1
|
Miao L, Xiao G, Chen W, Yang G, Hong D, Wang Z, Zhang L, Huang W. Non-invasive assessment of programmed cell death ligand-1 expression using 18F-FDG PET-CT imaging in esophageal squamous cell carcinoma. Sci Rep 2024; 14:26082. [PMID: 39478052 PMCID: PMC11525474 DOI: 10.1038/s41598-024-77680-4] [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: 02/23/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Programmed cell death ligand-1 (PD-L1) is an ideal checkpoint for immunohistochemical detection. The method of obtaining PD-L1 expression through biopsy can impact the accurate assessment of PD-L1 expression due to the spatial and temporal heterogeneity of tumors. Because of the limited sample size, biopsies often give only a localized picture of the tumor. In this retrospective study, a total of 2,386 metabolic tumor volume (MTV) features were extracted from 18F-FDG PET-CT images. A radiomics model was developed to holistically and non-invasively assess PD-L1 expression in patients with esophageal squamous cell carcinoma by identifying seven independent factors through feature screening. The radiomics model shows effective discrimination, with an area under the receiver operating characteristic curve of 0.888 [95% confidence interval (CI): 0.831-0.945] and 0.889 (95% CI: 0.706-1.000) for the training and validation cohorts, respectively. The results of the decision curve analysis demonstrated that utilizing the radiation model to forecast PD-L1 expression levels yielded more net benefits at threshold probabilities below 0.669. The clinical impact curves demonstrate that when the threshold probability is less than 0.501, the loss-to-benefit ratio is less than one in all cases.
Collapse
Affiliation(s)
- Liming Miao
- School of Computer and Information Engineering, Hanshan Normal University, Chaozhou, 521041, Guangdong, China
| | - Gang Xiao
- School of Mathematics and Statistics, Hanshan Normal University, Chaozhou, 521041, Guangdong, China
| | - Wanqi Chen
- Department of Nuclear Medicine, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China
| | - Guisheng Yang
- Department of Nuclear Medicine, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China
| | - Denghui Hong
- Department of Nuclear Medicine, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China
| | - Zhenshan Wang
- Department of Medical Imaging Center, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China
| | - Longsheng Zhang
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China.
| | - Weipeng Huang
- Department of Nuclear Medicine, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China.
| |
Collapse
|
2
|
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
|
3
|
Sigurjonsdottir G, De Marchi T, Ehinger A, Hartman J, Bosch A, Staaf J, Killander F, Niméus E. Comparison of SP142 and 22C3 PD-L1 assays in a population-based cohort of triple-negative breast cancer patients in the context of their clinically established scoring algorithms. Breast Cancer Res 2023; 25:123. [PMID: 37817263 PMCID: PMC10566164 DOI: 10.1186/s13058-023-01724-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Immunohistochemical (IHC) PD-L1 expression is commonly employed as predictive biomarker for checkpoint inhibitors in triple-negative breast cancer (TNBC). However, IHC evaluation methods are non-uniform and further studies are needed to optimize clinical utility. METHODS We compared the concordance, prognostic value and gene expression between PD-L1 IHC expression by SP142 immune cell (IC) score and 22C3 combined positive score (CPS; companion IHC diagnostic assays for atezolizumab and pembrolizumab, respectively) in a population-based cohort of 232 early-stage TNBC patients. RESULTS The expression rates of PD-L1 for SP142 IC ≥ 1%, 22C3 CPS ≥ 10, 22C3 CPS ≥ 1 and 22C3 IC ≥ 1% were 50.9%, 27.2%, 53.9% and 41.8%, respectively. The analytical concordance (kappa values) between SP142 IC+ and these three different 22C3 scorings were 73.7% (0.48, weak agreement), 81.5% (0.63) and 86.6% (0.73), respectively. The SP142 assay was better at identifying 22C3 positive tumors than the 22C3 assay was at detecting SP142 positive tumors. PD-L1 (CD274) gene expression (mRNA) showed a strong positive association with all two-categorical IHC scorings of the PD-L1 expression, irrespective of antibody and cut-off (Spearman Rho ranged from 0.59 to 0.62; all p-values < 0.001). PD-L1 IHC positivity and abundance of tumor infiltrating lymphocytes were of positive prognostic value in univariable regression analyses in patients treated with (neo)adjuvant chemotherapy, where it was strongest for 22C3 CPS ≥ 10 and distant relapse-free interval (HR = 0.18, p = 0.019). However, PD-L1 status was not independently prognostic when adjusting for abundance of tumor infiltrating lymphocytes in multivariable analyses. CONCLUSION Our findings support that the SP142 and 22C3 IHC assays, with their respective clinically applied scoring algorithms, are not analytically equivalent where they identify partially non-overlapping subpopulations of TNBC patients and cannot be substituted with one another regarding PD-L1 detection. Trial registration The Swedish Cancerome Analysis Network - Breast (SCAN-B) study, retrospectively registered 2nd Dec 2014 at ClinicalTrials.gov; ID NCT02306096.
Collapse
Affiliation(s)
- Gudbjörg Sigurjonsdottir
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Tommaso De Marchi
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anna Ehinger
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Laboratory Medicine, Region Skåne, Lund, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Ana Bosch
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Fredrika Killander
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Emma Niméus
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
- Divison of Surgery, Department of Clinical Sciences Lund, Lund University, Sölvegatan 19 - BMC I12, 22184, Lund, Sweden.
- Department of Surgery, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
4
|
Choi H, Ahn SG, Bae SJ, Kim JH, Eun NL, Lee Y, Nahm JH, Jeong J, Cha YJ. Comparison of Programmed Cell Death Ligand 1 Status between Core Needle Biopsy and Surgical Specimens of Triple-Negative Breast Cancer. Yonsei Med J 2023; 64:518-525. [PMID: 37488704 PMCID: PMC10375241 DOI: 10.3349/ymj.2023.0090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/26/2023] Open
Abstract
PURPOSE Pembrolizumab is currently used to treat advanced triple-negative breast cancer (TNBC) and high-risk early TNBC with neoadjuvant chemotherapy (NAC). The tumor-infiltrating lymphocyte (TIL) level and programmed cell death ligand 1 (PD-L1) status are predictors of response to NAC and immune checkpoint inhibitor treatment. We aimed to investigate whether the PD-L1 status in core needle biopsies (CNBs) could represent the whole tumor in TNBC. MATERIALS AND METHODS A total of 49 patients diagnosed with TNBC who received upfront surgery without NAC between January 2018 and March 2021 were included. The PD-L1 expression (SP142 and 22C3 clones) and TIL were evaluated in paired CNBs and resected specimens. The concordance PD-L1 status and TIL levels between CNBs and resected specimens were analyzed. RESULTS PD-L1 positivity was more frequently observed in resected specimens. The overall reliability of TIL level in the CNB was good [intraclass correlation coefficient (ICC)=0.847, p<0.001]. The agreements of PD-L1 status were good and fair, respectively (SP142, κ=0.503, p<0.001; 22C3, κ=0.380, p=0.010). As the core number of CNB increased, the reliability and agreement also improved, especially from five tumor cores (TIL, ICC=0.911, p<0.001; PD-L1 [22C3], κ=0.750, p=0.028). Regarding PD-L1 (SP142), no further improvement was observed with ≥5 tumor cores (κ=0.600, p=0.058). CONCLUSION CNBs with ≥5 tumor cores were sufficient to represent the TIL level and PD-L1 (22C3) status in TNBC.
