688. [PMID: 37298642 PMCID: PMC10253720 DOI: 10.3390/ijms24119688] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
The poor survival of triple-negative breast cancer (TNBC) is due to its aggressive behavior, large heterogeneity, and high risk of recurrence. A comprehensive molecular investigation of this type of breast cancer using high-throughput next-generation sequencing (NGS) methods may help to elucidate its potential progression and discover biomarkers related to patient survival. In this review, the NGS applications in TNBC research are described. Many NGS studies point to TP53 mutations, immunocheckpoint response genes, and aberrations in the PIK3CA and DNA repair pathways as recurrent pathogenic alterations in TNBC. Beyond their diagnostic and predictive/prognostic value, these findings suggest potential personalized treatments in PD -L1-positive TNBC or in TNBC with a homologous recombination deficit. Moreover, the comprehensive sequencing of large genomes with NGS has enabled the identification of novel markers with clinical value in TNBC, such as AURKA, MYC, and JARID2 mutations. In addition, NGS investigations to explore ethnicity-specific alterations have pointed to EZH2 overexpression, BRCA1 alterations, and a BRCA2-delaAAGA mutation as possible molecular signatures of African and African American TNBC. Finally, the development of long-read sequencing methods and their combination with optimized short-read techniques promise to improve the efficiency of NGS approaches for future massive clinical use.
Collapse
Affiliation(s)
- Domenico Tierno
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (D.T.); (G.G.)
| | - Gabriele Grassi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (D.T.); (G.G.)
| | - Serena Scomersi
- Breast Unit-Azienda Sanitaria Universitaria Integrata Giuliano Isontina ASUGI, University of Trieste, 34149 Trieste, Italy;
| | - Marina Bortul
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.B.); (D.G.); (F.Z.)
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.B.); (D.G.); (F.Z.)
- Azienda Socio-Sanitaria Territoriale di Cremona-ASST, Breast Cancer Unit and Translational Research Unit, 26100 Cremona, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.B.); (D.G.); (F.Z.)
| | - Bruna Scaggiante
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (D.T.); (G.G.)
| |
Collapse
|
8
|
Murazawa C, Hashimoto N, Kuraishi K, Motoyama M, Hashimoto SI, Ikeuchi M, Norimura S, Matsunaga T, Teramoto K, Haba R, Abe N, Yajima T, Kontani K. Status and prognostic value of immunological biomarkers of breast cancer. Oncol Lett 2023; 25:164. [PMID: 36960188 PMCID: PMC10028224 DOI: 10.3892/ol.2023.13750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/01/2023] [Indexed: 03/25/2023] Open
Abstract
The immune response to cancer serves an important role in disease progression and patient prognosis. For triple-negative breast cancer showing aggressive behavior, immunotherapy has a good efficacy because of the potent immunogenicity of this type of cancer. However, the dominant subtype, luminal human epidermal growth factor receptor-2 (HER2)-negative breast cancer, is less immunogenic. To determine whether luminal HER2-negative cancer reacts to the anticancer immune response, the present study analyzed the status and prognostic value of the principal immunological biomarkers of breast cancer, including tumor-infiltrating lymphocytes (TILs), CD8+ T lymphocytes, the major histocompatibility complex and programmed cell death ligand-1 (PD-L1). The biomarkers were compared between patients with luminal HER2-negative breast cancer and those with immunogenic subtypes including triple-negative and HER2-overexpressed breast cancer. A total of 71 patients with primary breast cancer were classified into the immunogenic non-luminal (n=23) and less immunogenic luminal HER2-negative groups (n=48) based on immunogenicity. In the luminal HER2-negative group, compared with patients with low TIL levels, those with high TIL levels were at an advanced stage of cancer (P=0.024) and showed worse relapse-free survival (P=0.057); however, the remaining biomarkers exhibited no association with cancer progression or prognosis. In the non-luminal group, patients with high TIL levels showed significantly better RFS than those with low TIL levels (P=0.014). Compared with non-luminal patients negative for PD-L1, those positive for PD-L1 exhibited better overall survival (P=0.064). Notably, TIL status was found to exhibit contrasting prognostic predictions based on immunogenicity. In conclusion, TILs are a strong candidate for prognostic prediction in breast cancer, regardless of the subtype. PD-L1 is a potential candidate for prognostic prediction in immunogenic breast cancers, but not in the luminal HER2-negative subtype.
