1
|
Schweihofer V, Bruss C, Seitz S, Glehr G, Hetterich M, Weber F, Hatzipanagiotou M, Álvarez MFP, Ortmann O, Brockhoff G, Bauer RJ, Wege AK. Breast cancer scoring based on a multiplexed profiling of soluble and cell-associated (immune) markers facilitates the prediction of pembrolizumab therapy. Cancer Cell Int 2025; 25:120. [PMID: 40148963 PMCID: PMC11948714 DOI: 10.1186/s12935-025-03729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The immune checkpoint targeting is nowadays an integral part of cancer therapies. However, only a minority of patients experience long-term benefits. Thus, the identification of predictive biomarkers contributing to therapy response is urgently needed. METHODS Here, we analyzed different immune and tumor specific expression and secretion profiles in the peripheral blood and tumor samples of 50 breast cancer patients by multicolor flow cytometry and bead-based immunoassays at the time of diagnosis. Due to individual phenotype variations, we quantitatively scored 25 expressed and secreted immune-associated (e.g., LAG-3, PD-1, TIM-3, CD27) and tumor relevant markers (e.g., PD-L1, CD44, MHC-I, MHC-II) in immune checkpoint-treated triple negative breast cancer patients based on the current literature. The calculated score divided the patients into individuals with predicted pCR (total score of > 0) or predicted residual disease (total score of ≤ 0). At the end of the neoadjuvant therapy, the truly achieved pathological complete response (pCR; end of observation) was determined. RESULTS The calculated score was 79% in accordance with the achieved pCR at the time of surgery. Moreover, the sensitivity was 83.3%, the specificity 76.9%, the positive predictive value 62.5%, and the negative predictive value 90.9%. In addition, we identified a correlation of PD-1 and LAG-3 expression between tumor-associated and peripheral immune cells, which was independent of the subtype. Overall, PD-1 was the most frequently expressed checkpoint. However, in a number of patient-derived tumors, additional checkpoints as LAG-3 and TIM-3 were substantially (co-)expressed, which potentially compromises anti-PD-(L)1 mono-therapy. CONCLUSIONS This study represents a proof-of-principle to identify potential checkpoint therapy responders in advance at the time of diagnosis. The work was based on a scoring derived from a multiplexed marker profiling. However, larger patient cohorts need to be prospectively evaluated for further validation.
Collapse
Affiliation(s)
- Verena Schweihofer
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Christina Bruss
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Gunther Glehr
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Madeleine Hetterich
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Maria Hatzipanagiotou
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Miriam Fernández-Pacheco Álvarez
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Richard J Bauer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Anja Kathrin Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany.
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany.
| |
Collapse
|
2
|
Tiedje V, Greenberg J, Qin T, Im SY, Krishnamoorthy GP, Boucai L, Xu B, French JD, Sherman EJ, Ho AL, de Stanchina E, Socci ND, Jin J, Ghossein R, Knauf JA, Koche RP, Fagin JA. Loss of tumor cell MHC Class II drives insensitivity of BRAF-mutant anaplastic thyroid cancers to MAPK inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635086. [PMID: 40093098 PMCID: PMC11908150 DOI: 10.1101/2025.01.27.635086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Cancer cells present neoantigens dominantly through MHC class I (MHCI) to drive tumor rejection through cytotoxic CD8+ T-cells. There is growing recognition that a subset of tumors express MHC class II (MHCII), causing recognition of antigens by TCRs of CD4+ T-cells that contribute to the anti-tumor response. We find that mouse Braf V600E -driven anaplastic thyroid cancers (ATC) respond markedly to the RAF + MEK inhibitors dabrafenib and trametinib (dab/tram) and that this is associated with upregulation of MhcII in cancer cells and increased CD4+ T-cell infiltration. A subset of recurrent tumors lose MhcII expression due to silencing of Ciita , the master transcriptional regulator of MhcII, despite preserved interferon gamma signal transduction, which can be rescued by EZH2 inhibition. Orthotopically-implanted Ciita -/- and H2-Ab1 -/- ATC cells into immune competent mice become unresponsive to the MAPK inhibitors. Moreover, depletion of CD4+, but not CD8+ T-cells, also abrogates response to dab/tram. These findings implicate MHCII-driven CD4+ T cell activation as a key determinant of the response of Braf-mutant ATCs to MAPK inhibition.
Collapse
|
3
|
Friedenson B. Identifying Safeguards Disabled by Epstein-Barr Virus Infections in Genomes From Patients With Breast Cancer: Chromosomal Bioinformatics Analysis. JMIRX MED 2025; 6:e50712. [PMID: 39885374 PMCID: PMC11796484 DOI: 10.2196/50712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 02/01/2025]
Abstract
Background The causes of breast cancer are poorly understood. A potential risk factor is Epstein-Barr virus (EBV), a lifelong infection nearly everyone acquires. EBV-transformed human mammary cells accelerate breast cancer when transplanted into immunosuppressed mice, but the virus can disappear as malignant cells reproduce. If this model applies to human breast cancers, then they should have genome damage characteristic of EBV infection. Objective This study tests the hypothesis that EBV infection predisposes one to breast cancer by causing permanent genome damage that compromises cancer safeguards. Methods Publicly available genome data from approximately 2100 breast cancers and 25 ovarian cancers were compared to cancers with proven associations to EBV, including 70 nasopharyngeal cancers, 90 Burkitt lymphomas, 88 diffuse large B-cell lymphomas, and 34 gastric cancers. Calculation algorithms to make these comparisons were developed. Results Chromosome breakpoints in breast and ovarian cancer clustered around breakpoints in EBV-associated cancers. Breakpoint distributions in breast and EBV-associated cancers on some chromosomes were not confidently distinguished (P>.05), but differed from controls unrelated to EBV infection. Viral breakpoint clusters occurred in high-risk, sporadic, and other breast cancer subgroups. Breakpoint clusters disrupted gene functions essential for cancer protection, which remain compromised even if EBV infection disappears. As CRISPR (clustered regularly interspaced short palindromic repeats)-like reminders of past infection during evolution, EBV genome fragments were found regularly interspaced between Piwi-interacting RNA (piRNA) genes on chromosome 6. Both breast and EBV-associated cancers had inactivated genes that guard piRNA defenses and the major histocompatibility complex (MHC) locus. Breast and EBV-associated cancer breakpoints and other variations converged around the highly polymorphic MHC. Not everyone develops cancer because MHC differences produce differing responses to EBV infection. Chromosome shattering and mutation hot spots in breast cancers preferentially occurred at incorporated viral sequences. On chromosome 17, breast cancer breakpoints that clustered around those in EBV-mediated cancers were linked to estrogen effects. Other breast cancer breaks affected sites where EBV inhibits JAK-STAT and SWI-SNF signaling pathways. A characteristic EBV-cancer gene deletion that shifts metabolism to favor tumors was also found in breast cancers. These changes push breast cancer into metastasis and then favor survival of metastatic cells. Conclusions EBV infection predisposes one to breast cancer and metastasis, even if the virus disappears. Identifying this pathogenic viral damage may improve screening, treatment, and prevention. Immunizing children against EBV may protect against breast, ovarian, other cancers, and potentially even chronic unexplained diseases.
Collapse
Affiliation(s)
- Bernard Friedenson
- Department of Biochemistry and Medical Genetics, Cancer Center, University of Illinois Chicago, 900 s Ashland, Chicago, IL, 60617, United States, 1 8479124216
| |
Collapse
|
4
|
Tian J, Li Y, Tong Y, Zhang Y, Zhao T, Kang Y, Bi Q. Uridine-cytidine kinase 2 is correlated with immune, DNA damage repair and promotion of cancer stemness in pan-cancer. Front Oncol 2025; 15:1503300. [PMID: 39931080 PMCID: PMC11807824 DOI: 10.3389/fonc.2025.1503300] [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: 09/28/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
Background UCK2 (Uridine-Cytidine Kinase 2) is a promising prognostic marker for malignant tumors, but its association with immune infiltration and cancer stemness in pan-cancer remains to be fully understood. we find that gene UCK2 is closed related to RNA stemness scores (RNAss) and DNA stemness scores (DNAss), which is measured the tumor stemness. We also discover an association between UCK2 expression and immune cells by CIBERSORT algorithm, ESTIMATE algorithm and ssGSEA algorithm, especially, related to T cell, monocytes, mast cells, and macrophages. This study aims to shed light on the role and possible mechanism of UCK2 in pan-cancer. Methods We used the R programming language for pan-cancer bulk sequencing data analysis, which were obtained from the University of California, Santa Cruz (UCSC) datasets. UCSC database is a very useful for explore data from TCGA and other cancer genomics datasets, The data we explored at the UCK2 transcriptome level came from TCGA data in the UCSC database. We explored differential UCK2 expression between tumor and normal samples. Immunohistochemistry (IHC) was utilized to validate the expression of UCK2 in different types cancers using tumor tissue chips. The correlations of UCK2 with prognosis, genetic instability, DNA repair, cancer stem cell characteristics, and immune cell infiltration were investigated. Furthermore, single-cell datasets, acquired from the Gene Expression Omnibus (GEO) database, were used to validate the relationship between UCK2 and immune cells. GEO is a famous public genomics database supporting freely disseminates microarray data. Finally, we analyzed the correlation between UCK2 and drug sensitivity. Results UCK2 expression was observed to be high in most cancers and was remarkably related to the prognosis of pan-cancers. We found that the increased UCK2 expression was associated with higher genetic instability. Additionally, positive relationships were observed between UCK2 expression and mismatch repair genes, homologous recombination repair genes, and cancer stemness across different cancer types. There were significant correlations between UCK2 and T cells, monocytes, mast cells, and macrophages. Moreover, as expected, the immune checkpoint human leucocyte antigen (HLA) was found to be negatively related to UCK2. Similarly, UCK2 was also observed to have a negative association with major histocompatibility complex (MHC) genes. We noted that UCK2 had significant correlations with the sensitivity to various anti-cancer drug. Conclusion We have observed that UCK2 plays pivotal roles in prognosis and tumor immunity, and it is associated with DNA repair and cancer stemness. The UCK2 gene exhibits a strong correlation with the immune checkpoints HLA. This study highlights its potential impact on drug sensitivity.
Collapse
Affiliation(s)
- Jinlong Tian
- Graduate School of Bengbu Medical University, Bengbu, Anhui, China
| | - Yanlei Li
- Graduate School of Bengbu Medical University, Bengbu, Anhui, China
| | - Yu Tong
- Sports Medicine Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yuan Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou, China
| | - Tingxiao Zhao
- Sports Medicine Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yao Kang
- Sports Medicine Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Qing Bi
- Graduate School of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
5
|
Cui Y, Zhang W, Zeng X, Yang Y, Park SJ, Nakai K. Computational analysis of the functional impact of MHC-II-expressing triple-negative breast cancer. Front Immunol 2024; 15:1497251. [PMID: 39664386 PMCID: PMC11631845 DOI: 10.3389/fimmu.2024.1497251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in tumor progression and immunoregulation. Major histocompatibility complex class II (MHC-II) is essential for immune surveillance within the TME. While MHC-II genes are typically expressed by professional antigen-presenting cells, they are also expressed in tumor cells, potentially facilitating antitumor immune responses. To understand the role of MHC-II-expressing tumor cells, we analyzed triple-negative breast cancer (TNBC), an aggressive subtype with poor prognosis and limited treatment options, using public bulk RNA-seq, single-cell RNA-seq, and spatial transcriptomics datasets. Our analysis revealed a distinct tumor subpopulation that upregulates MHC-II genes and actively interacts with immune cells. We implicated that this subpopulation is preferentially present in proximity to regions in immune infiltration of TNBC patient cohorts with a better prognosis, suggesting the functional importance of MHC-II-expressing tumor cells in modulating the immune landscape and influencing patient survival outcomes. Remarkably, we identified a prognostic signature comprising 40 significant genes in the MHC-II-expressing tumors in which machine leaning models with the signature successfully predicted patient survival outcomes and the degree of immune infiltration. This study advances our understanding of the immunological basis of cancer progression and suggests promising new directions for therapeutic strategies.
Collapse
Affiliation(s)
- Yang Cui
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Weihang Zhang
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Xin Zeng
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Yitao Yang
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Sung-Joon Park
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Zeng Q, Zeng S, Dai X, Ding Y, Huang C, Ruan R, Xiong J, Tang X, Deng J. MDM2 inhibitors in cancer immunotherapy: Current status and perspective. Genes Dis 2024; 11:101279. [PMID: 39263534 PMCID: PMC11388719 DOI: 10.1016/j.gendis.2024.101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 09/13/2024] Open
Abstract
Murine double minute 2 (MDM2) plays an essential role in the cell cycle, apoptosis, DNA repair, and oncogene activation through p53-dependent and p53-independent signaling pathways. Several preclinical studies have shown that MDM2 is involved in tumor immune evasion. Therefore, MDM2-based regulation of tumor cell-intrinsic immunoregulation and the immune microenvironment has attracted increasing research attention. In recent years, immune checkpoint inhibitors targeting PD-1/PD-L1 have been widely used in the clinic. However, the effectiveness of a single agent is only approximately 20%-40%, which may be related to primary and secondary drug resistance caused by the dysregulation of oncoproteins. Here, we reviewed the role of MDM2 in regulating the immune microenvironment, tumor immune evasion, and hyperprogression during immunotherapy. In addition, we summarized preclinical and clinical findings on the use of MDM2 inhibitors in combination with immunotherapy in tumors with MDM2 overexpression or amplification. The results reveal that the inhibition of MDM2 could be a promising strategy for enhancing immunotherapy.
