1
|
Hailin L, Yiting C, Yue W, Lijun L, Renlu Z, Yunhan C, Yanyang Z, Qiuyu Z. Ly6E on tumor cells impairs anti-tumor T-cell responses: a novel mechanism of tumor-induced immune exclusion. Cancer Immunol Immunother 2024; 74:4. [PMID: 39487896 DOI: 10.1007/s00262-024-03851-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Lymphocyte antigen 6 complex, locus E (Ly6E) has been initially demonstrated to involve in T cell activity and impair viral infectivity. Recently, high expression levels of Ly6E have been reported in tumor microenvironment (TME) of various types of cancers. However, the immunoregulatory mechanism of Ly6E manipulating TME remains unknown. METHODS TCGA database and Kaplan-Meier plotter database were used to evaluate the correlation between Ly6E expression levels and cancer patient survival. After analyzing Ly6E expression levels in human breast cancer tissues and tumor cell lines, we generated Ly6E knockout (KO) and overexpression (OE) mouse cell lines. Cell proliferation ability in vitro and the ability of growth and metastasis in mouse tumor models were compared between KO/OE and wild-type tumor cells. On day 7 after tumor implantation, tumor tissues were separated for flow cytometric assay, bulk RNA sequencing and single-cell RNA sequencing (ScRNA-seq). The role of Ly6E-expressing tumor cell on macrophage was analyzed in vitro. RESULTS Our result surprisingly found that high Ly6E expression levels were associated with CD8+ T cell exclusion in tumor tissues and resistance to immunotherapy. Our data showed that knockout of Ly6E in tumor cells prompts tumor regression and inhibits tumor metastases, and Ly6E-OE tumor cells vice versa. The enhanced anti-tumor effect of Ly6E knockout in tumor cells was dependent on T cell response and formed long-lasting memory. The increase in the CD8+ T-cell infiltration into the tumor islet of Ly6E-KO tumors confirmed the role of Ly6E on T cell exclusion. ScRNA-seq analysis showed that M2 macrophages are particularly abundant in the Ly6E-expressing tumor tissues, especially M2-4 macrophage cluster identified by high levels of Arg-1, indicates that Ly6E-expressing tumor cells might restrict T cell infiltration via M2 macrophages. Moreover, in vitro assay showed that cell culture media derived from Ly6E-positive tumor cells promoted macrophage migration and M2 polarization. CONCLUSION Our study illuminated that Ly6E-expressing tumor cells facilitated the accumulation of M2 macrophages in TME, which contributes to CD8+ T cell exclusion and provides new insights for improving efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Lan Hailin
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Chen Yiting
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Wu Yue
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Li Lijun
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
| | - Zhang Renlu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Chen Yunhan
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
| | - Zhu Yanyang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China
| | - Zhang Qiuyu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350122, China.
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
2
|
Fan Y, Xiao Y, Zhang Y, Chen J, Huang S, Bai W. Transcriptomic and multi-cytokines profile analysis revealed new insights into the integrating mechanisms of cyanidin-3-O-glucoside on male reproductive damage amelioration. Food Res Int 2024; 192:114802. [PMID: 39147501 DOI: 10.1016/j.foodres.2024.114802] [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: 05/10/2024] [Revised: 06/28/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Ulcerative colitis is a public health issue with a rising worldwide incidence. It has been found that current medications for treating UC may cause varying degrees of damage to male fertility. Our previous study demonstrated that cyanidin-3-O-glucoside (C3G) treatment could effectively restore reproductive damage in a mouse model of DSS induced colitis. However, the underlying mechanism of C3G alleviates UC induced male reproductive disorders remain scarce. The aim of this study is to discover the molecular mechanisms of C3G on the amelioration of UC stimulated reproductive disorders. The targeted genes toward UC-induced reproductive injury upon C3G treatments were explored by transcriptomic analysis. Hematological analysis, histopathological examination, and real time transcription-polymerase chain reaction (RT-PCR) analysis were applied for conjoined identification. Results showed that C3G may effectively target for reducing pro-inflammatory cytokine IL-6 in testis through cytokine-cytokine receptor interaction pathway. Transcriptome sequencing found that a series of genetic pathways involved in the protective effects of C3G on male reproduction were identified by gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Further results presented that C3G could effectively restore mRNA expression levels of Ly6a and Col1a1, closely linked with UC induced male reproductive damage pathways. Sufficient results implied that Ly6a and Col1a1 may be treated as the promising therapeutic targets for the mechanism of C3G in treating UC induced reproductive impairment. C3G administration might be an effective dietary supplementation strategy for male reproduction improvement.
Collapse
Affiliation(s)
- Yueyao Fan
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yuhang Xiao
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yingying Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiali Chen
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Sammi Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Weibin Bai
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
3
|
Sanvicente García A, Pedregal M, Paniagua-Herranz L, Díaz-Tejeiro C, Nieto-Jiménez C, Pérez Segura P, Munkácsy G, Győrffy B, Calvo E, Moreno V, Ocaña A. Clinical and Immunologic Characteristics of Colorectal Cancer Tumors Expressing LY6G6D. Int J Mol Sci 2024; 25:5345. [PMID: 38791382 PMCID: PMC11121234 DOI: 10.3390/ijms25105345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The identification of targets that are expressed on the cell membrane is a main goal in cancer research. The Lymphocyte Antigen 6 Family Member G6D (LY6G6D) gene codes for a protein that is mainly present on the surface of colorectal cancer (CRC) cells. Therapeutic strategies against this protein like the development of T cell engagers (TCE) are currently in the early clinical stage. In the present work, we interrogated public genomic datasets including TCGA to evaluate the genomic and immunologic cell profile present in tumors with high expression of LY6G6D. We used data from TCGA, among others, and the Tumor Immune Estimation Resource (TIMER2.0) platform for immune cell estimations and Spearman correlation tests. LY6G6D expression was exclusively present in CRC, particularly in the microsatellite stable (MSS) subtype, and was associated with left-side tumors and the canonical genomic subgroup. Tumors with mutations of APC and p53 expressed elevated levels of LY6G6D. This protein was expressed in tumors with an inert immune microenvironment with an absence of immune cells and co-inhibitory molecules. In conclusion, we described clinical, genomic and immune-pathologic characteristics that can be used to optimize the clinical development of agents against this target. Future studies should be performed to confirm these findings and potentially explore the suggested clinical development options.
Collapse
Affiliation(s)
- Adrián Sanvicente García
- Experimental Therapeutics in Cancer Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.S.G.); (L.P.-H.); (C.D.-T.); (C.N.-J.)
- Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Manuel Pedregal
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.P.); (E.C.); (V.M.)
| | - Lucía Paniagua-Herranz
- Experimental Therapeutics in Cancer Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.S.G.); (L.P.-H.); (C.D.-T.); (C.N.-J.)
| | - Cristina Díaz-Tejeiro
- Experimental Therapeutics in Cancer Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.S.G.); (L.P.-H.); (C.D.-T.); (C.N.-J.)
| | - Cristina Nieto-Jiménez
- Experimental Therapeutics in Cancer Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.S.G.); (L.P.-H.); (C.D.-T.); (C.N.-J.)
| | - Pedro Pérez Segura
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Gyöngyi Munkácsy
- Department of Bioinformatics, Semmelweis University, H-1094 Budapest, Hungary; (G.M.); (B.G.)
- Department of Pediatrics, Semmelweis University, H-1094 Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, H-1094 Budapest, Hungary; (G.M.); (B.G.)
- Department of Pediatrics, Semmelweis University, H-1094 Budapest, Hungary
- Research Centre for Natural Sciences, Institute of Enzymology, H-1117 Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Emiliano Calvo
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.P.); (E.C.); (V.M.)
- START Madrid-HM Centro Integral Oncológico Clara Campal (CIOCC), Early Phase Program, HM Sanchinarro University Hospital, 28050 Madrid, Spain
| | - Víctor Moreno
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.P.); (E.C.); (V.M.)
| | - Alberto Ocaña
- Experimental Therapeutics in Cancer Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.S.G.); (L.P.-H.); (C.D.-T.); (C.N.-J.)
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.P.); (E.C.); (V.M.)
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
4
|
Elez E, Cubillo A, Alfonso PG, Middleton MR, Chau I, Alkuzweny B, Alcasid A, Zhang X, Van Cutsem E. Binimetinib in combination with nivolumab or nivolumab and ipilimumab in patients with previously treated microsatellite-stable metastatic colorectal cancer with RAS mutations in an open-label phase 1b/2 study. BMC Cancer 2024; 24:446. [PMID: 38600471 PMCID: PMC11007903 DOI: 10.1186/s12885-024-12153-5] [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: 03/14/2023] [Accepted: 03/20/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND In patients with previously treated RAS-mutated microsatellite-stable (MSS) metastatic colorectal cancer (mCRC), a multicenter open-label phase 1b/2 trial was conducted to define the safety and efficacy of the MEK1/MEK2 inhibitor binimetinib in combination with the immune checkpoint inhibitor (ICI) nivolumab (anti-PD-1) or nivolumab and another ICI, ipilimumab (anti-CTLA4). METHODS In phase 1b, participants were randomly assigned to Arm 1A (binimetinib 45 mg twice daily [BID] plus nivolumab 480 mg once every 4 weeks [Q4W]) or Arm 1B (binimetinib 45 mg BID plus nivolumab 480 mg Q4W and ipilimumab 1 mg/kg once every 8 weeks [Q8W]) to determine the maximum tolerable dose (MTD) and recommended phase 2 dose (RP2D) of binimetinib. The MTD/RP2D was defined as the highest dosage combination that did not cause medically unacceptable dose-limiting toxicities in more than 35% of treated participants in Cycle 1. During phase 2, participants were randomly assigned to Arm 2A (binimetinib MTD/RP2D plus nivolumab) or Arm 2B (binimetinib MTD/RP2D plus nivolumab and ipilimumab) to assess the safety and clinical activity of these combinations. RESULTS In phase 1b, 21 participants were randomized to Arm 1A or Arm 1B; during phase 2, 54 participants were randomized to Arm 2A or Arm 2B. The binimetinib MTD/RP2D was determined to be 45 mg BID. In phase 2, no participants receiving binimetinib plus nivolumab achieved a response. Of the 27 participants receiving binimetinib, nivolumab, and ipilimumab, the overall response rate was 7.4% (90% CI: 1.3, 21.5). Out of 75 participants overall, 74 (98.7%) reported treatment-related adverse events (AEs), of whom 17 (22.7%) reported treatment-related serious AEs. CONCLUSIONS The RP2D binimetinib regimen had a safety profile similar to previous binimetinib studies or nivolumab and ipilimumab combination studies. There was a lack of clinical benefit with either drug combination. Therefore, these data do not support further development of binimetinib in combination with nivolumab or nivolumab and ipilimumab in RAS-mutated MSS mCRC. TRIAL REGISTRATION NCT03271047 (09/01/2017).
Collapse
Affiliation(s)
- Elena Elez
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Antonio Cubillo
- Centro Integral, Oncológico Clara Campal, HM CIOCC, Madrid, Spain
- Facultad HM Hospitales de Ciencias de La Salud UCJC, 28050, Madrid, Spain
| | - Pilar Garcia Alfonso
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Mark R Middleton
- Department of Oncology, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Ian Chau
- Gastrointestinal Unit, Royal Marsden Hospital, London & Surrey, UK
| | | | | | | | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Amniouel S, Jafri MS. High-accuracy prediction of colorectal cancer chemotherapy efficacy using machine learning applied to gene expression data. Front Physiol 2024; 14:1272206. [PMID: 38304289 PMCID: PMC10830836 DOI: 10.3389/fphys.2023.1272206] [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: 08/03/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024] Open
Abstract
Introduction: FOLFOX and FOLFIRI chemotherapy are considered standard first-line treatment options for colorectal cancer (CRC). However, the criteria for selecting the appropriate treatments have not been thoroughly analyzed. Methods: A newly developed machine learning model was applied on several gene expression data from the public repository GEO database to identify molecular signatures predictive of efficacy of 5-FU based combination chemotherapy (FOLFOX and FOLFIRI) in patients with CRC. The model was trained using 5-fold cross validation and multiple feature selection methods including LASSO and VarSelRF methods. Random Forest and support vector machine classifiers were applied to evaluate the performance of the models. Results and Discussion: For the CRC GEO dataset samples from patients who received either FOLFOX or FOLFIRI, validation and test sets were >90% correctly classified (accuracy), with specificity and sensitivity ranging between 85%-95%. In the datasets used from the GEO database, 28.6% of patients who failed the treatment therapy they received are predicted to benefit from the alternative treatment. Analysis of the gene signature suggests the mechanistic difference between colorectal cancers that respond and those that do not respond to FOLFOX and FOLFIRI. Application of this machine learning approach could lead to improvements in treatment outcomes for patients with CRC and other cancers after additional appropriate clinical validation.