Collapse
Affiliation(s)
- Hyungwook Choi
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Gwe Ahn
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soong Joon Bae
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Na Lae Eun
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Yangkyu Lee
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hae Nahm
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Joon Jeong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Jin Cha
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
5
|
Predictive Biomarkers for Response to Immunotherapy in Triple Negative Breast Cancer: Promises and Challenges. J Clin Med 2023; 12:jcm12030953. [PMID: 36769602 PMCID: PMC9917763 DOI: 10.3390/jcm12030953] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a highly heterogeneous disease with a poor prognosis and a paucity of therapeutic options. In recent years, immunotherapy has emerged as a new treatment option for patients with TNBC. However, this therapeutic evolution is paralleled by a growing need for biomarkers which allow for a better selection of patients who are most likely to benefit from this immune checkpoint inhibitor (ICI)-based regimen. These biomarkers will not only facilitate a better optimization of treatment strategies, but they will also avoid unnecessary side effects in non-responders, and limit the increasing financial toxicity linked to the use of these agents. Huge efforts have been deployed to identify predictive biomarkers for the ICI, but until now, the fruits of this labor remained largely unsatisfactory. Among clinically validated biomarkers, only programmed death-ligand 1 protein (PD-L1) expression has been prospectively assessed in TNBC trials. In addition to this, microsatellite instability and a high tumor mutational burden are approved as tumor agnostic biomarkers, but only a small percentage of TNBC fits this category. Furthermore, TNBC should no longer be approached as a single biological entity, but rather as a complex disease with different molecular, clinicopathological, and tumor microenvironment subgroups. This review provides an overview of the validated and evolving predictive biomarkers for a response to ICI in TNBC.
Collapse
|
6
|
Xin H, Zhou C, Wang G, Liu Y, Zhang J, Liu Y, Li B, Zhang J, Su M, Li Z, Wang G. Heterogeneity of PD-L1 expression and CD8 lymphocyte infiltration in metastatic colorectal cancer and their prognostic significance. Heliyon 2023; 9:e13048. [PMID: 36814622 PMCID: PMC9939551 DOI: 10.1016/j.heliyon.2023.e13048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Purpose In recent years, immune checkpoint inhibitors have become a major therapeutic method for the treatment of metastatic colorectal cancer (mCRC). Growing evidence indicates that tumour-infiltrating lymphocytes (TILs) in the tumour microenvironment are a prerequisite for the effectiveness of PD-1/PD-L1 blockade therapy. In this study, we aimed to compare PD-L1 expression and cluster of differentiation 4 (CD4) and CD8 TIL infiltration in primary tumours and paired metastases. Patients and methods Altogether, 111 patients with mCRC who underwent surgery at our hospital were included. PD-L1, CD4, and CD8 expression were detected by immunohistochemistry in a tissue microarray. PD-L1 expression was assessed using the combined positivity score (CPS), and a score ≥1 was judged as positive. The area proportion of TILs with positive staining ≥10% was classified as "high", while <10% was classified as "low". Results We observed the discordance of PD-L1 expression between primary tumours and paired metastases in 35/111 (31.5%) patients (κ = 0.137, P = 0.142). This heterogeneity was significantly correlated with discordance of CD8 TIL infiltration between primary tumours and paired metastases (P = 0.003). Compared with corresponding colorectal cancer tumours, lung metastases showed more CD8 TIL infiltration (P = 0.022, median: 8.5% vs. 5.0%), whereas liver metastases exhibited less CD8 TIL infiltration (P = 0.028, median: 3.0% vs. 5.0%). Area proportion of CD4+ and CD8+ TIL infiltration in lung metastases were all higher than those in liver metastases (P = 0.005, median: 15.0% vs. 9.0%; P = 0.001, median: 8.5% vs. 3.0%). Compared with p MMR (MSI-L/MS-S) subgroup, area proportion of CD8 TIL infiltration in primary tumours and CD4, CD8 TIL infiltration in paired metastases were all higher in d MMR (MSI-H) group (P = 0.026, median: 15.0% vs 5.0%; P = 0.039, median: 15.0% vs 9.0%; P = 0.015, median: 15.0% vs 5.0%). Preoperative chemo/radiotherapy may increase CD8 TIL infiltration in primary tumours (P = 0.045, median: 10.0% vs. 5.0%). CD8 TIL infiltration in primary tumours was an independent predictive factor for overall survival (HR 0.28, 95% CI 0.09-0.93, P = 0.038). Conclusion Heterogeneity in PD-L1 expression and CD8 TIL infiltration was found between primary tumours and paired metastases in mCRC. CD8 TIL infiltration in primary tumours could independently forecast the overall survival of patients with mCRC.
Collapse
Key Words
- CD8 tumour infiltrating lymphocytes (TILs)
- CD8, cluster of differentiation 8
- CPS, combined positivity score
- Heterogeneity
- MS-S, microsatellite stability
- MSI-H, microsatellite instability-high
- MSI-L, microsatellite instability-low
- Metastatic colorectal cancer (mCRC)
- PD-L1, programmed death-ligand 1
- Prognosis
- Programmed death-ligand 1 (PD-L1)
- TILs, tumour infiltrating lymphocytes
- dMMR, deficient mismatch repair
- mCRC, metastatic colorectal cancer
- pMMR, proficient mismatch repair
Collapse
Affiliation(s)
- Haisong Xin
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Chaoxi Zhou
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Guanglin Wang
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Yan Liu
- Department of Endocrinology, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Juan Zhang
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Youqiang Liu
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Baokun Li
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Jianfeng Zhang
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Mingming Su
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Zhihan Li
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Guiying Wang
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China,Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China,Corresponding author. Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050051, People’s Republic of China.
