1
|
Asare O, Shim L, Lee CJ, Delgado J, Quailes N, Zavala K, Park J, Hafeez BB, Cho YY, Chauhan SC, Kim DJ. Loss of TC-PTP in keratinocytes leads to increased UVB-induced autophagy. Cell Death Discov 2025; 11:80. [PMID: 40021617 PMCID: PMC11871011 DOI: 10.1038/s41420-025-02353-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/28/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
Ultraviolet B (UVB) radiation can distort cellular homeostasis and predispose the skin to carcinogenesis. Amongst the deteriorating effects of the sun's UVB radiation on cellular homeostasis is the formation of DNA photoproducts. These photoproducts can cause significant changes in the structure and conformation of DNA, inducing gene mutations which may accumulate to trigger the formation of skin cancer. Photoproducts are typically repaired by nucleotide excision repair. Notwithstanding, when the repair mechanism fails, apoptosis ensues to prevent the accumulation of mutations and to restore cellular homeostasis. This present study reports that T-cell protein tyrosine phosphatase (TC-PTP) can increase UVB-induced apoptosis by inhibiting autophagy-mediated cell survival of damaged keratinocytes. TC-PTP deficiency in 3PC mouse keratinocytes led to the formation of autophagic vacuoles and increased expression of LC3-II. We established human TC-PTP-deficient (TC-PTP/KO) HaCaT cells using the CRISPR/Cas9 system. TC-PTP/KO HaCaT cells exhibited increased cell survival upon UVB exposure, which was accompanied by increased expression of LC3-II and decreased expression of p62 compared to control cells. Pretreatment of TC-PTP/KO HaCaT cells with early-phase autophagy inhibitor, 3-methyladenine significantly decreased the expression of LC3-II and reduced cell survival in response to UVB irradiation in comparison with untreated TC-PTP/KO cells. Pretreatment of TC-PTP/KO HaCaT cells with late-phase inhibitor, chloroquine also significantly reduced cell viability with increased accumulation of LC3-II after UVB irradiation compared to untreated counterpart cells. While UVB significantly increased apoptosis in the engineered (Mock) cells, this was not observed in similarly treated TC-PTP/KO HaCaT cells. However, chloroquine treatment increased apoptosis in TC-PTP/KO HaCaT cells. Examination of human squamous cell carcinomas (SCCs) revealed that TC-PTP expression was inversely correlated with LC3 expression. Our findings suggest that TC-PTP negatively regulates autophagy-mediated survival of damaged cells following UVB exposure, which can contribute to remove damaged keratinocytes via apoptosis.
Collapse
Affiliation(s)
- Obed Asare
- Department of Medicine and Oncology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- Graduate Program in Biochemistry and Molecular Biology, University of Texas Rio Grande Valley, Edinburg, TX, USA
- Graduate Program in Cancer Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lindsey Shim
- Department of Medicine and Oncology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- Graduate Program in Biochemistry and Molecular Biology, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Cheol-Jung Lee
- Department of Medicine and Oncology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute, Cheongju-si, 28119, Republic of Korea
| | - Jose Delgado
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Natasha Quailes
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Klarissa Zavala
- Department of Health & Biomedical Sciences, College of Health Professions, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Bilal Bin Hafeez
- Department of Medicine and Oncology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- Graduate Program in Biochemistry and Molecular Biology, University of Texas Rio Grande Valley, Edinburg, TX, USA
- South Texas Center for Excellence in Cancer Research, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4TH, and RCD Control Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Subhash C Chauhan
- Department of Medicine and Oncology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- Graduate Program in Biochemistry and Molecular Biology, University of Texas Rio Grande Valley, Edinburg, TX, USA
- South Texas Center for Excellence in Cancer Research, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Dae Joon Kim
- Department of Medicine and Oncology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
- Graduate Program in Biochemistry and Molecular Biology, University of Texas Rio Grande Valley, Edinburg, TX, USA.
- South Texas Center for Excellence in Cancer Research, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
2
|
Tanwar AK, Sengar N, Mase N, Singh IP. Tetrahydroisoquinolines - an updated patent review for cancer treatment (2016 - present). Expert Opin Ther Pat 2024; 34:873-906. [PMID: 39126639 DOI: 10.1080/13543776.2024.2391288] [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: 05/01/2024] [Revised: 06/25/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Cancer is a prominent cause of death globally, triggered by both non-genetic and genetic alterations in genes influenced by various environmental factors. The tetrahydroisoquinoline (THIQ), specifically 1,2,3,4-tetrahydroisoquinoline serves as fundamental element in various alkaloids, prevalent in proximity to quinoline and indole alkaloids. AREA COVERED In this review, the therapeutic applications of THIQ derivatives as an anticancer agent from 2016 to 2024 have been examined. The patents were gathered through comprehensive searches of the Espacenet, Google patent, WIPO, and Sci Finder databases. The therapeutic areas encompassed in the patents include numerous targets of cancer. EXPERT OPINION THIQ analogues play a crucial role in medicinal chemistry, with many being integral to pharmacological processes and clinical trials. Numerous THIQ compounds have been synthesized for therapeutic purposes, notably in cancer treatment. They show great promise for developing anticancer drugs, demonstrating strong affinity and efficacy against various cancer targets. The creation of multi-target ligands is a compelling avenue for THIQ-based anticancer drug discovery.
Collapse
Affiliation(s)
- Ankur Kumar Tanwar
- Departments of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| | - Neha Sengar
- Departments of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| | - Nobuyuki Mase
- Research Institute of Green Science and Technology, Shizuoka University, Hamamatsu, Shizuoka, Japan
| | - Inder Pal Singh
- Departments of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| |
Collapse
|
3
|
Aslebagh R, Whitham D, Channaveerappa D, Lowe J, Pentecost BT, Arcaro KF, Darie CC. Proteomics analysis of human breast milk by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) coupled with mass spectrometry to assess breast cancer risk. Electrophoresis 2023; 44:1097-1113. [PMID: 36971330 PMCID: PMC10522790 DOI: 10.1002/elps.202300040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Breast cancer (BC) is one of the most common cancers and one of the most common causes for cancer-related mortality. Discovery of protein biomarkers associated with cancer is considered important for early diagnosis and prediction of the cancer risk. Protein biomarkers could be investigated by large-scale protein investigation or proteomics, using mass spectrometry (MS)-based techniques. Our group applies MS-based proteomics to study the protein pattern in human breast milk from women with BC and controls and investigates the alterations and dysregulations of breast milk proteins in comparison pairs of BC versus control. These dysregulated proteins might be considered potential future biomarkers of BC. Identification of potential biomarkers in breast milk may benefit young women without BC, but who could collect the milk for future assessment of BC risk. Previously we identified several dysregulated proteins in different sets of human breast milk samples from BC patients and controls using gel-based protein separation coupled with MS. Here, we performed 2D-PAGE coupled with nano-liquid chromatography-tandem MS (nanoLC-MS/MS) in a small-scale study on a set of six human breast milk pairs (three BC samples vs. three controls) and we identified several dysregulated proteins that have potential roles in cancer progression and might be considered potential BC biomarkers in the future.