Collapse
Affiliation(s)
- Qinru Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Shaocheng Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Xiaofeng Dai
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Yun Ding
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Xiaomei Tang
- Department of Oncology, Jiangxi Chest Hospital, Nanchang, Jiangxi 330006, China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
7
|
Naskar S, Sriraman N, Sarkar A, Mahajan N, Sarkar K. Tumor antigen presentation and the associated signal transduction during carcinogenesis. Pathol Res Pract 2024; 261:155485. [PMID: 39088877 DOI: 10.1016/j.prp.2024.155485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Numerous developments have been achieved in the study and treatment of cancer throughout the decades that it has been common. After decades of research, about 100 different kinds of cancer have been found, each with unique subgroups within certain organs. This has significantly expanded our understanding of the illness. A mix of genetic, environmental, and behavioral variables contribute to the complicated and diverse process of cancer formation. Mutations, or changes in the DNA sequence, are crucial to the development of cancer. These mutations have the ability to downregulate the expression and function of Major Histocompatibility Complex class I (MHC I) and MHCII receptors, as well as activate oncogenes and inactivate tumor suppressor genes. Cancer cells use this tactic to avoid being recognized by cytotoxic CD8+T lymphocytes, which causes issues with antigen presentation and processing. This review goes into great length into the PI3K pathway, changes to MHC I, and positive impacts of tsMHC-II on disease-free survival and overall survival and the involvement of dendritic cells (DCs) in different tumor microenvironments. The vital functions that the PI3K pathway and its link to the mTOR pathway are highlighted and difficulties in developing effective cancer targeted therapies and feedback systems has also been mentioned, where resistance mechanisms include RAS-mediated oncogenic changes and active PI3K signalling.
Collapse
Affiliation(s)
- Sohom Naskar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nawaneetan Sriraman
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ankita Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nitika Mahajan
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
8
|
Almeida-Nunes DL, Nunes M, Osório H, Ferreira V, Lobo C, Monteiro P, Abreu MH, Bartosch C, Silvestre R, Dinis-Oliveira RJ, Ricardo S. Ovarian cancer ascites proteomic profile reflects metabolic changes during disease progression. Biochem Biophys Rep 2024; 39:101755. [PMID: 38974022 PMCID: PMC11225207 DOI: 10.1016/j.bbrep.2024.101755] [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: 04/17/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 07/09/2024] Open
Abstract
Ovarian cancer (OC) patients develop ascites, an accumulation of ascitic fluid in the peritoneal cavity anda sign of tumour dissemination within the peritoneal cavity. This body fluid is under-researched, mainly regarding the ascites formed during tumour progression that have no diagnostic value and, therefore, are discarded. We performed a discovery proteomics study to identify new biomarkers in the ascites supernatant of OC patients. In this preliminary study, we analyzed a small amount of OC ascites to highlight the importance of not discarding such biological material during treatment, which could be valuable for OC management. Our findings reveal that OC malignant ascitic fluid (MAF) displays a proliferative environment that promotes the growth of OC cells that shift the metabolic pathway using alternative sources of nutrients, such as the cholesterol pathway. Also, OC ascites drained from patients during treatment showed an immunosuppressive environment, with up-regulation of proteins from the signaling pathways of IL-4 and IL-13 and down-regulation from the MHC-II. This preliminary study pinpointed a new protein (Transmembrane Protein 132A) in the OC context that deserves to be better explored in a more extensive cohort of patients' samples. The proteomic profile of MAF from OC patients provides a unique insight into the metabolic kinetics of cancer cells during disease progression, and this information can be used to develop more effective treatment strategies.
Collapse
Affiliation(s)
- Diana Luísa Almeida-Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
| | - Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313, Porto, Portugal
| | - Hugo Osório
- Proteomics Scientific Platform, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine from University of Porto (FMUP), 4200-319, Porto, Portugal
| | - Verónica Ferreira
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Cláudia Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Paula Monteiro
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Miguel Henriques Abreu
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Carla Bartosch
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (CI-IPO-Porto) / Health Research Network (RISE@CI-IPO-Porto), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine from University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- UCIBIO - Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
- FOREN – Forensic Science Experts, Dr. Mário Moutinho Avenue, No. 33-A, 1400-136, Lisbon, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
| |
Collapse
|
9
|
Tsutsumi E, Macy AM, LoBello J, Hastings KT, Kim S. Tumor immune microenvironment permissive to metastatic progression of ING4-deficient breast cancer. PLoS One 2024; 19:e0304194. [PMID: 38968186 PMCID: PMC11226078 DOI: 10.1371/journal.pone.0304194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/20/2024] [Indexed: 07/07/2024] Open
Abstract
Deficiencies in the ING4 tumor suppressor are associated with advanced stage tumors and poor patient survival in cancer. ING4 was shown to inhibit NF-kB in several cancers. As NF-kB is a key mediator of immune response, the ING4/NF-kB axis is likely to manifest in tumor-immune modulation but has not been investigated. To characterize the tumor immune microenvironment associated with ING4-deficient tumors, three approaches were employed in this study: First, tissue microarrays composed of 246 primary breast tumors including 97 ING4-deficient tumors were evaluated for the presence of selective immune markers, CD68, CD4, CD8, and PD-1, using immunohistochemical staining. Second, an immune-competent mouse model of ING4-deficient breast cancer was devised utilizing CRISPR-mediated deletion of Ing4 in a Tp53 deletion-derived mammary tumor cell line; mammary tumors were evaluated for immune markers using flow cytometry. Lastly, the METABRIC gene expression dataset was evaluated for patient survival related to the immune markers associated with Ing4-deleted tumors. The results showed that CD68, CD4, CD8, or PD-1, was not significantly associated with ING4-deficient breast tumors, indicating no enrichment of macrophages, T cells, or exhausted T cell types. In mice, Ing4-deleted mammary tumors had a growth rate comparable to Ing4-intact tumors but showed increased tumor penetrance and metastasis. Immune marker analyses of Ing4-deleted tumors revealed a significant increase in tumor-associated macrophages (Gr-1loCD11b+F4/80+) and a decrease in granzyme B-positive (GzmB+) CD4+ T cells, indicating a suppressive and/or less tumoricidal immune microenvironment. The METABRIC data analyses showed that low expression of GZMB was significantly associated with poor patient survival, as was ING4-low expression, in the basal subtype of breast cancer. Patients with GZMB-low/ING4-low tumors had the worst survival outcomes (HR = 2.80, 95% CI 1.36-5.75, p = 0.0004), supportive of the idea that the GZMB-low immune environment contributes to ING4-deficient tumor progression. Collectively, the study results demonstrate that ING4-deficient tumors harbor a microenvironment that contributes to immune evasion and metastasis.
Collapse
Affiliation(s)
- Emily Tsutsumi
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, Arizona, United States of America
| | - Anne M. Macy
- Department of Dermatology, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona, United States of America
| | - Janine LoBello
- Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Karen T. Hastings
- Department of Dermatology, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona, United States of America
| | - Suwon Kim
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, Arizona, United States of America
| |
Collapse
|
10
|
Velazquez-Caldelas TE, Zamora-Fuentes JM, Hernandez-Lemus E. Coordinated inflammation and immune response transcriptional regulation in breast cancer molecular subtypes. Front Immunol 2024; 15:1357726. [PMID: 38983850 PMCID: PMC11231215 DOI: 10.3389/fimmu.2024.1357726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
Breast cancer, characterized by its complexity and diversity, presents significant challenges in understanding its underlying biology. In this study, we employed gene co-expression network analysis to investigate the gene composition and functional patterns in breast cancer subtypes and normal breast tissue. Our objective was to elucidate the detailed immunological features distinguishing these tumors at the transcriptional level and to explore their implications for diagnosis and treatment. The analysis identified nine distinct gene module clusters, each representing unique transcriptional signatures within breast cancer subtypes and normal tissue. Interestingly, while some clusters exhibited high similarity in gene composition between normal tissue and certain subtypes, others showed lower similarity and shared traits. These clusters provided insights into the immune responses within breast cancer subtypes, revealing diverse immunological functions, including innate and adaptive immune responses. Our findings contribute to a deeper understanding of the molecular mechanisms underlying breast cancer subtypes and highlight their unique characteristics. The immunological signatures identified in this study hold potential implications for diagnostic and therapeutic strategies. Additionally, the network-based approach introduced herein presents a valuable framework for understanding the complexities of other diseases and elucidating their underlying biology.
Collapse
Affiliation(s)
| | | | - Enrique Hernandez-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
11
|
Xiang K, Zhang M, Yang B, Liu X, Wang Y, Liu H, Song Y, Yuan Y, Zhang L, Wen T, Zhang GW. TM-Score predicts immunotherapy efficacy and improves the performance of the machine learning prognostic model in gastric cancer. Int Immunopharmacol 2024; 134:112224. [PMID: 38723370 DOI: 10.1016/j.intimp.2024.112224] [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: 10/29/2023] [Revised: 04/13/2024] [Accepted: 05/05/2024] [Indexed: 06/03/2024]
Abstract
Immunotherapy is becoming increasingly important, but the overall response rate is relatively low in the treatment of gastric cancer (GC). The application of tumor mutational burden (TMB) in predicting immunotherapy efficacy in GC patients is limited and controversial, emphasizing the importance of optimizing TMB-based patient selection. By combining TMB and major histocompatibility complex (MHC) related hub genes, we established a novel TM-Score. This score showed superior performance for immunotherapeutic selection (AUC = 0.808) compared to TMB, MSI status, and EBV status. Additionally, it predicted the prognosis of GC patients. Subsequently, a machine learning model adjusted by the TM-Score further improved the accuracy of survival prediction (AUC > 0.8). Meanwhile, we found that GC patients with low TM-Score had a higher mutation frequency, higher expression of HLA genes and immune checkpoint genes, and higher infiltration of CD8+ T cells, CD4+ helper T cells, and M1 macrophages. This suggests that TM-Score is significantly associated with tumor immunogenicity and tumor immune environment. Notably, based on the RNA-seq and scRNA-seq, it was found that AKAP5, a key component gene of TM-Score, is involved in anti-tumor immunity by promoting the infiltration of CD4+ T cells, NK cells, and myeloid cells. Additionally, siAKAP5 significantly reduced MHC-II mRNA expression in the GC cell line. In addition, our immunohistochemistry assays confirmed a positive correlation between AKAP5 and human leukocyte antigen (HLA) expression. Furthermore, AKAP5 levels were higher in patients with longer survival and those who responded to immunotherapy in GC, indicating its potential value in predicting prognosis and immunotherapy outcomes. In conclusion, TM-Score, as an optimization of TMB, is a more precise biomarker for predicting the immunotherapy efficacy of the GC population. Additionally, AKAP5 shows promise as a therapeutic target for GC.
Collapse
Affiliation(s)
- Kanghui Xiang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Minghui Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bowen Yang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Liu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yusi Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hengxin Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yujia Song
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yonghui Yuan
- Liaoning Cancer Hospital & Institute, Clinical Research Center for Malignant Tumor of Liaoning Province, Cancer Hospital of China Medical University, Shenyang, China
| | - Lingyun Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China; Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Guang-Wei Zhang
- Smart Hospital Management Department, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
12
|
Wang B, Liu Y, Xiong F, Wang C. Improved Immunotherapy Outcomes via Cuproptosis Upregulation of HLA-DRA Expression: Promoting the Aggregation of CD4 + and CD8 +T Lymphocytes in Clear Cell Renal Cell Carcinoma. Pharmaceuticals (Basel) 2024; 17:678. [PMID: 38931345 PMCID: PMC11206763 DOI: 10.3390/ph17060678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024] Open
Abstract
Immunotherapy has shown promising clinical results in clear cell renal cell carcinoma (ccRCC), but low clinical target response rates due to dysfunction of the major histocompatibility complex (MHC) and an inhibitory tumor immune microenvironment (TIME) have largely limited the associated clinical benefits. In the present study, we explored the feasibility of enhancing tumor-specific-MHC-II-HLA-DRA expression, counteracting the TIME's suppressive effects, thereby improving the sensitivity of immune checkpoint inhibitor (ICI) therapy from the standpoint of cuproptosis. Immunohistochemical staining and in vitro experiments validated the expression of HLA-DRA in ccRCC and its positive impact on ICI therapy. Subsequently, we observed that cuproptosis upregulated HLA-DRA expression in a dose-dependent manner, further confirming the link between cuproptosis and HLA-DRA. In vivo experiments showed that cuproptosis increased the sensitivity to ICI treatment, and implementing cuproptosis alongside anti-PD-1 treatment curtailed tumor growth. Mechanistically, cuproptosis upregulates HLA-DRA expression at the transcriptional level in a dose-dependent manner by inducing the production of reactive oxygen species; high levels of HLA-DRA promote the expression of chemokines CCL5, CXCL9, and CXCL10 in the TIME, inhibiting the development of a pro-tumor microenvironment by promoting the infiltration of CD4+T and CD8+T cells, thereby synergizing ICI therapy and exerting anti-tumor effects. Taken together, this work highlights the role of cuproptosis in mediating TIME remodeling and synergistic immunotherapy, providing new evidence that cuproptosis can evoke effective anti-tumor immune responses.