| |
Collapse
|
7
|
Roesler AS, Malasi S, Koslosky L, Hartmayer P, Naab TJ, Carter JM, Zahrieh D, Hillman D, Leon-Ferre RA, Couch FJ, Goetz MP, Anderson KS, Pockaj BA, Barrett MT. PDJ amplicon in triple negative breast cancer. Sci Rep 2023; 13:618. [PMID: 36635351 PMCID: PMC9837184 DOI: 10.1038/s41598-023-27887-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Amplification of chromosome 9p24.1 targeting PD-L1, PD-L2, and JAK2 (PDJ amplicon) is present in subsets of triple negative breast cancers (TNBCs) and is associated with poor clinical outcomes. However, the prevalence of PDJ+ TNBCs varies extensively across studies applying different methods for interrogating samples of interest. To rigorously assess the prevalence of PDJ amplicons in TNBC, its prognostic value and whether it is enriched by chemotherapy, we interrogated 360 TNBC samples including 74 surgical resections from patients treated in the neoadjuvant setting, and tissue microarrays (TMAs) with 31 cases from African American women and 255 resected non-metastatic cases, with a 3 color fluorescence in situ hybridization (FISH) assay targeting the 9p24.1 PDJ amplicon, 9q24.3, and 9q34.1. Samples with mean PDJ signal of > 4.5 copies, and ratios of PDJ/9q24 ≥ 2 and/or PDJ/9q34.1 ≥ 2 were called amplified (PDJ+). Correlative analyses included the association of tumor infiltrating lymphocytes (TILs) with PDJ amplicons in TNBCs. In addition, we investigated intratumor copy number of PDJ amplicons in PDJ+ and PDJ- TNBCs. Matched pre- and post-neoadjuvant treatment biopsies were available from patients (n = 6) to evaluate the effects of therapy on PDJ status. Our study provides a rigorous analysis of the prevalence, distribution, and clinical correlatives of the PDJ amplicon in TNBC.
Collapse
Affiliation(s)
- Alexander S Roesler
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ, USA
- School of Medicine, Duke University, Durham, NC, USA
| | - Smriti Malasi
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ, USA
| | | | | | - Tammey J Naab
- Department of Pathology, Howard University Hospital, Washington, DC, USA
| | - Jodi M Carter
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA
| | - David Zahrieh
- Departments of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - David Hillman
- Departments of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Fergus J Couch
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Karen S Anderson
- Division of Hematology-Oncology, Mayo Clinic in Arizona, Scottsdale, AZ, USA
- Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Barbara A Pockaj
- Division of General Surgery, Section of Surgical Oncology, Mayo Clinic in Arizona, Phoenix, AZ, USA
| | - Michael T Barrett
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic in Arizona, Scottsdale, AZ, USA.
| |
Collapse
|
8
|
Li JJX, Tse GM. Marker assessments in ER-positive breast cancers: old markers, new applications? Histopathology 2023; 82:218-231. [PMID: 35945680 DOI: 10.1111/his.14767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/02/2022] [Accepted: 08/07/2022] [Indexed: 12/13/2022]
Abstract
Evaluation of oestrogen receptor (ER) expression by immunostaining is essential in the pathological assessment of breast cancer. Its expression is intercorrelated with clinicopathological features, molecular typing, and treatment selection. The development of novel therapeutic agents related to ER status, the recent ASCO introduction of an ER-low positive category of breast cancers, and the ever-increasing plethora of diagnostic and theragnostic markers call for a timely update. In this article we aim to review the clinicopathological features of ER-positive breast cancers, with an emphasis on ER-low positive breast cancers, and a focus on updating the (i) assessment, reporting and interpretation of ER immunohistochemical (IHC) staining, (ii) correlations of ER status with other diagnostic and theragnostic markers, and (iii) implications for treatment selection and response. In the face of the developments in IHC and molecular techniques and targeted therapy, ER immunostaining is still expected to remain as the core component of prognostic and theragnostic assessment of breast cancers.
Collapse
Affiliation(s)
- Joshua J X Li
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
9
|
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
|
10
|
Kelly RJ, Whitsett TG, Snipes GJ, Dobin SM, Finholt J, Settele N, Priest EL, Youens K, Wallace LB, Schwartz G, Wong L, Henderson SM, Gowan AC, Fonkem E, Juarez MI, Murray CE, Wu J, Van Keuren-Jensen K, Pirrotte P, Highlander S, Contente T, Baker A, Victorino J, Berens ME. The Texas Immuno-Oncology Biorepository, a statewide biospecimen collection and clinical informatics system to enable longitudinal tumor and immune profiling. Proc (Bayl Univ Med Cent) 2022; 36:1-7. [PMID: 36578607 PMCID: PMC9762845 DOI: 10.1080/08998280.2022.2114129] [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: 12/31/2022] Open
Abstract
A detailed understanding of the molecular and immunological changes that occur longitudinally across tumors exposed to immune checkpoint inhibitors is a significant knowledge gap in oncology. To address this unmet need, we created a statewide biospecimen collection and clinical informatics system to enable longitudinal tumor and immune profiling and to enhance translational research. The Texas Immuno-Oncology Biorepository (TIOB) consents patients to collect, process, store, and analyze serial biospecimens of tissue, blood, urine, and stool from a diverse population of over 100,000 cancer patients treated each year across the Baylor Scott & White Health system. Here we sought to demonstrate that these samples were fit for purpose with regard to downstream multi-omic assays. Plasma, urine, peripheral blood mononuclear cells, and stool samples from 11 enrolled patients were collected from various cancer types. RNA isolated from extracellular vesicles derived from plasma and urine was sufficient for transcriptomics. Peripheral blood mononuclear cells demonstrated excellent yield and viability. Ten of 11 stool samples produced RNA quality to enable microbiome characterization. Sample acquisition and processing methods are known to impact sample quality and performance. We demonstrate that consistent acquisition methodology, sample preparation, and sample storage employed by the TIOB can produce high-quality specimens, suited for employment in a wide array of multi-omic platforms, enabling comprehensive immune and molecular profiling.