Collapse
Affiliation(s)
- Roshanak Aslebagh
- Biochemistry and Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| | - Danielle Whitham
- Biochemistry and Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| | - Devika Channaveerappa
- Biochemistry and Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| | - James Lowe
- Biochemistry and Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| | - Brian T. Pentecost
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Kathleen F. Arcaro
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Costel C. Darie
- Biochemistry and Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| |
Collapse
|
4
|
Tang X, Sui X, Liu Y. Immune checkpoint PTPN2 predicts prognosis and immunotherapy response in human cancers. Heliyon 2023; 9:e12873. [PMID: 36685446 PMCID: PMC9852697 DOI: 10.1016/j.heliyon.2023.e12873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
Background PTPN2, a member of the non-receptor protein tyrosine phosphatases family, holds a crucial role in tumorigenesis and cancer immunotherapy. However, most studies on the role of PTPN2 in cancer are limited to specific cancer types. Therefore, this study aimed to investigate the prognostic significance of PTPN2 in human cancers and its function in the tumor microenvironment. Methods To shed light on this matter, we investigated the expression level, prognostic value, genomic alterations, molecular function, immune function, and immunotherapeutic predictive ability of PTPN2 in human cancers using the TCGA, GTEx, CGGA, GEO, cBioPortal, STRING, TISCH, TIMER2.0, ESTIMATE, and TIDE databases. Furthermore, the CCK-8 assay was utilized to detect the effect of PTPN2 on cell proliferation. Cell immunofluorescence analysis was performed to probe the cellular localization of PTPN2. Western blot was applied to examine the molecular targets downstream of PTPN2. Finally, a Nomogram model was constructed using the TCGA-LGG cohort and evaluated with calibration curves and time-dependent ROCs. Results PTPN2 was highly expressed in most cancers and was linked to poor prognosis in ACC, GBM, LGG, KICH, and PAAD, while the opposite was true in OV, SKCM, and THYM. PTPN2 knockdown promoted the proliferation of melanoma cells, while significantly inhibiting proliferation in colon cancer and glioblastoma cells. In addition, TC-PTP, encoded by the PTPN2 gene, was primarily localized in the nucleus and cytoplasm and could negatively regulate the JAK/STAT and MEK/ERK pathways. Strikingly, PTPN2 knockdown significantly enhanced the abundance of PD-L1. PTPN2 was abundantly expressed in Mono/Macro cells and positively correlated with multiple immune infiltrating cells, especially CD8+ T cells. Notably, DLBC, LAML, OV, and TGCT patients in the PTPN2-high group responded better to immunotherapy, while the opposite was true in ESCA, KIRC, KIRP, LIHC, and THCA. Finally, the construction of a Nomogram model on LGG exhibited a high prediction accuracy. Conclusion Immune checkpoint PTPN2 is a powerful biomarker for predicting prognosis and the efficacy of immunotherapy in cancers. Mechanistically, PTPN2 negatively regulates the JAK/STAT and MEK/ERK pathways and the abundance of PD-L1.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Xue Sui
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Yongshuo Liu
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China,Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China,Corresponding author. Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| |
Collapse
|
5
|
Zhu Z, Tang R, Huff S, Kummetha IR, Wang L, Li N, Rana TM. Small-molecule PTPN2 Inhibitors Sensitize Resistant Melanoma to Anti-PD-1 Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2023; 3:119-129. [PMID: 36968224 PMCID: PMC10035454 DOI: 10.1158/2767-9764.crc-21-0186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/23/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Although immune checkpoint inhibitors targeting T-cell immunoregulatory proteins have revolutionized cancer treatment, they are effective only in a limited number of patients, and new strategies are needed to enhance tumor responses to immunotherapies. Deletion of protein tyrosine phosphatase non-receptor type 2 (Ptpn2), a regulator of growth factor and cytokine signaling pathways, has been shown to sensitize murine B16F10 melanoma cells to IFNγ and anti-PD-1 immunotherapy. Here, we investigated the potential therapeutic utility of small-molecule PTPN2 inhibitors. Ten inhibitors were synthesized on the basis of in silico modeling and structure-based design and functionally tested in vitro and in vivo. We show that the inhibitors had little effect on B16F10 cells alone, but effectively sensitized the tumor cells to IFNγ treatment in vitro and to anti-PD-1 therapy in vivo. Under both conditions, Ptpn2 inhibitor cotreatment suppressed B16F10 cell growth and enhanced Stat1 phosphorylation and expression of IFNγ response genes. In vivo, PTPN2 inhibitor cotreatment significantly reduced melanoma and colorectal tumor growth and enhanced mouse survival compared with anti-PD-1 treatment alone, and this was accompanied by increased tumor infiltration by granzyme B+ CD8+ T cells. Similar results were obtained with representative murine and human colon cancer and lung cancer cell lines. Collectively, these results demonstrate that small-molecule inhibitors of PTPN2 may have clinical utility as sensitizing agents for immunotherapy-resistant cancers. Significance To enhance the effectiveness of immunotherapies in resistant or nonresponsive cancers, it is important to develop inhibitors of enzymes that negatively influence the outcome of treatments. We have designed and evaluated small-molecule inhibitors of PTPN2 demonstrating that these compounds may have clinical utility as sensitizing agents for immunotherapy-resistant cancers.