Collapse
Affiliation(s)
| | | | | | - Chunyang Wang
- Urology Surgery Department, The First Affiliated Hospital of Harbin Medical University, Youzheng Street #37, Nangang District, Harbin 150001, China
| |
Collapse
|
13
|
Sammut SJ, Galson JD, Minter R, Sun B, Chin SF, De Mattos-Arruda L, Finch DK, Schätzle S, Dias J, Rueda OM, Seoane J, Osbourn J, Caldas C, Bashford-Rogers RJM. Predictability of B cell clonal persistence and immunosurveillance in breast cancer. Nat Immunol 2024; 25:916-924. [PMID: 38698238 PMCID: PMC11065701 DOI: 10.1038/s41590-024-01821-0] [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: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 05/05/2024]
Abstract
B cells and T cells are important components of the adaptive immune system and mediate anticancer immunity. The T cell landscape in cancer is well characterized, but the contribution of B cells to anticancer immunosurveillance is less well explored. Here we show an integrative analysis of the B cell and T cell receptor repertoire from individuals with metastatic breast cancer and individuals with early breast cancer during neoadjuvant therapy. Using immune receptor, RNA and whole-exome sequencing, we show that both B cell and T cell responses seem to coevolve with the metastatic cancer genomes and mirror tumor mutational and neoantigen architecture. B cell clones associated with metastatic immunosurveillance and temporal persistence were more expanded and distinct from site-specific clones. B cell clonal immunosurveillance and temporal persistence are predictable from the clonal structure, with higher-centrality B cell antigen receptors more likely to be detected across multiple metastases or across time. This predictability was generalizable across other immune-mediated disorders. This work lays a foundation for prioritizing antibody sequences for therapeutic targeting in cancer.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/immunology
- B-Lymphocytes/immunology
- Immunologic Surveillance
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- T-Lymphocytes/immunology
- Monitoring, Immunologic
- Exome Sequencing
- Antigens, Neoplasm/immunology
- Neoplasm Metastasis
- Clone Cells
Collapse
Affiliation(s)
- Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- The Royal Marsden Hospital NHS Foundation Trust, London, UK.
| | | | | | - Bo Sun
- Wellcome Centre for Human Genetics, Oxford, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Leticia De Mattos-Arruda
- IrsiCaixa, Germans Trias i Pujol University Hospital, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | | | | | | | - Oscar M Rueda
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Institució Catalana de Recerca i Estudis Avançats (ICREA), Universitat Autònoma de Barcelona (UAB), CIBERONC, Barcelona, Spain
| | | | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| | - Rachael J M Bashford-Rogers
- Wellcome Centre for Human Genetics, Oxford, UK.
- Department of Biochemistry, University of Oxford, Oxford, UK.
- Oxford Cancer Centre, Oxford, UK.
| |
Collapse
|
14
|
Mei SQ, Liu JQ, Huang ZJ, Luo WC, Peng YL, Chen ZH, Deng Y, Xu CR, Zhou Q. Identification of a risk score model based on tertiary lymphoid structure-related genes for predicting immunotherapy efficacy in non-small cell lung cancer. Thorac Cancer 2024; 15:1119-1131. [PMID: 38558529 DOI: 10.1111/1759-7714.15299] [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: 01/24/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) affect the prognosis and efficacy of immunotherapy in patients with non-small cell lung cancer (NSCLC), but the underlying mechanisms are not well understood. METHODS TLSs were identified and categorized online from the Cancer Digital Slide Archive (CDSA). Overall survival (OS) and disease-free survival (DFS) were analyzed. GSE111414 and GSE136961 datasets were downloaded from the GEO database. GSVA, GO and KEGG were used to explore the signaling pathways. Immune cell infiltration was analyzed by xCell, ssGSEA and MCP-counter. The analysis of WGCNA, Lasso and multivariate cox regression were conducted to develop a gene risk score model based on the SU2C-MARK cohort. RESULTS TLS-positive was a protective factor for OS according to multivariate cox regression analysis (p = 0.029). Both the TLS-positive and TLS-mature groups exhibited genes enrichment in immune activation pathways. The TLS-mature group showed more activated dendritic cell infiltration than the TLS-immature group. We screened TLS-related genes using WGCNA. Lasso and multivariate cox regression analysis were used to construct a five-genes (RGS8, RUF4, HLA-DQB2, THEMIS, and TRBV12-5) risk score model, the progression free survival (PFS) and OS of patients in the low-risk group were markedly superior to those in the high-risk group (p < 0.0001; p = 0.0015, respectively). Calibration and ROC curves indicated that the combined model with gene risk score and clinical features could predict the PFS of patients who have received immunotherapy more accurately than a single clinical factor. CONCLUSIONS Our data suggested a pivotal role of TLSs formation in survival outcome and immunotherapy response of NSCLC patients. Tumors with mature TLS formation showed more activated immune microenvironment. In addition, the model constructed by TLS-related genes could predict the response to immunotherapy and is meaningful for clinical decision-making.
Collapse
Affiliation(s)
- Shi-Qi Mei
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jia-Qi Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zi-Jian Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wei-Chi Luo
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying-Long Peng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine South China University of Technology, Guangzhou, China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Gao Y, Zhou L, Su Q, Li Q. Identification of Lung Adenocarcinoma Subtypes Based on MHC-II Gene Expression Profile and Immunological Analysis. Int Arch Allergy Immunol 2024; 185:884-899. [PMID: 38636483 DOI: 10.1159/000538056] [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: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 04/20/2024] Open
Abstract
INTRODUCTION Major histocompatibility complex class II molecule (MHC-II) is pivotal in anti-tumor immunity, and targeting MHC-II in tumors may help improve patient survival. But function of MHC-II in the immunotherapy and prognosis of lung adenocarcinoma (LUAD) patients has not been thoroughly studied and reported. METHODS We selected LUAD-related MHC-II genes from public databases based on previous literature reports. We identified different subtypes according to expression differences of these genes in different LUAD samples through cluster analysis. We used R package to conduct a series of analyses on different subtypes, exploring their survival differences, gene expression differences, pathway enrichment differences, and differences in immune characteristics and immune therapy. Finally, we screened potential drugs from the cMAP database. RESULTS We identified two MHC-II-related LUAD subtypes. Our analyses presented that patients with cluster2 subtype showed better prognosis, higher immune scores, higher levels of immune cell infiltration and immune function activation. In addition, patients with this subtype had higher immunophenoscore, lower TIDE scores, and DEPTH scores. We also identified 10 small molecule drugs, such as lenalidomide, VX-745, and tyrphostin-AG-1295. CONCLUSION Overall, MHC-II is not only a potential biomarker for accurately distinguishing LUAD subtypes but also a predictive factor for their survival. Our study offers novel insights into understanding of impact of MHC-II in LUAD and offers a new perspective for improving the accurate classification of LUAD patients and enhancing drug treatment.
Collapse
Affiliation(s)
- Yongcai Gao
- Department of Respiratory Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, China
| | - Lingli Zhou
- Department of Respiratory Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, China
| | - Qiong Su
- Department of Respiratory Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, China
| | - Qiang Li
- Department of Neurosurgery Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, China
| |
Collapse
|
16
|
Jafari N, Khajenabi F, Masumi N, Abediankenari S, Ranjbaran H. Evaluation of HLA-DR and HLA-DQ expression in gastric cancer tissues. J Cancer Res Ther 2024; 20:204-210. [PMID: 38554322 DOI: 10.4103/jcrt.jcrt_144_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 09/15/2022] [Indexed: 04/01/2024]
Abstract
BACKGROUND AND OBJECTIVES Despite recent advances in understanding the gastric cancer (GC) biology, the precise molecular mechanism of gastric carcinogenesis and role of deregulated immune responses in GC progression are still not well understood. In this study, mRNA levels of human leukocyte antigen (HLA)-DRA and -DQA1 were assessed in GC patients to find a potential association between expression of these HLA-II molecules and gastric carcinogenesis. METHODS Using quantitative real-time (qRT)-PCR, mRNA levels of HLA-DRA and -DQA1 were assessed in 20 pairs of matched GC and normal tissues. RESULTS Our results showed that overall mRNA level of HLA-DRA was decreased in the tumor samples relative to control tissues (median fold change [FC] = 0.693; P = 0.445). Overall HLA-DQA1 level was increased in the tumor samples relative to control tissues (median FC = 1.659; P = 0.5117). However, the mentioned data were not statistically significant. Meanwhile, using a ≥ 2.5 FC as the cutoff to determine upregulation or downregulation, 35% of patients showed a downregulated expression of HLA-DRA, while 10% of those showed upregulation in HLA-DRA expression. Upregulation and downregulation of HLA-DQA1 expression were detected, respectively, in 35% and 25% of samples. A strong positive correlation was determined between HLA-DRA and HLA-DQA1 levels in tumor tissues (r = 0.7298; P = 0.0003). CONCLUSION The results reported here along with future studies can be useful to understand the interplay between immune system and GC, therefore, may be helpful to design an effective immune-based therapy.
Collapse
Affiliation(s)
- Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Khajenabi
- Department of Medical Laboratory Sciences, Khatam Al-Anbia Hospital, Behshahr, Iran
| | - Nastaran Masumi
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Ranjbaran
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
17
|
Song F, Tarantino P, Garrido-Castro A, Lynce F, Tolaney SM, Schlam I. Immunotherapy for Early-Stage Triple Negative Breast Cancer: Is Earlier Better? Curr Oncol Rep 2024; 26:21-33. [PMID: 38198112 DOI: 10.1007/s11912-023-01487-1] [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] [Accepted: 12/11/2023] [Indexed: 01/11/2024]
Abstract
PURPOSE OF REVIEW In this narrative review, we discuss the optimal timing of immune checkpoint inhibitors (ICI) in early triple negative breast cancer (TNBC), the landscape of predictive biomarkers for the use of immunotherapy, and the mounting literature suggesting a benefit for an early use of ICI. RECENT FINDINGS TNBC is associated with a poor prognosis relative to other breast cancer subtypes, and until recently, the treatment of TNBC was limited to cytotoxic chemotherapy. In 2021, the immune-checkpoint inhibitor, pembrolizumab, was approved in combination with neoadjuvant chemotherapy for patients with high-risk early stage TNBC. This approval changed the treatment paradigm of early TNBC concomitantly raised several challenges in clinical practice, pertaining to patient selection, toxicity management, and post-neoadjuvant treatment, among others. The introduction of neoadjuvant chemoimmunotherapy has transformed the treatment landscape for early TNBC. However, several challenges, including patient selection, toxicity management, and the identification of predictive biomarkers, need to be addressed. Future research should focus on refining the timing and duration of immunotherapy, optimizing the chemotherapy partner, and exploring novel predictive biomarkers of response or toxicity.
Collapse
Affiliation(s)
- Fei Song
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Ana Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Filipa Lynce
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ilana Schlam
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA.
- Tufts University, Boston, MA, USA.
| |
Collapse
|
18
|
Nersesian S, Carter EB, Lee SN, Westhaver LP, Boudreau JE. Killer instincts: natural killer cells as multifactorial cancer immunotherapy. Front Immunol 2023; 14:1269614. [PMID: 38090565 PMCID: PMC10715270 DOI: 10.3389/fimmu.2023.1269614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Natural killer (NK) cells integrate heterogeneous signals for activation and inhibition using germline-encoded receptors. These receptors are stochastically co-expressed, and their concurrent engagement and signaling can adjust the sensitivity of individual cells to putative targets. Against cancers, which mutate and evolve under therapeutic and immunologic pressure, the diversity for recognition provided by NK cells may be key to comprehensive cancer control. NK cells are already being trialled as adoptive cell therapy and targets for immunotherapeutic agents. However, strategies to leverage their naturally occurring diversity and agility have not yet been developed. In this review, we discuss the receptors and signaling pathways through which signals for activation or inhibition are generated in NK cells, focusing on their roles in cancer and potential as targets for immunotherapies. Finally, we consider the impacts of receptor co-expression and the potential to engage multiple pathways of NK cell reactivity to maximize the scope and strength of antitumor activities.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Emily B. Carter
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stacey N. Lee
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | - Jeanette E. Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
19
|
Nilsson JB, Kaabinejadian S, Yari H, Kester MG, van Balen P, Hildebrand WH, Nielsen M. Accurate prediction of HLA class II antigen presentation across all loci using tailored data acquisition and refined machine learning. SCIENCE ADVANCES 2023; 9:eadj6367. [PMID: 38000035 PMCID: PMC10672173 DOI: 10.1126/sciadv.adj6367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023]
Abstract
Accurate prediction of antigen presentation by human leukocyte antigen (HLA) class II molecules is crucial for rational development of immunotherapies and vaccines targeting CD4+ T cell activation. So far, most prediction methods for HLA class II antigen presentation have focused on HLA-DR because of limited availability of immunopeptidomics data for HLA-DQ and HLA-DP while not taking into account alternative peptide binding modes. We present an update to the NetMHCIIpan prediction method, which closes the performance gap between all three HLA class II loci. We accomplish this by first integrating large immunopeptidomics datasets describing the HLA class II specificity space across all loci using a refined machine learning framework that accommodates inverted peptide binders. Next, we apply targeted immunopeptidomics assays to generate data that covers additional HLA-DP specificities. The final method, NetMHCIIpan-4.3, achieves high accuracy and molecular coverage across all HLA class II allotypes.