Collapse
Affiliation(s)
- Soukaina Amniouel
- School of Systems Biology, George Mason University, Fairfax, VA, United States
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA, United States
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Giordano G, Pancione M. MHC class III lymphocyte antigens 6 as endogenous immunotoxins: Unlocking immunotherapy in proficient mismatch repair colorectal cancer. WIREs Mech Dis 2024; 16:e1631. [PMID: 37818781 DOI: 10.1002/wsbm.1631] [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: 10/09/2022] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 10/13/2023]
Abstract
A majority of cancers, including colorectal cancer (CRC) with intact DNA mismatch repair, exhibit a paralyzed antitumor immune response and resistance to immune checkpoint inhibitors. Members of MHC class III lymphocyte antigen 6G (LY6G) encode glycosylphosphatidylinositol (GPI) proteins anchored to the membrane. Snake venom neurotoxins and LY6G proteins share a three-finger (3F) folding domain. LY6 proteins such as LY6G6D are gaining a reputation as excellent tumor-associated antigens that can potently inhibit anti-tumor immunity in cancers with proficient mismatch repair. Thus, we called MHC class III LY6G endogenous immunotoxins. Since the discovery of LY6G6D as a tumor-associated antigen, T-cell engagers (TcEs) have been developed to simultaneously bind LY6G6D on cancer cells and CD3 on T cells, improving the treatment of metastatic solid tumors that are resistant to ICIs. We present a current understanding of how alterations in MHC class III genes inhibit antitumor immunity, and how these understandings can be turned into effective treatments for patients who are refractory to standard immunotherapy. This article is categorized under: Cancer > Genetics/Genomics/Epigenetics Cancer > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
7
|
Borcherding DC, Amin NV, He K, Zhang X, Lyu Y, Dehner C, Bhatia H, Gothra A, Daud L, Ruminski P, Pratilas CA, Pollard K, Sundby T, Widemann BC, Hirbe AC. MEK Inhibition Synergizes with TYK2 Inhibitors in NF1-Associated Malignant Peripheral Nerve Sheath Tumors. Clin Cancer Res 2023; 29:1592-1604. [PMID: 36799629 PMCID: PMC10102849 DOI: 10.1158/1078-0432.ccr-22-3722] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/23/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas with limited treatment options and poor survival rates. About half of MPNST cases are associated with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Overexpression of TYK2 occurs in the majority of MPNST, implicating TYK2 as a therapeutic target. EXPERIMENTAL DESIGN The effects of pharmacologic TYK2 inhibition on MPNST cell proliferation and survival were examined using IncuCyte live cell assays in vitro, and downstream actions were analyzed using RNA-sequencing (RNA-seq), qPCR arrays, and validation of protein changes with the WES automated Western system. Inhibition of TYK2 alone and in combination with MEK inhibition was evaluated in vivo using both murine and human MPNST cell lines, as well as MPNST PDX. RESULTS Pharmacologic inhibition of TYK2 dose-dependently decreased proliferation and induced apoptosis over time. RNA-seq pathway analysis on TYK2 inhibitor-treated MPNST demonstrated decreased expression of cell cycle, mitotic, and glycolysis pathways. TYK2 inhibition resulted in upregulation of the MEK/ERK pathway gene expression, by both RNA-seq and qPCR array, as well as increased pERK1/2 levels by the WES Western system. The compensatory response was tested with dual treatment with TYK2 and MEK inhibitors, which synergistically decreased proliferation and increased apoptosis in vitro. Finally, combination therapy was shown to inhibit growth of MPNST in multiple in vivo models. CONCLUSIONS These data provide the preclinical rationale for the development of a phase I clinical trial of deucravacitinib and mirdametinib in NF1-assosciated MPNST.
Collapse
Affiliation(s)
- Dana C. Borcherding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Neha V. Amin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin He
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaochun Zhang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yang Lyu
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Carina Dehner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Himanshi Bhatia
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Angad Gothra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Layla Daud
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Peter Ruminski
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Christine A. Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Kai Pollard
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Taylor Sundby
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Brigitte C. Widemann
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Angela C. Hirbe
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
8
|
Liu HT, Chen SY, Peng LL, Zhong L, Zhou L, Liao SQ, Chen ZJ, Wang QL, He S, Zhou ZH. Spatially resolved transcriptomics revealed local invasion-related genes in colorectal cancer. Front Oncol 2023; 13:1089090. [PMID: 36816947 PMCID: PMC9928961 DOI: 10.3389/fonc.2023.1089090] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Objective Local invasion is the first step of metastasis, the main cause of colorectal cancer (CRC)-related death. Recent studies have revealed extensive intertumoral and intratumoral heterogeneity. Here, we focused on revealing local invasion-related genes in CRC. Methods We used spatial transcriptomic techniques to study the process of local invasion in four CRC tissues. First, we compared the pre-cancerous, cancer center, and invasive margin in one section (S115) and used pseudo-time analysis to reveal the differentiation trajectories from cancer center to invasive margin. Next, we performed immunohistochemical staining for RPL5, STC1, AKR1B1, CD47, and HLA-A on CRC samples. Moreover, we knocked down AKR1B1 in CRC cell lines and performed CCK-8, wound healing, and transwell assays to assess cell proliferation, migration, and invasion. Results We demonstrated that 13 genes were overexpressed in invasive clusters, among which the expression of CSTB and TM4SF1 was correlated with poor PFS in CRC patients. The ribosome pathway was increased, while the antigen processing and presentation pathway was decreased along CRC progression. RPL5 was upregulated, while HLA-A was downregulated along cancer invasion in CRC samples. Pseudo-time analysis revealed that STC1, AKR1B1, SIRPA, C4orf3, EDNRA, CES1, PRRX1, EMP1, PPIB, PLTP, SULF2, and EGFL6 were unpregulated along the trajectories. Immunohistochemic3al staining showed the expression of STC1, AKR1B1, and CD47 was increased along cancer invasion in CRC samples. Knockdown of AKR1B1 inhibited CRC cells' proliferation, migration, and invasion. Conclusions We revealed the spatial heterogeneity within CRC tissues and uncovered some novel genes that were associated with CRC invasion.
Collapse
Affiliation(s)
- Hong-Tao Liu
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si-Yuan Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling-Long Peng
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhong
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si-Qi Liao
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Ji Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing-Liang Wang
- Department of Pathology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song He
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Zhi-Hang Zhou, ; Song He,
| | - Zhi-Hang Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Zhi-Hang Zhou, ; Song He,
| |
Collapse
|
9
|
Shen M, Kang Y. Cancer fitness genes: emerging therapeutic targets for metastasis. Trends Cancer 2023; 9:69-82. [PMID: 36184492 DOI: 10.1016/j.trecan.2022.08.007] [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: 07/07/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 12/31/2022]
Abstract
Development of cancer therapeutics has traditionally focused on targeting driver oncogenes. Such an approach is limited by toxicity to normal tissues and treatment resistance. A class of 'cancer fitness genes' with crucial roles in metastasis have been identified. Elevated or altered activities of these genes do not directly cause cancer; instead, they relieve the stresses that tumor cells encounter and help them adapt to a changing microenvironment, thus facilitating tumor progression and metastasis. Importantly, as normal cells do not experience high levels of stress under physiological conditions, targeting cancer fitness genes is less likely to cause toxicity to noncancerous tissues. Here, we summarize the key features and function of cancer fitness genes and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Minhong Shen
- Department of Pharmacology, Wayne State University School of Medicine, Michigan, MI, USA; Department of Oncology, Wayne State University School of Medicine and Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Michigan, MI, USA.