Collapse
Affiliation(s)
- Zhouting Zhu
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Rachel Tang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Sarah Huff
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Indrasena Reddy Kummetha
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Lingling Wang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Na Li
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Tariq M. Rana
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
- San Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California San Diego, La Jolla, California
| |
Collapse
|
6
|
Song J, Lan J, Tang J, Luo N. PTPN2 in the Immunity and Tumor Immunotherapy: A Concise Review. Int J Mol Sci 2022; 23:ijms231710025. [PMID: 36077422 PMCID: PMC9456094 DOI: 10.3390/ijms231710025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
PTPN2 (protein tyrosine phosphatase non-receptor 2), also called TCPTP (T cell protein tyrosine phosphatase), is a member of the PTP family signaling proteins. Phosphotyrosine-based signaling of this non-transmembrane protein is essential for regulating cell growth, development, differentiation, survival, and migration. In particular, PTPN2 received researchers’ attention when Manguso et al. identified PTPN2 as a cancer immunotherapy target using in vivo CRISPR library screening. In this review, we attempt to summarize the important functions of PTPN2 in terms of its structural and functional properties, inflammatory reactions, immunomodulatory properties, and tumor immunity. PTPN2 exerts synergistic anti-inflammatory effects in various inflammatory cells and regulates the developmental differentiation of immune cells. The diversity of PTPN2 effects in different types of tumors makes it a potential target for tumor immunotherapy.
Collapse
|
7
|
Asare O, Ayala Y, Hafeez BB, Ramirez-Correa GA, Cho YY, Kim DJ. Ultraviolet Radiation Exposure and its Impacts on Cutaneous Phosphorylation Signaling in Carcinogenesis: Focusing on Protein Tyrosine Phosphatases †. Photochem Photobiol 2022; 99:344-355. [PMID: 36029171 DOI: 10.1111/php.13703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/23/2022] [Indexed: 01/14/2023]
Abstract
Sunlight exposure is a significant risk factor for UV-induced deteriorating transformations of epidermal homeostasis leading to skin carcinogenesis. The ability of UVB radiation to cause melanoma, as well as basal and squamous cell carcinomas, makes UVB the most harmful among the three known UV ranges. UVB-induced DNA mutations and dysregulation of signaling pathways contribute to skin cancer formation. Among various signaling pathways modulated by UVB, tyrosine phosphorylation signaling which is mediated by the action of protein tyrosine kinases (PTKs) on specific tyrosine residues is highly implicated in photocarcinogenesis. Following UVB irradiation, PTKs get activated and their downstream signaling pathways contribute to photocarcinogenesis by promoting the survival of damaged keratinocytes and increasing cell proliferation. While UVB activates oncogenic signaling pathways, it can also activate tumor suppressive signaling pathways as initial protective mechanisms to maintain epidermal homeostasis. Tyrosine dephosphorylation is one of the protective mechanisms and is mediated by the action of protein tyrosine phosphatases (PTPs). PTP can counteract UVB-mediated PTK activation and downregulate oncogenic signaling pathways. However, PTPs have not been studied extensively in photocarcinogenesis with previous studies regarding their inactivation induced by UVB. This current review will summarize the recent progress in the protective function of PTPs in epidermal photocarcinogenesis.
Collapse
Affiliation(s)
- Obed Asare
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX
| | - Yasmin Ayala
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX
| | - Bilal Bin Hafeez
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX.,South Texas Center for Excellence in Cancer Research, University of Texas Rio Grande Valley, Edinburg, TX
| | - Genaro A Ramirez-Correa
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Korea
| | - Dae Joon Kim
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX.,South Texas Center for Excellence in Cancer Research, University of Texas Rio Grande Valley, Edinburg, TX.,Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX
| |
Collapse
|
8
|
Tang X, Qi C, Zhou H, Liu Y. Critical roles of PTPN family members regulated by non-coding RNAs in tumorigenesis and immunotherapy. Front Oncol 2022; 12:972906. [PMID: 35957898 PMCID: PMC9360549 DOI: 10.3389/fonc.2022.972906] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/04/2022] [Indexed: 12/22/2022] Open
Abstract
Since tyrosine phosphorylation is reversible and dynamic in vivo, the phosphorylation state of proteins is controlled by the opposing roles of protein tyrosine kinases (PTKs) and protein tyrosine phosphatase (PTPs), both of which perform critical roles in signal transduction. Of these, intracellular non-receptor PTPs (PTPNs), which belong to the largest class I cysteine PTP family, are essential for the regulation of a variety of biological processes, including but not limited to hematopoiesis, inflammatory response, immune system, and glucose homeostasis. Additionally, a substantial amount of PTPNs have been identified to hold crucial roles in tumorigenesis, progression, metastasis, and drug resistance, and inhibitors of PTPNs have promising applications due to striking efficacy in antitumor therapy. Hence, the aim of this review is to summarize the role played by PTPNs, including PTPN1/PTP1B, PTPN2/TC-PTP, PTPN3/PTP-H1, PTPN4/PTPMEG, PTPN6/SHP-1, PTPN9/PTPMEG2, PTPN11/SHP-2, PTPN12/PTP-PEST, PTPN13/PTPL1, PTPN14/PEZ, PTPN18/PTP-HSCF, PTPN22/LYP, and PTPN23/HD-PTP, in human cancer and immunotherapy and to comprehensively describe the molecular pathways in which they are implicated. Given the specific roles of PTPNs, identifying potential regulators of PTPNs is significant for understanding the mechanisms of antitumor therapy. Consequently, this work also provides a review on the role of non-coding RNAs (ncRNAs) in regulating PTPNs in tumorigenesis and progression, which may help us to find effective therapeutic agents for tumor therapy.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, China
| | - Chumei Qi
- Department of Clinical Laboratory, Dazhou Women and Children’s Hospital, Dazhou, China
| | - Honghong Zhou
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| |
Collapse
|
9
|
Huang X, Lai S, Qu F, Li Z, Fu X, Li Q, Zhong X, Wang C, Li H. CCL18 promotes breast cancer progression by exosomal miR-760 activation of ARF6/Src/PI3K/Akt pathway. Mol Ther Oncolytics 2022; 25:1-15. [PMID: 35399607 PMCID: PMC8971730 DOI: 10.1016/j.omto.2022.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/13/2022] [Indexed: 11/02/2022] Open
Abstract