Collapse
Affiliation(s)
- Jonas B. Nilsson
- Department of Health Technology, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | - Saghar Kaabinejadian
- Pure MHC LLC, Oklahoma City, OK, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hooman Yari
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michel G. D. Kester
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - William H. Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, DK-2800 Lyngby, Denmark
| |
Collapse
|
20
|
Topchyan P, Lin S, Cui W. The Role of CD4 T Cell Help in CD8 T Cell Differentiation and Function During Chronic Infection and Cancer. Immune Netw 2023; 23:e41. [PMID: 37970230 PMCID: PMC10643329 DOI: 10.4110/in.2023.23.e41] [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: 06/05/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 11/17/2023] Open
Abstract
CD4 and CD8 T cells are key players in the immune response against both pathogenic infections and cancer. CD4 T cells provide help to CD8 T cells via multiple mechanisms, including licensing dendritic cells (DCs), co-stimulation, and cytokine production. During acute infection and vaccination, CD4 T cell help is important for the development of CD8 T cell memory. However, during chronic viral infection and cancer, CD4 helper T cells are critical for the sustained effector CD8 T cell response, through a variety of mechanisms. In this review, we focus on T cell responses in conditions of chronic Ag stimulation, such as chronic viral infection and cancer. In particular, we address the significant role of CD4 T cell help in promoting effector CD8 T cell responses, emerging techniques that can be utilized to further our understanding of how these interactions may take place in the context of tertiary lymphoid structures, and how this key information can be harnessed for therapeutic utility against cancer.
Collapse
Affiliation(s)
- Paytsar Topchyan
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Siying Lin
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Weiguo Cui
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
21
|
Ji H, Hu C, Yang X, Liu Y, Ji G, Ge S, Wang X, Wang M. Lymph node metastasis in cancer progression: molecular mechanisms, clinical significance and therapeutic interventions. Signal Transduct Target Ther 2023; 8:367. [PMID: 37752146 PMCID: PMC10522642 DOI: 10.1038/s41392-023-01576-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 07/04/2023] [Accepted: 07/26/2023] [Indexed: 09/28/2023] Open
Abstract
Lymph nodes (LNs) are important hubs for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites through a series of mechanisms, and it has been proved that lymph node metastasis (LNM) is an essential prognostic indicator in many different types of cancer. Therefore, it is important for oncologists to understand the mechanisms of tumor cells to metastasize to LNs, as well as how LNM affects the prognosis and therapy of patients with cancer in order to provide patients with accurate disease assessment and effective treatment strategies. In recent years, with the updates in both basic and clinical studies on LNM and the application of advanced medical technologies, much progress has been made in the understanding of the mechanisms of LNM and the strategies for diagnosis and treatment of LNM. In this review, current knowledge of the anatomical and physiological characteristics of LNs, as well as the molecular mechanisms of LNM, are described. The clinical significance of LNM in different anatomical sites is summarized, including the roles of LNM playing in staging, prognostic prediction, and treatment selection for patients with various types of cancers. And the novel exploration and academic disputes of strategies for recognition, diagnosis, and therapeutic interventions of metastatic LNs are also discussed.
Collapse
Affiliation(s)
- Haoran Ji
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chuang Hu
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuhui Yang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yuanhao Liu
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guangyu Ji
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiansong Wang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Mingsong Wang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
22
|
Borella R, Paolini A, Aramini B, Gibellini L, Masciale V, Lo Tartaro D, Dominici M, De Biasi S, Cossarizza A. Multiparametric analysis of tumor infiltrating lymphocytes in solid tumors. Methods Cell Biol 2023; 195:39-70. [PMID: 40180454 DOI: 10.1016/bs.mcb.2023.03.006] [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: 04/05/2025]
Abstract
The use of single-cell technologies in characterizing the interactions between immune and cancer cells is in continuous expansion. Indeed, the combination of different single-cell approaches enables the definition of novel phenotypic and functional aspects of the immune cells infiltrating the tumor microenvironment (TME). This approach is promoting the discovery of relevant and reliable predictive biomarkers, along with the development of new promising treatments. In this chapter, we describe the main subsets of tumor-infiltrating lymphocytes from a phenotypical and functional point of view and discuss the use of single-cell technologies used to characterize these cell populations within TME.
Collapse
Affiliation(s)
- Rebecca Borella
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum-University of Bologna and G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy.
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy; Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| |
Collapse
|
23
|
Wen M, Li Y, Qin X, Qin B, Wang Q. Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota. Cells 2023; 12:1882. [PMID: 37508545 PMCID: PMC10378520 DOI: 10.3390/cells12141882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/16/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer cells circumvent immune surveillance via diverse strategies. In accordance, a large number of complex studies of the immune system focusing on tumor cell recognition have revealed new insights and strategies developed, largely through major histocompatibility complexes (MHCs). As one of them, tumor-specific MHC-II expression (tsMHC-II) can facilitate immune surveillance to detect tumor antigens, and thereby has been used in immunotherapy, including superior cancer prognosis, clinical sensitivity to immune checkpoint inhibition (ICI) therapy and tumor-bearing rejection in mice. NK cells play a unique role in enhancing innate immune responses, accounting for part of the response including immunosurveillance and immunoregulation. NK cells are also capable of initiating the response of the adaptive immune system to cancer immunotherapy independent of cytotoxic T cells, clearly demonstrating a link between NK cell function and the efficacy of cancer immunotherapies. Eosinophils were shown to feature pleiotropic activities against a variety of solid tumor types, including direct interactions with tumor cells, and accessorily affect immunotherapeutic response through intricating cross-talk with lymphocytes. Additionally, microbial sequencing and reconstitution revealed that commensal microbiota might be involved in the modulation of cancer progression, including positive and negative regulatory bacteria. They may play functional roles in not only mucosal modulation, but also systemic immune responses. Here, we present a panorama of the cancer immune network mediated by MHCI/II molecules, immune cells and commensal microbiota and a discussion of prospective relevant intervening mechanisms involved in cancer immunotherapies.
Collapse
Affiliation(s)
- Minting Wen
- School of Life Science, Guangzhou University, Guangzhou 510006, China
| | - Yingjing Li
- School of Life Science, Guangzhou University, Guangzhou 510006, China
| | - Xiaonan Qin
- School of Life Science, Guangzhou University, Guangzhou 510006, China
| | - Bing Qin
- School of Life Science, Guangzhou University, Guangzhou 510006, China
| | - Qiong Wang
- School of Life Science, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
24
|
Senosain MF, Zou Y, Patel K, Zhao S, Coullomb A, Rowe DJ, Lehman JM, Irish JM, Maldonado F, Kammer MN, Pancaldi V, Lopez CF. Integrated Multi-omics Analysis of Early Lung Adenocarcinoma Links Tumor Biological Features with Predicted Indolence or Aggressiveness. CANCER RESEARCH COMMUNICATIONS 2023; 3:1350-1365. [PMID: 37501683 PMCID: PMC10370362 DOI: 10.1158/2767-9764.crc-22-0373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 05/01/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023]
Abstract
Lung adenocarcinoma (LUAD) is a heterogeneous group of tumors associated with different survival rates, even when detected at an early stage. Here, we aim to investigate the biological determinants of early LUAD indolence or aggressiveness using radiomics as a surrogate of behavior. We present a set of 92 patients with LUAD with data collected across different methodologies. Patients were risk-stratified using the CT-based Score Indicative of Lung cancer Aggression (SILA) tool (0 = least aggressive, 1 = most aggressive). We grouped the patients as indolent (x ≤ 0.4, n = 14), intermediate (0.4 > x ≤ 0.6, n = 27), and aggressive (0.6 > x ≤ 1, n = 52). Using Cytometry by time of flight (CyTOF), we identified subpopulations with high HLA-DR expression that were associated with indolent behavior. In the RNA sequencing (RNA-seq) dataset, pathways related to immune response were associated with indolent behavior, while pathways associated with cell cycle and proliferation were associated with aggressive behavior. We extracted quantitative radiomics features from the CT scans of the patients. Integrating these datasets, we identified four feature signatures and four patient clusters that were associated with survival. Using single-cell RNA-seq, we found that indolent tumors had significantly more T cells and less B cells than aggressive tumors, and that the latter had a higher abundance of regulatory T cells and Th cells. In conclusion, we were able to uncover a correspondence between radiomics and tumor biology, which could improve the discrimination between indolent and aggressive LUAD tumors, enhance our knowledge in the biology of these tumors, and offer novel and personalized avenues for intervention. Significance This study provides a comprehensive profiling of LUAD indolence and aggressiveness at the biological bulk and single-cell levels, as well as at the clinical and radiomics levels. This hypothesis generating study uncovers several potential future research avenues. It also highlights the importance and power of data integration to improve our systemic understanding of LUAD and to help reduce the gap between basic science research and clinical practice.
Collapse
Affiliation(s)
- Maria-Fernanda Senosain
- Cancer Biology Graduate Program, Vanderbilt University, Nashville, Tennessee
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Yong Zou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Khushbu Patel
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Shilin Zhao
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexis Coullomb
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Dianna J. Rowe
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Jonathan M. Lehman
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan M. Irish
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fabien Maldonado
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael N. Kammer
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Barcelona Supercomputing Center, Carrer de Jordi Girona, 29, 31, 08034 Barcelona, Spain
| | - Carlos F. Lopez
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
25
|
Kvokačková B, Fedr R, Kužílková D, Stuchlý J, Vávrová A, Navrátil J, Fabian P, Ondruššek R, Ovesná P, Remšík J, Bouchal J, Kalina T, Souček K. Single-cell protein profiling defines cell populations associated with triple-negative breast cancer aggressiveness. Mol Oncol 2023; 17:1024-1040. [PMID: 36550781 PMCID: PMC10257414 DOI: 10.1002/1878-0261.13365] [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: 07/01/2022] [Revised: 11/22/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and complex subtype of breast cancer that lacks targeted therapy. TNBC manifests characteristic, extensive intratumoral heterogeneity that promotes disease progression and influences drug response. Single-cell techniques in combination with next-generation computation provide an unprecedented opportunity to identify molecular events with therapeutic potential. Here, we describe the generation of a comprehensive mass cytometry panel for multiparametric detection of 23 phenotypic markers and 13 signaling molecules. This single-cell proteomic approach allowed us to explore the landscape of TNBC heterogeneity, with particular emphasis on the tumor microenvironment. We prospectively profiled freshly resected tumors from 26 TNBC patients. These tumors contained phenotypically distinct subpopulations of cancer and stromal cells that were associated with the patient's clinical status at the time of surgery. We further classified the epithelial-mesenchymal plasticity of tumor cells, and molecularly defined phenotypically diverse populations of tumor-associated stroma. Furthermore, in a retrospective tissue-microarray TNBC cohort, we showed that the level of CD97 at the time of surgery has prognostic potential.