Collapse
Affiliation(s)
- Chisa Murazawa
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Nozomi Hashimoto
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Kana Kuraishi
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Mutsumi Motoyama
- Department of Diagnostic Pathology, Kagawa University Hospital, Kagawa 761-0793, Japan
| | - Shin-Ichiro Hashimoto
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Mayumi Ikeuchi
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Shoko Norimura
- Department of Surgery, Takamatsu Red Cross Hospital, Takamatsu, Kagawa 760-0017, Japan
| | - Toru Matsunaga
- Department of Diagnostic Pathology, Kagawa University Hospital, Kagawa 761-0793, Japan
| | - Koji Teramoto
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga 520-2191, Japan
| | - Reiji Haba
- Department of Diagnostic Pathology, Kagawa University Hospital, Kagawa 761-0793, Japan
| | - Noriko Abe
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Toshiki Yajima
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Keiichi Kontani
- Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
- Correspondence to: Dr Keiichi Kontani, Department of Thoracic, Breast and Endocrine Surgery, Faculty of Medicine, Kagawa University, 1750-1 Miki-cho, Kita-gun, Kagawa 761-0793, Japan, E-mail:
| |
Collapse
|
9
|
Möller K, Knöll M, Bady E, Schmerder MJ, Rico SD, Kluth M, Hube-Magg C, Blessin NC, Mandelkow T, Lennartz M, Menz A, Luebke AM, Höflmayer D, Fraune C, Bernreuther C, Lebok P, Uhlig R, Contreras H, Weidemann S, Gorbokon N, Jacobsen F, Clauditz TS, Steurer S, Burandt E, Minner S, Sauter G, Simon R, Marx AH, Krech T. PD-L1 expression and CD8 positive lymphocytes in human neoplasms: A tissue microarray study on 11,838 tumor samples. Cancer Biomark 2023; 36:177-191. [PMID: 36683495 PMCID: PMC9986704 DOI: 10.3233/cbm-220030] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Programmed death ligand 1 (PD-L1) is the target of immune checkpoint inhibitor therapies in a growing number of tumor types, but a unanimous picture on PD-L1 expression across cancer types is lacking. MATERIALS AND METHODS We analyzed immunohistochemical PD-L1 expression in 11,838 samples from 118 human tumor types and its relationship with tumor infiltrating CD8 positive lymphocytes. RESULTS At a cut-off level of 10% positive tumor cells, PD-L1 positivity was seen in 85 of 118 (72%) tumor types, including thymoma (100% positive), Hodgkin's lymphoma (93%), anaplastic thyroid carcinoma (76%), Kaposi sarcoma (71%), sarcomatoid urothelial carcinoma (71%), and squamous cell carcinoma of the penis (67%), cervix (65%), floor of the mouth (61%), the lung (53%), and pharynx (50%). In immune cells, PD-L1 positivity was detectable in 103 (87%) tumor types, including tumors of haematopoetic and lymphoid tissues (75% to 100%), Warthin tumors of the parotid glands (95%) and Merkel cell carcinoma (82%). PD-L1 positivity in tumor cells was significantly correlated with the number of intratumoral CD8 positive lymphocytes across all tumor types as well as in individual tumor types, including serous carcinoma of the ovary, invasive breast carcinoma of no special type, intestinal gastric adenocarcinoma, and liposarcoma (p< 0.0001 each). CONCLUSIONS PD-L1 expression in tumor and inflammatory cells is found in a wide range of human tumor types. Higher rates of tumor infiltrating CD8 positive lymphocytes in PD-L1 positive than in PD-L1 negative cancers suggest that the antitumor immune response may trigger tumoral PD-L1 expression.
Collapse
Affiliation(s)
- Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madeleine Knöll
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Bady
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niclas C Blessin
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Mandelkow
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hendrina Contreras
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| |
Collapse
|
10
|
Molecular Pathways of Breast Cancer in Systemic Sclerosis: Exploratory Immunohistochemical Analysis from the Sclero-Breast Study. J Pers Med 2022; 12:jpm12122007. [PMID: 36556228 PMCID: PMC9780893 DOI: 10.3390/jpm12122007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/25/2022] [Indexed: 12/11/2022] Open
Abstract
Several authors reported an increased risk of cancer in SSc patients, including breast cancer (BC). Nevertheless, the mechanisms underlying this association have not yet been clarified. SSc and BC share several molecular pathways, which seem to play a common etiopathogenetic role. The previously published Sclero-Breast study demonstrated the development of BC with a good prognosis among these patients, which could be explained by an autoimmune background as a possible mechanism for limiting tumor extension. Here, we report the results of an IHC analysis of molecular pathways known to be common drivers for both diseases, with the aim to better define the mechanisms underlying a good prognosis of BC in patients affected by SSc. The analysis demonstrated higher TILs rates in all BC subgroups, with a high rate of PD-L1 expression especially in TNBC and HER2-positive BC, suggesting a less aggressive behavior in these patients compared to the general population. These results support a possible de-escalation strategy of cancer therapies in these fragile patients. These data could represent a starting point for future prospective studies based on the clinical application of these biomarkers with a larger sample size to promote a personalized and targeted oncological treatment for this specific subset of patients.
Collapse
|
11
|
Deutschmann C, Bartsch R, Singer CF, Gschwantler-Kaulich D, Seifert M, Leser C, Marhold M, Bago-Horvath Z, Pfeiler G. Atezolizumab plus nab-paclitaxel for unresectable, locally advanced or metastatic breast cancer: real-world results from a single academic center in Austria. BMC Cancer 2022; 22:1099. [PMID: 36289467 PMCID: PMC9609239 DOI: 10.1186/s12885-022-10168-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022] Open
Abstract
Purpose IMpassion130 led to the approval of atezolizumab plus nab-paclitaxel as first-line treatment for patients with unresectable locally advanced or metastatic triple-negative, PD-L1 immune-cell positive breast cancer (BC) by the European Medicines Agency (EMA). The objective of the present study was to investigate the implementation, safety and efficacy of this combination in the initial phase after approval. Methods A retrospective data analysis including all BC patients who received atezolizumab and nab-paclitaxel between 1.1.2019 and 31.10.2020 at the Department of Obstetrics and Gynecology and the Department of Medicine 1, respectively, at the Medical University of Vienna, Austria, was performed. Progression-free survival (PFS) and overall survival (OS) were estimated with the Kaplan-Maier product-limit method. Owing to the retrospective nature of this study, all statistics must be considered exploratory. Results In total 20 patients were included in the study. Median follow-up was 7.1 months (IQR 5.2–9.1). Median PFS was 3.0 months (SE = .24; 95% CI [2.5; 3.5]). Median OS was 8.94 months (SE = 2.34, 95%CI [4.35; 13.53]). No new safety signals were observed. Conclusion The present study showed a considerably shorter PFS (3.0 vs. 7.5 months) and OS (8.94 vs. 25.0 months) than IMpassion130 putatively owing to the use of atezolizumab in later treatment lines, more aggressive tumors and a study population with higher morbidity compared to the pivotal trial.