The small GTPase ADP-ribosylation factor 6 (ARF6) mediates chemokine (C-C motif) ligand 18 (CCL18)-induced activation of breast cancer (BC) metastasis through its downstream effector AMAP1. However, the molecular mechanisms underlying CCL18 up-regulating ARF6 remain largely unclear. Here, microRNAs (miRNAs) that target ARF6 were predicted and selected in high metastatic BC cells treated with CCL18. Next, we assessed the role of exosomal miR-760 in vitro and in vivo. We further analyzed the expression of ARF6, AMAP1, and phosphorylated (p)-AMAP1 in tumor and adjacent normal tissues. We first observed that CCL18 increased the expression of ARF6 and p-AMAP1 and activated the Src/phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. ARF6 knockdown significantly impaired CCL18-induced malignant cellular behaviors and the Src/PI3K/Akt signaling pathway. Next, ARF6 was confirmed as a target gene of miR-760 in exosomes derived from CCL18-stimulated high metastatic BC cells. Moreover, recipient MCF-7 cells could effectively uptake these miR-760-rich exosomes that significantly promoted proliferation, tumor growth in vivo, migration, invasion, and chemoresistance by activating ARF6-mediated Src/PI3K/Akt signaling and the epithelial-mesenchymal transition (EMT) pathway. Together, our results support that exosomal miR-760 secreted by CCL18-stimulated high metastatic BC cells promoted the malignant behaviors in low metastatic BC cells by up-regulating the ARF6-mediated Src/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xiaojia Huang
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Shengqing Lai
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Fanli Qu
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Zongyan Li
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Xiaoyan Fu
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Qian Li
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Xiaofang Zhong
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| | - Chao Wang
- Department of Pathology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Haiyan Li
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong 510655, China
| |
Collapse
|
10
|
Dal Berto M, Dos Santos GT, Dos Santos AV, Silva AO, Vargas JE, Alves RJV, Barbisan F, da Cruz IBM, Bica CG. Molecular markers associated with the outcome of tamoxifen treatment in estrogen receptor-positive breast cancer patients: scoping review and in silico analysis. Discov Oncol 2021; 12:37. [PMID: 35201456 PMCID: PMC8777552 DOI: 10.1007/s12672-021-00432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Tamoxifen (TMX) is used as adjuvant therapy for estrogen receptor-positive (ER+) breast cancer cases due to its affinity and inhibitory effects. However, about 30% of cases show drug resistance, resulting in recurrence and metastasis, the leading causes of death. A literature review can help to elucidate the main cellular processes involved in TMX resistance. A scoping review was performed to find clinical studies investigating the association of expression of molecular markers profiles with long-term outcomes in ER+ patients treated with TMX. In silico analysis was performed to assess the interrelationship among the selected markers, evaluating the joint involvement with the biological processes. Forty-five studies were selected according to the inclusion and exclusion criteria. After clustering and gene ontology analysis, 23 molecular markers were significantly associated, forming three clusters of strong correlation with cell cycle regulation, signal transduction of proliferative stimuli, and hormone response involved in morphogenesis and differentiation of mammary gland. Also, it was found that overexpression of markers in selected clusters is a significant indicator of poor overall survival. The proposed review offered a better understanding of independent data from the literature, revealing an integrative network of markers involved in cellular processes that could modulate the response of TMX. Analysis of these mechanisms and their molecular components could improve the effectiveness of TMX.
Collapse
Affiliation(s)
- Maiquidieli Dal Berto
- Laboratory of Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245,Sarmento Leite street, Porto Alegre, RS, 90050-170, Brazil
| | - Giovana Tavares Dos Santos
- Laboratory of Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245,Sarmento Leite street, Porto Alegre, RS, 90050-170, Brazil
| | - Aniúsca Vieira Dos Santos
- Laboratory of Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245,Sarmento Leite street, Porto Alegre, RS, 90050-170, Brazil
| | - Andrew Oliveira Silva
- Laboratory of Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245,Sarmento Leite street, Porto Alegre, RS, 90050-170, Brazil
| | - José Eduardo Vargas
- Institute of Biological Sciences, University of Passo Fundo (UPF), 285, Brazil Avenue, Passo Fundo, RS, 99052-900, Brazil
| | - Rafael José Vargas Alves
- Department of Clinical Medicine, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245, Sarmento Leite street, Porto Alegre, RS, 90050-170, Brazil
| | - Fernanda Barbisan
- Graduate Program in Gerontology, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | | | - Claudia Giuliano Bica
- Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245, Sarmento Leite street., Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
11
|
Turan T, Kongpachith S, Halliwill K, Roelands J, Hendrickx W, Marincola FM, Hudson TJ, Jacob HJ, Bedognetti D, Samayoa J, Ceccarelli M. A balance score between immune stimulatory and suppressive microenvironments identifies mediators of tumour immunity and predicts pan-cancer survival. Br J Cancer 2020; 124:760-769. [PMID: 33139798 PMCID: PMC7884411 DOI: 10.1038/s41416-020-01145-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/26/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The balance between immune-stimulatory and immune-suppressive mechanisms in the tumour microenvironment is associated with tumour rejection and can predict the efficacy of immune checkpoint-inhibition therapies. METHODS We consider the observed differences between the transcriptional programmes associated with cancer types where the levels of immune infiltration predict a favourable prognosis versus those in which the immune infiltration predicts an unfavourable prognosis and defined a score named Mediators of Immune Response Against Cancer in soLid microEnvironments (MIRACLE). MIRACLE deconvolves T cell infiltration, from inhibitory mechanisms, such as TGFβ, EMT and PI3Kγ signatures. RESULTS Our score outperforms current state-of-the-art immune signatures as a predictive marker of survival in TCGA (n = 9305, HR: 0.043, p value: 6.7 × 10-36). In a validation cohort (n = 7623), MIRACLE predicts better survival compared to other immune metrics (HR: 0.1985, p value: 2.73 × 10-38). MIRACLE also predicts response to checkpoint-inhibitor therapies (n = 333). The tumour-intrinsic factors inversely associated with the reported score such as EGFR, PRKAR1A and MAP3K1 are frequently associated with immune-suppressive phenotypes. CONCLUSIONS The association of cancer outcome with the level of infiltrating immune cells is mediated by the balance of activatory and suppressive factors. MIRACLE accounts for this balance and predicts favourable cancer outcomes.