Collapse
Affiliation(s)
- Barbora Kvokačková
- Department of CytokineticsInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Radek Fedr
- Department of CytokineticsInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Daniela Kužílková
- Childhood Leukaemia Investigation PragueCzech Republic
- Department of Pediatric Haematology and Oncology, 2nd Faculty of MedicineCharles University Prague and University Hospital MotolCzech Republic
| | - Jan Stuchlý
- Childhood Leukaemia Investigation PragueCzech Republic
- Department of Pediatric Haematology and Oncology, 2nd Faculty of MedicineCharles University Prague and University Hospital MotolCzech Republic
| | - Adéla Vávrová
- Childhood Leukaemia Investigation PragueCzech Republic
- Faculty of ScienceCharles University PragueCzech Republic
| | - Jiří Navrátil
- Department of Comprehensive Cancer CareMasaryk Memorial Cancer InstituteBrnoCzech Republic
| | - Pavel Fabian
- Department of Oncological PathologyMasaryk Memorial Cancer InstituteBrnoCzech Republic
| | - Róbert Ondruššek
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and DentistryPalacký University and University HospitalOlomoucCzech Republic
- Department of PathologyEUC Laboratoře CGB a.s.OstravaCzech Republic
| | - Petra Ovesná
- Institute of Biostatistics and Analyses, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Ján Remšík
- Human Oncology and Pathogenesis ProgramMemorial Sloan Kettering Cancer CenterNew York CityNYUSA
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and DentistryPalacký University and University HospitalOlomoucCzech Republic
| | - Tomáš Kalina
- Childhood Leukaemia Investigation PragueCzech Republic
- Department of Pediatric Haematology and Oncology, 2nd Faculty of MedicineCharles University Prague and University Hospital MotolCzech Republic
| | - Karel Souček
- Department of CytokineticsInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| |
Collapse
|
26
|
Long F, Ma H, Hao Y, Tian L, Li Y, Li B, Chen J, Tang Y, Li J, Deng L, Xie G, Liu M. A novel exosome-derived prognostic signature and risk stratification for breast cancer based on multi-omics and systematic biological heterogeneity. Comput Struct Biotechnol J 2023; 21:3010-3023. [PMID: 37273850 PMCID: PMC10232662 DOI: 10.1016/j.csbj.2023.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/29/2023] [Accepted: 05/11/2023] [Indexed: 06/06/2023] Open
Abstract
Tumor heterogeneity remains a major challenge for disease subtyping, risk stratification, and accurate clinical management. Exosome-based liquid biopsy can effectively overcome the limitations of tissue biopsy, achieving minimal invasion, multi-point dynamic monitoring, and good prognosis assessment, and has broad clinical prospects. However, there is still lacking comprehensive analysis of tumor-derived exosome (TDE)-based stratification of risk patients and prognostic assessment for breast cancer with systematic dissection of biological heterogeneity. In this study, the robust corroborative analysis for biomarker discovery (RCABD) strategy was used for the identification of exosome molecules, differential expression verification, risk prediction modeling, heterogenous dissection with multi-ome (6101 molecules), our ExoBCD database (306 molecules), and 53 independent studies (481 molecules). Our results showed that a 10-molecule exosome-derived signature (exoSIG) could successfully fulfill breast cancer risk stratification, making it a novel and accurate exosome prognostic indicator (Cox P = 9.9E-04, HR = 3.3, 95% CI 1.6-6.8). Interestingly, HLA-DQB2 and COL17A1, closely related to tumor metastasis, achieved high performance in prognosis prediction (86.35% contribution) and accuracy (Log-rank P = 0.028, AUC = 85.42%). With the combined information of patient age and tumor stage, they formed a bimolecular risk signature (Clinmin-exoSIG) and a convenient nomogram as operable tools for clinical applications. In conclusion, as an extension of ExoBCD, this study conducted systematic analyses to identify prognostic multi-molecular panel and risk signature, stratify patients and dissect biological heterogeneity based on breast cancer exosomes from a multi-omics perspective. Our results provide an important reference for in-depth exploration of the "biological heterogeneity - risk stratification - prognosis prediction".
Collapse
Affiliation(s)
- Fei Long
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Haodong Ma
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Luyao Tian
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Yinghong Li
- Key Laboratory on Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, PR China
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Juan Chen
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Ying Tang
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Li
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Lili Deng
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Guoming Xie
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Mingwei Liu
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| |
Collapse
|
27
|
Heng Y, Zhu X, Wu Q, Lin H, Ding X, Tao L, Lu L. High Expression of Tumor HLA-DR Predicts Better Prognosis and Response to Anti-PD-1 Therapy in Laryngeal Squamous Cell Carcinoma. Transl Oncol 2023; 33:101678. [PMID: 37149969 DOI: 10.1016/j.tranon.2023.101678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND HLA-DR is expressed in epithelial and several types of tumor cells. However, the correlation between tumor-expressed HLA-DR (teHLA-DR) and patient outcome as well as its regulation on the tumor microenvironment (TME) of laryngeal squamous cell carcinoma (LSCC) are yet to be elucidated. METHODS Hematoxylin and eosin (HE) staining were performed to define the tumor nest and stroma of LSCC tissue microarrays. teHLA-DR tumor cell, CD4+ and CD8+ tumor-infiltrating T lymphocytes (TITLs) were obtained and analyzed through double-labeling immunofluorescence and immunohistochemical staining. The recurrence-free (RFS) and overall survival (OS) curves were plotted using the Kaplan-Meier method and tested by the log-rank test method. Expression of teHLA-DR+ tumor cells and infiltration of T lymphocytes and their corresponding subgroups were analyzed by flow cytometry using fresh LSCC tissue samples. RESULTS Our research discovered elevated expressions of multiple MHC-II-related genes in tumor compared to the adjacent normal tissue samples of LSCC patients. We also found that patients in the teHLA-DR high-expression group (teHLA-DRhigh) tend to have less tumor recurrence and better survival outcomes compared to those in the teHLA-DRlow group. Intriguingly, teHLA-DR+ tumor cells had significantly higher PD-L1 and PD-L2 expression and their TME showed increased infiltrated T lymphocytes (TITLs). Flow cytometry analysis and IHC staining indicated that CD4+ TITLs but not CD3+ total TITLs or CD8+ TITLs were significantly enriched in teHLA-DR+ tumors. CONCLUSIONS teHLA-DR may be a predictive marker for favorable prognosis and response to anti-PD-1/PD-L1 therapy of LSCC, possibly due to the increased CD4+ TITLs in the TME.
Collapse
Affiliation(s)
- Yu Heng
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Xiaoke Zhu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qian Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hanqing Lin
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Xuping Ding
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Tao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China.
| | - Liming Lu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
28
|
Carter JM, Chumsri S, Hinerfeld DA, Ma Y, Wang X, Zahrieh D, Hillman DW, Tenner KS, Kachergus JM, Brauer HA, Warren SE, Henderson D, Shi J, Liu Y, Joensuu H, Lindman H, Leon-Ferre RA, Boughey JC, Liu MC, Ingle JN, Kalari KR, Couch FJ, Knutson KL, Goetz MP, Perez EA, Thompson EA. Distinct spatial immune microlandscapes are independently associated with outcomes in triple-negative breast cancer. Nat Commun 2023; 14:2215. [PMID: 37072398 PMCID: PMC10113250 DOI: 10.1038/s41467-023-37806-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 03/30/2023] [Indexed: 04/20/2023] Open
Abstract
The utility of spatial immunobiomarker quantitation in prognostication and therapeutic prediction is actively being investigated in triple-negative breast cancer (TNBC). Here, with high-plex quantitative digital spatial profiling, we map and quantitate intraepithelial and adjacent stromal tumor immune protein microenvironments in systemic treatment-naïve (female only) TNBC to assess the spatial context in immunobiomarker-based prediction of outcome. Immune protein profiles of CD45-rich and CD68-rich stromal microenvironments differ significantly. While they typically mirror adjacent, intraepithelial microenvironments, this is not uniformly true. In two TNBC cohorts, intraepithelial CD40 or HLA-DR enrichment associates with better outcomes, independently of stromal immune protein profiles or stromal TILs and other established prognostic variables. In contrast, intraepithelial or stromal microenvironment enrichment with IDO1 associates with improved survival irrespective of its spatial location. Antigen-presenting and T-cell activation states are inferred from eigenprotein scores. Such scores within the intraepithelial compartment interact with PD-L1 and IDO1 in ways that suggest prognostic and/or therapeutic potential. This characterization of the intrinsic spatial immunobiology of treatment-naïve TNBC highlights the importance of spatial microenvironments for biomarker quantitation to resolve intrinsic prognostic and predictive immune features and ultimately inform therapeutic strategies for clinically actionable immune biomarkers.
Collapse
Affiliation(s)
- Jodi M Carter
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Saranya Chumsri
- Department of Medicine, Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Yaohua Ma
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL, USA
| | - Xue Wang
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL, USA
| | - David Zahrieh
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - David W Hillman
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Kathleen S Tenner
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Ji Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Yi Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Heikki Joensuu
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Henrik Lindman
- Department of Oncology, University of Uppsala, Uppsala, Sweden
| | - Roberto A Leon-Ferre
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | | | | | - James N Ingle
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Krishna R Kalari
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
| | - Matthew P Goetz
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Edith A Perez
- Department of Medicine, Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
29
|
Jeong S, Jang N, Kim M, Choi IK. CD4 + cytotoxic T cells: an emerging effector arm of anti-tumor immunity. BMB Rep 2023; 56:140-144. [PMID: 36863358 PMCID: PMC10068340 DOI: 10.5483/bmbrep.2023-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 04/29/2024] Open
Abstract
While CD8+ cytotoxic T cells have long been considered the primary effector in controlling tumors, the involvement of CD4+ "helper" T cells in anti-tumor immunity has been underappreciated. The investigations of intra-tumoral T cells, fueled by the recent advances in genomic technologies, have led to a rethinking of the indirect role of CD4+ T cells that have traditionally been described as a "helper". Accumulating evidence from preclinical and clinical studies indicates that CD4+ T cells can acquire intrinsic cytotoxic properties and directly kill various types of tumor cells in a major histocompatibility complex class II (MHC-II)-dependent manner, as opposed to the indirect "helper" function, thus underscoring a potentially critical contribution of CD4+ cytotoxic T cells to immune responses against a wide range of tumor types. Here, we discuss the biological properties of anti-tumor CD4+ T cells with cytotoxic capability and highlight the emerging observations suggesting their more significant role in anti-tumor immunity than previously appreciated. [BMB Reports 2023; 56(3): 140-144].
Collapse
Affiliation(s)
- Seongmin Jeong
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Nawon Jang
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Minchae Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Il-Kyu Choi
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- New Biology Research Center (NBRC), Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
30
|
Liu C, Li X, Huang Q, Zhang M, Lei T, Wang F, Zou W, Huang R, Hu X, Wang C, Zhang X, Sun B, Xing L, Yue J, Yu J. Single-cell RNA-sequencing reveals radiochemotherapy-induced innate immune activation and MHC-II upregulation in cervical cancer. Signal Transduct Target Ther 2023; 8:44. [PMID: 36710358 PMCID: PMC9884664 DOI: 10.1038/s41392-022-01264-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 01/31/2023] Open
Abstract
Radiochemotherapy (RCT) is a powerful treatment for cervical cancer, which affects not only malignant cells but also the immune and stromal compartments of the tumor. Understanding the remodeling of the local ecosystem induced by RCT would provide valuable insights into improving treatment strategies for cervical cancer. In this study, we applied single-cell RNA-sequencing to paired pre- and post-RCT tumor biopsies from patients with cervical cancer and adjacent normal cervical tissues. We found that the residual population of epithelial cells post-RCT showed upregulated expression of MHC class II genes. Moreover, RCT led to the accumulation of monocytic myeloid-derived suppressor cells with increased pro-inflammatory features and CD16+ NK cells with a higher cytotoxic gene expression signature. However, subclusters of T cells showed no significant increase in the expression of cytotoxic features post-RCT. These results reveal the complex responses of the tumor ecosystem to RCT, providing evidence of activation of innate immunity and MHC-II upregulation in cervical cancer.
Collapse
Affiliation(s)
- Chao Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, 250117, China
| | - Xiaohui Li
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qingyu Huang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Min Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Fuhao Wang
- School of Clinical Medicine, Weifang Medical University, Weifang, 261053, China
| | - Wenxue Zou
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Rui Huang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xiaoyu Hu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Cong Wang
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xiaoling Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Bing Sun
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Ligang Xing
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, 250117, China.
| | - Jinbo Yue
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, 250117, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
31
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
32
|
Mechanisms and Strategies to Overcome PD-1/PD-L1 Blockade Resistance in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 15:cancers15010104. [PMID: 36612100 PMCID: PMC9817764 DOI: 10.3390/cancers15010104] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a high rate of systemic metastasis, insensitivity to conventional treatment and susceptibility to drug resistance, resulting in a poor patient prognosis. The immune checkpoint inhibitors (ICIs) represented by antibodies of programmed death receptor 1 (PD-1) and programmed death receptor ligand 1 (PD-L1) have provided new therapeutic options for TNBC. However, the efficacy of PD-1/PD-L1 blockade monotherapy is suboptimal immune response, which may be caused by reduced antigen presentation, immunosuppressive tumor microenvironment, interplay with other immune checkpoints and aberrant activation of oncological signaling in tumor cells. Therefore, to improve the sensitivity of TNBC to ICIs, suitable patients are selected based on reliable predictive markers and treated with a combination of ICIs with other therapies such as chemotherapy, radiotherapy, targeted therapy, oncologic virus and neoantigen-based therapies. This review discusses the current mechanisms underlying the resistance of TNBC to PD-1/PD-L1 inhibitors, the potential biomarkers for predicting the efficacy of anti-PD-1/PD-L1 immunotherapy and recent advances in the combination therapies to increase response rates, the depth of remission and the durability of the benefit of TNBC to ICIs.
Collapse
|
33
|
McKeown BT, Relja NJ, Hall SR, Gebremeskel S, MacLeod JM, Veinotte CJ, Bennett LG, Ohlund LB, Sleno L, Jakeman DL, Berman JN, Johnston B, Goralski KB. Pilot study of jadomycin B pharmacokinetics and anti-tumoral effects in zebrafish larvae and mouse breast cancer xenograft models. Can J Physiol Pharmacol 2022; 100:1065-1076. [PMID: 35985040 DOI: 10.1139/cjpp-2022-0152] [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: 01/26/2023]
Abstract
Despite numerous therapeutic options, multidrug resistance (MDR) remains an obstacle to successful breast cancer therapy. Jadomycin B, a natural product derived from Streptomyces venezuelae ISP5230, maintains cytotoxicity in MDR human breast cancer cells. Our objectives were to evaluate the pharmacokinetics, toxicity, anti-tumoral, and anti-metastatic effects of jadomycin B in zebrafish larvae and mice. In a zebrafish larval xenograft model, jadomycin B significantly reduced the proliferation of human MDA-MB-231 cells at or below its maximum tolerated dose (40 µm). In female Balb/C mice, a single intraperitoneal dose (6 mg/kg) was rapidly absorbed with a maximum serum concentration of 3.4 ± 0.27 µm. Jadomycin B concentrations declined biphasically with an elimination half-life of 1.7 ± 0.058 h. In the 4T1 mouse mammary carcinoma model, jadomycin B (12 mg/kg every 12 h from day 6 to 15 after tumor cell injection) decreased primary tumor volume compared to vehicle control. Jadomycin B-treated mice did not exhibit weight loss, nor significant increases in biomarkers of impaired hepatic (alanine aminotransferase) and renal (creatinine) function. In conclusion, jadomycin B demonstrated a good safety profile and provided partial anti-tumoral effects, warranting further dose-escalation safety and efficacy studies in MDR breast cancer models.