Collapse
Affiliation(s)
- Ronan J. Kelly
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas,Corresponding author: Ronan J. Kelly, MD, MBA, Charles A. Sammons Cancer Center, Baylor University Medical Center, 3420 Worth Street, Suite 550, Dallas, TX75246 (e-mail: ); Michael E. Berens, PhD, Cancer & Cell Biology Division, Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ85004 (e-mail: )
| | - Timothy G. Whitsett
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | - G. Jackson Snipes
- Department of Pathology, Baylor University Medical Center, Dallas, Texas
| | - Sheila M. Dobin
- Department of Pathology, Baylor Scott & White Medical Center – Temple, Temple, Texas
| | | | | | | | - Kenneth Youens
- Department of Pathology, Baylor University Medical Center, Dallas, Texas
| | - Lucy B. Wallace
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas,Texas A&M Health Science Center, Dallas, Texas
| | - Gary Schwartz
- Department of Thoracic Surgery, Baylor University Medical Center, Dallas, Texas
| | - Lucas Wong
- Texas A&M Health Science Center, Dallas, Texas,Department of Hematology and Medical Oncology, Baylor Scott & White Medical Center – Temple, Temple, Texas
| | | | - Alan C. Gowan
- Baylor Scott & White Vasicek Cancer Treatment Center – Temple, Temple, Texas
| | - Ekokobe Fonkem
- Texas A&M Health Science Center, Dallas, Texas,Department of Neurosurgery, Baylor Scott & White Medical Center – Temple, Temple, Texas
| | - Maria I. Juarez
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Christal E. Murray
- Department of Hematology and Medical Oncology, Baylor Scott & White Medical Center – Temple, Temple, Texas,Baylor Scott & White Cancer Center – Round Rock, Round Rock, Texas
| | - Jeffrey Wu
- Department of Cardiac and Thoracic Surgery, Baylor Scott & White All Saints Medical Center, Fort Worth, Texas
| | | | - Patrick Pirrotte
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Sarah Highlander
- Pathogen and Microbiome Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Tania Contente
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Angela Baker
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jose Victorino
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Michael E. Berens
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona,Corresponding author: Ronan J. Kelly, MD, MBA, Charles A. Sammons Cancer Center, Baylor University Medical Center, 3420 Worth Street, Suite 550, Dallas, TX75246 (e-mail: ); Michael E. Berens, PhD, Cancer & Cell Biology Division, Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ85004 (e-mail: )
| |
Collapse
|
11
|
Shenasa E, Stovgaard ES, Jensen MB, Asleh K, Riaz N, Gao D, Leung S, Ejlertsen B, Laenkholm AV, Nielsen TO. Neither Tumor-Infiltrating Lymphocytes nor Cytotoxic T Cells Predict Enhanced Benefit from Chemotherapy in the DBCG77B Phase III Clinical Trial. Cancers (Basel) 2022; 14:cancers14153808. [PMID: 35954471 PMCID: PMC9367267 DOI: 10.3390/cancers14153808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Apart from the direct killing of cancer cells, cyclophosphamide-based chemotherapy has been shown to induce an antitumor immune response, and is being used in combination with immunotherapies in cancer care. We assessed the interaction of chemotherapy with immune biomarkers expressed on primary tumor tissue from a randomized phase III clinical trial, and confirmed that the presence of tumor-infiltrating lymphocytes is linked to improved survival in premenopausal women with high-risk breast cancer, regardless of their treatment allocation. However, immune biomarkers including tumor-infiltrating lymphocytes do not predict extra benefit from cyclophosphamide chemotherapy. This finding applies across the major molecular subgroups, including non-luminal and basal breast cancers that tend to be more immunogenic, and are often considered the most suitable subsets for receiving immunotherapy. Abstract Recent studies have shown that immune infiltrates in the tumor microenvironment play a role in response to therapy, with some suggesting that patients with immunogenic tumors may receive increased benefit from chemotherapies. We evaluated this hypothesis in early breast cancer by testing the interaction between immune biomarkers and chemotherapy using materials from DBCG77B, a phase III clinical trial where high-risk premenopausal women were randomized to receive chemotherapy or no chemotherapy. Tissue microarrays were evaluated for tumor-infiltrating lymphocytes (TILs) assessed morphologically on hematoxylin and eosin-stained slides, and by immunohistochemistry for CD8, FOXP3, LAG-3, PD-1 and PD-L1. Following REMARK reporting guidelines, data analyses were performed according to a prespecified statistical plan, using 10-year invasive disease-free survival as the endpoint. Differences in survival probabilities between biomarker groups were evaluated by Kaplan–Meier and Cox proportional hazard ratio analyses and prediction for treatment benefit by an interaction test. Our results showed that stromal TILs were associated with an improved prognosis (HR = 0.93; p-value = 0.03), consistent with previous studies. However, none of the immune biomarkers predicted benefit from chemotherapy in the full study set nor within major breast cancer subtypes. Our study indicates that primary tumors with higher immune infiltration do not derive extra benefit from cyclophosphamide-based cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Elahe Shenasa
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver V6H 3Z6, BC, Canada
| | | | - Maj-Britt Jensen
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Karama Asleh
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver V6H 3Z6, BC, Canada
| | - Nazia Riaz
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver V6H 3Z6, BC, Canada
- Centre for Regenerative Medicine and Stem Cell Research, Aga Khan University, Karachi 74800, Pakistan
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver V6H 3Z6, BC, Canada
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver V6H 3Z6, BC, Canada
| | - Bent Ejlertsen
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Anne-Vibeke Laenkholm
- Department of Surgical Pathology, Zealand University Hospital, 4000 Roskilde, Denmark
| | - Torsten O. Nielsen
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver V6H 3Z6, BC, Canada
- Correspondence:
| |
Collapse
|
12
|
Zhao X, Bao Y, Meng B, Xu Z, Li S, Wang X, Hou R, Ma W, Liu D, Zheng J, Shi M. From rough to precise: PD-L1 evaluation for predicting the efficacy of PD-1/PD-L1 blockades. Front Immunol 2022; 13:920021. [PMID: 35990664 PMCID: PMC9382880 DOI: 10.3389/fimmu.2022.920021] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Developing biomarkers for accurately predicting the efficacy of immune checkpoint inhibitor (ICI) therapies is conducive to avoiding unwanted side effects and economic burden. At the moment, the quantification of programmed cell death ligand 1 (PD-L1) in tumor tissues is clinically used as one of the combined diagnostic assays of response to anti-PD-1/PD-L1 therapy. However, the current assays for evaluating PD-L1 remain imperfect. Recent studies are promoting the methodologies of PD-L1 evaluation from rough to precise. Standardization of PD-L1 immunohistochemistry tests is being promoted by using optimized reagents, platforms, and cutoff values. Combining novel in vivo probes with PET or SPECT will probably be of benefit to map the spatio-temporal heterogeneity of PD-L1 expression. The dynamic change of PD-L1 in the circulatory system can also be realized by liquid biopsy. Consider PD-L1 expressed on non-tumor (immune and non-immune) cells, and optimized combination detection indexes are further improving the accuracy of PD-L1 in predicting the efficacy of ICIs. The combinations of artificial intelligence with novel technologies are conducive to the intelligence of PD-L1 as a predictive biomarker. In this review, we will provide an overview of the recent progress in this rapidly growing area and discuss the clinical and technical challenges.