Collapse
Affiliation(s)
- Tolga Turan
- Computational Immunology and Oncology (CIAO), AbbVie, Redwood City, CA, USA
| | - Sarah Kongpachith
- Computational Immunology and Oncology (CIAO), AbbVie, Redwood City, CA, USA
| | - Kyle Halliwill
- Computational Immunology and Oncology (CIAO), AbbVie, Redwood City, CA, USA
| | - Jessica Roelands
- Cancer Research Department, Sidra Medicine, Doha, Qatar.,Department of Surgery, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | | | | | - Thomas J Hudson
- Computational Immunology and Oncology (CIAO), AbbVie, Redwood City, CA, USA
| | - Howard J Jacob
- Genomics Research Center (GRC), AbbVie, Lake County, IL, USA
| | - Davide Bedognetti
- Cancer Research Department, Sidra Medicine, Doha, Qatar. .,Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, Genova, Italy.
| | - Josue Samayoa
- Computational Immunology and Oncology (CIAO), AbbVie, Redwood City, CA, USA.
| | - Michele Ceccarelli
- Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Naples, Italy. .,Istituto di Ricerche Genetiche "G. Salvatore", Biogem s.c.ar.l, 83031, Ariano Irpino, Italy.
| |
Collapse
|
12
|
Pardella E, Pranzini E, Leo A, Taddei ML, Paoli P, Raugei G. Oncogenic Tyrosine Phosphatases: Novel Therapeutic Targets for Melanoma Treatment. Cancers (Basel) 2020; 12:E2799. [PMID: 33003469 PMCID: PMC7599540 DOI: 10.3390/cancers12102799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a large number of therapeutic options available, malignant melanoma remains a highly fatal disease, especially in its metastatic forms. The oncogenic role of protein tyrosine phosphatases (PTPs) is becoming increasingly clear, paving the way for novel antitumor treatments based on their inhibition. In this review, we present the oncogenic PTPs contributing to melanoma progression and we provide, where available, a description of new inhibitory strategies designed against these enzymes and possibly useful in melanoma treatment. Considering the relevance of the immune infiltrate in supporting melanoma progression, we also focus on the role of PTPs in modulating immune cell activity, identifying interesting therapeutic options that may support the currently applied immunomodulating approaches. Collectively, this information highlights the value of going further in the development of new strategies targeting oncogenic PTPs to improve the efficacy of melanoma treatment.
Collapse
Affiliation(s)
- Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (E.P.); (E.P.); (A.L.); (G.R.)
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (E.P.); (E.P.); (A.L.); (G.R.)
| | - Angela Leo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (E.P.); (E.P.); (A.L.); (G.R.)
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (E.P.); (E.P.); (A.L.); (G.R.)
| | - Giovanni Raugei
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (E.P.); (E.P.); (A.L.); (G.R.)
| |
Collapse
|
13
|
Kim M, Morales LD, Lee CJ, Olivarez SA, Kim WJ, Hernandez J, Mummidi S, Jenkinson C, Tsin AT, Jang IS, Slaga TJ, Kim DJ. Overexpression of TC-PTP in murine epidermis attenuates skin tumor formation. Oncogene 2020; 39:4241-4256. [PMID: 32286519 PMCID: PMC7244373 DOI: 10.1038/s41388-020-1282-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 03/18/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023]
Abstract
T-cell protein tyrosine phosphatase (TC-PTP), encoded by Ptpn2, has been shown to function as a tumor suppressor during skin carcinogenesis. In the current study, we generated a novel epidermal-specific TC-PTP-overexpressing (K5HA.Ptpn2) mouse model to show that TC-PTP contributes to the attenuation of chemically induced skin carcinogenesis through the synergistic regulation of STAT1, STAT3, STAT5, and PI3K/AKT signaling. We found overexpression of TC-PTP increased epidermal sensitivity to DMBA-induced apoptosis and it decreased TPA-mediated hyperproliferation, coinciding with reduced epidermal thickness. Inhibition of STAT1, STAT3, STAT5, or AKT reversed the effects of TC-PTP overexpression on epidermal survival and proliferation. Mice overexpressing TC-PTP in the epidermis developed significantly reduced numbers of tumors during skin carcinogenesis and presented a prolonged latency of tumor initiation. Examination of human papillomas and squamous cell carcinomas (SCCs) revealed that TC-PTP expression was significantly reduced and TC-PTP expression was inversely correlated with the increased grade of SCCs. Our findings demonstrate that TC-PTP is a potential therapeutic target for the prevention of human skin cancer given that it is a major negative regulator of oncogenic signaling.
Collapse
Affiliation(s)
- Mihwa Kim
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Liza D Morales
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Cheol Jung Lee
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Serena A Olivarez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Woo Jin Kim
- School of Mathematical and Statistical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Joselin Hernandez
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Srinivas Mummidi
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Christopher Jenkinson
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Andrew T Tsin
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Ik-Soon Jang
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Daejeon, 305-333, Republic of Korea
| | - Thomas J Slaga
- Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Dae Joon Kim
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA.
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA.
| |
Collapse
|
14
|
Xia Z, Su Y, Petersen P, Qi L, Kim AE, Figueiredo JC, Lin Y, Nan H, Sakoda LC, Albanes D, Berndt SI, Bézieau S, Bien S, Buchanan DD, Casey G, Chan AT, Conti DV, Drew DA, Gallinger SJ, Gauderman WJ, Giles GG, Gruber SB, Gunter MJ, Hoffmeister M, Jenkins MA, Joshi AD, Le Marchand L, Lewinger JP, Li L, Lindor NM, Moreno V, Murphy N, Nassir R, Newcomb PA, Ogino S, Rennert G, Song M, Wang X, Wolk A, Woods MO, Brenner H, White E, Slattery ML, Giovannucci EL, Chang‐Claude J, Pharoah PDP, Hsu L, Campbell PT, Peters U. Functional informed genome-wide interaction analysis of body mass index, diabetes and colorectal cancer risk. Cancer Med 2020; 9:3563-3573. [PMID: 32207560 PMCID: PMC7221445 DOI: 10.1002/cam4.2971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/28/2020] [Accepted: 02/21/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Body mass index (BMI) and diabetes are established risk factors for colorectal cancer (CRC), likely through perturbations in metabolic traits (e.g. insulin resistance and glucose homeostasis). Identification of interactions between variation in genes and these metabolic risk factors may identify novel biologic insights into CRC etiology. METHODS To improve statistical power and interpretation for gene-environment interaction (G × E) testing, we tested genetic variants that regulate expression of a gene together for interaction with BMI (kg/m2 ) and diabetes on CRC risk among 26 017 cases and 20 692 controls. Each variant was weighted based on PrediXcan analysis of gene expression data from colon tissue generated in the Genotype-Tissue Expression Project for all genes with heritability ≥1%. We used a mixed-effects model to jointly measure the G × E interaction in a gene by partitioning the interactions into the predicted gene expression levels (fixed effects), and residual G × E effects (random effects). G × BMI analyses were stratified by sex as BMI-CRC associations differ by sex. We used false discovery rates to account for multiple comparisons and reported all results with FDR <0.2. RESULTS Among 4839 genes tested, genetically predicted expressions of FOXA1 (P = 3.15 × 10-5 ), PSMC5 (P = 4.51 × 10-4 ) and CD33 (P = 2.71 × 10-4 ) modified the association of BMI on CRC risk for men; KIAA0753 (P = 2.29 × 10-5 ) and SCN1B (P = 2.76 × 10-4 ) modified the association of BMI on CRC risk for women; and PTPN2 modified the association between diabetes and CRC risk in both sexes (P = 2.31 × 10-5 ). CONCLUSIONS Aggregating G × E interactions and incorporating functional information, we discovered novel genes that may interact with BMI and diabetes on CRC risk.
