51
|
Li M, Ma Y, Zhong Y, Liu Q, Chen C, Qiang L, Wang X. KALRN mutations promote antitumor immunity and immunotherapy response in cancer. J Immunother Cancer 2021; 8:jitc-2019-000293. [PMID: 33037113 PMCID: PMC7549479 DOI: 10.1136/jitc-2019-000293] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/26/2022] Open
Abstract
Background kalirin RhoGEF kinase (KALRN) is mutated in a wide range of cancers. Nevertheless, the association between KALRN mutations and the pathogenesis of cancer remains unexplored. Identification of biomarkers for cancer immunotherapy response is crucial because immunotherapies only show beneficial effects in a subset of patients with cancer. Methods We explored the correlation between KALRN mutations and antitumor immunity in 10 cancer cohorts from The Cancer Genome Atlas program by the bioinformatics approach. Moreover, we verified the findings from the bioinformatics analysis with in vitro and in vivo experiments. We explored the correlation between KALRN mutations and immunotherapy response in five cancer cohorts receiving immune checkpoint blockade therapy. Results Antitumor immune signatures were more enriched in KALRN-mutated than KALRN-wildtype cancers. Moreover, KALRN mutations displayed significant correlations with increased tumor mutation burden and the microsatellite instability or DNA damage repair deficiency genomic properties, which may explain the high antitumor immunity in KALRN-mutated cancers. Also, programmed cell death 1 ligand (PD-L1) expression was markedly upregulated in KALRN-mutated versus KALRN-wildtype cancers. The increased antitumor immune signatures and PD-L1 expression in KALRN-mutated cancers may favor the response to immune checkpoint blockade therapy in this cancer subtype, as evidenced in five cancer cohorts receiving antiprogrammed cell death protein 1 (PD-1)/PD-L1/cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immunotherapy. Furthermore, the significant association between KALRN mutations and increased antitumor immunity was associated with the fact that KALRN mutations compromised the function of KALRN in targeting Rho GTPases for the regulation of DNA damage repair pathways. In vitro and in vivo experiments validated the association of KALRN deficiency with antitumor immunity and the response to immune checkpoint inhibitors. Conclusions The KALRN mutation is a useful biomarker for predicting the response to immunotherapy in patients with cancer.
Collapse
Affiliation(s)
- Mengyuan Li
- Biomedical Informatics Research Lab,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| | - Yuxiang Ma
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - You Zhong
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qian Liu
- Biomedical Informatics Research Lab,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| | - Canping Chen
- Biomedical Informatics Research Lab,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| | - Lei Qiang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China .,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
52
|
Xing Z, Ma B, Sun W, Sun Y, Liu C. Comprehensive characterization and clinical relevance of the SWI/SNF copy number aberrations across human cancers. Hereditas 2021; 158:38. [PMID: 34598711 PMCID: PMC8487138 DOI: 10.1186/s41065-021-00203-y] [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: 04/09/2021] [Accepted: 09/18/2021] [Indexed: 01/04/2023] Open
Abstract
Background Alterations in genes encoding chromatin regulatory proteins are prevalent in cancers and may confer oncogenic properties and molecular changes linked to therapy resistance. However, the impact of copy number alterations (CNAs) of the SWItch/Sucrose NonFermentable (SWI/SNF) complex on the oncogenic and immunologic properties has not been systematically explored across human cancer types. Methods We comprehensively analyzed the genomic, transcriptomic and clinical data of The Cancer Genome Atlas (TCGA) dataset across 33 solid cancers. Results CNAs of the SWI/SNF components were identified in more than 25% of all queried cancers, and tumors harboring SWI/SNF CNAs demonstrated a worse overall survival (OS) than others in several cancer types. Mechanistically, the SCNA events in the SWI/SNF complex are correlated with dysregulated genomic features and oncogenic pathways, including the cell cycle, DNA damage and repair. Notably, the SWI/SNF CNAs were associated with homologous recombination deficiency (HRD) and improved clinical outcomes of platinum-treated ovarian cancer. Furthermore, we observed distinct immune infiltrating patterns and immunophenotypes associated with SWI/SNF CNAs in different cancer types. Conclusion The CNA events of the SWI/SNF components are a key process linked to oncogenesis, immune infiltration and therapeutic responsiveness across human cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00203-y.
Collapse
Affiliation(s)
- Zhiwei Xing
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Buhuan Ma
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Weiting Sun
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Yimin Sun
- National Engineering Research Center for Beijing Biochip Technology, Beijing, 102206, China.,Department of Biomedical Engineering, Medical Systems Biology Research Center, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Caixia Liu
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China.
| |
Collapse
|
53
|
Feng Q, Song D, Wang X. Pan-cancer analysis reveals that neurotrophin signaling correlates positively with anti-tumor immunity, clinical outcomes, and response to targeted therapies and immunotherapies in cancer. Life Sci 2021; 282:119848. [PMID: 34293398 DOI: 10.1016/j.lfs.2021.119848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022]
Abstract
AIMS The crosstalk between cancer cells and nerves plays an important role in tumor biology. However, the correlation between the neurotrophin signaling (NS) and anti-tumor immunity and immunotherapy response in cancer remains unexplored. MATERIALS AND METHODS We analyzed associations of NS with anti-tumor immune signatures, tumor immunity-related molecular and genomic features, and clinical features in 33 TCGA cancer types. We also explored the association between NS and the response to immune checkpoint inhibitors (ICIs) in four cancer cohorts. KEY FINDINGS NS scores had significant positive correlations with the enrichment scores of anti-tumor immune signatures, including CD8+ T cells, interferon response, natural killer cells, Toll-like receptor and NOD-like receptor signaling pathways in most cancer types. NS scores were inversely correlated with the scores of DNA damage repair pathways, tumor mutation burden, copy number alterations, intra-tumor heterogeneity, and tumor stemness in diverse cancers. In contrast, NS scores were significantly and positively correlated with the apoptosis pathway's scores in 32 of the 33 cancer types. NS scores were significantly lower in early-stage versus late-stage and in primary versus metastatic tumors in diverse cancers. Higher NS scores were correlated with better survival in pan-cancer and in eight individual cancer types. Moreover, the response rate to ICIs was higher in higher-NS-score than in lower-NS-score tumors in four cancer cohorts. Elevated NS was correlated with increased drug sensitivity for numerous anti-tumor targeted drugs. SIGNIFICANCE NS is a positive biomarker for anti-tumor immune response, prognosis, and the response to targeted and immunotherapeutic drugs in cancer.