Collapse
Affiliation(s)
- Brendan T McKeown
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, NS, B3H 4R2, Canada
| | - Nicholas J Relja
- Faculty of Health, College of Pharmacy, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Steven R Hall
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Simon Gebremeskel
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Jeanna M MacLeod
- Faculty of Health, College of Pharmacy, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Chansey J Veinotte
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, IWK Health Centre, Halifax, NS, B3K 6R8, Canada
| | - Leah G Bennett
- Faculty of Health, College of Pharmacy, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Leanne B Ohlund
- Chemistry department/CERMO-FC, Faculty of Sciences, Université du Québec à Montréal, Montréal, QC, H2X 2J6, Canada
| | - Lekha Sleno
- Chemistry department/CERMO-FC, Faculty of Sciences, Université du Québec à Montréal, Montréal, QC, H2X 2J6, Canada
| | - David L Jakeman
- Faculty of Health, College of Pharmacy, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Chemistry, Faculty of Science, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Jason N Berman
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, IWK Health Centre, Halifax, NS, B3K 6R8, Canada.,Children's Hospital of Eastern Ontario Research Institute and Department of Pediatrics, University of Ottawa, Ottawa, ON, K1H 5B2, Canada.,Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Brent Johnston
- Beatrice Hunter Cancer Research Institute, Halifax, NS, B3H 4R2, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Kerry B Goralski
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, NS, B3H 4R2, Canada.,Faculty of Health, College of Pharmacy, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, IWK Health Centre, Halifax, NS, B3K 6R8, Canada
| |
Collapse
|
34
|
Chan KL, Gomez J, Cardinez C, Kumari N, Sparbier CE, Lam EYN, Yeung MM, Garciaz S, Kuzich JA, Ong DM, Brown FC, Chan YC, Vassiliadis D, Wainwright EN, Motazedian A, Gillespie A, Fennell KA, Lai J, House IG, Macpherson L, Ang CS, Dawson SJ, Beavis PA, Wei AH, Burr ML, Dawson MA. Inhibition of the CtBP complex and FBXO11 enhances MHC class II expression and anti-cancer immune responses. Cancer Cell 2022; 40:1190-1206.e9. [PMID: 36179686 PMCID: PMC7615013 DOI: 10.1016/j.ccell.2022.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/22/2022] [Accepted: 09/04/2022] [Indexed: 11/24/2022]
Abstract
There is increasing recognition of the prognostic significance of tumor cell major histocompatibility complex (MHC) class II expression in anti-cancer immunity. Relapse of acute myeloid leukemia (AML) following allogeneic stem cell transplantation (alloSCT) has recently been linked to MHC class II silencing in leukemic blasts; however, the regulation of MHC class II expression remains incompletely understood. Utilizing unbiased CRISPR-Cas9 screens, we identify that the C-terminal binding protein (CtBP) complex transcriptionally represses MHC class II pathway genes, while the E3 ubiquitin ligase complex component FBXO11 mediates degradation of CIITA, the principal transcription factor regulating MHC class II expression. Targeting these repressive mechanisms selectively induces MHC class II upregulation across a range of AML cell lines. Functionally, MHC class II+ leukemic blasts stimulate antigen-dependent CD4+ T cell activation and potent anti-tumor immune responses, providing fundamental insights into the graft-versus-leukemia effect. These findings establish the rationale for therapeutic strategies aimed at restoring tumor-specific MHC class II expression to salvage AML relapse post-alloSCT and also potentially to enhance immunotherapy outcomes in non-myeloid malignancies.
Collapse
Affiliation(s)
- Kah Lok Chan
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Juliana Gomez
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Division of Genome Science and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Chelisa Cardinez
- Division of Genome Science and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Nishi Kumari
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Christina E Sparbier
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Enid Y N Lam
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Miriam M Yeung
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Sylvain Garciaz
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Aix-Marseille University, INSERM U1068, CNRS, Institut Paoli-Calmettes, 13009 Marseille, France
| | - James A Kuzich
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Doen Ming Ong
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia; Department of Haematology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Fiona C Brown
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia
| | - Yih-Chih Chan
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Dane Vassiliadis
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elanor N Wainwright
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ali Motazedian
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | | | - Katie A Fennell
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Junyun Lai
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Imran G House
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Laura Macpherson
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Centre for Cancer Research, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Paul A Beavis
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew H Wei
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia; Department of Haematology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Marian L Burr
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Genome Science and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; Department of Anatomical Pathology, ACT Pathology, Canberra Health Services, Canberra, ACT 2606, Australia.
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; Centre for Cancer Research, The University of Melbourne, Parkville, VIC 3000, Australia.
| |
Collapse
|
35
|
Wu G, Xiao G, Yan Y, Guo C, Hu N, Shen S. Bioinformatics analysis of the clinical significance of HLA class II in breast cancer. Medicine (Baltimore) 2022; 101:e31071. [PMID: 36221383 PMCID: PMC9543021 DOI: 10.1097/md.0000000000031071] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human leukocyte antigen (HLA) class II plays critical roles in antigen presentation and the initiation of immune responses. However, the correlation between the HLA class II gene expression level and the survival of patients with breast cancer is still under investigation. We analyzed microarray and RNA-Seq data of breast cancer from the cancer genome atlas (TCGA), genotype-tissue expression (GTEx) and Oncomine databases by using bioinformatics tools. The expression of the HLA-DQA1, HLA-DQA2, and HLA-DQB2 genes was significantly upregulated in breast cancer. Higher expression levels of HLA class II genes in breast cancer, especially HLA-DOB and HLA-DQB2, were significantly associated with better overall survival. Furthermore, the expression of HLA class II genes was more closely associated with survival in breast cancer than in other cancer types. CD48 coexpressed with both HLA-DOB and HLA-DQB2 was also positively associated with the overall survival of breast cancer patients. The results indicated that HLA class II and CD48 may enhance antitumor immunity, and their expression patterns may serve as potential prognostic biomarkers and therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- Guihua Wu
- Finance Section, Yuebei People’s Hospital, Shantou University, Shaoguan, China
| | - Gaofang Xiao
- Department of Pathology, Yuebei People’s Hospital, Shantou University, Shaoguan, China
| | - Yuhang Yan
- Breast Surgery, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Chengwei Guo
- Department of Radiology, 82 Group Hospital of PLA, Baoding, China
| | - Ningdong Hu
- Thoracic surgery, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Sandi Shen
- Breast Surgery, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
- *Correspondence: Sandi Shen, Breast Surgery, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, 21 Yinquan South Road, Qingyuan 511518, China (e-mail: )
| |
Collapse
|
36
|
Massa D, Tosi A, Rosato A, Guarneri V, Dieci MV. Multiplexed In Situ Spatial Protein Profiling in the Pursuit of Precision Immuno-Oncology for Patients with Breast Cancer. Cancers (Basel) 2022; 14:4885. [PMID: 36230808 PMCID: PMC9562913 DOI: 10.3390/cancers14194885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many solid tumors. In breast cancer (BC), immunotherapy is currently approved in combination with chemotherapy, albeit only in triple-negative breast cancer. Unfortunately, most patients only derive limited benefit from ICIs, progressing either upfront or after an initial response. Therapeutics must engage with a heterogeneous network of complex stromal-cancer interactions that can fail at imposing cancer immune control in multiple domains, such as in the genomic, epigenomic, transcriptomic, proteomic, and metabolomic domains. To overcome these types of heterogeneous resistance phenotypes, several combinatorial strategies are underway. Still, they can be predicted to be effective only in the subgroups of patients in which those specific resistance mechanisms are effectively in place. As single biomarker predictive performances are necessarily suboptimal at capturing the complexity of this articulate network, precision immune-oncology calls for multi-omics tumor microenvironment profiling in order to identify unique predictive patterns and to proactively tailor combinatorial treatments. Multiplexed single-cell spatially resolved tissue analysis, through precise epitope colocalization, allows one to infer cellular functional states in view of their spatial organization. In this review, we discuss-through the lens of the cancer-immunity cycle-selected, established, and emerging markers that may be evaluated in multiplexed spatial protein panels to help identify prognostic and predictive patterns in BC.
Collapse
Affiliation(s)
- Davide Massa
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| |
Collapse
|
37
|
Barkley D, Moncada R, Pour M, Liberman DA, Dryg I, Werba G, Wang W, Baron M, Rao A, Xia B, França GS, Weil A, Delair DF, Hajdu C, Lund AW, Osman I, Yanai I. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat Genet 2022; 54:1192-1201. [PMID: 35931863 PMCID: PMC9886402 DOI: 10.1038/s41588-022-01141-9] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/22/2022] [Indexed: 02/01/2023]
Abstract
Transcriptional heterogeneity among malignant cells of a tumor has been studied in individual cancer types and shown to be organized into cancer cell states; however, it remains unclear to what extent these states span tumor types, constituting general features of cancer. Here, we perform a pan-cancer single-cell RNA-sequencing analysis across 15 cancer types and identify a catalog of gene modules whose expression defines recurrent cancer cell states including 'stress', 'interferon response', 'epithelial-mesenchymal transition', 'metal response', 'basal' and 'ciliated'. Spatial transcriptomic analysis linked the interferon response in cancer cells to T cells and macrophages in the tumor microenvironment. Using mouse models, we further found that induction of the interferon response module varies by tumor location and is diminished upon elimination of lymphocytes. Our work provides a framework for studying how cancer cell states interact with the tumor microenvironment to form organized systems capable of immune evasion, drug resistance and metastasis.
Collapse
Affiliation(s)
- Dalia Barkley
- Institute for Computational Medicine, New York, NY, USA
| | | | - Maayan Pour
- Institute for Computational Medicine, New York, NY, USA
| | | | - Ian Dryg
- Department of Dermatology, NYU School of Medicine, New York, NY, USA
| | - Gregor Werba
- Department of Surgery, NYU School of Medicine, New York, NY, USA,Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Wei Wang
- Department of Surgery, NYU School of Medicine, New York, NY, USA,Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Maayan Baron
- Institute for Computational Medicine, New York, NY, USA
| | - Anjali Rao
- Institute for Computational Medicine, New York, NY, USA
| | - Bo Xia
- Institute for Computational Medicine, New York, NY, USA
| | | | - Alejandro Weil
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | | | - Cristina Hajdu
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Amanda W. Lund
- Department of Dermatology, NYU School of Medicine, New York, NY, USA,Department of Surgery, NYU School of Medicine, New York, NY, USA,Perlmutter Cancer Center NYU School of Medicine, New York, NY, USA
| | - Iman Osman
- Department of Dermatology, NYU School of Medicine, New York, NY, USA,Department of Pathology, NYU School of Medicine, New York, NY, USA,Perlmutter Cancer Center NYU School of Medicine, New York, NY, USA
| | - Itai Yanai
- Institute for Computational Medicine, New York, NY, USA,Perlmutter Cancer Center NYU School of Medicine, New York, NY, USA,Corresponding author:
| |
Collapse
|
38
|
Yao L, Jia G, Lu L, Ma W. Breast Cancer Patients: Who Would Benefit from Neoadjuvant Chemotherapies? Curr Oncol 2022; 29:4902-4913. [PMID: 35877249 PMCID: PMC9320700 DOI: 10.3390/curroncol29070389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 11/17/2022] Open
Abstract
Neoadjuvant chemotherapy (NACT) was developed with the aims of shrinking tumors or stopping cancer cells from spreading before surgery. Unfortunately, not all breast cancer patients will benefit from NACT, and thus, patients must weigh the risks and benefits of treatment prior to the initiation of therapy. Currently, the data for predicting the efficacy of NACT is limited. Molecular testing, such as Oncotype DX, MammaPrint, and Curebest 95GC, have been developed to assist which breast cancer patients will benefit from the treatment. Patients with an increased level of Human Leukocyte Antigen-DR isotype, tumor-infiltrating lymphocytes, Fizzy-related protein homolog, and a decreased level of tumor-associated macrophages appear to benefit most from NACT.