Collapse
Affiliation(s)
- Xuan Zhao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yulin Bao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Bi Meng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Zijian Xu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Sijin Li
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xu Wang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Rui Hou
- College of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Wen Ma
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Dan Liu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dan Liu, ; Junnian Zheng, ; Ming Shi,
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dan Liu, ; Junnian Zheng, ; Ming Shi,
| | - Ming Shi
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dan Liu, ; Junnian Zheng, ; Ming Shi,
| |
Collapse
|
13
|
CDK Inhibition Primes for Anti-PD-L1 Treatment in Triple-Negative Breast Cancer Models. Cancers (Basel) 2022; 14:cancers14143361. [PMID: 35884422 PMCID: PMC9322647 DOI: 10.3390/cancers14143361] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancers (TNBC) expressing PD-L1 qualify for checkpoint inhibitor immunotherapy. Cyclin E/CDK2 is a potential target axis in TNBC; however, small-molecule drugs at efficacious doses may be associated with toxicity, and treatment alongside immunotherapy requires investigation. We evaluated CDK inhibition at suboptimal levels and its anti-tumor and immunomodulatory effects. Transcriptomic analyses of primary breast cancers confirmed higher cyclin E/CDK2 expression in TNBC compared with non-TNBC. Out of the three CDK2-targeting inhibitors tested, the CDK 2, 7 and 9 inhibitor SNS-032 was the most potent in reducing TNBC cell viability and exerted cytotoxicity against all eight TNBC cell lines evaluated in vitro. Suboptimal SNS-032 dosing elevated cell surface PD-L1 expression in surviving TNBC cells. In mice engrafted with human immune cells and challenged with human MDA-MB-231 TNBC xenografts in mammary fat pads, suboptimal SNS-032 dosing partially restricted tumor growth, enhanced the tumor infiltration of human CD45+ immune cells and elevated cell surface PD-L1 expression in surviving cancer cells. In tumor-bearing mice engrafted with human immune cells, the anti-PD-L1 antibody avelumab, given sequentially following suboptimal SNS-032 dosing, reduced tumor growth compared with SNS-032 alone or with avelumab without prior SNS-032 priming. CDK inhibition at suboptimal doses promotes immune cell recruitment to tumors, PD-L1 expression by surviving TNBC cells and may complement immunotherapy.
Collapse
|
14
|
Mukherji R, Yin C, Hameed R, Alqahtani AZ, Kulasekaran M, He AR, Weinberg BA, Marshall JL, Hartley ML, Noel MS. The current state of molecular profiling in gastrointestinal malignancies. Biol Direct 2022; 17:15. [PMID: 35668531 PMCID: PMC9172079 DOI: 10.1186/s13062-022-00322-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/19/2022] [Indexed: 11/10/2022] Open
Abstract
This is a review of the current state of molecular profiling in gastrointestinal (GI) cancers and what to expect from this evolving field in the future. Individualized medicine is moving from broad panel testing of numerous genes or gene products in tumor biopsy samples, identifying biomarkers of prognosis and treatment response, to relatively noninvasive liquid biopsy assays, building on what we have learned in our tumor analysis and growing into its own evolving predictive and prognostic subspecialty. Hence, the field of GI precision oncology is exploding, and this review endeavors to summarize where we are now in preparation for the journey ahead.
Collapse
Affiliation(s)
- Reetu Mukherji
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Chao Yin
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Rumaisa Hameed
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Ali Z Alqahtani
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Monika Kulasekaran
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Aiwu R He
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Benjamin A Weinberg
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - John L Marshall
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Marion L Hartley
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Marcus S Noel
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA.
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA.
| |
Collapse
|
15
|
Reznitsky FM, Jensen JD, Knoop A, Laenkholm AV. Inter-observer agreement of tumor infiltrating lymphocytes in primary HER2-positive breast cancer and correlation between tissue microarray and full tumor-sections. APMIS 2022; 130:545-550. [PMID: 35639634 DOI: 10.1111/apm.13251] [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: 05/30/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Tumor infiltrating lymphocytes (TILs) have predictive and prognostic potential in HER2-positive breast cancer (HER2 + BC). Due to tumor heterogeneity, guidelines recommend evaluation on full tumor-sections over biopsies, but aren't clear regarding tissue microarrays (TMAs). Herein, we investigate the inter-observer agreement of TILs assessment in HER2 + BC on full-sections and TMAs using a standardized method. Two pathologists assessed TILs using HE full-sections and TMAs from 244 patients with HER2 + BC. TILs were assessed in 10% intervals; values <10% evaluated as 0%, 1% or 5%. Levels of agreement were evaluated using intraclass-coefficient (ICC), Kappa statistics and concordance analysis. For inter-observer agreement for full-sections, ICC was 0.93 (95% CI 0.89-0.95) and Kappa was 0.75, corresponding to acceptable and moderate agreement respectively. For TMAs, ICC was 0.73 (95% CI: 0.62-0.81) and Kappa 0.33, corresponding to unacceptable agreement. For association in matched TMA and full-sections, ICC was 0.64 (95% CI 0.55-0.71) and Lin's concordance correlation coefficient was 0.63 (95% CI 0.55-0.71), corresponding to unacceptable agreement. There is acceptable inter-observer agreement of TILs assessment on full-sections but not TMAs and discrepancy between full-sections and TMAs. TMA preparation must include consideration for representation of both entire tumor area and tumor-microenvironment to correctly define prognostic and predictive values of potential immuno-related biomarkers.
Collapse
Affiliation(s)
- Frances Mary Reznitsky
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark.,Department of Pathology, Herlev and Gentofte Hospital, Herlev, Denmark
| | | | - Ann Knoop
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
16
|
Ben Dori S, Aizic A, Zubkov A, Tsuriel S, Sabo E, Hershkovitz D. The risk of PD-L1 expression misclassification in triple-negative breast cancer. Breast Cancer Res Treat 2022; 194:297-305. [PMID: 35622241 PMCID: PMC9239943 DOI: 10.1007/s10549-022-06630-3] [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: 01/11/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022]
Abstract
Purpose Stratification of patients with triple-negative breast cancer (TNBC) for anti-PD-L1 therapy is based on PD-L1 expression in tumor biopsies. This study sought to evaluate the risk of PD-L1 misclassification. Methods We conducted a high-resolution analysis on ten surgical specimens of TNBC. First, we determined PD-L1 expression pattern distribution via manual segmentation and measurement of 6666 microscopic clusters of positive PD-L1 immunohistochemical staining. Then, based on these results, we generated a computer model to calculate the effect of the positive PD-L1 fraction, aggregate size, and distribution of PD-L1 positive cells on the diagnostic accuracy. Results Our computer-based model showed that larger aggregates of PD-L1 positive cells and smaller biopsy size were associated with higher fraction of false results (P < 0.001, P < 0.001, respectively). Additionally, our model showed a significant increase in error rate when the fraction of PD-L1 expression was close to the cut-off (error rate of 12.1%, 0.84%, and 0.65% for PD-L1 positivity of 0.5–1.5%, ≤ 0.5% ,and ≥ 1.5%, respectively, P < 0.0001). Interestingly, false positive results were significantly higher than false negative results (0.51–22.62%, with an average of 6.31% versus 0.11–11.36% with an average of 1.58% for false positive and false negative results, respectively, P < 0.05). Furthermore, heterogeneous tumors with different aggregate sizes in the same tumor, were associated with increased rate of false results in comparison to homogenous tumors (P < 0.001). Conclusion Our model can be used to estimate the risk of PD-L1 misclassification in biopsies, with potential implications for treatment decisions. Supplementary Information The online version contains supplementary material available at 10.1007/s10549-022-06630-3.