Collapse
Affiliation(s)
- Christine Deutschmann
- grid.22937.3d0000 0000 9259 8492Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Rupert Bartsch
- grid.22937.3d0000 0000 9259 8492Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Christian F Singer
- grid.22937.3d0000 0000 9259 8492Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Daphne Gschwantler-Kaulich
- grid.22937.3d0000 0000 9259 8492Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Michael Seifert
- grid.22937.3d0000 0000 9259 8492Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Carmen Leser
- grid.22937.3d0000 0000 9259 8492Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Maximilian Marhold
- grid.22937.3d0000 0000 9259 8492Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- grid.22937.3d0000 0000 9259 8492Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Georg Pfeiler
- grid.22937.3d0000 0000 9259 8492Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
12
|
Chen C, Ma X, Li Y, Ma J, Yang W, Shui R. Concordance of PD-L1 expression in triple-negative breast cancers in Chinese patients: A retrospective and pathologist-based study. Pathol Res Pract 2022; 238:154137. [PMID: 36152566 DOI: 10.1016/j.prp.2022.154137] [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: 05/20/2022] [Revised: 09/07/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVE To compare the expression of programmed cell death ligand 1 (PD-L1) in different paraffin blocks from the same triple-negative breast cancers (TNBC) specimen and between matched primary tumors and lymph node metastases (LNMets). We also aim to determine the interobserver agreement between pathologists trained on PD-L1 (SP142) assay in assessing TNBC. METHODS 426 histologically confirmed TNBC cases, in which 85 have LNMets, were included in this study. A PD-L1 (SP142) assay was used to identify PD-L1 expression on tumor infiltrating immune cells (IC) and also on tumor cells (TC) in primary tumors and LNMets of TNBC by two trained pathologists. PD-L1 scoring and assessment were based on criteria in IMpassion 130 trial criteria. Concordance of PD-L1 expression in TNBC were analyzed using Kappa-test and assessed by the Kappa value. RESULTS Prevalence of positive PD-L1 expression (PD-L1 +) on tumor-infiltrating immune cells (PD-L1 IC+) (IC≥1%) in LNMets (49.4%) was higher than in the matched primary tumors (38.9%). Concordance of PD-L1 expression on IC between the two paraffin blocks from the same primary tumor specimen was substantial (P < 0.000, Kappa = 0.627) and was identified in 83.1% (108/130) of the selected cases. For TNBC cases with matched primary and LNMets blocks, the concordance of PD-L1IC scoring between the two blocks was moderate (P < 0.000, Kappa = 0.434). Interobserver agreement of PD-L1 assessment was 78.2% (P < 0.000, Kappa = 0.567) in primary tumors and 61.4% (P < 0.000, Kappa = 0.253) in the matched LNets. CONCLUSION Substantial intratumor concordance of PD-L1 scoring of the primary tumors in TNBC patients was determined, implying that immunohistochemically detection using one representative block of the primary tumor should be enough to assign the expression status of PD-L1 in clinical practice. The prevalence of PD-L1 + in lymph node metastases (LNMets) was higher than in the matched primary tumors, implying that PD-L1 detection in LNMets may provide additional PD-L1 expression information, especially in TNBC cases with PD-L1- in the matched primary breast tumors. Interobserver agreement of PD-L1 scoring in primary tumors was moderate while only fair in LNMets, implying that the additional training for PD-L1 assessment of TNBC LNMets specimens is recommended to enhance interobserver agreement. DATA AVAILABILITY The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Chen Chen
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Xiaoxi Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Yanping Li
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Jing Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China.
| | - Ruohong Shui
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China.
| |
Collapse
|
13
|
Huertas-Caro CA, Ramirez MA, Gonzalez-Torres HJ, Sanabria-Salas MC, Serrano-Gómez SJ. Immune Lymphocyte Infiltrate and its Prognostic Value in Triple-Negative Breast Cancer. Front Oncol 2022; 12:910976. [PMID: 35924147 PMCID: PMC9342669 DOI: 10.3389/fonc.2022.910976] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) occurs more frequently in young (<50 years) non-Hispanic black and Hispanic/Latina women. It is considered the most aggressive subtype of breast cancer, although, recently, immune infiltrate has been associated with long-term survival, lower risk of death and recurrence, and response to neoadjuvant chemotherapy. The aim of this review was to evaluate the clinical impact of the immune infiltrate in TNBC by discussing whether its prognostic value varies across different populations. A comprehensive systematic search in databases such as PubMed and Web of Science was conducted to include papers focused on tumor-infiltrating lymphocytes (TILs) in TNBC in different population groups and that were published before January 2021. TNBC patients with higher levels of TILs had longer overall survival and disease-free survival times compared with TNBC patients with low TIL levels. Similar results were observed for CD4+, CD8+ TIL populations. On the other hand, patients with high TIL levels showed a higher rate of pathological complete response regardless of the population group (Asian, European, and American). These results altogether suggest that TIL subpopulations might have a prognostic role in TNBC, but the underlying mechanism needs to be elucidated. Although the prognosis value of TILs was not found different between the population groups analyzed in the revised literature, further studies including underrepresented populations with different genetic ancestries are still necessary to conclude in this regard.