Collapse
Affiliation(s)
- Qiushi Feng
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Dandan Song
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
54
|
Sun D, Zhu Y, Zhao H, Bian T, Li T, Liu K, Feng L, Li H, Hou H. Loss of ARID1A expression promotes lung adenocarcinoma metastasis and predicts a poor prognosis. Cell Oncol (Dordr) 2021; 44:1019-1034. [PMID: 34109546 DOI: 10.1007/s13402-021-00616-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/26/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND ARID1A is an essential subunit of SWI/SNF chromatin remodeling complexes. ARID1A gene mutations and loss of ARID1A expression have been observed in a variety of cancers, and to be correlated with invasion, immune escape and synthetic lethality. As yet, however, the biological effect of ARID1A expression and its role in the prognosis of lung adenocarcinoma (LUAD) patients have remained unclear. In this study we aimed to further elucidate the role of ARID1A expression in LUAD in vitro and in vivo and to assess its effect on the clinical prognosis of LUAD patients. METHODS ARID1A expression was detected by IHC in tissue samples from LUAD patients. After regular culturing of LUAD cell lines and constructing stable ARID1A knockdown lines, wound healing and Transwell assays were used to assess the role of ARID1A in cell migration and invasion. The effect of ARID1A knockdown on metastasis was verified in vivo. Western blotting was used to examine the expression of target proteins. Univariate and multivariate analyses were performed to assess survival and to provide variables for nomogram construction. In addition, we used the "rms" package to construct a prognostic nomogram based on a Cox regression model. RESULTS We found that ARID1A expression serves as an effective prognostic marker for LUAD patients. Loss of ARID1A expression correlated with a poor prognosis, as verified with a nomogram based on a Cox regression model. In addition, we found that ARID1A knockdown promoted LUAD cell proliferation, migration and invasion in vitro and enhanced LUAD metastasis in vivo by activating the Akt signaling pathway. CONCLUSIONS Our data indicate that loss of ARID1A expression promotes LUAD metastasis and predicts a poor prognosis in LUAD patients.
Collapse
Affiliation(s)
- Dantong Sun
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Shandong, 266000, Qingdao, China
| | - Yan Zhu
- Department of Medical Oncology, The Municipal Hospital of Qingdao, 266000, Qingdao, China
| | - Han Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, 266000, Qingdao, China
| | - Tiantian Bian
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao University, 266100, Qingdao, China
| | - Tianjun Li
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Shandong, 266000, Qingdao, China
| | - Kewei Liu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Shandong, 266000, Qingdao, China
| | - Lizong Feng
- Department of General Surgery, Qingdao Eighth People's Hospital, 266041, Qingdao, China
| | - Hong Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| | - Helei Hou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Shandong, 266000, Qingdao, China.
| |
Collapse
|
55
|
Mutations in Epigenetic Regulation Genes in Gastric Cancer. Cancers (Basel) 2021; 13:cancers13184586. [PMID: 34572812 PMCID: PMC8467700 DOI: 10.3390/cancers13184586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Epigenetic mechanisms, such as DNA methylation/demethylation, covalent modifications of histone proteins, and chromatin remodeling, create specific patterns of gene expression. Epigenetic deregulations are associated with oncogenesis, relapse of the disease and metastases, and can serve as a useful clinical marker. We assessed the clinical relevance of integrity of the genes coding for epigenetic regulator proteins by mutational profiling of 25 genes in 135 gastric cancer (GC) samples. Overall, mutations in the epigenetic regulation genes were found to be significantly associated with reduced overall survival of patients in the group with metastases and in the group with tumors with signet ring cells. We have also discovered mutual exclusivity of somatic mutations in the KMT2D, KMT2C, ARID1A, and CHD7 genes in our cohort. Our results suggest that mutations in epigenetic regulation genes may be valuable clinical markers and deserve further exploration in independent cohorts. Abstract We have performed mutational profiling of 25 genes involved in epigenetic processes on 135 gastric cancer (GC) samples. In total, we identified 79 somatic mutations in 49/135 (36%) samples. The minority (n = 8) of mutations was identified in DNA methylation/demethylation genes, while the majority (n = 41), in histone modifier genes, among which mutations were most commonly found in KMT2D and KMT2C. Somatic mutations in KMT2D, KMT2C, ARID1A and CHD7 were mutually exclusive (p = 0.038). Mutations in ARID1A were associated with distant metastases (p = 0.03). The overall survival of patients in the group with metastases and in the group with tumors with signet ring cells was significantly reduced in the presence of mutations in epigenetic regulation genes (p = 0.036 and p = 0.041, respectively). Separately, somatic mutations in chromatin remodeling genes correlate with low survival rate of patients without distant metastasis (p = 0.045) and in the presence of signet ring cells (p = 0.0014). Our results suggest that mutations in epigenetic regulation genes may be valuable clinical markers and deserve further exploration in independent cohorts.
Collapse
|
56
|
Cao W, Ma X, Fischer JV, Sun C, Kong B, Zhang Q. Immunotherapy in endometrial cancer: rationale, practice and perspectives. Biomark Res 2021; 9:49. [PMID: 34134781 PMCID: PMC8207707 DOI: 10.1186/s40364-021-00301-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy has attracted more and more attention nowadays, and multiple clinical trials have confirmed its effect in a variety of solid tumors. Immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell transfer (ACT), and lymphocyte-promoting cytokines are the main immunotherapy methods. Endometrial cancer (EC) is one of the most frequent tumors in women and the prognosis of recurrent or metastatic EC is poor. Since molecular classification has been applied to EC, immunotherapy for different EC subtypes (especially POLE and MSI-H) has gradually attracted attention. In this review, we focus on the expression and molecular basis of the main biomarkers in the immunotherapy of EC firstly, as well as their clinical application significance and limitations. Blocking tumor immune checkpoints is one of the most effective strategies for cancer treatment in recent years, and has now become the focus in the field of tumor research and treatment. We summarized clinical date of planned and ongoing clinical trials and introduced other common immunotherapy methods in EC, such as cancer vaccine and ACT. Hormone aberrations, metabolic syndrome (MetS) and p53 mutant and that affect the immunotherapy of endometrial cancer will also be discussed in this review.
Collapse
Affiliation(s)
- Wenyu Cao
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Xinyue Ma
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Jean Victoria Fischer
- Department of Pathology, Northwestern Medicine, Gynecologic Pathology Fellow, Chicago, Illinois, USA
| | - Chenggong Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China.