Collapse
|
15
|
Abstract
This volume is intended to review the methods used to identify biomarkers predictive of cancer responsiveness to immunotherapy. The successful development of clinically actionable biomarkers depends upon three features: (a) their biological role with respect to malignant transformation and tumor progression; (b) the ability to detect them with robust, reliable, and clinically applicable assays; and (c) their prognostic or predictive value, as validated in clinical trials.Identifying biomarkers that have predictive value for patient selection based on the likelihood of benefiting from anticancer immunotherapy is a lengthy and complex process. To date, few predictive biomarkers for anticancer immunotherapy have been robustly analytically and clinically validated (i.e., PD-L1 expression as measured by IHC assays and microsatellite instability (MSI)/dMMR as measured by PCR or IHC, respectively).This introductory chapter to this book focuses on scientific and technical aspects relevant to the identification and validation of predictive biomarkers for immunotherapy. We emphasize that methods should address both the biology of the tumor and the tumor microenvironment. Moreover, the identification of biomarkers requires highly sensitive, multiplexed, comprehensive techniques, especially for application in clinical care. Thus, in this chapter, we will define the outstanding questions related to the immune biology of cancer as a base for development of the biomarkers and assays using diverse methodologies. These biomarkers will likely be identified through research that integrates conventional immunological approaches along with high-throughput genomic and proteomic screening and the host immune response of individual patients that relates to individual tumor biology and immune drugs' mechanism of action.Checkpoint inhibitor therapy (CIT) is by now an accepted modality of cancer treatment. However, immune resistance is common, and most patients do not benefit from the treatment. The reasons for resistance are diverse, and approaches to circumvent it need to consider genetic, biologic, and environmental factors that affect anticancer immune response. Here, we propose to systematically address fundamental concepts based on the premise that malignant cells orchestrate their surroundings by interacting with innate and adaptive immune sensors. This principle applies to most cancers and governs their evolution in the immune-competent host. Understanding the basic requirement(s) for this evolutionary process will guide biomarker discovery and validation and ultimately guide to effective therapeutic choices. This volume will also discuss novel biomarker approaches aimed at informing an effective assay development from a mechanistic point of view, as well as the clinical implementation (i.e., patient enrichment) for immune therapies.
Collapse
Affiliation(s)
- Alessandra Cesano
- ESSA Pharma, South San Francisco, CA, USA.
- Nanostring Technologies, Seattle, WA, USA.
| | | | - Magdalena Thurin
- Cancer Diagnosis Program, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
16
|
Huang L, Tang X, Shi X, Su L. miR-532-5p promotes breast cancer proliferation and migration by targeting RERG. Exp Ther Med 2019; 19:400-408. [PMID: 31853317 DOI: 10.3892/etm.2019.8186] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/04/2019] [Indexed: 02/06/2023] Open
Abstract
Aberrant expression of microRNAs (miRNAs/miRs) mediates the initiation and progression of breast cancer. Therefore, it is important to investigate the molecular mechanisms of miRNAs and their effects on breast cancer progression. In the present study, miR-532-5p was highly expressed in breast cancer tissues compared with normal tissues. In addition, expression of ras-related and estrogen-regulated growth inhibitor (RERG), a tumor suppressor in breast cancer, was negatively correlated with miR-532-5p expression. Inhibition of miR-532-5p significantly elevated RERG at both mRNA and protein levels and inactivated the mitogen-activated protein kinase (MAPK)/ERK signaling pathway. Overexpression of miR-532-5p decreased RERG expression and activated the MAPK/ERK signaling in breast cancer cell line MDA-MB-231. Bioinformatic analysis indicated that RERG 3'-untraslated region contained a putative binding site for miR-532-5p. Dual luciferase assay further validated RERG as a target gene of miR-532-5p. Notably, downregulation of miR-532-5p inhibited MDA-MB-231 cell proliferation and migration, which was partially attenuated upon RERG knockdown. In conclusion, the current study revealed an oncogenic role of miR-532-5p in breast cancer cells via direct targeting of RERG expression.
Collapse
Affiliation(s)
- Lei Huang
- Department of Breast and Thyroid Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoqiao Tang
- Department of Breast and Thyroid Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xianbiao Shi
- Department of Breast and Thyroid Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Lei Su
- Department of Breast and Thyroid Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
17
|
Wu L, Wang F, Xu J, Chen Z. PTPN2 induced by inflammatory response and oxidative stress contributed to glioma progression. J Cell Biochem 2019; 120:19044-19051. [PMID: 31241223 DOI: 10.1002/jcb.29227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
Malignant glioma remains the most frequent form of primary brain tumors all over the world. The gliomagenesis is characterized by various molecular processes such as neoplastic transformation, dysregulation of the cell cycle, and angiogenesis. Among these biomolecular events, the existence of inflammation and oxidative stress pathways in the development of glioma has been reported. PTPN2 is associated with several inflammatory disorders. However, the biological role of PTPN2 in inflammation responses and oxidative stress pathways involved in glioma remains poorly known. Here, we focused on its function in glioma development. Here, we observed that PTPN2 was significantly increased in glioma especially in a grade-dependent manner. Meanwhile, interferon-γ and tumor necrosis factor-α, which have been identified as crucial inflammation cytokines, were able to trigger PTPN2 expression in a dose-dependent course in T98G cells. Then, we found that PTPN2 was oxidated and inactivated by H2 O2 . Meanwhile, H2 O2 induced glioma cell colony formation capacity and increased ki-67 expression confirmed by flow cytometry assay. Finally, T98G cells were transfected with PTPN2 shRNA and it was shown that knockdown of PTPN2 obviously inhibited T98G cell colony formation and induced cell apoptosis. In summary, our findings indicated that PTPN2 could be induced by inflammatory response and oxidative stress and its deficiency depressed glioma cell growth.