Collapse
Affiliation(s)
| | - Mayra Alejandra Ramirez
- Grupo de investigación en biología del cáncer, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Henry J. Gonzalez-Torres
- Doctorado en Ciencias Biomédicas, Universidad del Valle, Cali, Colombia
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | | | - Silvia J. Serrano-Gómez
- Grupo de apoyo y seguimiento para la investigación, Instituto Nacional de Cancerología, Bogotá, Colombia
| |
Collapse
|
14
|
Shafi S, Parwani AV, Li Z. PD-L1 (SP142 and 22C3) Immunohistochemistry in Clinical Metastatic Triple Negative or Low Hormone Receptor Breast Carcinomas: Experience from a Large Academic Institution. Hum Pathol 2022; 126:100-107. [PMID: 35623466 DOI: 10.1016/j.humpath.2022.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 12/31/2022]
Abstract
The discovery of new immune checkpoint molecules has led to the emergence of new treatment for metastatic breast carcinoma. We aimed to investigate PD-L1 (SP142 and 22C3) expression in 77 clinical metastatic triple negative or low hormone receptor (<10%) breast carcinomas (TNBC/LHRBC). SP142 was positive in 23.4% (18/77) of cases. SP142 positive (SP142+) cases showed lower liver metastasis and androgen receptor expression, but increased tumor infiltrating lymphocytes than SP142 negative (SP142-) cases in univariate analysis, but only increased tumor infiltrating lymphocyte in multivariate analysis. 22C3 testing was available in 21 cases including 14 with combined positive score (CPS) ≥10 (9 SP142- and 5 SP142+) and 7 with CPS <10 (7 SP142-). Ten (13%) patients received immunotherapy including 8 SP142+ (7 with atezolizumab, 1 with pembrolizumab) and 2 SP142- cases (2 with pembrolizumab). Survival data showed a trend of increased survival rate in SP142+ (72.2%) when comparing to SP142- patients (55.9%). Our study provides unique insights into the distribution of PD-L1 staining in metastatic breast carcinomas in a real-world clinical setting.
Collapse
Affiliation(s)
- Saba Shafi
- Department of Pathology, Wexner Medical Center at The Ohio State University, 410 W. 10th Ave, Columbus, OH 43210
| | - Anil V Parwani
- Department of Pathology, Wexner Medical Center at The Ohio State University, 410 W. 10th Ave, Columbus, OH 43210
| | - Zaibo Li
- Department of Pathology, Wexner Medical Center at The Ohio State University, 410 W. 10th Ave, Columbus, OH 43210.
| |
Collapse
|
15
|
Immunoarchitectural patterns as potential prognostic factors for invasive ductal breast cancer. NPJ Breast Cancer 2022; 8:26. [PMID: 35228530 PMCID: PMC8885796 DOI: 10.1038/s41523-022-00389-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 01/21/2022] [Indexed: 12/17/2022] Open
Abstract
Currently, tumor-infiltrating lymphocytes (TILs) in invasive breast cancers are assessed solely on the basis of their number, whereas their spatial distribution is rarely investigated. Therefore, we evaluated TILs in 579 patients with invasive breast cancer of no special type (IBC-NST) with a focus on their spatial distributions in tumor center (TC) and invasive margin (IM). We also assessed a new factor, namely para-tumor infiltrating lymphocytes (PILs) in the para-tumor lobular area (Para). Five immunoarchitectural patterns (IPs) were observed, which were significantly associated with clinicopathological features, especially molecular subtypes, histological grades, clinical stages, and programmed death-ligand 1 (PD-L1) expression. High-TIL density (IP1/2) correlated with favorable disease-free survival (DFS) in TNBC patients (p = 0.04), but opposite results were observed for luminal B subtype patients (both the lowest TIL and PIL densities (IP5) correlated with good DFS, p = 0.013). Luminal B patients with high TILs in the IM and low TILs in the TC (IP3) exhibited the worst DFS, whereas those with low TILs (similar to IP5) and high PILs (IP4) exhibited poor DFS. We also identified TIL subpopulations with significantly different IPs. Our findings suggest that IP can be a potential prognostic factor for tumor immunity in IBC.