Collapse
Affiliation(s)
- Shani Ben Dori
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Asaf Aizic
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel
| | - Asia Zubkov
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel
| | - Shlomo Tsuriel
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel
| | - Edmond Sabo
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Institute of Pathology, Carmel Medical Center, Haifa, Israel
| | - Dov Hershkovitz
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
17
|
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
|
18
|
Discordance of PD-L1 Expression at the Protein and RNA Levels in Early Breast Cancer. Cancers (Basel) 2021; 13:cancers13184655. [PMID: 34572882 PMCID: PMC8467035 DOI: 10.3390/cancers13184655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Despite the increasing use of checkpoint inhibitors for early and metastatic breast cancer, Programmed Death Ligand 1 (PD-L1) remains the only validated albeit imperfect predictive biomarker. Significant discordance in PD-L1 protein expression depending on the antibody used has been demonstrated, while the weak correlation and discordant prognostic information between protein and gene expression underscore its biologic heterogeneity. In this study, we use material from two patient cohorts of early breast cancer and multiple methodologies (immunohistochemistry, RNA fluorescent in situ hybridization, immunofluorescence, bulk gene expression, and multiplex fluorescent immunohistochemistry) to demonstrate the significant discordance in PD-L1 expression among various methods and between different areas of the same tumor, which hints toward the presence of spatial, intratumoral and biological heterogeneity. Abstract We aimed to assess if the discrepant prognostic information between Programmed Death Ligand 1 (PD-L1) protein versus mRNA expression in early breast cancer (BC) could be attributed to heterogeneity in its expression. PD-L1 protein and mRNA expression in BC tissue microarrays from two clinical patient cohorts were evaluated (105 patients; cohort 1: untreated; cohort 2: neoadjuvant chemotherapy-treated). Immunohistochemistry (IHC) with SP142, SP263 was performed. PD-L1 mRNA was evaluated using bulk gene expression and RNA-FISH RNAscope®, the latter scored in a semi-quantitative manner and combined with immunofluorescence (IF) staining for the simultaneous detection of PD-L1 protein expression. PD-L1 expression was assessed in cores as a whole and in two regions of interest (ROI) from the same core. The cell origin of PD-L1 expression was evaluated using multiplex fluorescent IHC. IHC PD-L1 expression between SP142 and SP263 was concordant in 86.7% of cores (p < 0.001). PD-L1 IF/IHC was weakly correlated with spatial mRNA expression (concordance 54.6–71.2%). PD-L1 was mostly expressed by lymphocytes intra-tumorally, while its stromal expression was mostly observed in macrophages. Our results demonstrate only moderate concordance between the various methods of assessing PD-L1 expression at the protein and mRNA levels, which may be attributed to both analytical performance and spatial heterogeneity.
Collapse
|
19
|
Pang JMB, Castles B, Byrne DJ, Button P, Hendry S, Lakhani SR, Sivasubramaniam V, Cooper WA, Armes J, Millar EK, Raymond W, Roberts-Thomson S, Kumar B, Burr M, Selinger C, Harvey K, Chan C, Beith J, Clouston D, O’Toole SA, Fox SB. SP142 PD-L1 Scoring Shows High Interobserver and Intraobserver Agreement in Triple-negative Breast Carcinoma But Overall Low Percentage Agreement With Other PD-L1 Clones SP263 and 22C3. Am J Surg Pathol 2021; 45:1108-1117. [PMID: 34232604 PMCID: PMC8277187 DOI: 10.1097/pas.0000000000001701] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
SP142 programmed cell death ligand 1 (PD-L1) status predicts response to atezolizumab in triple-negative breast carcinoma (TNBC). Prevalence of VENTANA PD-L1 (SP142) Assay positivity, concordance with the VENTANA PD-L1 (SP263) Assay and Dako PD-L1 IHC 22C3 pharmDx assay, and association with clinicopathologic features were assessed in 447 TNBCs. SP142 PD-L1 intraobserver and interobserver agreement was investigated in a subset of 60 TNBCs, with scores enriched around the 1% cutoff. The effect of a 1-hour training video on pretraining and posttraining scores was ascertained. At a 1% cutoff, 34.2% of tumors were SP142 PD-L1 positive. SP142 PD-L1 positivity was significantly associated with tumor-infiltrating lymphocytes (P <0.01), and node negativity (P=0.02), but not with tumor grade (P=0.35), tumor size (P=0.58), or BRCA mutation (P=0.53). Overall percentage agreement (OPA) for intraobserver and interobserver agreement was 95.0% and 93.7%, respectively, among 5 pathologists trained in TNBC SP142 PD-L1 scoring. In 5 TNBC SP142 PD-L1-naive pathologists, significantly higher OPA to the reference score was achieved after video training (posttraining OPA 85.7%, pretraining OPA 81.5%, P<0.05). PD-L1 status at a 1% cutoff was assessed by SP142 and SP263 in 420 cases, and by SP142 and 22C3 in 423 cases, with OPA of 88.1% and 85.8%, respectively. The VENTANA PD-L1 (SP142) Assay is reproducible for classifying TNBC PD-L1 status by trained observers; however, it is not analytically equivalent to the VENTANA PD-L1 (SP263) Assay and Dako PD-L1 IHC 22C3 pharmDx assay.