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
10
|
Corrales L, Hipp S, Martin K, Sabarth N, Tirapu I, Fuchs K, Thaler B, Walterskirchen C, Bauer K, Fabits M, Bergmann M, Binder C, Chetta PML, Vogt AB, Adam PJ. LY6G6D is a selectively expressed colorectal cancer antigen that can be used for targeting a therapeutic T-cell response by a T-cell engager. Front Immunol 2022; 13:1008764. [PMID: 36159851 PMCID: PMC9493073 DOI: 10.3389/fimmu.2022.1008764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and demands more effective treatments. We sought to identify tumor selective CRC antigens and their therapeutic potential for cytotoxic T-cell targeting by transcriptomic and immunohistochemical analysis. LY6G6D was identified as a tumor selectively expressed CRC antigen, mainly in the microsatellite stable (MSS) subtype. A specific anti LY6G6D/CD3 T cell engager (TcE) was generated and demonstrated potent tumor cell killing and T cell activation in vitro. Ex vivo treatment of primary patient-derived CRC tumor slice cultures with the LY6G6D/CD3 TcE led to IFNγ secretion in LY6G6D positive tumor samples. In vivo, LY6G6D/CD3 TcE monotherapy demonstrated tumor regressions in pre-clinical mouse models of engrafted human CRC tumor cells and PBMCs. Lastly, 2D and 3D cocultures of LY6G6D positive and negative cells were used to explore the bystander killing of LY6G6D negative cells after specific activation of T cells by LY6G6D positive cells. LY6G6D/CD3 TcE treatment was shown to lyse target negative cells in the vicinity of target positive cells through a combined effect of IFNγ, TNFα and Fas/FasL. In summary, LY6G6D was identified as a selectively expressed CRC antigen that can be utilized to potently re-direct and activate cytotoxic T-cells to lyse LY6G6D expressing CRC using a TcE. This effect can be spread to target negative neighboring tumor cells, potentially leading to improved therapeutic efficacy.
Collapse
Affiliation(s)
- Leticia Corrales
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
- *Correspondence: Leticia Corrales,
| | - Susanne Hipp
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
- Boehringer Ingelheim Pharmaceuticals, Inc., Translational Medicine and Clinical Pharmacology, Ridgefield, CT, United States
| | - Katharina Martin
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Nicolas Sabarth
- Boehringer Ingelheim Regional Center Vienna (RCV) GmbH & Co KG., Biotherapeutics Discovery, Vienna, Austria
| | - Iñigo Tirapu
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Klaus Fuchs
- Boehringer Ingelheim Pharma, GmbH & Co KG, Biotherapeutics Discovery, Biberach, Germany
| | - Barbara Thaler
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Christian Walterskirchen
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Kathrin Bauer
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Markus Fabits
- Division of Visceral Surgery, Department of General Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Michael Bergmann
- Division of Visceral Surgery, Department of General Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Carina Binder
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Paolo ML. Chetta
- Boehringer Ingelheim RCV, GmbH & Co KG., Oncology Translational Science, Vienna, Austria
| | - Anne B. Vogt
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Paul J. Adam
- Boehringer Ingelheim Regional Center Vienna (RCV), GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| |
Collapse
|
11
|
Caruso FP, D'Andrea MR, Coppola L, Landriscina M, Condelli V, Cerulo L, Giordano G, Porras A, Pancione M. Lymphocyte antigen 6G6D-mediated modulation through p38α MAPK and DNA methylation in colorectal cancer. Cancer Cell Int 2022; 22:253. [PMID: 35953834 PMCID: PMC9373545 DOI: 10.1186/s12935-022-02672-1] [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: 02/19/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
In addition to being novel biomarkers for poor cancer prognosis, members of Lymphocyte antigen-6 (Ly6) gene family also play a crucial role in avoiding immune responses to tumors. However, it has not been possible to identify the underlying mechanism of how Ly6 gene regulation operates in human cancers. Transcriptome, epigenome and proteomic data from independent cancer databases were analyzed in silico and validated independently in 334 colorectal cancer tissues (CRC). RNA mediated gene silencing of regulatory genes, and treatment with MEK and p38 MAPK inhibitors were also tested in vitro. We report here that the Lymphocyte antigen 6G6D is universally downregulated in mucinous CRC, while its activation progresses through the classical adenoma-carcinoma sequence. The DNA methylation changes in LY6G6D promoter are intimately related to its transcript regulation, epigenomic and histological subtypes. Depletion of DNA methyltransferase 1 (DNMT1), which maintains DNA methylation, results in the derepression of LY6G6D expression. RNA-mediated gene silencing of p38α MAPK or its selective chemical inhibition, however, reduces LY6G6D expression, reducing trametinib’s anti-inflammatory effects. Patients treated with FOLFOX-based first-line therapy experienced decreased survival due to hypermethylation of the LY6G6D promoter and decreased p38α MAPK signaling. We found that cancer-specific immunodominant epitopes are controlled by p38α MAPKs signaling and suppressed by DNA methylation in histological variants with Mucinous differentiation. This work provides a promising prospective for clinical application in diagnosis and personalized therapeutic strategies of colorectal cancer.