Collapse
|
16
|
Ni Y, Tsang JY, Shao Y, Poon IK, Tam F, Shea KH, Tse GM. OUP accepted manuscript. Oncologist 2022; 27:e313-e327. [PMID: 35380716 PMCID: PMC8982370 DOI: 10.1093/oncolo/oyab063] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 12/18/2021] [Indexed: 11/22/2022] Open
Abstract
Background PD-L1 has been used as a biomarker to select patients for treatment of PD-1/PD-L1 inhibitors. Materials and Methods In this study, we assessed the clinicopathological features of breast cancers that are associated with PD-L1 expression, as well as its relationship with other immune components and its prognostic significance. Results Totally 1752 cases were included in this cohort. PD-L1 expression in tumor-infiltrating immune cells (PD-L1-IC) expression and in tumor cells (PD-L1-TC) expression were identified in 34.2% and 10.1% of cases, respectively, and they showed a positive correlation with higher tumor grade, morphological apocrine features, presence of necrosis, and higher stromal tumor-infiltrating lymphocytes (sTIL). PD-L1-IC and PD-L1-TC expression correlated positively with each other, and both of them were negatively associated with estrogen receptor and progesterone receptor and positively associated with Ki67, HER2, EGFR, p63, and p-cadherin. In survival analysis, PD-L1-IC expression was associated with better disease-free survival (DFS) and breast cancer-specific survival (BCSS) in HER2-overexpressed (HER2-OE) cancers and high–grade luminal B cancers. In triple–negative breast cancers (TNBC) and HER2–OE cancers, compared with sTIL low PD-L1-IC negative cases, sTIL high cases showed significantly better DFS independent of PD-L1-IC status. sTIL low PD-L1-IC positive cases also demonstrated a better DFS in HER2–OE cancers. In high–grade luminal B cancers, sTIL high PD-L1-IC positive cases showed the best BCSS. Conclusion The data suggested that the combining analysis of sTIL and PD-L1-IC expression refined the prognostication of breast cancer subtypes. Cases with high TIL and PD-LI-IC expression appear to be more immune active.
Collapse
Affiliation(s)
- Yunbi Ni
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Ngan Shing Street, Shatin, NT, Hong Kong
| | - Julia Y Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Ngan Shing Street, Shatin, NT, Hong Kong
| | - Yan Shao
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Ngan Shing Street, Shatin, NT, Hong Kong
| | - Ivan K Poon
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Ngan Shing Street, Shatin, NT, Hong Kong
| | - Fiona Tam
- Department of Pathology, Kwong Wah Hospital, Hong Kong
| | - Ka-Ho Shea
- Department of Pathology, Tuen Mun Hospital, Hong Kong
| | - Gary M Tse
- Department of Pathology, Kwong Wah Hospital, Hong Kong
- Corresponding author: Gary M. Tse, FRCPC, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Ngan Shing Street, Shatin, Hong Kong. Tel: 852 35052359;
| |
Collapse
|
17
|
Salisbury T, Abozina A, Zhang C, Mao E, Banyi N, Leo J, Ionescu D, Zhou C, Wang G. Histological subtype is associated with PD-L1 expression and CD8+ T-cell infiltrates in triple-negative breast carcinoma. Ann Diagn Pathol 2022; 57:151901. [DOI: 10.1016/j.anndiagpath.2022.151901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/29/2021] [Accepted: 01/14/2022] [Indexed: 12/22/2022]
|
18
|
Lo Gullo R, Wen H, Reiner JS, Hoda R, Sevilimedu V, Martinez DF, Thakur SB, Jochelson MS, Gibbs P, Pinker K. Assessing PD-L1 Expression Status Using Radiomic Features from Contrast-Enhanced Breast MRI in Breast Cancer Patients: Initial Results. Cancers (Basel) 2021; 13:cancers13246273. [PMID: 34944898 PMCID: PMC8699819 DOI: 10.3390/cancers13246273] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary To our knowledge, this is the first study assessing radiomics coupled with machine learning from MRI-derived features to predict PD-L1 expression status in biopsy-proven triple negative breast cancers and comparing the performance of this approach with the performance of qualitative assessment by two radiologists. This pilot study shows that radiomics analysis coupled with machine learning of DCE-MRI is a promising approach to derive prognostic and predictive information and to select patients who could benefit from anti-PD-1/PD-L1 treatment. This technique could also be used to monitor PD-L1 expression, as it can vary over time and between different regions of the tumor, thus avoiding repeated biopsies. Abstract The purpose of this retrospective study was to assess whether radiomics analysis coupled with machine learning (ML) based on standard-of-care dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can predict PD-L1 expression status in patients with triple negative breast cancer, and to compare the performance of this approach with radiologist review. Patients with biopsy-proven triple negative breast cancer who underwent pre-treatment breast MRI and whose PD-L1 status was available were included. Following 3D tumor segmentation and extraction of radiomic features, radiomic features with significant differences between PD-L1+ and PD-L1− patients were determined, and a final predictive model to predict PD-L1 status was developed using a coarse decision tree and five-fold cross-validation. Separately, all lesions were qualitatively assessed by two radiologists independently according to the BI-RADS lexicon. Of 62 women (mean age 47, range 31–81), 27 had PD-L1− tumors and 35 had PD-L1+ tumors. The final radiomics model to predict PD-L1 status utilized three MRI parameters, i.e., variance (FO), run length variance (RLM), and large zone low grey level emphasis (LZLGLE), for a sensitivity of 90.7%, specificity of 85.1%, and diagnostic accuracy of 88.2%. There were no significant associations between qualitative assessed DCE-MRI imaging features and PD-L1 status. Thus, radiomics analysis coupled with ML based on standard-of-care DCE-MRI is a promising approach to derive prognostic and predictive information and to select patients who could benefit from anti-PD-1/PD-L1 treatment.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
| | - Hannah Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.W.); (R.H.)
| | - Jeffrey S. Reiner
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
| | - Raza Hoda
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.W.); (R.H.)
| | - Varadan Sevilimedu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10017, USA;
| | - Danny F. Martinez
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
| | - Sunitha B. Thakur
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maxine S. Jochelson
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
| | - Peter Gibbs
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Katja Pinker
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.L.G.); (J.S.R.); (D.F.M.); (S.B.T.); (M.S.J.); (P.G.)