| |
Collapse
|
57
|
Kamori T, Oki E, Shimada Y, Hu Q, Hisamatsu Y, Ando K, Shimokawa M, Wakai T, Oda Y, Mori M. The effects of ARID1A mutations on colorectal cancer and associations with PD-L1 expression by stromal cells. Cancer Rep (Hoboken) 2021; 5:e1420. [PMID: 34042312 PMCID: PMC8789618 DOI: 10.1002/cnr2.1420] [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: 01/27/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022] Open
Abstract
Background ARID1A is a component of the SWI/SNF complex, which controls the accessibility of proteins to DNA. ARID1A mutations are frequently observed in colorectal cancers (CRCs) and have been reported to be associated with high mutational burden and tumor PD‐L1 expression in vitro. Aim To clarify the role of ARID1A mutation in CRC. Method and results We used next generation sequencing (NGS) and immunohistochemistry on clinically obtained samples. A total of 201 CRC tissues from Niigata University and Niigata Center Hospital were processed by NGS using the CANCERPLEX panel. Immunohistochemistry for ARID1A, PD‐L1, MLH1, and MSH2 was performed on 66 propensity‐matched (33 microsatellite instability‐high [MSI‐H] and 33 microsatellite‐stable [MSS]) cases among 499 cases from Kyushu University. TCGA data were downloaded from cBioPortal. NGS showed significantly more mutations in ARID1A mutated CRCs (p = 0.01), and the trend was stronger for right‐sided CRCs than left‐sided. TCGA data confirmed these findings (p < 0.01). BRAF V600E and ATM mutations were also found at higher frequencies. Immunohistochemistry showed that 30% of MSI‐H CRCs had ARID1A loss, while this was true in only 6% of MSS CRCs. In both MSI‐H and MSS, PD‐L1 expression by stromal cells was enhanced in the ARID1A‐mutant groups (90% vs 39% in MSI‐H, 100% vs 26% in MSS). Conclusion We found a higher mutational burden in ARID1A‐mutant CRCs, and IHC study showed that ARID1A loss was correlated with high PD‐L1 expression in stromal cells regardless of MSI status. These data support the idea that mutant ARID1A is a potential biomarker for CRCs.
Collapse
Affiliation(s)
- Tomohiro Kamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Qingjiang Hu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Hisamatsu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
58
|
Zhang L, Wang Y, Li Z, Lin D, Liu Y, Zhou L, Wang D, Wu A, Li Z. Clinicopathological features of tumor mutation burden, Epstein-Barr virus infection, microsatellite instability and PD-L1 status in Chinese patients with gastric cancer. Diagn Pathol 2021; 16:38. [PMID: 33933102 PMCID: PMC8088709 DOI: 10.1186/s13000-021-01099-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/15/2021] [Indexed: 01/10/2023] Open
Abstract
Objectives Gastric cancer (GC) is the 4th most common type of cancer worldwide. Different GC subtypes have unique molecular features that may have different therapeutic methods. The aim of the present study was to investigate Epstein-Barr virus (EBV) infection, microsatellite instability (MSI) status, the expression of programmed death-ligand 1 (PD-L1) and gene mutations in GC patients. Methods The data of 2504 GC patients, who underwent curative gastrectomy with lymphadenectomy at Peking University Cancer Hospital between 2013 and 2018, were reviewed. We analyzed the clinicopathological factors associated with the immunohistochemistry (IHC) profiles of these patients, and genetic alterations were analyzed using next generation sequencing (NGS). Results Mismatch repair-deficient (d-MMR) GC patients were found to have a higher probability of expressing PD-L1 (p = 0.000, PD-L1 cutoff value = 1%). In addition, 4 and 6.9% of the 2504 gastric cancer patients were EBV-positive and d-MMR, respectively. The number of MLH1/PMS2-negative cases was 126 (6%), and the number of MSH2/MSH6-negative cases was 14 (0.9%). d-MMR status was associated with a intestinal group (p = 0.012), but not with tumor differentiation. Furthermore, MSI and d-MMR GC status (detected by NGS and IHC, respectively) were consistently high, and the rate of MSI was higher in patients with d-MMR GC. A number of genes associated with DNA damage repair were detected in GC patients with MSI, including POLE, ETV6, BRCA and RNF43. In patients with a high tumor mutation burden, the most significantly mutated genes were LRP1B (79.07%), ARID1A (74.42%), RNF43 (69.77%), ZFHX3 (65.12%), TP53 (58.14%), GANS (51.16%), BRCA2 (51.16%), PIK3CA (51.16%), NOTCH1 (51.16%), SMARCA4 (48.84%), ATR (46.51%), POLE (41.86%) and ATM (39.53%). Conclusions Using IHC and NGS, MSI status, protein expression, tumor mutation burden (TMB) and genetic alterations were identified in patients with GC, which provides a theoretical basis for the future clinical treatment of GC.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China
| | - Yinkui Wang
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China
| | - Zhongwu Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China
| | - Dongmei Lin
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China
| | - Yiqiang Liu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China.,Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China
| | - Linxin Zhou
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China
| | - Dongliang Wang
- ChosenMed, Beijing Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Aiwen Wu
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China.
| | - Ziyu Li
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital, No.52 Fucheng Road Haidian District, Beijing, 100142, People's Republic of China.
| |
Collapse
|
59
|
Durán M, Faull I, Lastra E, Laes JF, Rodrigo AB, Sánchez-Escribano R. ARID1A genomic alterations driving microsatellite instability through somatic MLH1 methylation with response to immunotherapy in metastatic lung adenocarcinoma: a case report. J Med Case Rep 2021; 15:89. [PMID: 33608032 PMCID: PMC7896399 DOI: 10.1186/s13256-020-02589-1] [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: 09/21/2020] [Accepted: 11/17/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Tumor molecular screening allows categorization of molecular alterations to select the best therapeutic strategy. AT-rich interactive domain-containing 1A (ARID1A) gene mutations are present in gastric, endometrial, and clear cell ovarian tumors. Inactivation of this gene impairs mismatch repair (MMR) machinery leading to an increased mutation burden that correlates with microsatellite instability (MSI), associated with tumor-infiltrating lymphocytes and programmed death ligand 1 (PD-L1) expression. This is the first case report in lung adenocarcinoma of ARID1A gene alterations leading to sporadic MSI, through somatic mutL homolog 1 (MLH1) promoter methylation, with an MLH1 gene mutation as the second somatic hit. CASE PRESENTATION A 50-year-old never-smoker Bulgarian woman, with no comorbidities and no family history of cancer, was diagnosed with metastatic lung adenocarcinoma. PD-L1 immunohistochemistry (IHC) of tissue biopsies on right groin adenopathies resulted in 30% positivity. Liquid biopsy test reported actionable alterations in ARID1A gene, rearranged during transfection (RET) gene fusions, epidermal growth factor receptor (EGFR) gene R776H mutation, breast cancer (BRCA) genes 1/2, and cyclin-dependent kinase inhibitor 2A (CDKN2A) gene mutations. The patient was treated with immunotherapy, and showed a treatment response lasting for 19 months until a new metastasis appeared at the right deltoid muscle. Genomic analysis of a sample of this metastasis confirmed PD-L1 positivity of greater than 50% with CD8+ T cells expression and showed MSI with a deleterious c.298C>T (p.R100*) MLH1 gene mutation. Multiplex ligation-dependent probe amplification (MLPA) of this sample unveiled MLH1 gene promoter methylation. The MLH1 gene mutation and the MLH1 gene methylation were not present at the germline setting. CONCLUSIONS In this particular case, we show that ARID1A gene mutations with sporadic MSI due to somatic MLH1 gene promoter methylation and MLH1 gene mutation could change the prognosis and define the response to immunotherapy in a patient with lung adenocarcinoma. Comprehensive solid and liquid biopsy tests are useful to find out resistance mechanisms to immune checkpoint inhibitors. Our data encourages the development of new therapies against ARID1A mutations and epigenomic methylation when involved in MSI neoplasms.