Collapse
Affiliation(s)
| | | | - Luigi Coppola
- UOC Anatomia ed Istologia Patologica e Citologia Diagnostica, Dipartimento dei Servizi Diagnostici e della Farmaceutica, Ospedale Sandro Pertini, ASL Roma 2, 00157, Rome, Italy
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata (CROB), Rionero in Vulture, Potenza, Italy
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata (CROB), Rionero in Vulture, Potenza, Italy
| | - Luigi Cerulo
- Bioinformatics Laboratory, BIOGEM scrl, Ariano Irpino, Avellino, Italy
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, 71122, Foggia, Italy.
| | - Almudena Porras
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University Madrid, 28040, Madrid, Spain. .,Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy.
| |
Collapse
|
12
|
Wang P, Sun LL, Clark R, Hristopoulos M, Chiu CP, Dillon M, Lin W, Lo AA, Chalsani S, Das Thakur M, Zimmerman Savill KM, Rougé L, Lupardus P, Piskol R, Husain B, Ellerman D, Shivva V, Leong SR, Ovacik M, Totpal K, Wu Y, Spiess C, Lee G, Leipold DD, Polson AG. Novel Anti-LY6G6D/CD3 T-Cell-Dependent Bispecific Antibody for the Treatment of Colorectal Cancer. Mol Cancer Ther 2022; 21:974-985. [PMID: 35364611 PMCID: PMC9381132 DOI: 10.1158/1535-7163.mct-21-0599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/21/2021] [Accepted: 03/21/2022] [Indexed: 01/07/2023]
Abstract
New therapeutics and combination regimens have led to marked clinical improvements for the treatment of a subset of colorectal cancer. Immune checkpoint inhibitors have shown clinical efficacy in patients with mismatch-repair-deficient or microsatellite instability-high (MSI-H) metastatic colorectal cancer (mCRC). However, patients with microsatellite-stable (MSS) or low levels of microsatellite instable (MSI-L) colorectal cancer have not benefited from these immune modulators, and the survival outcome remains poor for the majority of patients diagnosed with mCRC. In this article, we describe the discovery of a novel T-cell-dependent bispecific antibody (TDB) targeting tumor-associated antigen LY6G6D, LY6G6D-TDB, for the treatment of colorectal cancer. RNAseq analysis showed that LY6G6D was differentially expressed in colorectal cancer with high prevalence in MSS and MSI-L subsets, whereas LY6G6D expression in normal tissues was limited. IHC confirmed the elevated expression of LY6G6D in primary and metastatic colorectal tumors, whereas minimal or no expression was observed in most normal tissue samples. The optimized LY6G6D-TDB, which targets a membrane-proximal epitope of LY6G6D and binds to CD3 with high affinity, exhibits potent antitumor activity both in vitro and in vivo. In vitro functional assays show that LY6G6D-TDB-mediated T-cell activation and cytotoxicity are conditional and target dependent. In mouse xenograft tumor models, LY6G6D-TDB demonstrates antitumor efficacy as a single agent against established colorectal tumors, and enhanced efficacy can be achieved when LY6G6D-TDB is combined with PD-1 blockade. Our studies provide evidence for the therapeutic potential of LY6G6D-TDB as an effective treatment option for patients with colorectal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew G. Polson
- Corresponding Author: Andrew G. Polson, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080. Phone: 650-225-5134; Fax: 650-225-6240; E-mail:
| |
Collapse
|
13
|
Yang L, WenTao T, ZhiYuan Z, Qi L, YuXiang L, Peng Z, Ke L, XiaoNa J, YuZhi P, MeiLing J, QingYang F, GuoDong H, YueXiang W, JianMin X. Cullin-9/p53 mediates HNRNPC degradation to inhibit erastin-induced ferroptosis and is blocked by MDM2 inhibition in colorectal cancer. Oncogene 2022; 41:3210-3221. [PMID: 35505093 DOI: 10.1038/s41388-022-02284-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 02/27/2022] [Accepted: 03/17/2022] [Indexed: 01/05/2023]
Abstract
Colorectal cancer (CRC) is the leading cause of cancer associated death worldwide. Ferroptosis is a newly defined form of regulated cell death characterized by the accumulation of lipid hydroperoxides and exerts an increased attention for cancer treatment. However, little is known about ferroptosis in CRC. In this study, through whole genome sequencing and external differential differentiated expression analysis, we identify CUL9 as a novel important modulator for ferroptosis in CRC. Here we demonstrated that CUL9 can binds p53 to ubiquitylate heterogeneous nuclear ribonucleoprotein C for degradation. Overexpression of CUL9 increases resistance to erastin-induced ferroptosis. Then, we discovered this resistance was mediated by CUL9-HNRNPC-MATE1 negative loop, which can provide us with a novel target to overcome drug resistance to ferroptosis activators. Finally, we found that targeting MDM2 was developed as an effective strategy to destroy precious drug-resistant CRC cells.
Collapse
Affiliation(s)
- Lv Yang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tang WenTao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - Zhang ZhiYuan
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - Lin Qi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - Luo YuXiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zheng Peng
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - Li Ke
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia XiaoNa
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pang YuZhi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ji MeiLing
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - Feng QingYang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - He GuoDong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China
| | - Wang YueXiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Xu JianMin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Surgery, Shanghai, China.
| |
Collapse
|
14
|
Seo MK, Kang H, Kim S. Tumor microenvironment-aware, single-transcriptome prediction of microsatellite instability in colorectal cancer using meta-analysis. Sci Rep 2022; 12:6283. [PMID: 35428835 PMCID: PMC9012745 DOI: 10.1038/s41598-022-10182-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Detecting microsatellite instability (MSI) in colorectal cancers (CRCs) is essential because it is the determinant of treatment strategies, including immunotherapy and chemotherapy. Yet, no attempt has been made to exploit transcriptomic profile and tumor microenvironment (TME) of it to unveil MSI status in CRC. Hence, we developed a novel TME-aware, single-transcriptome predictor of MSI for CRC, called MAP (Microsatellite instability Absolute single sample Predictor). MAP was developed utilizing recursive feature elimination-random forest with 466 CRC samples from The Cancer Genome Atlas, and its performance was validated in independent cohorts, including 1118 samples. MAP showed robustness and predictive power in predicting MSI status in CRC. Additional advantages for MAP were demonstrated through comparative analysis with existing MSI classifier and other cancer types. Our novel approach will provide access to untouched vast amounts of publicly available transcriptomic data and widen the door for MSI CRC research and be useful for gaining insights to help with translational medicine.