- Correspondence: ; Tel.: +1-646-888-5200
| |
Collapse
|
19
|
Schwartz CJ, da Silva EM, Marra A, Gazzo AM, Selenica P, Rai VK, Mandelker D, Pareja F, Misyura M, D'Alfonso TM, Brogi E, Drullinsky P, Razavi P, Robson ME, Drago JZ, Wen HY, Zhang L, Weigelt B, Shia J, Reis-Filho JS, Zhang H. Morphological and genomic characteristics of breast cancers occurring in individuals with Lynch Syndrome. Clin Cancer Res 2021; 28:404-413. [PMID: 34667028 DOI: 10.1158/1078-0432.ccr-21-2027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/11/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Lynch syndrome (LS) is defined by germline pathogenic mutations involving DNA Mismatch Repair (MMR) genes and linked with the development of MMR-deficient (MMRd) colon and endometrial cancers. Whether breast cancers (BC) developing in context of LS are causally related to MMR deficiency (MMRd), remains controversial. Thus, we explored the morphological and genomic characteristics of BCs occurring in LS individuals. EXPERIMENTAL DESIGN A retrospective analysis of 20,110 cancer patients who underwent multigene panel genetic testing was performed to identify individuals with a likely pathogenic/pathogenic germline variant in MLH1, MSH2, MSH6 or PMS2 who developed BCs. The histological characteristics and immunohistochemical (IHC) assessment of BCs for MMR proteins and programmed death-ligand 1 (PD-L1) expression were assessed on cases with available materials. DNA samples from paired tumors and blood were sequenced with MSK-IMPACT ({greater than or equal to}468 key cancer genes). MSI status was assessed utilizing MSISensor. Mutational signatures were defined using SigMA. RESULTS 272 LS individuals were identified, 13 (5%) of whom had primary BCs. The majority of BCs (92%) were hormone receptor positive tumors. Five (42%) of 12 BCs displayed loss of MMR proteins by IHC. Four (36%) of 11 BCs subjected to tumor-normal sequencing showed dominant microsatellite instability mutational signatures, high tumor mutational burden and indeterminate (27%) or high MSISensor scores (9%). One patient with metastatic MMRd BC received anti-PD1 therapy and achieved a robust and durable response. CONCLUSIONS A subset of BCs developing in LS individuals are etiologically linked to MMRd and may benefit from anti-PD1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
| | | | - Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS
| | - Andrea M Gazzo
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | | | | | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | | | | | - Edi Brogi
- Memorial Sloan Kettering Cancer Center
| | | | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Mark E Robson
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | - Liying Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | | | - Hong Zhang
- Pathology, Memorial Sloan Kettering Cancer Center
| |
Collapse
|
20
|
Carter JM, Polley MYC, Leon-Ferre RA, Sinnwell J, Thompson KJ, Wang X, Ma Y, Zahrieh D, Kachergus JM, Solanki M, Boughey JC, Liu MC, Ingle JN, Kalari KR, Couch FJ, Thompson EA, Goetz MP. Characteristics and Spatially Defined Immune (micro)landscapes of Early-stage PD-L1-positive Triple-negative Breast Cancer. Clin Cancer Res 2021; 27:5628-5637. [PMID: 34108182 PMCID: PMC8808363 DOI: 10.1158/1078-0432.ccr-21-0343] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Programmed death ligand 1 [PD-(L)1]-targeted therapies have shown modest survival benefit in triple-negative breast cancer (TNBC). PD-L1+ microenvironments in TNBC are not well characterized and may inform combinatorial immune therapies. Herein, we characterized clinicopathologic features, RNA-based immune signatures, and spatially defined protein-based tumor-immune microenvironments (TIME) in early-stage PD-L1+ and PD-L1- TNBC. EXPERIMENTAL DESIGN From a large cohort of chemotherapy-naïve TNBC, clinicopathologic features, deconvoluted RNA immune signatures, and intraepithelial and stromal TIME (Nanostring GeoMX) were identified in subsets of PD-L1+ and PD-L1- TNBC, as defined by FDA-approved PD-L1 companion assays. RESULTS 228 of 499 (46%) TNBC were PD-L1+ (SP142: ≥1% immune cells-positive). Using PD-L1 22C3, 46% had combined positive score (CPS) ≥ 1 and 16% had CPS ≥10. PD-L1+ TNBC were higher grade with higher tumor-infiltrating lymphocytes (TIL; P < 0.05). PD-L1 was not associated with improved survival following adjustment for TILs and other variables. RNA profiles of PD-L1+ TNBC had increased dendritic cell, macrophage, and T/B cell subset features; and decreased myeloid-derived suppressor cells. PD-L1+ stromal and intraepithelial TIMEs were highly enriched in IDO-1, HLA-DR, CD40, and CD163 compared with PD-L1-TIME, with spatially specific alterations in CTLA-4, Stimulator of Interferon Genes (STING), and fibronectin. Macrophage- and antigen presentation-related proteins correlated most strongly with PD-L1 protein. CONCLUSIONS In this early-stage TNBC cohort, nearly 50% were PD-L1+ (SP142 companion assay) while 16% were PD-L1+ with the 22C3 companion assay. PD-L1+ TNBC had specific myeloid-derived and lymphoid features. Spatially defined PD-L1+ TIME were enriched in several clinically actionable immune proteins. These data may inform future studies on combinatorial immunotherapies for patients with PD-L1+ TNBC.See related commentary by Symmans, p. 5446.