Collapse
Affiliation(s)
- Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Hospital of Xuzhou Medical University, Huai'an, China
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Szijgyarto Z, Flach KD, Opdam M, Palmieri C, Linn SC, Wesseling J, Ali S, Bliss JM, Cheang MCU, Zwart W, Coombes RC. Dissecting the predictive value of MAPK/AKT/estrogen-receptor phosphorylation axis in primary breast cancer to treatment response for tamoxifen over exemestane: a Translational Report of the Intergroup Exemestane Study (IES)-PathIES. Breast Cancer Res Treat 2019; 175:149-163. [PMID: 30680659 PMCID: PMC6491661 DOI: 10.1007/s10549-018-05110-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/18/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE The prognostic and predictive values of the MAPK/AKT/ERα phosphorylation axis (pT202/T204MAPK, pT308AKT, pS473AKT, pS118ERα and pS167ERα) in primary tumours were assessed to determine whether these markers can differentiate between patient responses for switching adjuvant endocrine therapy after 2-3 years from tamoxifen to exemestane and continued tamoxifen monotherapy in the Intergroup Exemestane Study (IES). METHODS Of the 4724 patients in IES, 1506 were managed in a subset of centres (N = 89) participating in PathIES. These centres recruited 1282 (85%, 1282/1506) women into PathIES of whom 1036 had phospho-marker data. All phospho-markers were analysed by immunohistochemistry staining. Multivariable Cox proportional hazards models of the phospho-markers for disease-free survival (DFS) and overall survival (OS) were adjusted for clinicopathological factors. Treatment effects on the biomarker expression were determined by interaction tests. Benjamini-Hochberg adjustment for multiple testing with a false discovery rate of 10% was applied (pBH). RESULTS Phospho-T202/T204MAPK, pS118ERα and pS167ERα were all found to be correlated (pBH = 0.0002). These markers were not associated with either DFS or OS when controlling for the established clinicopathological factors. Interaction terms between the phospho-markers and treatment strategies for either DFS or OS were not statistically significant (pBH > 0.05 for all). CONCLUSIONS This PathIES study confirmed previously described associations between the phosphorylation site markers of AKT, MAPK and ERα activity in postmenopausal breast cancer patients. No prognostic correlations between the phosphorylation markers and clinical outcome were found, nor were they predictive for clinical outcomes among patients who switched therapy over those treated with tamoxifen alone.
Collapse
Affiliation(s)
- Zsolt Szijgyarto
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG UK
| | - Koen D. Flach
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Carlo Palmieri
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3BX UK
- Academic Department of Medical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, CH63 4JY UK
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN UK
| | - Sabine C. Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Department of Medical Onology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Simak Ali
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN UK
| | - Judith M. Bliss
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG UK
| | - Maggie Chon U. Cheang
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG UK
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - R. Charles Coombes
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN UK
| |
Collapse
|
19
|
Veenstra C, Karlsson E, Mirwani SM, Nordenskjöld B, Fornander T, Pérez-Tenorio G, Stål O. The effects of PTPN2 loss on cell signalling and clinical outcome in relation to breast cancer subtype. J Cancer Res Clin Oncol 2019; 145:1845-1856. [PMID: 31025094 PMCID: PMC6571101 DOI: 10.1007/s00432-019-02918-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/15/2019] [Indexed: 11/28/2022]
Abstract
Purpose The protein tyrosine phosphatase PTPN2 dephosphorylates several tyrosine kinases in cancer-related signalling pathways and is thought to be a tumour suppressor. As PTPN2 is not frequently studied in breast cancer, we aimed to explore the role of PTPN2 and the effects of its loss in breast cancer. Methods Protein expression and gene copy number of PTPN2 were analysed in a cohort of pre-menopausal breast cancer patients with immunohistochemistry and droplet digital PCR, respectively. PTPN2 was knocked down in three cell lines, representing different breast cancer subtypes, with siRNA transfection. Several proteins related to PTPN2 were analysed with Western blot. Results Low PTPN2 protein expression was found in 50.2% of the tumours (110/219), gene copy loss in 15.4% (33/214). Low protein expression was associated with a higher relapse rate in patients with Luminal A and HER2-positive tumours, but not triple-negative tumours. In vitro studies further suggested a subtype-specific role of PTPN2. Knockdown of PTPN2 had no effect on the triple-negative cell line, whilst knockdown in MCF7 inhibited phosphorylation of Met and promoted that of Akt. Knockdown in SKBR3 led to increased Met phosphorylation and decreased Erk phosphorylation as well as EGF-mediated STAT3 activation. Conclusion We confirm previous studies showing that the PTPN2 protein is lost in half of the breast cancer cases and gene deletion occurs in 15–18% of the cases. Furthermore, the results suggest that the role of PTPN2 is subtype-related and should be further investigated to assess how this could affect breast cancer prognosis and treatment response.
Collapse
Affiliation(s)
- Cynthia Veenstra
- Division of Clinical Sciences, Department of Clinical and Experimental Medicine and Department of Oncology, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden.