Collapse
Affiliation(s)
- Mercedes Durán
- Instituto de Biología Y Genética Molecular, IBGM University of Valladolid, Sanz Y Fores Street, 3, 47003, Valladolid, Spain
| | - Iris Faull
- Guardant Health, 505 Penobscot Dr, Redwood, CA, 94063, USA
| | - Enrique Lastra
- Molecular Tumor Board, Genetic Counselling Unit, Medical Oncology Department, Hospital Universitario de Burgos, Av. Islas Baleares, 3, 09006, Burgos, Spain.
| | | | | | - Ricardo Sánchez-Escribano
- Medical Oncology Department, Hospital Clínico Universitario De Valladolid, Av. Ramón Y Cajal, 3, 47003, Valladolid, Spain
| |
Collapse
|
60
|
Cormedi MCV, Van Allen EM, Colli LM. Predicting immunotherapy response through genomics. Curr Opin Genet Dev 2021; 66:1-9. [DOI: 10.1016/j.gde.2020.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/20/2020] [Accepted: 11/01/2020] [Indexed: 12/21/2022]
|
61
|
Mehrvarz Sarshekeh A, Alshenaifi J, Roszik J, Manyam GC, Advani SM, Katkhuda R, Verma A, Lam M, Willis J, Shen JP, Morris J, Davis JS, Loree JM, Lee HM, Ajani JA, Maru DM, Overman MJ, Kopetz S. ARID1A Mutation May Define an Immunologically Active Subgroup in Patients with Microsatellite Stable Colorectal Cancer. Clin Cancer Res 2021; 27:1663-1670. [PMID: 33414133 DOI: 10.1158/1078-0432.ccr-20-2404] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 11/08/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE AT-rich interactive domain 1A (ARID1A) is commonly mutated in colorectal cancer, frequently resulting in truncation and loss of protein expression. ARID1A recruits MSH2 for mismatch repair during DNA replication. ARID1A deficiency promotes hypermutability and immune activation in preclinical models, but its role in patients with colorectal cancer is being explored. EXPERIMENTAL DESIGN The DNA sequencing and gene expression profiling of patients with colorectal cancer were extracted from The Cancer Genome Atlas and MD Anderson Cancer Center databases, with validation utilizing external databases, and correlation between ARID1A and immunologic features. IHC for T-cell markers was performed on a separate cohort of patients. RESULTS Twenty-eight of 417 patients with microsatellite stable (MSS) colorectal cancer (6.7%) had ARID1A mutation. Among 58 genes most commonly mutated in colorectal cancer, ARID1A mutation had the highest increase with frameshift mutation rates in MSS cases (8-fold, P < 0.001). In MSS, ARID1A mutation was enriched in immune subtype (CMS1) and had a strong correlation with IFNγ expression (Δz score +1.91, P < 0.001). Compared with ARID1A wild-type, statistically significant higher expression for key checkpoint genes (e.g., PD-L1, CTLA4, and PDCD1) and gene sets (e.g., antigen presentation, cytotoxic T-cell function, and immune checkpoints) was observed in mutant cases. This was validated by unsupervised differential expression of genes related to immune response and further confirmed by higher infiltration of T cells in IHC of tumors with ARID1A mutation (P = 0.01). CONCLUSIONS The immunogenicity of ARID1A-mutant cases is likely due to an increased level of neoantigens resulting from increased tumor mutational burden and frameshift mutations. Tumors with ARID1A mutation may be more susceptible to immune therapy-based treatment strategies and should be recognized as a unique molecular subgroup in future immune therapy trials.
Collapse
Affiliation(s)
- Amir Mehrvarz Sarshekeh
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jumanah Alshenaifi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ganiraju C Manyam
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Riham Katkhuda
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anuj Verma
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Lam
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Willis
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Morris
- Department of Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer S Davis
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jonathan M Loree
- Division of Medical Oncology, BC Cancer, Vancouver, British Columbia
| | - Hey Min Lee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dipen M Maru
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
62
|
Xu S, Tang C. The Role of ARID1A in Tumors: Tumor Initiation or Tumor Suppression? Front Oncol 2021; 11:745187. [PMID: 34671561 PMCID: PMC8521028 DOI: 10.3389/fonc.2021.745187] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/08/2021] [Indexed: 01/08/2023] Open
Abstract
Genes encoding subunits of SWItch/Sucrose Non-Fermenting (SWI/SNF) chromatin remodeling complexes are collectively mutated in 20% of all human cancers, among which the AT-rich interacting domain-containing protein 1A (ARID1A, also known as BAF250a, B120, C1orf4, Osa1) that encodes protein ARID1A is the most frequently mutated, and mutations in ARID1A have been found in various types of cancer. ARID1A is thought to play a significant role both in tumor initiation and in tumor suppression, which is highly dependent upon context. Recent molecular mechanistic research has revealed that ARID1A participates in tumor progression through its effects on control of cell cycle, modulation of cellular functions such as EMT, and regulation of various signaling pathways. In this review, we synthesize a mechanistic understanding of the role of ARID1A in human tumor initiation as well as in tumor suppression and further discuss the implications of these new discoveries for potential cancer intervention. We also highlight the mechanisms by which mutations affecting the subunits in SWI/SNF complexes promote cancer.
Collapse
|
63
|
Zhang Y, Lin A, Li Y, Ding W, Meng H, Luo P, Zhang J. Age and Mutations as Predictors of the Response to Immunotherapy in Head and Neck Squamous Cell Cancer. Front Cell Dev Biol 2020; 8:608969. [PMID: 33363171 PMCID: PMC7755718 DOI: 10.3389/fcell.2020.608969] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022] Open
Abstract
The immunosuppressive tumor microenvironment plays an essential role in the treatment of head and neck squamous cell carcinoma (HNSC). Compared to traditional chemoradiotherapy, immune checkpoint inhibitors (ICIs) have become increasingly important in HNSC therapy. Prior studies linked the efficacy of ICIs to PD-L1, microsatellite instability (MSI), HPV infection, tumor mutation burden (TMB), and tumor lymphocyte infiltration in patients with HNSC, but further verification is needed. Additional predictors are needed to recognize HNSC patients with a good response to ICIs. We collected the clinical information and mutation data of HNSC patients from Memorial Sloan Kettering Cancer Center (MSKCC) and The Cancer Genome Atlas (TCGA) databases to generate two clinical cohorts. The MSKCC cohort was used to recognize predictors related to the efficacy of ICIs, and the TCGA cohort was used to further examine the immune microenvironment features and signaling pathways that are significantly enriched in the subgroups of predictors. Multivariate Cox regression analysis indicated that age (HR = 0.50, p = 0.014) and ARID1A (HR = 0.13, p = 0.048), PIK3CA (HR = 0.45, p = 0.021), and TP53 (HR = 1.82, p = 0.035) mutations were potential predictors for ICI efficacy in HNSC patients. Age > 65 years and ARID1A or PIK3CA mutations correlated with good overall survival (OS). TP53 mutant-type (MT) patients experienced a worse prognosis than TP53 wild-type (WT) patients. The subgroups associated with a good prognosis (age > 65 years, ARID1A-MT, and PIK3CA-MT) universally had a high TMB and increased expression of immune checkpoint molecules. Although TP53-MT was associated with a high TMB, the expression of most immune checkpoint molecules and immune-related genes was lower in TP53-MT patients than TP53-WT patients, which may reflect low immunogenicity. Pathways related to the immunosuppressive tumor microenvironment were mostly enriched in the subgroups associated with a poor prognosis (age ≤ 65 years, low TMB, ARID1A-WT, PIK3CA-WT, and TP53-MT). In conclusion, the factors age > 65 years, PIK3CA-MT, and ARID1A-MT predicted favorable efficacy for ICI treatment in HNSC patients, and TP53 mutation was a negative predictor.