Collapse
Affiliation(s)
| | | | | | | | | | - Sunil R. Lakhani
- University of Queensland Centre for Clinical Research
- Pathology Queensland, Brisbane
| | | | - Wendy A. Cooper
- Sydney Medical School, The University of Sydney
- Department of Tissue Pathology, Royal Prince Alfred Hospital, NSW Health Pathology
- Western Sydney University, Campbelltown
| | - Jane Armes
- Pathology Queensland, Sunshine Coast, QLD
| | - Ewan K.A. Millar
- NSW Health Pathology, St George Hospital
- St. George and Sutherland Clinical School, University of New South Wales, Kogarah
| | - Wendy Raymond
- Flinders Medical Centre, Flinders University of South Australia
- Clinpath Laboratories, Adelaide, SA, Australia
| | | | | | - Marian Burr
- Royal Melbourne Hospital
- Sir Peter MacCallum Department of Oncology, University of Melbourne
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Cambridge, UK
| | | | - Kate Harvey
- The Garvan Institute of Medical Research, Darlinghurst
| | - Charles Chan
- Concord Clinical School, The University of Sydney, Sydney
- Concord Repatriation General Hospital, Concord, NSW
| | - Jane Beith
- Sydney Medical School, The University of Sydney
- Chris O’Brien Lifehouse, Camperdown
| | | | - Sandra A. O’Toole
- Sydney Medical School, The University of Sydney
- The Garvan Institute of Medical Research, Darlinghurst
- Department of Tissue Pathology, Royal Prince Alfred Hospital, NSW Health Pathology
- Western Sydney University, Campbelltown
| | - Stephen B. Fox
- Peter MacCallum Cancer Centre
- Sir Peter MacCallum Department of Oncology, University of Melbourne
| |
Collapse
|
20
|
Longitudinal assessment of PD-L1 expression and gene expression profiles in patients with head and neck cancer reveals temporal heterogeneity. Oral Oncol 2021; 119:105368. [PMID: 34111704 DOI: 10.1016/j.oraloncology.2021.105368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/29/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) is the most validated predictive biomarker used for the treatment of head and neck squamous cell carcinoma (HNSCC) with immune checkpoint inhibitors (ICI). Several gene expression-based signatures surrogate of the activation of IFN-gamma pathway and of the presence of tertiary lymphoid structures (TLS) have also been proposed as potential biomarkers. While they may have a potential therapeutic implication, the longitudinal changes of either PD-L1 or gene expression profiles between the initial and recurrent HNSCC lesions is unknown. METHODS PD-L1 immunohistochemistry (IHC) and targeted RNA-sequencing of 2,549 transcripts were analyzed on paired specimens from the initial diagnosis and recurrent HNSCC. PD-L1 status was defined using the combined positive score (CPS). PD-L1 mRNA levels were compared with protein expression levels by IHC. Enrichment scores of surrogate signatures for TLS and IFN-gamma (IFN-γ) pathway activation were computed using the single sample gene set enrichment analysis (ssGSEA). RESULTS PD-L1 status was 64% (21/33) concordant between the initial and recurrent lesions using a CPS 1 threshold and 67% (22/33) concordant using a CPS 20 threshold. CPS score was associated with PD-L1 gene expression levels. There was a 43% (15/35) and 66% (23/35) concordance for the IFN-γ and TLS signature scores, respectively. CONCLUSION Our study reveals temporal heterogeneity of PD-L1 status and TLS/IFN-γ gene expression surrogates in HNSCC that need to be considered when interpreting biomarker studies.
Collapse
|
21
|
Prognostic and Clinicopathologic Associations of LAG-3 Expression in Triple-negative Breast Cancer. Appl Immunohistochem Mol Morphol 2021; 30:62-71. [PMID: 34081635 DOI: 10.1097/pai.0000000000000954] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/04/2021] [Indexed: 11/26/2022]
Abstract
The immune checkpoint molecule lymphocyte activation gene 3 (LAG-3) is currently being investigated as a possible target for immunotherapy in triple-negative breast cancer (TNBC), frequently as an addition to treatment with programmed cell death protein 1/programmed death ligand 1 (PD-L1) inhibition. However, expression of LAG-3, the frequency of coexpression with PD-L1, and the prognostic significance of this marker have not been studied extensively in TNBC. For this study, tissue microarrays (TMAs) were constructed from surgical specimens of 514 patients with TNBC. TMAs were stained immunohistochemically for LAG-3 and PD-L1 expression. Tumor-infiltrating lymphocytes (TILs) were evaluated on full glass slides. LAG-3 expression was significantly associated with improved overall survival and relapse-free survival. When adjusted for clinicopathologic factors, each increment of 10 LAG-3-positive intratumoral lymphocytes per TMA core was associated with improved overall survival (hazard ratio=0.93, 95% confidence interval: 0.89-0.97, P=0.002), and recurrence-free survival (hazard ratio=0.91, 95% confidence interval: 0.85-0.97, P=0.002). PD-L1 expression on immune cells and PD-L1 expression evaluated with the combined positive score and TILs were also associated with improved survival in both univariate and multivariate analyses. PD-L1 expression on tumor cells was only associated with improved survival in univariate analysis. LAG-3 expression was associated with both TILs and PD-L1 expression. Coexpression of LAG-3 and PD-L1 did not confer additional survival benefits. In conclusion, LAG-3 expression is associated with improved survival in TNBC. LAG-3 is often coexpressed with PD-L1, confirming that TNBC is likely a suitable candidate for cotreatment with LAG-3 and programmed cell death protein 1/PD-L1 inhibitors. However, coexpression does not confer additional survival benefits.
Collapse
|
22
|
Determining Factors in the Therapeutic Success of Checkpoint Immunotherapies against PD-L1 in Breast Cancer: A Focus on Epithelial-Mesenchymal Transition Activation. J Immunol Res 2021; 2021:6668573. [PMID: 33506060 PMCID: PMC7808819 DOI: 10.1155/2021/6668573] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common neoplasm diagnosed in women around the world. Checkpoint inhibitors, targeting the programmed death receptor-1 or ligand-1 (PD-1/PD-L1) axis, have dramatically changed the outcome of cancer treatment. These therapies have been recently considered as alternatives for treatment of breast cancers, in particular those with the triple-negative phenotype (TNBC). A further understanding of the regulatory mechanisms of PD-L1 expression is required to increase the benefit of PD-L1/PD-1 checkpoint immunotherapy in breast cancer patients. In this review, we will compile the most recent studies evaluating PD-1/PD-L1 checkpoint inhibitors in breast cancer. We review factors that determine the therapeutic success of PD-1/PD-L1 immunotherapies in this pathology. In particular, we focus on pathways that interconnect the epithelial-mesenchymal transition (EMT) with regulation of PD-L1 expression. We also discuss the relationship between cellular metabolic pathways and PD-L1 expression that are involved in the promotion of resistance in TNBC.
Collapse
|
23
|
Zhou KI, Peterson B, Serritella A, Thomas J, Reizine N, Moya S, Tan C, Wang Y, Catenacci DVT. Spatial and Temporal Heterogeneity of PD-L1 Expression and Tumor Mutational Burden in Gastroesophageal Adenocarcinoma at Baseline Diagnosis and after Chemotherapy. Clin Cancer Res 2020; 26:6453-6463. [PMID: 32820017 PMCID: PMC7744325 DOI: 10.1158/1078-0432.ccr-20-2085] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Intrapatient heterogeneity of programmed death ligand 1 (PD-L1) expression and tumor mutational burden (TMB) in gastroesophageal adenocarcinoma (GEA) could influence their roles as predictive biomarkers for response to immune checkpoint inhibitors (ICI). In this retrospective analysis, we evaluated the spatiotemporal heterogeneity and prognostic relevance of PD-L1 expression and TMB in GEA. EXPERIMENTAL DESIGN A cohort of 211 patients with stage II-IV GEA was retrospectively reviewed for a total of 407 tumor samples with PD-L1 expression data and 319 tumor samples with TMB data. PD-L1 status was defined as positive if combined positive score (CPS) ≥1 using the 22C3 pharmDx assay. TMB levels were categorized as low, intermediate, or high (≤5, 5-15, or >15 mutations/Mb), or using a single threshold (<10 or ≥10 mutation/Mb), determined by next-generation sequencing using a targeted gene panel. RESULTS Of 407 tumors, 56% were PD-L1 negative and 44% PD-L1 positive. Of 319 tumors, 50% were TMB-low, 45% TMB-intermediate, and 5% TMB-high; 86% had <10 and 14% ≥10 mutations/Mb. TMB level was significantly associated with MSI-status. PD-L1 expression and TMB exhibited marked spatial heterogeneity between baseline primary and metastatic tumors (61% and 69% concordance), and temporal heterogeneity between tumors before and after chemotherapy (57%-63% and 73%-75% concordance). PD-L1 expression and TMB were not significantly associated with overall survival. CONCLUSIONS PD-L1 expression and TMB exhibit marked spatial and temporal heterogeneity in GEA. This heterogeneity should be considered when obtaining tumor samples for molecular testing and when deciding whether ICI therapy is appropriate.See related commentary by Klempner et al., p. 6401.