Collapse
Affiliation(s)
- Mi-Kyoung Seo
- Department of Biomedical Systems Informatics, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyundeok Kang
- Department of Biomedical Systems Informatics, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
15
|
Luo C, He S, Zhang H, He S, Qi H, Wei A. Clinical and Biological Significance of DNA Methylation-Driven Differentially Expressed Genes in Biochemical Recurrence After Radical Prostatectomy. Front Genet 2022; 13:727307. [PMID: 35186021 PMCID: PMC8847683 DOI: 10.3389/fgene.2022.727307] [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: 06/18/2021] [Accepted: 01/13/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Biochemical recurrence (BCR) after radical prostatectomy indicates poor prognosis in patients with prostate cancer (PCA). DNA methylation (DNAm) is a critical factor in tumorigenesis and has attracted attention as a biomarker for the diagnosis, treatment, and prognosis of PCA. However, the predictive value of DNAm-derived differentially expressed genes (DMGs) in PCA with BCR remains elusive. Methods: We filtered the methylated genes and the differentially expressed genes (DGEs) for more than 1,000 clinical samples from the TCGA cohort using the chAMP and DESeq2 packages of R language, respectively. Next, we integrated the DNAm beta value and gene expression data with the Mithymix package of R language to obtain the DMGs. Then, 1,000 times Cox LASSO regression with 10-fold cross validation was performed to screen signature DMGs and establish a predictive classifier. Univariate and multivariate cox regressive analyses were used to identify the prognostic factors to build a predictive model, and its performance was measured by receiver operating characteristic, calibration curves, and Harrell’s concordance index (C-index). Additionally, a GEO dataset was used to validate the prognostic classifier. Results: One hundred DMGs were mined using the chAMP and Methymix packages of R language. Of these, seven DMGs (CCK, CD38, CYP27A1, EID3, HABP2, LRRC4, and LY6G6D) were identified to build the prognostic classifier (Classifier) through LASSO analysis. Moreover, univariate and multivariate Cox regression analysis determined that the Classifier and pathological T stage (pathological_T) were independent predictors of BCR (hazard ratio (HR 2.2), (95% CI 1.4–3.5), p < 0.0012, and (HR 1.8), (95% CI 1.0–3.2), p < 0.046). A nomogram based on the Classifier was constructed, with high prediction accuracy for BCR-free survival in TCGA and GEO datasets. GSEA enrichment analysis showed that the DMGs were mainly enriched in the metabolism pathways. Conclusion: We identified and validated the nomogram of BCR-free survival for PCA patients, which has the potential to guide treatment decisions for patients at differing risks of BCR. Our study deepens the understanding of DMGs in the pathogenesis of PCA.
Collapse
Affiliation(s)
- Chao Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Songzhe He
- Department of Laboratory Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Haibo Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuhua He
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huan Qi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Anyang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Lin XT, Wu QN, Qin S, Fan DJ, Lv MY, Chen X, Cai JW, Weng JR, Zou YF, Rong YM, Gao F. Identification of an Autophagy-Related Gene Signature for the Prediction of Prognosis in Early-Stage Colorectal Cancer. Front Genet 2021; 12:755789. [PMID: 34899841 PMCID: PMC8657766 DOI: 10.3389/fgene.2021.755789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/19/2021] [Indexed: 01/01/2023] Open
Abstract
Purpose: A certain number of early-stage colorectal cancer (CRC) patients suffer tumor recurrence after initial curative resection. In this context, an effective prognostic biomarker model is constantly in need. Autophagy exhibits a dual role in tumorigenesis. Our study aims to develop an autophagy-related gene (ATG) signature-based on high-throughput data analysis for disease-free survival (DFS) prognosis of patients with stage I/II CRC. Methods: Gene expression profiles and clinical information of CRC patients extracted from four public datasets were distributed to discovery and training cohort (GSE39582), validation cohort (TCGA CRC, n = 624), and meta-validation cohort (GSE37892 and GSE14333, n = 420). Autophagy genes significantly associated with prognosis were identified. Results: Among 655 autophagy-related genes, a 10-gene ATG signature, which was significantly associated with DFS in the training cohort (HR, 2.76[1.56–4.82]; p = 2.06 × 10–4), was constructed. The ATG signature, stratifying patients into high and low autophagy risk groups, was validated in the validation (HR, 2.29[1.15–4.55]; p = 1.5 × 10–2) and meta-validation cohorts (HR, 2.5[1.03–6.06]; p = 3.63 × 10–2) and proved to be prognostic in a multivariate analysis. Functional analysis revealed enrichment of several immune/inflammatory pathways in the high autophagy risk group, where increased infiltration of T regulatory cells (Tregs) and decreased infiltration of M1 macrophages were observed. Conclusion: Our study established a prognostic ATG signature that effectively predicted DFS for early-stage CRC patients. Meanwhile, the study also revealed the possible relationship among autophagy process, immune/inflammatory response, and tumorigenesis.
Collapse
Affiliation(s)
- Xu-Tao Lin
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiu-Ning Wu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Si Qin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Medical Ultrasonics, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - De-Jun Fan
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Min-Yi Lv
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xi Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia-Wei Cai
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing-Rong Weng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi-Feng Zou
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu-Ming Rong
- Department of VIP Region, Cancer Center of Sun Yat-sen University, Guangzhou, China
| | - Feng Gao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Wang Y, Yang C, Li W, Shen Y, Deng J, Lu W, Jin J, Liu Y, Liu Q. Identification of colon tumor marker NKD1 via integrated bioinformatics analysis and experimental validation. Cancer Med 2021; 10:7383-7394. [PMID: 34547189 PMCID: PMC8525156 DOI: 10.1002/cam4.4224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022] Open
Abstract
Background Colorectal cancer is an important death‐related disease in the worldwide. However, specific colon cancer tumor markers currently used for diagnosis and treatment are few. The purpose of this study is to screen the potential colon cancer markers by bioinformatics and verify the results with experiments. Methods Gene expression data were downloaded from two different databases: TCGA database and GEO datasets, which were then analyzed by two different methods (difference analysis and WGCNA method). Venn and PPI analysis obtained the potential core genes, which were then performed the GO enrichment and KEGG pathway analysis. Expressions levels of NKD1 in colon carcinoma tissues were further confirmed by immunohistochemical staining and western blot assays. Moreover, the function was measured by MTT, clone formation, and tumor transplantation experiments. Importantly, co‐immunoprecipitation, immunofluorescence, and protein stability assays were further performed to explore the underlying mechanism of NKD1 promoting cell proliferation. Results Nine potential core genes highly expressed in colon cancer samples were screened out by bioinformatics analysis. NKD1, one of the hub genes, highly expressed in the colon carcinoma tissues could enhance the proliferation of colon cancer cells. Mechanism research demonstrated that NKD1 was essential for the combination between Wnt signalosome (DVL) and β‐catenin, and that NKD1 knockout remarkably decreased the β‐catenin expression. Immunofluorescence assays further implied that NKD1 knockout significantly inhibited β‐catenin nuclear accumulation. Importantly, the stability of β‐catenin proteins was maintained by NKD1 in the colon cancer cells. Conclusion We believe that NKD1 well expressed in the colorectal carcinoma tissues can enhance the proliferation of colon cancer cells. Furthermore, the functions that NKD1 may have in colon cancer cells should be different from that NKD1 has played in the zebrafish. Thus, NKD1 could be a specific colorectal cancer marker.