Collapse
Affiliation(s)
- Yueming Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yonghe Li
- Department of Otolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weimin Ding
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Meng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
64
|
Fang J, Hu M, Sun Y, Zhou S, Li H. Expression Profile Analysis of m6A RNA Methylation Regulators Indicates They Are Immune Signature Associated and Can Predict Survival in Kidney Renal Cell Carcinoma. DNA Cell Biol 2020; 39:2194-2211. [PMID: 33085515 DOI: 10.1089/dna.2020.5767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
N6-Methyladenosine (m6A) refers to the methylation modification occurring at the nitrogen-6 position of adenosine. Many human physiological processes such as modulation of spermatogenesis are caused by m6A RNA modifications. However, the relationship between m6A RNA methylation regulators and kidney renal clear cell carcinoma (KIRC) remains rarely investigated. This work aimed to explore the influence of m6A RNA methylation regulators in KIRC. We examined abnormally expressed m6A RNA methylation regulators among different clinicopathological features of KIRC. We recognized three subgroups (KIRC1, KIRC2, and KIRC3) with significant differences in overall survival through consensus clustering of m6A RNA methylation regulators. Surprisingly, KIRC2 displayed elevated immune activity, but high proportions of immune-inhibitory cells (Tregs and myeloid-derived suppressor cell) based on single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT analysis. Moreover, the KIRC2 subgroup had the lowest tumor mutation burden levels and the highest expression levels of 80% (12/15) of co-inhibitory molecules. Next, correlation analysis indicated that RBM15B expression was negatively correlated with multiple immune signatures, which was verified by ssGSEA and CIBERSORT analyses. Multiple immune-related and cancer-related pathways were enriched in the group with high RBM15B expression. Furthermore, a four-m6A RNA methylation regulator-based risk signature was constructed based on an ArrayExpress (E-MTAB-3267) dataset and confirmed in the The Cancer Genome Atlas (TCGA) testing cohort. In conclusion, our study successfully classified TCGA samples into three subgroups with different immune signatures, and suggested that the worse prognosis of KIRC2 is probably mediated by immune evasion. These findings will facilitate personalized immunotherapy in patients with KIRC. In addition, the risk score system was revealed as an independent prognostic marker that can predict survival in KIRC patients.
Collapse
Affiliation(s)
- Jiuyuan Fang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Mingyang Hu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yan Sun
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Sijie Zhou
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Huixiang Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
65
|
Nemtsova MV, Mikhaylenko DS, Kuznetsova EB, Bykov II, Zamyatnin AA. Inactivation of Epigenetic Regulators due to Mutations in Solid Tumors. BIOCHEMISTRY (MOSCOW) 2020; 85:735-748. [PMID: 33040718 DOI: 10.1134/s0006297920070020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Main factors involved in carcinogenesis are associated with somatic mutations in oncogenes and tumor suppressor genes representing changes in the DNA nucleotide sequence. Epigenetic changes, such as aberrant DNA methylation, modifications of histone proteins, and chromatin remodeling, are equally important in the development of human neoplasms. From this perspective, mutations in the genes encoding key participants of epigenetic regulation are of particular interest including enzymes that methylate/demethylate DNA, enzymes that covalently attach or remove regulatory signals from histones, components of nucleosome remodeling multiprotein complexes, auxiliary proteins and cofactors of the above-mentioned molecules. This review describes both germline and somatic mutations in the key epigenetic regulators with emphasis on the latter ones in the solid human tumors, as well as considers functional consequences of these mutations on the cellular level. In addition, clinical associations of the somatic mutations in epigenetic regulators are presented, as well as DNA diagnostics of hereditary cancer syndromes due to germline mutations in the SMARC proteins and chemotherapy drugs directly affecting the altered epigenetic mechanisms for treatment of patients with solid neoplasms. The review is intended for a wide range of molecular biologists, geneticists, oncologists, and associated specialists.
Collapse
Affiliation(s)
- M V Nemtsova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia.,Research Centre for Medical Genetics, Moscow, 115478, Russia
| | - D S Mikhaylenko
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia. .,Research Centre for Medical Genetics, Moscow, 115478, Russia
| | - E B Kuznetsova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| | - I I Bykov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| | - A A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
66
|
Yan H, Chen Y, Wang K, Yu L, Huang X, Li Q, Xie Y, Lin J, He Y, Yi X, Wang Y, Chen L, Ding Y, Li Y. Identification of immune landscape signatures associated with clinical and prognostic features of hepatocellular carcinoma. Aging (Albany NY) 2020; 12:19641-19659. [PMID: 33049716 PMCID: PMC7732284 DOI: 10.18632/aging.103977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 08/14/2020] [Indexed: 01/24/2023]
Abstract
While cancer immunotherapy has been remarkably successful in some malignancies, some cancers derive limited benefit from current immunotherapies. Here, we combined immune landscape signatures with hepatocellular carcinoma clinical and prognostic features to classify them into distinct subtypes. The immunogenomic profiles, stromal cell features and immune cell composition of the subtypes were then systematically analyzed. Two independent prognostic indexes were established based on 6 immune-related genes and 17 differentially expressed genes associated with stromal cell content. These indexes were significantly correlated with tumor mutation burden, deficient DNA mismatch repair and microsatellite instability. In addition, tumor-infiltrating lymphocytes, including activated NK cells, resting memory CD4 T-cells, eosinophils, and activated mast cells were significantly correlated with hepatocellular carcinoma survival. In conclusion, we have comprehensively described the immune landscape signatures and identified prognostic immune-associated biomarkers of hepatocellular carcinoma. Our findings highlight potential novel avenues for improving responses to immunotherapy.