Collapse
Affiliation(s)
- Katherine I Zhou
- Medical Scientist Training Program, University of Chicago, Chicago, Illinois
| | - Bryan Peterson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Anthony Serritella
- Department of Medicine, University of Chicago Medical Center, Chicago, Illinois
| | - Joseph Thomas
- Department of Medicine, University of Chicago Medical Center, Chicago, Illinois
| | - Natalie Reizine
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Stephanie Moya
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Carol Tan
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Yan Wang
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Daniel V T Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.
| |
Collapse
|
24
|
Sood R, Kumar S, Laroiya I, Khare S, Das A, Singh G, Bal A. Assessment of PD-L1 expression and tumor-infiltrating lymphocytes (TILs) across molecular subtypes of triple-negative breast cancer. Breast J 2020; 26:2424-2427. [PMID: 33314356 DOI: 10.1111/tbj.14110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 01/27/2023]
Affiliation(s)
- Ridhi Sood
- Departments of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sandeep Kumar
- Departments of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ishita Laroiya
- Departments of General Surgery, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Siddhant Khare
- Departments of General Surgery, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ashim Das
- Departments of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Gurpreet Singh
- Departments of General Surgery, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amanjit Bal
- Departments of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
25
|
Sinclair W, Kobalka P, Ren R, Beshai B, Lott Limbach AA, Wei L, Mei P, Li Z. Interobserver agreement in programmed cell death-ligand 1 immunohistochemistry scoring in nonsmall cell lung carcinoma cytologic specimens. Diagn Cytopathol 2020; 49:219-225. [PMID: 33104298 DOI: 10.1002/dc.24651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/28/2020] [Accepted: 10/14/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The evaluation of PD-L1 expression in nonsmall cell lung carcinoma (NSCLC) is becoming increasingly important given the effectiveness of PD-L1 inhibitors. Although cytologic specimens have been shown to be compatible with surgical specimens to evaluate PD-L1 immunohistochemistry (IHC), evidence of the reproducibility of PD-L1 in cytologic specimens is lacking. The aim of this study is to evaluate interobserver agreement in PD-L1 IHC in cytologic specimens. METHODS PD-L1 IHC was performed on 86 NSCLC cytology specimens using Dako PD-L1 IHC 22C3 pharmDx. The digitally scanned whole slide images (WSI) were read by five pathologists. Each case was given a Tumor Proportion Score (TPS) and the results were compared between the observers. The interobserver concordance was assessed using 1% and 50% as cutoffs. RESULTS TPSs were highly correlated among observers (Spearman correlation coefficient, 0.86-0.94). Using greater than 1% as a cutoff, interobserver agreement measured by Fleiss Kappa was 0.74 for all pathologists and Cohen's Kappa coefficient ranged from 0.49 to 0.83, consistent with moderate to substantial agreement. With a cutoff of greater than 50%, Fleiss Kappa was 0.79 for all pathologists and the kappa values ranged from 0.63 to 0.90, consistent with substantial to almost perfect agreement. Several pitfalls were identified by reviewing discordant cases, including staining in macrophages, stromal cells, and intratumoral heterogeneity. CONCLUSION Our data suggest that TPS of PD-L1 IHC on cytology specimens is reproducible, with a better agreement when using 50% as the cutoff value. However, special attention is required when the TPS is near the 1% cutoff.
Collapse
Affiliation(s)
- William Sinclair
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, Ohio, USA
| | - Peter Kobalka
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, Ohio, USA
| | - Rongqin Ren
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, Ohio, USA
| | - Boulos Beshai
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Abberly A Lott Limbach
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, Ohio, USA
| | - Lai Wei
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ping Mei
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zaibo Li
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
26
|
Zhai X, Yang Z, Liu X, Dong Z, Zhou D. Identification of NUF2 and FAM83D as potential biomarkers in triple-negative breast cancer. PeerJ 2020; 8:e9975. [PMID: 33005492 PMCID: PMC7513746 DOI: 10.7717/peerj.9975] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Background Breast cancer is a heterogeneous disease. Compared with other subtypes of breast cancer, triple-negative breast cancer (TNBC) is easy to metastasize and has a short survival time, less choice of treatment options. Here, we aimed to identify the potential biomarkers to TNBC diagnosis and prognosis. Material/Methods Three independent data sets (GSE45827, GSE38959, GSE65194) were downloaded from the Gene Expression Omnibus (GEO). The R software packages were used to integrate the gene profiles and identify differentially expressed genes (DEGs). A variety of bioinformatics tools were used to explore the hub genes, including the DAVID database, STRING database and Cytoscape software. Reverse transcription quantitative PCR (RT-qPCR) was used to verify the hub genes in 14 pairs of TNBC paired tissues. Results In this study, we screened out 161 DEGs between 222 non-TNBC and 126 TNBC samples, of which 105 genes were up-regulated and 56 were down-regulated. These DEGs were enriched for 27 GO terms and two pathways. GO analysis enriched mainly in “cell division”, “chromosome, centromeric region” and “microtubule motor activity”. KEGG pathway analysis enriched mostly in “Cell cycle” and “Oocyte meiosis”. PPI network was constructed and then 10 top hub genes were screened. According to the analysis results of the Kaplan-Meier survival curve, the expression levels of only NUF2, FAM83D and CENPH were associated with the recurrence-free survival in TNBC samples (P < 0.05). RT-qPCR confirmed that the expression levels of NUF2 and FAM83D in TNBC tissues were indeed up-regulated significantly. Conclusions The comprehensive analysis showed that NUF2 and FAM83D could be used as potential biomarkers for diagnosis and prognosis of TNBC.
Collapse
Affiliation(s)
- Xiuming Zhai
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaowei Yang
- Department of Breast and Thyroid, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xiji Liu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zihe Dong
- Department of Laboratory Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Dandan Zhou
- Department of Laboratory Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|