Collapse
Affiliation(s)
- Liqin Yao
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, School of Medicine, Huzhou University, Huzhou 313000, China;
| | - Gang Jia
- Department of Medical Oncology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China;
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, School of Medicine, Yale School of Public Health, New Haven, CT 06520, USA;
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
- Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
| | - Wenxue Ma
- Sanford Stem Cell Clinical Center, Moores Cancer Center, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
39
|
Wang B, Liu J, Han Y, Deng Y, Li J, Jiang Y. The Presence of Tertiary Lymphoid Structures Provides New Insight Into the Clinicopathological Features and Prognosis of Patients With Breast Cancer. Front Immunol 2022; 13:868155. [PMID: 35664009 PMCID: PMC9161084 DOI: 10.3389/fimmu.2022.868155] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/19/2022] [Indexed: 02/05/2023] Open
Abstract
Background Tertiary lymphoid structures (TLSs) have been proven to be predictive biomarkers of favorable clinical outcomes and response to immunotherapies in several solid malignancies. Nevertheless, the effect of TLSs in patients with breast cancer (BC) remains controversial. The objective of the current study is to investigate the clinicopathological and prognostic significance of TLSs in BC. Given the unique difficulties for detecting and quantifying TLSs, a TLS-associated gene signature based on The Cancer Genome Atlas (TCGA) BC cohort was used to validate and supplement our results. Methods Electronic platforms (PubMed, Web of Science, EMBASE, the Cochrane Library, CNKI, and Wanfang) were searched systematically to identify relevant studies as of January 11, 2022. We calculated combined odds ratios (ORs) with 95% confidence intervals (CIs) to determine the relationship between clinicopathological parameters and TLSs. The pooled hazard ratios (HRs) and 95% CIs were also calculated to evaluate the prognostic significance of TLSs. The TLS signature based on the TCGA BC cohort was applied to validate and supplement our results. Results Fifteen studies with 3,898 patients were eligible for enrollment in our study. The combined analysis indicated that the presence of TLSs was related to improved disease-free survival (DFS) (HR = 0.61, 95% CI: 0.41-0.90, p < 0.05) and overall survival (OS) (HR = 1.66, 95% CI: 1.26-2.20, p < 0.001). Additionally, the presence of TLSs was positively correlated with early tumor TNM stage and high tumor-infiltrating lymphocytes. TLS presence was positively related to human epidermal growth factor receptor 2 (HER-2) and Ki-67 but inversely correlated with the status of estrogen and progesterone receptor. Simultaneously, our study found that tumor immune microenvironment was more favorable in the high-TLS signature group than in the low-TLS signature group. Consistently, BC patients in the high-TLS signature group exhibited better survival outcomes compared to those in the low-TLS signature group, suggesting that TLSs might be favorable prognostic biomarkers. Conclusions TLS presence provides new insight into the clinicopathological features and prognosis of patients with BC, whereas the factors discussed limited the evidence quality of this study. We look forward to consistent methods to define and characterize TLSs, and more high-quality prospective clinical trials designed to validate the value of TLSs alone or in combination with other markers.
Collapse
Affiliation(s)
- Bin Wang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yin Han
- Department of Pathology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Yaotiao Deng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jinze Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Jiang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Gene signatures in patients with early breast cancer and relapse despite pathologic complete response. NPJ Breast Cancer 2022; 8:42. [PMID: 35351903 PMCID: PMC8964729 DOI: 10.1038/s41523-022-00403-3] [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: 08/02/2021] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
A substantial minority of early breast cancer (EBC) patients relapse despite their tumors achieving pathologic complete response (pCR) after neoadjuvant therapy. We compared gene expression (BC360; nCounter® platform; NanoString) between primary tumors of patients with post-pCR relapse (N = 14) with: (i) matched recurrent tumors from same patient (intraindividual analysis); and (ii) primary tumors from matched controls with pCR and no relapse (N = 41; interindividual analysis). Intraindividual analysis showed lower estrogen receptor signaling signature expression in recurrent tumors versus primaries (logFC = −0.595; P = 0.022). Recurrent tumors in patients with distant metastases also exhibited reduced expression of immune-related expression parameters. In interindividual analyses, primary tumor major histocompatibility complex class II expression was lower versus controls in patients with any relapse (logFC = −0.819; P = 0.030) or distant relapse (logFC = −1.151; P = 0.013). Primaries with later distant relapse also had greater homologous recombination deficiency than controls (logFC = 0.649; P = 0.026). Although no associations remained statistically significant following adjustment for false discovery rate, our results show that transcriptomic analyses have potential for prognostic value and may help in selecting optimal treatment regimens for EBC at risk of relapse and warrant further investigation.
Collapse
|
41
|
Valencia GA, Rioja P, Morante Z, Ruiz R, Fuentes H, Castaneda CA, Vidaurre T, Neciosup S, Gomez HL. Immunotherapy in triple-negative breast cancer: A literature review and new advances. World J Clin Oncol 2022; 13:219-236. [PMID: 35433291 PMCID: PMC8966508 DOI: 10.5306/wjco.v13.i3.219] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/23/2021] [Accepted: 02/19/2022] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly complex, heterogeneous disease and historically has limited treatment options. It has a high probability of disease recurrence and rapid disease progression despite adequate systemic treatment. Immunotherapy has emerged as an important alternative in the management of this malignancy, showing an impact on progression-free survival and overall survival in selected populations. In this review we focused on immunotherapy and its current relevance in the management of TNBC, including various scenarios (metastatic and early -neoadjuvant, adjuvant-), new advances in this subtype and the research of potential predictive biomarkers of response to treatment.
Collapse
Affiliation(s)
| | - Patricia Rioja
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Zaida Morante
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Rossana Ruiz
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Hugo Fuentes
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Carlos A Castaneda
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Tatiana Vidaurre
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Silvia Neciosup
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Henry L Gomez
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| |
Collapse
|
42
|
Asleh K, Negri GL, Spencer Miko SE, Colborne S, Hughes CS, Wang XQ, Gao D, Gilks CB, Chia SKL, Nielsen TO, Morin GB. Proteomic analysis of archival breast cancer clinical specimens identifies biological subtypes with distinct survival outcomes. Nat Commun 2022; 13:896. [PMID: 35173148 PMCID: PMC8850446 DOI: 10.1038/s41467-022-28524-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Despite advances in genomic classification of breast cancer, current clinical tests and treatment decisions are commonly based on protein level information. Formalin-fixed paraffin-embedded (FFPE) tissue specimens with extended clinical outcomes are widely available. Here, we perform comprehensive proteomic profiling of 300 FFPE breast cancer surgical specimens, 75 of each PAM50 subtype, from patients diagnosed in 2008-2013 (n = 178) and 1986-1992 (n = 122) with linked clinical outcomes. These two cohorts are analyzed separately, and we quantify 4214 proteins across all 300 samples. Within the aggressive PAM50-classified basal-like cases, proteomic profiling reveals two groups with one having characteristic immune hot expression features and highly favorable survival. Her2-Enriched cases separate into heterogeneous groups differing by extracellular matrix, lipid metabolism, and immune-response features. Within 88 triple-negative breast cancers, four proteomic clusters display features of basal-immune hot, basal-immune cold, mesenchymal, and luminal with disparate survival outcomes. Our proteomic analysis characterizes the heterogeneity of breast cancer in a clinically-applicable manner, identifies potential biomarkers and therapeutic targets, and provides a resource for clinical breast cancer classification.
Collapse
Affiliation(s)
- Karama Asleh
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gian Luca Negri
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Sandra E Spencer Miko
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Shane Colborne
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Christopher S Hughes
- Department of Molecular Oncology, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Xiu Q Wang
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - C Blake Gilks
- Division of Anatomical Pathology, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
- Canadian Immunohistochemistry Quality Control, University of British Columbia, Vancouver, BC, Canada
| | - Stephen K L Chia
- Division of Medical Oncology, British Columbia Cancer Centre, University of British Columbia, Vancouver, BC, Canada
| | - Torsten O Nielsen
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of Anatomical Pathology, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Gregg B Morin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
43
|
Balakrishnan CK, Tye GJ, Balasubramaniam SD, Kaur G. CD74 and HLA-DRA in Cervical Carcinogenesis: Potential Targets for Antitumour Therapy. Medicina (B Aires) 2022; 58:medicina58020190. [PMID: 35208514 PMCID: PMC8877221 DOI: 10.3390/medicina58020190] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/21/2022] Open
Abstract
Background and Objectives: Abnormal expressions of CD74 and human leukocyte antigen-DR alpha (HLA-DRA) have been reported in various cancers, though their roles in cervical cancer remain unclear. This study aimed to evaluate the gene and protein expressions of CD74 and HLA-DRA in the progression from normal cervix to precancerous cervical intraepithelial neoplasia (CIN) and finally to squamous cell carcinoma (SCC). Materials and Methods: The gene expression profiles of CD74 and HLA-DRA were determined in formalin-fixed paraffin-embedded tissues, with three samples each from normal cervixes, human papillomavirus type 16/18-positive, low-grade CIN (LGCIN), high-grade CIN (HGCIN), and squamous cell carcinoma (SCC) using Human Transcriptome Array 2.0. Immunohistochemical expression of the proteins was semi-quantitatively assessed in another cohort of tissue microarray samples comprising 7 normal cervix cases, 10 LGCIN, 10 HGCIN, and 95 SCC. Results: The transcriptomics profile and proteins’ expression demonstrated similar trends of upregulation of CD74 and HLA-DRA from normal cervix to CIN and highest in SCC. There was a significant difference in both proteins’ expression between the histological groups (p = 0.0001). CD74 and HLA-DRA expressions were significantly associated with CIN grade (p = 0.001 and p = 0.030, respectively) but not with the subjects’ age or SCC stage. Further analysis revealed a positive correlation between CD74 and HLA-DRA proteins. Conclusions: CD74 appears to promote cervical carcinogenesis via oncogenic signalling mechanisms and may serve as a potential antitumour target. Additionally, the upregulation of HLA-DRA, often associated with stronger immunogenicity, could be a promising biomarker for developing immunotherapies.
Collapse
Affiliation(s)
- Carol K. Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia; (C.K.B.); (G.J.T.); (S.D.B.)
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia; (C.K.B.); (G.J.T.); (S.D.B.)
| | - Shandra Devi Balasubramaniam
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia; (C.K.B.); (G.J.T.); (S.D.B.)
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong 08100, Semeling, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia; (C.K.B.); (G.J.T.); (S.D.B.)
- Correspondence: ; Tel.: +604-6534865
| |
Collapse
|
44
|
Nanamori H, Sawada Y. Epigenetic Modification of PD-1/PD-L1-Mediated Cancer Immunotherapy against Melanoma. Int J Mol Sci 2022; 23:ijms23031119. [PMID: 35163049 PMCID: PMC8835029 DOI: 10.3390/ijms23031119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Malignant melanoma is one of the representative skin cancers with unfavorable clinical behavior. Immunotherapy is currently used for the treatment, and it dramatically improves clinical outcomes in patients with advanced malignant melanoma. On the other hand, not all these patients can obtain therapeutic efficacy. To overcome this limitation of current immunotherapy, epigenetic modification is a highlighted issue for clinicians. Epigenetic modification is involved in various physiological and pathological conditions in the skin. Recent studies identified that skin cancer, especially malignant melanoma, has advantages in tumor development, indicating that epigenetic manipulation for regulation of gene expression in the tumor can be expected to result in additional therapeutic efficacy during immunotherapy. In this review, we focus on the detailed molecular mechanism of epigenetic modification in immunotherapy, especially anti-PD-1/PD-L1 antibody treatment for malignant melanoma.
Collapse
|
45
|
Harnessing Antitumor CD4 + T Cells for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14010260. [PMID: 35008422 PMCID: PMC8750687 DOI: 10.3390/cancers14010260] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Diverse evidence revealed that CD4+ T cells play an important role in antitumor immunity by promoting or suppressing cytotoxic T cell responses. This review outlines the role of CD4+ T subsets within the tumor microenvironment and summarizes the latest progress regarding their potentials in cancer immunotherapy and methods for improving outcomes in cancer strategies by modulating CD4+ T responses. Abstract Over the past decades, CD4+ T cells have been considered as a supporting actor in the fields of cancer immunotherapy. Until recently, accumulating evidence has demonstrated the critical role of CD4+ T cells during antitumor immunity. CD4+ T cells can either suppress or promote the antitumor cytotoxic CD8+ T cell responses, either in secondary lymphoid organs or in the tumor. In this review, we provide an overview of the multifaceted role of different CD4+ T cell subsets in cancer immune response and their contribution during cancer therapies. Specifically, we focus on the latest progress regarding the impact of CD4+ T cell modulation on immunotherapies and other cancer therapies and discuss the prospect for harnessing CD4+ T cells to control tumor progression and prevent recurrence in patients.
Collapse
|
46
|
Comprehensive Transcriptome and Pathway Analyses Revealed Central Role for Fascin in Promoting Triple-Negative Breast Cancer Progression. Pharmaceuticals (Basel) 2021; 14:ph14121228. [PMID: 34959629 PMCID: PMC8708558 DOI: 10.3390/ph14121228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
Recent years have witnessed major progress in development of novel therapeutic agents such as chemotherapy, targeted therapy and immune checkpoint inhibitors for breast cancer. However, cancer-related death remains high especially in triple-negative breast cancer (TNBC) due limited therapeutic options. Development of targeted therapies for TNBC requires better understanding of biology and signaling networks that promote disease progression. Fascin, an actin bundling protein, was identified as a key regulator of many signaling pathways that contribute to breast cancer progression. Herein, fascin ShRNA was used to generate stable fascin knockdown (FSCN1KD) in the MDA-MB-231 TNBC cell line and then were subjected to comprehensive mRNA and miRNA transcriptome analysis. We identified 129 upregulated and 114 downregulated mRNA transcripts, while 14 miRNAs were differentially expressed in FSCN1KD. Ingenuity pathway analysis (IPA) was used to predict the impact of differentially expressed transcripts on signaling pathways and functional categories and to construct miRNA-mRNA regulatory networks in the context of FSCN1 knockdown. Compared to FSCN1KD, fascin-positive (FSCN1CON) breast cancer cells showed enrichment in genes promoting cellular proliferation, migration, survival, DNA replication and repair. Expression of FSCN1high (identified in BRCA dataset from TCGA) in conjunction with elevated expression of the top 10 upregulated or decreased expression of the top 10 downregulated genes (identified in our FSCN1CON vs. FSCN1KD) correlates with worst survival outcome. Taken together, these data confirmed fascin's role in promoting TNBC progression, and identified a novel opportunity for therapeutic interventions via targeting those FSCN1-related transcripts.