Collapse
Affiliation(s)
- Hongmei Yan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuchuan Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kai Wang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lu Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xixin Huang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qianyu Li
- Medical Imaging Specialty, the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yuwen Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China,Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiayu Lin
- Clinical Medicine Specialty, the First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yueyun He
- Medical Imaging Specialty, the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xinyu Yi
- Clinical Medicine Specialty, the First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yanzhi Wang
- Medical Imaging Specialty, the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Longhua Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yiyi Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
67
|
Wang T, Gao X, Zhou K, Jiang T, Gao S, Liu P, Zuo X, Shi X. Role of ARID1A in epithelial‑mesenchymal transition in breast cancer and its effect on cell sensitivity to 5‑FU. Int J Mol Med 2020; 46:1683-1694. [PMID: 33000179 PMCID: PMC7521577 DOI: 10.3892/ijmm.2020.4727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
Abstract
The loss of function mutation of AT‑rich interactive domain 1A (ARID1A) often occurs in patients with breast cancer. It has been found that ARID1A knockout can enhance both the migratory activity of renal carcinoma cells and their sensitivity to therapeutic drugs by promoting epithelial-mesenchymal transition (EMT); however, its mechanisms of action in breast cancer remain unclear. In the present study, immunohistochemistry and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) revealed that the expression of ARID1A in breast cancer tissues was significantly lower than that in paracancerous tissues, and patients with a low ARID1A expression had a lower survival rate. ARID1A was expressed at low levels in breast cancer cells. In addition, siRNA targeting ARID1A (siARID1A) and ARID1A overexpression vector were transfected into MCF7 and MDA‑MB‑231 cells, respectively. Proliferation assay revealed that ARID1A silencing increased cell viability and partially reversed the inhibitory effects of 5‑fluorouracil (5‑FU) on the MCF7 cells, while ARID1A overexpression exerted an opposite effect on the MDA‑MB‑231 cells. ARID1A silencing promoted proliferation, migration, invasion and angiogenesis, and partly reversed the inhibitory effects of 5‑FU on cell biological behaviors, while the overexpression of ARID1A further enhanced the inhibitory effect of 5‑FU on the cells. Furthermore, ARID1A regulated the migration and invasion of breast cancer cells through EMT. On the whole, the findings of the present study demonstrate that ARID1A exerts an antitumor effect on breast cancer, and its overexpression can enhance the sensitivity of breast cancer cells to 5‑FU.
Collapse
Affiliation(s)
- Tangshun Wang
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Xiang Gao
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Kexin Zhou
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Tao Jiang
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Shuang Gao
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Pengzhou Liu
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Ximeng Zuo
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Xiaoguang Shi
- Department of General Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| |
Collapse
|
68
|
Biomarkers for immune checkpoint therapy targeting programmed death 1 and programmed death ligand 1. Biomed Pharmacother 2020; 130:110621. [PMID: 34321165 DOI: 10.1016/j.biopha.2020.110621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Rapidly increasing usages of immune checkpoint therapy for cancer treatment, particularly monoclonal antibodies that target programmed cell death-1 (PD-1) and its ligand PD-L1, have been achieved due to startling durable therapeutic efficacy with limited toxicity. The therapeutics significantly prolonged the overall survival and progression free survival of patients across multiple cancer types. However, the objective response rate of patients receiving this kind of treatment is substantially low. Therefore, it is of great importance to exploit reliable biomarkers that can robustly predict the therapeutic effects. Several biomarkers have been characterized for the selection of patients, which is mainly based on immunological and genetic criteria. Herein, we focus on the current progress regarding the biomarkers for anti-PD-1/PD-L1 therapy.
Collapse
|
69
|
The Clinicopathological Characteristics And Genetic Alterations of Signet-ring Cell Carcinoma in Gastric Cancer. Cancers (Basel) 2020; 12:cancers12082318. [PMID: 32824568 PMCID: PMC7463705 DOI: 10.3390/cancers12082318] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/04/2020] [Accepted: 08/12/2020] [Indexed: 02/08/2023] Open
Abstract
Signet-ring cell carcinoma (SRC) in advanced gastric cancer (GC) is often associated with more invasiveness and a worse prognosis than other cell types. The genetic alterations associated with gastric carcinogenesis in SRC are still unclear. In this study, 441 GC patients receiving curative surgery for GC between 2005 and 2013 were enrolled. The clinicopathological characteristics and genetic alterations of GC patients with and without SRC were compared. Among the 441 GC patients, 181 had SRC. For early GC, patients with SRC had more tumors located in the middle and lower stomach, more infiltrating tumors and better overall survival (OS) rates than those without SRC. For advanced GC, patients with SRC had more scirrhous type tumors, more PIK3CA amplifications, fewer microsatellite instability-high (MSI-H) tumors, more peritoneal recurrences and worse 5-year OS rates than those without SRC. For advanced GC with SRC, patients with peritoneal recurrence tended to have PD-L1 expression. For advanced GC without SRC, patients with liver metastasis tended to have PD-L1 expression, PI3K/AKT pathway mutations, TP53 mutations and MSI-H tumors. For advanced GC, PD-L1 expression was associated with peritoneal recurrence in SRC tumors, while non-SRC tumors with liver metastasis were likely to have PI3K/AKT pathway mutations, TP53 mutations and PD-L1 expression; immunotherapy and targeted therapy may be beneficial for these patients.
Collapse
|
70
|
Sun D, Tian L, Zhu Y, Wo Y, Liu Q, Liu S, Li H, Hou H. Subunits of ARID1 serve as novel biomarkers for the sensitivity to immune checkpoint inhibitors and prognosis of advanced non-small cell lung cancer. Mol Med 2020; 26:78. [PMID: 32791957 PMCID: PMC7425138 DOI: 10.1186/s10020-020-00208-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction Patients with advanced non-small cell lung cancer (NSCLC) benefit from treatment with immune checkpoint inhibitors (ICIs). Biomarkers such as programmed death-ligand 1 (PD-L1), the tumor mutational burden (TMB) and the mismatch repair (MMR) status are used to predict the prognosis of ICIs therapy. Nevertheless, novel biomarkers need to be further investigated, and a systematic prognostic model is needed for the evaluation of the survival risks of ICIs treatment. Methods A cohort of 240 patients who received ICIs from the cBioPortal for Cancer Genomics was evaluated in this research. Clinical information and targeted sequencing data were acquired for analyses. The Kaplan-Meier plot method was used to perform survival analyses, and selected variables were then confirmed by a novel nomogram constructed by the “rms” package of R software. Results Seven percent of the NSCLC patients harbored ARID1A mutations, while 4% of the NSCLC patients harbored ARID1B mutations. Mutations in ARID1A and ARID1B were confirmed to be associated with sensitivity to ICIs. Patients harboring these mutations were found to have a better response to treatment (ARID1A: P = 0.045; ARID1B: P = 0.034) and prolonged progression-free survival (ARID1B: P = 0.032). Here, a novel nomogram was constructed to predict the prognosis of ICIs treatment. Elevation of the TMB, enhanced expression of PD-L1 and activation of the antigen presentation process and cellular immunity were found to be correlated with ARID1A and ARID1B mutations. Conclusion ARID1A and ARID1B could serve as novel biomarkers for the prognosis and sensitivity to ICIs of advanced NSCLC.