Collapse
Affiliation(s)
- Yue Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.,Clinical Oncology Laboratory, Changzhou Tumor Hospital Affiliated to Soochow University, Changzhou, Changzhou, China.,Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Chunxia Yang
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Wenjing Li
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Ying Shen
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Jianzhong Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| | - Yongping Liu
- The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.,Clinical Oncology Laboratory, Changzhou Tumor Hospital Affiliated to Soochow University, Changzhou, Changzhou, China
| | - Qian Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, China
| |
Collapse
|
18
|
Borcherding DC, He K, Amin NV, Hirbe AC. TYK2 in Cancer Metastases: Genomic and Proteomic Discovery. Cancers (Basel) 2021; 13:4171. [PMID: 34439323 PMCID: PMC8393599 DOI: 10.3390/cancers13164171] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Advances in genomic analysis and proteomic tools have rapidly expanded identification of biomarkers and molecular targets important to cancer development and metastasis. On an individual basis, personalized medicine approaches allow better characterization of tumors and patient prognosis, leading to more targeted treatments by detection of specific gene mutations, overexpression, or activity. Genomic and proteomic screens by our lab and others have revealed tyrosine kinase 2 (TYK2) as an oncogene promoting progression and metastases of many types of carcinomas, sarcomas, and hematologic cancers. TYK2 is a Janus kinase (JAK) that acts as an intermediary between cytokine receptors and STAT transcription factors. TYK2 signals to stimulate proliferation and metastasis while inhibiting apoptosis of cancer cells. This review focuses on the growing evidence from genomic and proteomic screens, as well as molecular studies that link TYK2 to cancer prevalence, prognosis, and metastasis. In addition, pharmacological inhibition of TYK2 is currently used clinically for autoimmune diseases, and now provides promising treatment modalities as effective therapeutic agents against multiple types of cancer.
Collapse
Affiliation(s)
- Dana C. Borcherding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
| | - Kevin He
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
| | - Neha V. Amin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
| | - Angela C. Hirbe
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
19
|
Ma YS, Liu JB, Yang XL, Xin R, Shi Y, Zhang DD, Wang HM, Wang PY, Lin QL, Li W, Fu D. Basic approaches, challenges and opportunities for the discovery of small molecule anti-tumor drugs. Am J Cancer Res 2021; 11:2386-2400. [PMID: 34249406 PMCID: PMC8263657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/15/2021] [Indexed: 06/13/2023] Open
Abstract
Chemotherapy is one of the main treatments for cancer, especially for advanced cancer patients. In the past decade, significant progress has been made with the research into the molecular mechanisms of cancer cells and the precision medicine. The treatment on cancer patients has gradually changed from cytotoxic chemotherapy to precise treatment strategy. Research into anticancer drugs has also changed from killing effects on all cells to targeting drugs for target genes. Besides, researchers have developed the understanding of the abnormal physiological function, related genomics, epigenetics, and proteomics of cancer cells with cancer genome sequencing, epigenetic research, and proteomic research. These technologies and related research have accelerated the development of related cancer drugs. In this review, we summarize the research progress of anticancer drugs, the current challenges, and future opportunities.
Collapse
Affiliation(s)
- Yu-Shui Ma
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Xiao-Li Yang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Rui Xin
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Yi Shi
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Dan-Dan Zhang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Hui-Min Wang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Pei-Yao Wang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Qin-Lu Lin
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
| | - Wen Li
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
| | - Da Fu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| |
Collapse
|
20
|
Wu TM, Liu JB, Liu Y, Shi Y, Li W, Wang GR, Ma YS, Fu D. Power and Promise of Next-Generation Sequencing in Liquid Biopsies and Cancer Control. Cancer Control 2021; 27:1073274820934805. [PMID: 32806937 PMCID: PMC7791471 DOI: 10.1177/1073274820934805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traditional methods of cancer treatment are usually based on the morphological
and histological diagnosis of tumors, and they are not optimized according to
the specific situation. Precision medicine adjusts the existing treatment
regimen based on the patient’s genomic information to make it most suitable for
patients. Detection of genetic mutations in tumors is the basis of precise
cancer medicine. Through the analysis of genetic mutations in patients with
cancer, we can tailor the treatment plan for each patient with cancer to
maximize the curative effect, minimize damage to healthy tissues, and optimize
resources. In recent years, next-generation sequencing technology has developed
rapidly and has become the core technology of precise targeted therapy and
immunotherapy for cancer. From early cancer screening to treatment guidance for
patients with advanced cancer, liquid biopsy is increasingly used in cancer
management. This is as a result of the development of better noninvasive,
repeatable, sensitive, and accurate tools used in early screening, diagnosis,
evaluation, and monitoring of patients. Cell-free DNA, which is a new
noninvasive molecular pathological detection method, often carries
tumor-specific gene changes. It plays an important role in optimizing treatment
and evaluating the efficacy of different treatment options in clinical trials,
and it has broad clinical applications.
Collapse
Affiliation(s)
- Ting-Miao Wu
- Department of Radiology, 12485The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ji-Bin Liu
- Cancer Institute, 377323Nantong Tumor Hospital, Nantong, China
| | - Yu Liu
- National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, 12571Central South University of Forestry and Technology, Chaha, China
| | - Yi Shi
- National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, 12571Central South University of Forestry and Technology, Chaha, China
| | - Wen Li
- National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, 12571Central South University of Forestry and Technology, Chaha, China
| | - Gao-Ren Wang
- Cancer Institute, 377323Nantong Tumor Hospital, Nantong, China
| | - Yu-Shui Ma
- Cancer Institute, 377323Nantong Tumor Hospital, Nantong, China.,Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, 12476Tongji University School of Medicine, Shanghai, China
| | - Da Fu
- Department of Radiology, 12485The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China.,Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, 12476Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Ma YS, Xin R, Yang XL, Shi Y, Zhang DD, Wang HM, Wang PY, Liu JB, Chu KJ, Fu D. Paving the way for small-molecule drug discovery. Am J Transl Res 2021; 13:853-870. [PMID: 33841626 PMCID: PMC8014367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/18/2020] [Indexed: 06/12/2023]
Abstract
Small-molecule drugs are organic compounds affecting molecular pathways by targeting important proteins, which have a low molecular weight, making them penetrate cells easily. Small-molecule drugs can be developed from leads derived from rational drug design or isolated from natural resources. As commonly used medications, small-molecule drugs can be taken orally, which enter cells to act on intracellular targets. These characteristics make small-molecule drugs promising candidates for drug development, and they are increasingly favored in the pharmaceutical market. Despite the advancements in molecular genetics and effective new processes in drug development, the drugs currently used in clinical practice are inadequate due to their poor efficacy or severe side effects. Therefore, developing new safe and efficient drugs is a top priority for disease control and curing.