Collapse
|
47
|
Beyaz S, Chung C, Mou H, Bauer-Rowe KE, Xifaras ME, Ergin I, Dohnalova L, Biton M, Shekhar K, Eskiocak O, Papciak K, Ozler K, Almeqdadi M, Yueh B, Fein M, Annamalai D, Valle-Encinas E, Erdemir A, Dogum K, Shah V, Alici-Garipcan A, Meyer HV, Özata DM, Elinav E, Kucukural A, Kumar P, McAleer JP, Fox JG, Thaiss CA, Regev A, Roper J, Orkin SH, Yilmaz ÖH. Dietary suppression of MHC class II expression in intestinal epithelial cells enhances intestinal tumorigenesis. Cell Stem Cell 2021; 28:1922-1935.e5. [PMID: 34529935 PMCID: PMC8650761 DOI: 10.1016/j.stem.2021.08.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/25/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
Little is known about how interactions of diet, intestinal stem cells (ISCs), and immune cells affect early-stage intestinal tumorigenesis. We show that a high-fat diet (HFD) reduces the expression of the major histocompatibility complex class II (MHC class II) genes in intestinal epithelial cells, including ISCs. This decline in epithelial MHC class II expression in a HFD correlates with reduced intestinal microbiome diversity. Microbial community transfer experiments suggest that epithelial MHC class II expression is regulated by intestinal flora. Mechanistically, pattern recognition receptor (PRR) and interferon-gamma (IFNγ) signaling regulates epithelial MHC class II expression. MHC class II-negative (MHC-II-) ISCs exhibit greater tumor-initiating capacity than their MHC class II-positive (MHC-II+) counterparts upon loss of the tumor suppressor Apc coupled with a HFD, suggesting a role for epithelial MHC class II-mediated immune surveillance in suppressing tumorigenesis. ISC-specific genetic ablation of MHC class II increases tumor burden cell autonomously. Thus, HFD perturbs a microbiome-stem cell-immune cell interaction that contributes to tumor initiation in the intestine.
Collapse
Affiliation(s)
- Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA; The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA.
| | - Charlie Chung
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Haiwei Mou
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Khristian E Bauer-Rowe
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Michael E Xifaras
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Ilgin Ergin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lenka Dohnalova
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moshe Biton
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; The Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Karthik Shekhar
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemical and Biomolecular Engineering, Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Kadir Ozler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Mohammad Almeqdadi
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Brian Yueh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Miriam Fein
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Damodaran Annamalai
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eider Valle-Encinas
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Aysegul Erdemir
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Karoline Dogum
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Vyom Shah
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Hannah V Meyer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Deniz M Özata
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alper Kucukural
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Pawan Kumar
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jeremy P McAleer
- Department of Pharmaceutical Science and Research, Marshall University School of Pharmacy, Huntington, WV 25701, USA
| | - James G Fox
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aviv Regev
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA; Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02140, USA
| | - Jatin Roper
- Department of Medicine, Division of Gastroenterology, Duke University, Durham, NC 27710, USA
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
48
|
Tariq F, Khan W, Ahmad W, Riaz SK, Khan M, Sherwani S, Haque S, Malik MFA, Iftikhar MJ, Khan S, Haq F. Effect of MHC Linked 7-Gene Signature on Delayed Hepatocellular Carcinoma Recurrence. J Pers Med 2021; 11:jpm11111129. [PMID: 34834481 PMCID: PMC8625636 DOI: 10.3390/jpm11111129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Dysregulated immune response significantly affects hepatocellular carcinoma's (HCC) prognosis. Human Leukocyte Antigens are key in devising immune responses against HCC. Here, we investigated how HLAs modulate HCC development at the transcriptomic level. RNA-seq data of 576 patients from two independent cohorts was retrieved. The clinicopathological relevance of all HLA genes was investigated using Fisher-Exact, correlation, and Kaplan-Meier and cox regression survival tests. Clustering of ~800 immune-related genes against HLAs was completed using a ward-agglomerative method. Networks were generated using 40 HLA associated unique genes and hub genes were investigated. HLAs including HLA-DMA, HLA-DMB, HLA-DOA and HLA-DRB6 were associated with delayed recurrence in both discovery (204 HCC cases) and validation (372 HCC cases) cohorts. Clustering analyses revealed 40 genes associated with these four HLAs in both cohorts. A set of seven genes (NCF4, TYROBP, LCP2, ZAP70, PTPRC, FYN and WAS) was found co-expressed at gene-gene interaction level in both cohorts. Furthermore, survival analysis revealed seven HLA-linked genes as predictors of delayed recurrence. Multivariate analysis also predicted that mean expression of 7-gene is an independent predictor of delayed recurrence in both cohorts. We conclude that the expression of 7-gene signature may lead to improved patient prognosis. Further studies are required for consideration in clinical practice.
Collapse
Affiliation(s)
- Fomaz Tariq
- Department of Biosciences, COMSATS University, Islamabad 44000, Pakistan; (F.T.); (W.K.); (W.A.); (S.K.R.); (M.F.A.M.)
| | - Walizeb Khan
- Department of Biosciences, COMSATS University, Islamabad 44000, Pakistan; (F.T.); (W.K.); (W.A.); (S.K.R.); (M.F.A.M.)
| | - Washaakh Ahmad
- Department of Biosciences, COMSATS University, Islamabad 44000, Pakistan; (F.T.); (W.K.); (W.A.); (S.K.R.); (M.F.A.M.)
| | - Syeda Kiran Riaz
- Department of Biosciences, COMSATS University, Islamabad 44000, Pakistan; (F.T.); (W.K.); (W.A.); (S.K.R.); (M.F.A.M.)
- Department of Molecular Biology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad 44000, Pakistan
- College of Medicine, Texas A&M University, College Station, TX 77840-77845, USA
| | - Mahvish Khan
- Department of Biology, College of Science, Ha’il University, Ha’il 55211, Saudi Arabia; (M.K.); (S.S.)
| | - Subuhi Sherwani
- Department of Biology, College of Science, Ha’il University, Ha’il 55211, Saudi Arabia; (M.K.); (S.S.)
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Faculty of Medicine, Görükle Campus, Bursa Uludağ University, Bursa 16059, Turkey
| | - Muhammad Faraz Arshad Malik
- Department of Biosciences, COMSATS University, Islamabad 44000, Pakistan; (F.T.); (W.K.); (W.A.); (S.K.R.); (M.F.A.M.)
| | | | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, Ha’il University, Ha’il 55211, Saudi Arabia
- Correspondence: (S.K.); (F.H.)
| | - Farhan Haq
- Department of Biosciences, COMSATS University, Islamabad 44000, Pakistan; (F.T.); (W.K.); (W.A.); (S.K.R.); (M.F.A.M.)
- Correspondence: (S.K.); (F.H.)
| |
Collapse
|
49
|
de Sousa E, Lérias JR, Beltran A, Paraschoudi G, Condeço C, Kamiki J, António PA, Figueiredo N, Carvalho C, Castillo-Martin M, Wang Z, Ligeiro D, Rao M, Maeurer M. Targeting Neoepitopes to Treat Solid Malignancies: Immunosurgery. Front Immunol 2021; 12:592031. [PMID: 34335558 PMCID: PMC8320363 DOI: 10.3389/fimmu.2021.592031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 05/07/2021] [Indexed: 12/26/2022] Open
Abstract
Successful outcome of immune checkpoint blockade in patients with solid cancers is in part associated with a high tumor mutational burden (TMB) and the recognition of private neoantigens by T-cells. The quality and quantity of target recognition is determined by the repertoire of ‘neoepitope’-specific T-cell receptors (TCRs) in tumor-infiltrating lymphocytes (TIL), or peripheral T-cells. Interferon gamma (IFN-γ), produced by T-cells and other immune cells, is essential for controlling proliferation of transformed cells, induction of apoptosis and enhancing human leukocyte antigen (HLA) expression, thereby increasing immunogenicity of cancer cells. TCR αβ-dependent therapies should account for tumor heterogeneity and availability of the TCR repertoire capable of reacting to neoepitopes and functional HLA pathways. Immunogenic epitopes in the tumor-stroma may also be targeted to achieve tumor-containment by changing the immune-contexture in the tumor microenvironment (TME). Non protein-coding regions of the tumor-cell genome may also contain many aberrantly expressed, non-mutated tumor-associated antigens (TAAs) capable of eliciting productive anti-tumor immune responses. Whole-exome sequencing (WES) and/or RNA sequencing (RNA-Seq) of cancer tissue, combined with several layers of bioinformatic analysis is commonly used to predict possible neoepitopes present in clinical samples. At the ImmunoSurgery Unit of the Champalimaud Centre for the Unknown (CCU), a pipeline combining several tools is used for predicting private mutations from WES and RNA-Seq data followed by the construction of synthetic peptides tailored for immunological response assessment reflecting the patient’s tumor mutations, guided by MHC typing. Subsequent immunoassays allow the detection of differential IFN-γ production patterns associated with (intra-tumoral) spatiotemporal differences in TIL or peripheral T-cells versus TIL. These bioinformatics tools, in addition to histopathological assessment, immunological readouts from functional bioassays and deep T-cell ‘adaptome’ analyses, are expected to advance discovery and development of next-generation personalized precision medicine strategies to improve clinical outcomes in cancer in the context of i) anti-tumor vaccination strategies, ii) gauging mutation-reactive T-cell responses in biological therapies and iii) expansion of tumor-reactive T-cells for the cellular treatment of patients with cancer.
Collapse
Affiliation(s)
- Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana R Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Antonio Beltran
- Department of Pathology, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jéssica Kamiki
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Nuno Figueiredo
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Zhe Wang
- Jiangsu Industrial Technology Research Institute (JITRI), Applied Adaptome Immunology Institute, Nanjing, China
| | - Dário Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação (IPST), Lisbon, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,I Medical Clinic, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
50
|
Senosain MF, Zou Y, Novitskaya T, Vasiukov G, Balar AB, Rowe DJ, Doxie DB, Lehman JM, Eisenberg R, Maldonado F, Zijlstra A, Novitskiy SV, Irish JM, Massion PP. HLA-DR cancer cells expression correlates with T cell infiltration and is enriched in lung adenocarcinoma with indolent behavior. Sci Rep 2021; 11:14424. [PMID: 34257356 PMCID: PMC8277797 DOI: 10.1038/s41598-021-93807-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/25/2021] [Indexed: 11/16/2022] Open
Abstract
Lung adenocarcinoma (ADC) is a heterogeneous group of tumors associated with different survival rates, even when detected at an early stage. Here, we aim to investigate whether CyTOF identifies cellular and molecular predictors of tumor behavior. We developed and validated a CyTOF panel of 34 antibodies in four ADC cell lines and PBMC. We tested our panel in a set of 10 ADCs, classified into long- (LPS) (n = 4) and short-predicted survival (SPS) (n = 6) based on radiomics features. We identified cellular subpopulations of epithelial cancer cells (ECC) and their microenvironment and validated our results by multiplex immunofluorescence (mIF) applied to a tissue microarray (TMA) of LPS and SPS ADCs. The antibody panel captured the phenotypical differences in ADC cell lines and PBMC. LPS ADCs had a higher proportion of immune cells. ECC clusters (ECCc) were identified and uncovered two ADC groups. ECCc with high HLA-DR expression were correlated with CD4+ and CD8+ T cells, with LPS samples being enriched for those clusters. We confirmed a positive correlation between HLA-DR expression on ECC and T cell number by mIF staining on TMA slides. Spatial analysis demonstrated shorter distances from T cells to the nearest ECC in LPS. Our results demonstrate a distinctive cellular profile of ECC and their microenvironment in ADC. We showed that HLA-DR expression in ECC is correlated with T cell infiltration, and that a set of ADCs with high abundance of HLA-DR+ ECCc and T cells is enriched in LPS samples. This suggests new insights into the role of antigen presenting tumor cells in tumorigenesis.
Collapse
Affiliation(s)
- Maria-Fernanda Senosain
- Cancer Biology Graduate Program, Vanderbilt University, Nashville, TN, USA.
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Yong Zou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tatiana Novitskaya
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Georgii Vasiukov
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aneri B Balar
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dianna J Rowe
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Deon B Doxie
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Jonathan M Lehman
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rosana Eisenberg
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fabien Maldonado
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sergey V Novitskiy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan M Irish
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pierre P Massion
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
- US Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|