Collapse
Affiliation(s)
- Dantong Sun
- Precision Medicine Center of Oncology, the Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China
| | - Lu Tian
- College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China
| | - Yan Zhu
- Department of Medical Oncology, the Municipal Hospital of Qingdao, Qingdao, 266000, China
| | - Yang Wo
- Department of Thoracic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Qiaoling Liu
- Department of Medical Oncology, Qingdao West Coast New Area Central Hospital, Qingdao, 266555, China
| | - Shihai Liu
- Medical Animal Laboratory, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Hong Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| | - Helei Hou
- Precision Medicine Center of Oncology, the Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China.
| |
Collapse
|
71
|
Wang L, Qu J, Zhou N, Hou H, Jiang M, Zhang X. Effect and biomarker of immune checkpoint blockade therapy for ARID1A deficiency cancers. Biomed Pharmacother 2020; 130:110626. [PMID: 32791396 DOI: 10.1016/j.biopha.2020.110626] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022] Open
Abstract
The AT-rich interaction domain 1A (ARID1A) are frequently mutates across a broad spectrum of cancers. The majority of ARID1A mutations are inactivating mutations and lead to loss expression of the ARID1A protein. To date, clinical applicable targeted cancer therapy based on ARID1A mutational status has not been described. With increasing number of studies reported that the ARID1A deficiency may be a novel predictive biomarker for immune checkpoint blockade (ICB) treatment. ARID1A deficiency would compromise mismatch repair pathway and increase the number of tumor-infiltrating lymphocytes, tumor mutation burden and expression of programmed cell death ligand 1 (PD-L1) in some cancers, which would suggested cooperate with ICB treatment. In this review, we summarize the relationship between ARID1A deficiency and ICB treatment including potential mechanisms, potential therapeutic combination, and the biomarker value of ARID1A deficiency.
Collapse
Affiliation(s)
- Li Wang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Jialin Qu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Na Zhou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Helei Hou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Man Jiang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Xiaochun Zhang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China.
| |
Collapse
|
72
|
Pinto MP, Córdova-Delgado M, Retamal IN, Muñoz-Medel M, Bravo ML, Durán D, Villanueva F, Sanchez C, Acevedo F, Mondaca S, Koch E, Ibañez C, Galindo H, Madrid J, Nervi B, Peña J, Torres J, Owen GI, Corvalán AH, Armisén R, Garrido M. A Molecular Stratification of Chilean Gastric Cancer Patients with Potential Clinical Applicability. Cancers (Basel) 2020; 12:E1863. [PMID: 32664343 PMCID: PMC7408697 DOI: 10.3390/cancers12071863] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a complex and heterogeneous disease. In recent decades, The Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG) defined GC molecular subtypes. Unfortunately, these systems require high-cost and complex techniques and consequently their impact in the clinic has remained limited. Additionally, most of these studies are based on European, Asian, or North American GC cohorts. Herein, we report a molecular classification of Chilean GC patients into five subtypes, based on immunohistochemical (IHC) and in situ hybridization (ISH) methods. These were Epstein-Barr virus positive (EBV+), mismatch repair-deficient (MMR-D), epithelial to mesenchymal transition (EMT)-like, and accumulated (p53+) or undetected p53 (p53-). Given its lower costs this system has the potential for clinical applicability. Our results confirm relevant molecular alterations previously reported by TCGA and ACRG. We confirm EBV+ and MMR-D patients had the best prognosis and could be candidates for immunotherapy. Conversely, EMT-like displayed the poorest prognosis; our data suggest FGFR2 or KRAS could serve as potential actionable targets for these patients. Finally, we propose a low-cost step-by-step stratification system for GC patients. To the best of our knowledge, this is the first Latin American report on a molecular classification for GC. Pending further validation, this stratification system could be implemented into the routine clinic.
Collapse
Affiliation(s)
- Mauricio P. Pinto
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Miguel Córdova-Delgado
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Faculty of Chemical & Pharmaceutical Sciences, Universidad de Chile, Santiago 8380494, Chile
| | - Ignacio N. Retamal
- Faculty of Dentistry, Universidad de los Andes, Santiago 7620001, Chile;
| | - Matías Muñoz-Medel
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - M. Loreto Bravo
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Doris Durán
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago 7510157, Chile;
| | - Francisco Villanueva
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - César Sanchez
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Francisco Acevedo
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Sebastián Mondaca
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Erica Koch
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Carolina Ibañez
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Héctor Galindo
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Jorge Madrid
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Bruno Nervi
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - José Peña
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| | - Javiera Torres
- Department of Pathology, Faculty of Medicine Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
| | - Gareth I. Owen
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8330034, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Alejandro H. Corvalán
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8330034, Chile
| | - Ricardo Armisén
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7590943, Chile;
| | - Marcelo Garrido
- Department of Hematology & Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile; (M.P.P.); (M.C.-D.); (M.M.-M.); (M.L.B.); (F.V.); (C.S.); (F.A.); (S.M.); (E.K.); (C.I.); (H.G.); (J.M.); (B.N.); (J.P.); (G.I.O.); (A.H.C.)
| |
Collapse
|
73
|
Li Z, Lin J, Zhang L, Li J, Zhang Y, Zhao C, Wang H. Comprehensive analysis of multiple parameters associated with tumor immune microenvironment in ARID1A mutant cancers. Future Oncol 2020; 16:2295-2306. [PMID: 32639175 DOI: 10.2217/fon-2020-0243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To verify the relationship between ARID1A and tumor immune microenvironment thus immune checkpoint inhibitors (ICIs) response. Material & methods: Several public databases were used to characterize the association between ARID1A gene alteration and tumor immunity. Results: The gene mutation frequency was 8.2% in all cancer types. The ARID1A-mutated cancers have higher scores of mutation count, tumor mutational burden, neoantigen load (p < 0.001) and T cell repertoire, B cell repertoire diversity (p < 0.05). The gene mutation has tight association with multiple-activated immune cells. Survival analysis suggested that patients with ARID1A mutant cancers benefit more from ICIs treatment (p = 0.013). Conclusion: The ARID1A gene mutation was correlated with higher tumor immunogenicity and activated antitumor immune microenvironment, resulting in superior cohort that respond well to ICIs.