Collapse
Affiliation(s)
- Yu-Shui Ma
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Rui Xin
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Xiao-Li Yang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Yi Shi
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Dan-Dan Zhang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Hui-Min Wang
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Pei-Yao Wang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Kai-Jian Chu
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical UniversityShanghai 200438, China
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| |
Collapse
|
22
|
JAK3 and TYK2 Serve as Prognostic Biomarkers and Are Associated with Immune Infiltration in Stomach Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7973568. [PMID: 33083484 PMCID: PMC7559258 DOI: 10.1155/2020/7973568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/19/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Background Stomach adenocarcinoma (STAD) is one of the most common malignant tumors. The Janus kinases (JAKs) play a significant part in cellular biological process, inflammation, and immunity. The roles of JAKs in STAD are still not systematically described. Methods A series of bioinformatics tools were used to clarify the role of JAKs in STAD. Results JAK3/TYK2 levels were significantly increased in STAD during subgroup analyses based on gender, tumor grade, cancer stages, and nodal metastasis status. STAD patients with high levels of JAK3/TYK2 had poor overall survival, postprogression survival, and first progression. Immune infiltration revealed a significant correlation between JAK3/TYK2 expression and the abundance of immune cells as well as immune biomarker expression in STAD. JAK3/TYK2 was associated with the adaptive immune response, chemokine signaling pathway, and JAK-STAT signaling pathway. Conclusions JAK3 and TYK2 serve as prognostic biomarkers and are associated with immune infiltration in STAD.
Collapse
|
23
|
Qureshy Z, Johnson DE, Grandis JR. Targeting the JAK/STAT pathway in solid tumors. JOURNAL OF CANCER METASTASIS AND TREATMENT 2020; 6:27. [PMID: 33521321 PMCID: PMC7845926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Aberrant activation of signal transducer and activator of transcription (STAT) proteins is associated with the development and progression of solid tumors. However, as transcription factors, these proteins are difficult to target directly. In this review, we summarize the role of targeting Janus kinases (JAKs), upstream activators of STATs, as a strategy for decreasing STAT activation in solid tumors. Preclinical studies in solid tumor cell line models show that JAK inhibitors decrease STAT activation, cell proliferation, and cell survival; in in vivo models, they also inhibit tumor growth. JAK inhibitors, particularly the JAK1/2 inhibitor ruxolitinib, sensitize cell lines and murine models to chemotherapy, immunotherapy, and oncolytic viral therapy. Ten JAK inhibitors have been or are actively being tested in clinical trials as monotherapy or in combination with other agents in patients with solid tumors; two of these inhibitors are already Food and Drug Administration (FDA) approved for the treatment of myeloproliferative disorders and rheumatoid arthritis, making them attractive agents for use in patients with solid tumors as they are known to be well-tolerated. Four JAK inhibitors (two of which are FDA approved for other indications) have exhibited promising anti-cancer effects in preclinical studies; however, clinical studies specifically assessing their activity against the JAK/STAT pathway in solid tumors have not yet been conducted. In summary, JAK inhibition is a viable option for targeting the JAK/STAT pathway in solid tumors and merits further testing in clinical trials.
Collapse
Affiliation(s)
- Zoya Qureshy
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco 94158, USA
| | - Daniel E Johnson
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco 94158, USA
| | - Jennifer R Grandis
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco 94158, USA
| |
Collapse
|
24
|
Ma YS, Li W, Liu Y, Shi Y, Lin QL, Fu D. Targeting Colorectal Cancer Stem Cells as an Effective Treatment for Colorectal Cancer. Technol Cancer Res Treat 2020; 19:1533033819892261. [PMID: 32748700 PMCID: PMC7785997 DOI: 10.1177/1533033819892261] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
As one of the common cancers that threaten human life, the recurrence and metastasis of colorectal cancer seriously affect the prognosis of patients. Although new drugs and comprehensive treatments have been adopted, the current treatment effect on this tumor, especially in advanced colorectal cancer, is still not satisfactory. More and more evidence shows that tumors are likely to be a stem cell disease. In recent years, the rise of cancer stem cell theory has provided a new way for cancer treatment. Studies have found that a small number of special cells in colorectal cancer tissues that induce tumorigenesis, proliferation, and promote tumor migration and metastasis, namely, colorectal cancer stem cells. Colorectal cancer stem cells are defined with a group of cell-surface markers, such as CD44, CD133, CD24, epithelial cell adhesion factor molecule, LGR5, and acetaldehyde dehydrogenase. They are highly tumorigenic, aggressive, and chemoresistant and thus are critical in the metastasis and recurrence of colorectal cancer. Therefore, targeting colorectal cancer stem cells may become an important research direction for the future cure of colorectal cancer.
Collapse
Affiliation(s)
- Yu-Shui Ma
- National Engineering Laboratory for Rice and By-Product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China.,Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Li
- National Engineering Laboratory for Rice and By-Product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Yu Liu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Shi
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qin-Lu Lin
- National Engineering Laboratory for Rice and By-Product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Da Fu
- National Engineering Laboratory for Rice and By-Product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China.,Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Giordano G, Remo A, Porras A, Pancione M. Immune Resistance and EGFR Antagonists in Colorectal Cancer. Cancers (Basel) 2019; 11:E1089. [PMID: 31370270 PMCID: PMC6721348 DOI: 10.3390/cancers11081089] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 02/05/2023] Open
Abstract
: Targeting the epidermal growth factor receptor (EGFR) either alone or in combination with chemotherapy in patients with RAS wild type metastatic colorectal cancer (mCRC) has revolutionized the treatment of CRC, but with less results than initially envisaged. In recent years, the discovery of multiple pathways leading to the escape from anti-EGFR therapy has revealed an enormous complexity and heterogeneity of human CRC due to the intrinsic genomic instability and immune/cancer cell interaction. Therefore, understanding the mechanistic basis of acquired resistance to targeted therapies represents a major challenge to improve the clinical outcomes of patients with CRC. The latest findings strongly suggest that complex molecular alterations coupled with changes of the immune tumor microenvironment may substantially contribute to the clinical efficacy of EGFR antagonist. In this review, we discuss the most recent findings that contribute to both primary and acquired anti-EGFR therapy resistance. In addition, we analyze how strategies aiming to enhance the favorable effects in the tumor microenvironment may contribute to overcome resistance to EGFR therapies.
Collapse
Affiliation(s)
- Guido Giordano
- U.O.C. Medical Oncology, Ospedali Riuniti, Azienda Ospedaliero Universitaria, 251 Foggia, Italy
| | - Andrea Remo
- Pathology Unit, Mater Salutis Hospital AULSS9, "Scaligera", 37122 Verona, Italy
| | - Almudena Porras
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University Madrid, 28040 Madrid, Spain.
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Massimo Pancione
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University Madrid, 28040 Madrid, Spain.
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| |
Collapse
|