Collapse
Affiliation(s)
- Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Mei-Yin C Polley
- Department of Public Health Sciences, The University of Chicago, Chicago, Illinois
| | | | - Jason Sinnwell
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Kevin J Thompson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - Yaohua Ma
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - David Zahrieh
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Malvika Solanki
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Minetta C Liu
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | | |
Collapse
|
21
|
Grabenstetter A, Jungbluth AA, Frosina D, Hoda R, Dos Anjos CH, Patil S, Sevilimedu V, Weigelt B, Reis-Filho JS, Zhang H, Traina T, Robson ME, Brogi E, Wen HY. PD-L1 Expression in Metaplastic Breast Carcinoma Using the PD-L1 SP142 Assay and Concordance Among PD-L1 Immunohistochemical Assays. Am J Surg Pathol 2021; 45:1274-1281. [PMID: 34115674 PMCID: PMC9437740 DOI: 10.1097/pas.0000000000001760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Immunotherapy for the treatment of programmed death-ligand 1 (PD-L1) positive locally advanced or metastatic triple negative breast cancer may benefit patients with metaplastic breast cancer (MpBC). Previous study of PD-L1 in MpBC scored tumor cells (TCs), different from Food and Drug Administration-approved scoring methods. We sought to define PD-L1 expression in MpBCs and to evaluate concordance of 3 PD-L1 assays. Primary, treatment naive MpBC treated at our Center from 1998 to 2019 were identified. PD-L1 expression was assessed using SP142, E1L3n, and 73-10. We evaluated PD-L1 expression on tumor infiltrating immune cells (IC) and also in TCs. For each assay, we scored PD-L1 expression using ≥1% IC expression according to the IMpassion130 trial criteria and using combined positive score (CPS) ≥10 according to the KEYNOTE-355 trial cutoff. A total of 42 MpBCs were identified. Most MpBC had PD-L1 positivity in ≥1% IC with all 3 assays (95%, 95%, 86%) in contrast to a maximum 71% with a CPS ≥10. PD-L1 IC expression was comparable between the SP142 and 73-10 assays and was lowest with E1L3n. PD-L1 TC expression was lowest using SP142. The overall concordance for IC scoring was 88% while 62% had concordant CPS. For each assay, the results of the 2 scoring algorithms were not interchangeable. The SP142 assay showed distinct expression patterns between IC (granular, dot-like) and TC (membranous) while 73-10 and E1L3n showed membranous and/or cytoplasmic expression in both IC and TC. Most MpBC in our cohort were positive for PD-L1 indicating eligibility for anti-PD-L1/programmed death-1 immunotherapy.
Collapse
Affiliation(s)
- Anne Grabenstetter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Achim A. Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Denise Frosina
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Raza Hoda
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Carlos H. Dos Anjos
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Sujata Patil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Varadan Sevilimedu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Hong Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Tiffany Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Mark E. Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Hannah Y. Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| |
Collapse
|
22
|
Emens LA, Adams S, Cimino-Mathews A, Disis ML, Gatti-Mays ME, Ho AY, Kalinsky K, McArthur HL, Mittendorf EA, Nanda R, Page DB, Rugo HS, Rubin KM, Soliman H, Spears PA, Tolaney SM, Litton JK. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of breast cancer. J Immunother Cancer 2021; 9:e002597. [PMID: 34389617 PMCID: PMC8365813 DOI: 10.1136/jitc-2021-002597] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer has historically been a disease for which immunotherapy was largely unavailable. Recently, the use of immune checkpoint inhibitors (ICIs) in combination with chemotherapy for the treatment of advanced/metastatic triple-negative breast cancer (TNBC) has demonstrated efficacy, including longer progression-free survival and increased overall survival in subsets of patients. Based on clinical benefit in randomized trials, ICIs in combination with chemotherapy for the treatment of some patients with advanced/metastatic TNBC have been approved by the United States (US) Food and Drug Administration (FDA), expanding options for patients. Ongoing questions remain, however, about the optimal chemotherapy backbone for immunotherapy, appropriate biomarker-based selection of patients for treatment, the optimal strategy for immunotherapy treatment in earlier stage disease, and potential use in histological subtypes other than TNBC. To provide guidance to the oncology community on these and other important concerns, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel of experts to develop a clinical practice guideline (CPG). The expert panel drew upon the published literature as well as their clinical experience to develop recommendations for healthcare professionals on these important aspects of immunotherapeutic treatment for breast cancer, including diagnostic testing, treatment planning, immune-related adverse events (irAEs), and patient quality of life (QOL) considerations. The evidence-based and consensus-based recommendations in this CPG are intended to give guidance to cancer care providers treating patients with breast cancer.