Collapse
Affiliation(s)
- Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Jiamao Lin
- Department of Medical Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Lijuan Zhang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, PR China
| | - Jingchao Li
- Department of Radiation Oncology, The People's Hospital of Zhangqiu Area, Jinan 250200, China
| | - Yingyun Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Chenglong Zhao
- Department of pathology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Haiyong Wang
- Department of Medical Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| |
Collapse
|
74
|
Correlation of PD-L1 expression with immunohistochemically determined molecular profile in endometrial carcinomas. Virchows Arch 2020; 477:845-856. [PMID: 32594230 DOI: 10.1007/s00428-020-02867-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/23/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022]
Abstract
Endometrial carcinoma programmed death-ligand 1 (PD-L1) expression in tumor cells (TCs) and tumor-associated inflammatory cells (ICs) have recently been reported in several studies which vary in terms of their cohort size, design, and methodology. We aimed to assess PD-L1 staining in endometrial carcinomas and correlate this with clinical and pathological factors and PTEN, ARID1A, p53, and MMR protein expression. PD-L1 immunohistochemistry was performed on whole tissue sections of all tumor blocks of 59 consecutive unselected endometrial carcinomas between November 2018 and September 2019. TC and IC PD-L1 positivity with a 1% cut-off value was observed in 10.2% and 67.8% of cases, respectively, and with a 5% cut-off value in 3.4% and 42.4% of cases, respectively. TC PD-L1 positivity with both 1% and 5% cut-off values was significantly related to ARID1A loss (p = 0.001 and p = 0.046, respectively). IC PD-L1 positivity with 1% and 5% cut-off values and combined score were significantly associated with MMR protein deficiency (p = 0.041, p = 0.031, and p = 0.028, respectively). Advanced stage tumors exhibited more frequent PD-L1 expression in ICs (p = 0.039). MELF-type myometrial invasion pattern was more common in tumors with ARID1A loss (p = 0.047). We observed higher rates of IC PD-L1 positivity in endometrial carcinomas than documented in prior studies; this may be related to our usage of "recent" paraffin blocks and whole tissue sections of all tumor blocks. There was a much higher PD-L1 expression in the ICs compared to TCs in our cases. We confirm a previously documented association between MMR deficiency and PD-L1 expression and show a novel association between ARID1A loss and PD-L1 expression in endometrial carcinomas. ARID1A loss represents a potential biomarker of immune checkpoint inhibitor response in endometrial carcinoma.
Collapse
|
75
|
Chen B, Li L, Li M, Wang X. HIF1A expression correlates with increased tumor immune and stromal signatures and aggressive phenotypes in human cancers. Cell Oncol (Dordr) 2020; 43:877-888. [PMID: 32488852 DOI: 10.1007/s13402-020-00534-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although many studies have revealed essential roles of HIF1A (hypoxia inducible factor 1 subunit alpha) in regulating various pathways associated with cancer development, a systematic investigation of associations of HIF1A expression with tumor immunity, the tumor microenvironment and other tumor phenotypes in pan-cancer is lacking. METHODS Using cancer genomics datasets of 10 cancer cohorts from the Cancer Genome Atlas (TCGA) program, we investigated associations of HIF1A expression with tumor immune and stromal signatures, and various tumor phenotypes, including cell proliferation, stemness, epithelial-mesenchymal transition (EMT), tumor purity, oncogenic signaling, and clinical outcomes. RESULTS HIF1A upregulation was found to be associated with increased immune and stromal signatures, aggressive phenotypes, and worse survival rates in various cancers. Moreover, HIF1A upregulation was not only associated with activation of various oncogenic signaling pathways, but also with increased tumor suppressive signatures, including apoptosis and anti-tumor immune response, indicative of a dual role of HIF1A in cancer development. However, HIF1A expression showed a stronger correlation with immune-inhibiting signatures than with immune-promoting signatures. Furthermore, HIF1A expression was found to be positively associated with both tumor immune infiltration and PD-L1 expression, indicating that tumors with elevated expression of HIF1A may exhibit a more active immunotherapy response. This indication was substantiated in a kidney cancer cohort receiving anti-PD-1/PD-L1 immunotherapy. CONCLUSIONS HIF1A upregulation correlates with increased tumor immune and stromal signatures and aggressive phenotypes in human cancers. Our analysis may provide new insights into the role of HIF1A in tumor biology and clinical management.
Collapse
Affiliation(s)
- Baotao Chen
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Lin Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Mengyuan Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China. .,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China. .,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
76
|
Abou Alaiwi S, Nassar AH, Xie W, Bakouny Z, Berchuck JE, Braun DA, Baca SC, Nuzzo PV, Flippot R, Mouhieddine TH, Spurr LF, Li YY, Li T, Flaifel A, Steinharter JA, Margolis CA, Vokes NI, Du H, Shukla SA, Cherniack AD, Sonpavde G, Haddad RI, Awad MM, Giannakis M, Hodi FS, Liu XS, Signoretti S, Kadoch C, Freedman ML, Kwiatkowski DJ, Van Allen EM, Choueiri TK. Mammalian SWI/SNF Complex Genomic Alterations and Immune Checkpoint Blockade in Solid Tumors. Cancer Immunol Res 2020; 8:1075-1084. [PMID: 32321774 DOI: 10.1158/2326-6066.cir-19-0866] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/10/2020] [Accepted: 04/16/2020] [Indexed: 02/05/2023]
Abstract
Prior data have variably implicated the inactivation of the mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) complex with increased tumor sensitivity to immune checkpoint inhibitors (ICI). Herein, we examined the association between mSWI/SNF variants and clinical outcomes to ICIs. We correlated somatic loss-of-function (LOF) variants in a predefined set of mSWI/SNF genes (ARID1A, ARID1B, SMARCA4, SMARCB1, PBRM1, and ARID2) with clinical outcomes in patients with cancer treated with systemic ICIs. We identified 676 patients from Dana-Farber Cancer Institute (DFCI, Boston, MA) and 848 patients from a publicly available database from Memorial Sloan Kettering Cancer Center (MSKCC, New York, NY) who met the inclusion criteria. Multivariable analyses were conducted and adjusted for available baseline factors and tumor mutational burden. Median follow-up was 19.6 (17.6-22.0) months and 28.0 (25.0-29.0) months for the DFCI and MSKCC cohorts, respectively. Seven solid tumor subtypes were examined. In the DFCI cohort, LOF variants of mSWI/SNF did not predict improved overall survival (OS), time-to-treatment failure (TTF), or disease control rate. Only patients with renal cell carcinoma with mSWI/SNF LOF showed significantly improved OS and TTF with adjusted HRs (95% confidence interval) of 0.33 (0.16-0.7) and 0.49 (0.27-0.88), respectively, and this was mostly driven by PRBM1 In the MSKCC cohort, where only OS was captured, LOF mSWI/SNF did not correlate with improved outcomes across any tumor subtype. We did not find a consistent association between mSWI/SNF LOF variants and improved clinical outcomes to ICIs, suggesting that mSWI/SNF variants should not be considered as biomarkers of response to ICIs.
Collapse
Affiliation(s)
- Sarah Abou Alaiwi
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amin H Nassar
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Wanling Xie
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ziad Bakouny
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacob E Berchuck
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David A Braun
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sylvan C Baca
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Pier Vitale Nuzzo
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genoa, Italy
| | - Ronan Flippot
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Tarek H Mouhieddine
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Liam F Spurr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yvonne Y Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Taiwen Li
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Abdallah Flaifel
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - John A Steinharter
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Claire A Margolis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Natalie I Vokes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Heng Du
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sachet A Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Andrew D Cherniack
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Guru Sonpavde
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert I Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Melanoma Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - X Shirley Liu
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Cigall Kadoch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|