Collapse
Affiliation(s)
- Leisha A Emens
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sylvia Adams
- Perlmutter Cancer Center, New York University Langone, New York, New York, USA
| | - Ashley Cimino-Mathews
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, Washington, USA
| | - Margaret E Gatti-Mays
- Pelotonia Institute for Immuno-Oncology, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Alice Y Ho
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kevin Kalinsky
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | | | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Breast Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rita Nanda
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| | - David B Page
- Earle A Chiles Research Institute, Portland, Oregon, USA
| | - Hope S Rugo
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Krista M Rubin
- Center for Melanoma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Hatem Soliman
- Department of Breast Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Patricia A Spears
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
23
|
Boman C, Zerdes I, Mårtensson K, Bergh J, Foukakis T, Valachis A, Matikas A. Discordance of PD-L1 status between primary and metastatic breast cancer: A systematic review and meta-analysis. Cancer Treat Rev 2021; 99:102257. [PMID: 34237488 DOI: 10.1016/j.ctrv.2021.102257] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Programmed cell death ligand 1 (PD-L1) expression is predictive for benefit from immunotherapy in several human malignancies including triple negative breast cancer. Lower positivity rates but a larger relative benefit from atezolizumab has been implied when PD-L1 status is assessed at metastatic sites. We aimed to study the discordance of PD-L1 expression between primary tumor and metastasis in breast cancer due to its potential clinical utility. METHODS Cochrane Library, Embase, Medline and Web of science were searched for studies reporting on PD-L1 expression in primary and metastatic breast cancer, followed by data extraction. Outcomes included pooled PD-L1 positivity rates in tumor cells, immune cells or both in primary tumor and metastasis, PD-L1 discordance between matched primary tumors and metastasis and direction of discordance. RESULTS Of 2552 identified entries following de-duplication, 20 studies fulfilled the predefined inclusion criteria. Pooled PD-L1 positivity rate was higher in primary tumors compared to metastasis when assessed in immune cells (51.2% vs 37.1% p < 0.001) and tumor/immune cells (30.1% vs 14.6% p < 0.001), but not in tumor cells (18.7% vs 17.8% p = 0.65). PD-L1 positivity was lowest when assessed in bone metastases (12%) and highest in lymph nodes (60%). Discordance between primary tumors and metastasis was bidirectional, with higher pooled discordance rates when PD-L1 expression was assessed in immune compared to tumor cells (39.5% vs 13.6%, p < 0.001). CONCLUSION The observed considerable discordance between PD-L1 status in primary and metastatic breast cancer emphasizes the importance of appropriate tissue sampling when selecting patients for immunotherapy.
Collapse
Affiliation(s)
- Caroline Boman
- Department of Oncology-Pathology, Karolinska Institutet Visionsgatan 4, Bioclinicum, 171 74 Stockholm, Sweden; Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Gävlegatan 55, 171 64 Solna, Sweden.
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institutet Visionsgatan 4, Bioclinicum, 171 74 Stockholm, Sweden; Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Gävlegatan 55, 171 64 Solna, Sweden
| | - Kira Mårtensson
- Department of Clinical Pathology and Cytology, Karolinska University Laboratory, 171 76 Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet Visionsgatan 4, Bioclinicum, 171 74 Stockholm, Sweden; Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Gävlegatan 55, 171 64 Solna, Sweden
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institutet Visionsgatan 4, Bioclinicum, 171 74 Stockholm, Sweden; Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Gävlegatan 55, 171 64 Solna, Sweden
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Alexios Matikas
- Department of Oncology-Pathology, Karolinska Institutet Visionsgatan 4, Bioclinicum, 171 74 Stockholm, Sweden; Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Gävlegatan 55, 171 64 Solna, Sweden
| |
Collapse
|
24
|
Kockx MM, McCleland M, Koeppen H. Microenvironmental regulation of tumour immunity and response to immunotherapy. J Pathol 2021; 254:374-383. [PMID: 33846997 PMCID: PMC8252752 DOI: 10.1002/path.5681] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/25/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
The confluence of immunology and oncology has led to a lot of uncertainty and questions about relevant biomarkers. Despite the complexity of the tumour microenvironment, most clinical studies have relied on a single‐parameter immunohistochemical assay to prospectively select patients for checkpoint inhibitor therapy; the results of this strategy have been highly variable and often less than optimal. While great efforts have been made to identify additional or alternative biomarkers, pathologists, drug developers, and clinicians alike have faced technical, logistical, and regulatory challenges on how to implement them successfully. In this review, we will discuss these challenges; we will also highlight recent advances in dissecting the functional diversity of immune cell populations within the tumour microenvironment and their potential for improved, biomarker‐driven therapeutic strategies. The dynamic nature and cellular diversity of the tumour microenvironment may challenge past models of a single biomarker predicting patient response and clinical outcome. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
|
25
|
Zhang Q, Chen Y, Bai X, Liang T. Immune Checkpoint Blockade Therapy for Hepatocellular Carcinoma: Clinical Challenges and Considerations. Front Oncol 2020; 10:590058. [PMID: 33178615 PMCID: PMC7593704 DOI: 10.3389/fonc.2020.590058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
Although many approaches have been developed for the treatment of hepatocellular carcinoma (HCC) that has both high incidence and high mortality especially in Asian countries, the prognosis of HCC patients is still dismal. Immunotherapy, particularly immune checkpoint inhibitors show encouraging efficacy and have already been widely applied in clinic. However, in contrast to traditional therapies, immunotherapy brings many challenges when using in a real world, including biomarker discovery, response evaluation, adverse event treatment, etc. In this review, we proposed some important and intractable issues in current clinical practice regarding the strategy of immune checkpoint blockade, collected current evidence, and discuss the critical challenges and possible approaches to a bright future.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|