| | - Elin Karlsson
- Division of Clinical Sciences, Department of Clinical and Experimental Medicine and Department of Oncology, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Sanam Mirwani Mirwani
- Division of Clinical Sciences, Department of Clinical and Experimental Medicine and Department of Oncology, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Bo Nordenskjöld
- Division of Clinical Sciences, Department of Clinical and Experimental Medicine and Department of Oncology, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Tommy Fornander
- Department of Oncology-Pathology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Gizeh Pérez-Tenorio
- Division of Clinical Sciences, Department of Clinical and Experimental Medicine and Department of Oncology, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Olle Stål
- Division of Clinical Sciences, Department of Clinical and Experimental Medicine and Department of Oncology, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden
| |
Collapse
|
20
|
Ellegård S, Veenstra C, Pérez-Tenorio G, Fagerström V, Gårsjö J, Gert K, Sundquist M, Malmström A, Wingren S, Elander NO, Hallbeck AL, Stål O. ERBB2 and PTPN2 gene copy numbers as prognostic factors in HER2-positive metastatic breast cancer treated with trastuzumab. Oncol Lett 2019; 17:3371-3381. [PMID: 30867772 PMCID: PMC6396168 DOI: 10.3892/ol.2019.9998] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 12/25/2022] Open
Abstract
Trastuzumab has markedly improved the treatment and long-term prognosis of patients with HER2-positive breast cancer. A frequent clinical challenge in patients with relapsing and/or metastatic disease is de novo or acquired trastuzumab resistance, and to date no predictive biomarkers for palliative trastuzumab have been established. In the present study, the prognostic values of factors involved in the HER2-associated PI3K/Akt signalling pathway were explored. The first 46 consecutive patients treated at the Department of Oncology, Linköping University Hospital between 2000 and 2007 with trastuzumab for HER2-positive metastatic breast cancer were retrospectively included. The gene copy number variation and protein expression of several components of the PI3K/Akt pathway were assessed in the tumour tissue and biopsy samples using droplet digital polymerase chain reaction and immunohistochemistry. Patients with tumours displaying a high-grade ERBB2 (HER2) amplification level of ≥6 copies had a significantly improved overall survival hazard ratio [(HR)=0.4; 95%, confidence interval (CI): 0.2–0.9] and progression-free survival (HR=0.3; 95% CI: 0.1–0.7) compared with patients with tumours harbouring fewer ERBB2 copies. High-grade ERBB2 amplification was significantly associated with the development of central nervous system metastases during palliative treatment. Copy gain (≥3 copies) of the gene encoding the tyrosine phosphatase PTPN2 was associated with a shorter overall survival (HR=2.0; 95% CI: 1.0–4.0) and shorter progression-free survival (HR=2.1; 95% CI: 1.0–4.1). In conclusion, high ERBB2 amplification level is a potential positive prognostic factor in trastuzumab-treated HER2-positive metastatic breast cancer, whereas PTPN2 gain is a potential negative prognostic factor. Further studies are warranted on the role of PTPN2 in HER2 signalling.
Collapse
Affiliation(s)
- Sander Ellegård
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Cynthia Veenstra
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Gizeh Pérez-Tenorio
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Victor Fagerström
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden.,Department of Surgery, Kalmar Hospital, SE-392 44 Kalmar, Sweden
| | - Jon Gårsjö
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Krista Gert
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Marie Sundquist
- Department of Surgery, Kalmar Hospital, SE-392 44 Kalmar, Sweden
| | - Annika Malmström
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Sten Wingren
- Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, SE-701 82 Örebro, Sweden
| | - Nils O Elander
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Anna-Lotta Hallbeck
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| | - Olle Stål
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE-581 85 Linköping, Sweden
| |
Collapse
|
21
|
Karlsson E, Veenstra C, Gårsjö J, Nordenskjöld B, Fornander T, Stål O. PTPN2 deficiency along with activation of nuclear Akt predict endocrine resistance in breast cancer. J Cancer Res Clin Oncol 2018; 145:599-607. [PMID: 30515568 PMCID: PMC6394658 DOI: 10.1007/s00432-018-2810-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 11/30/2018] [Indexed: 01/18/2023]
Abstract
Purpose The protein tyrosine phosphatase, non-receptor type 2 (PTNP2) regulates receptor tyrosine kinase signalling, preventing downstream activation of intracellular pathways like the PI3K/Akt pathway. The gene encoding the protein is located on chromosome 18p11; the 18p region is commonly deleted in breast cancer. In this study, we aimed to evaluate PTPN2 protein expression in a large breast cancer cohort, its possible associations to PTPN2 gene copy loss, Akt activation, and the potential use as a clinical marker in breast cancer. Methods PTPN2 protein expression was analysed by immunohistochemistry in 664 node-negative breast tumours from patients enrolled in a randomised tamoxifen trial. DNA was available for 146 patients, PTPN2 gene copy number was determined by real-time PCR. Results PTPN2 gene loss was detected in 17.8% of the tumours. Low PTPN2 protein expression was associated with higher levels of nuclear-activated Akt (pAkt-n). Low PTPN2 as well as the combination variable low PTPN2/high pAkt-n could be used as predictive markers of poor tamoxifen response. Conclusion PTPN2 negatively regulates Akt signalling and loss of PTPN2 protein along with increased pAkt-n is a new potential clinical marker of endocrine treatment efficacy, which may allow for further tailored patient therapies.
Collapse
Affiliation(s)
- Elin Karlsson
- Department of Clinical and Experimental Medicine, Department of Oncology, Linköping University, 58185, Linköping, Sweden
| | - Cynthia Veenstra
- Department of Clinical and Experimental Medicine, Department of Oncology, Linköping University, 58185, Linköping, Sweden.
| | - Jon Gårsjö
- Department of Clinical and Experimental Medicine, Department of Oncology, Linköping University, 58185, Linköping, Sweden
| | - Bo Nordenskjöld
- Department of Clinical and Experimental Medicine, Department of Oncology, Linköping University, 58185, Linköping, Sweden
| | - Tommy Fornander
- Department of Oncology, Karolinska University Hospital and Karolinska Institute, 17176, Stockholm, Sweden
| | - Olle Stål
- Department of Clinical and Experimental Medicine, Department of Oncology, Linköping University, 58185, Linköping, Sweden
| |
Collapse
|
22
|
Immune oncology, immune responsiveness and the theory of everything. J Immunother Cancer 2018; 6:50. [PMID: 29871670 PMCID: PMC5989400 DOI: 10.1186/s40425-018-0355-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/11/2018] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer immunotherapy is encountering its own checkpoint. Responses are dramatic and long lasting but occur in a subset of tumors and are largely dependent upon the pre-existing immune contexture of individual cancers. Available data suggest that three landscapes best define the cancer microenvironment: immune-active, immune-deserted and immune-excluded. This trichotomy is observable across most solid tumors (although the frequency of each landscape varies depending on tumor tissue of origin) and is associated with cancer prognosis and response to checkpoint inhibitor therapy (CIT). Various gene signatures (e.g. Immunological Constant of Rejection - ICR and Tumor Inflammation Signature - TIS) that delineate these landscapes have been described by different groups. In an effort to explain the mechanisms of cancer immune responsiveness or resistance to CIT, several models have been proposed that are loosely associated with the three landscapes. Here, we propose a strategy to integrate compelling data from various paradigms into a “Theory of Everything”. Founded upon this unified theory, we also propose the creation of a task force led by the Society for Immunotherapy of Cancer (SITC) aimed at systematically addressing salient questions relevant to cancer immune responsiveness and immune evasion. This multidisciplinary effort will encompass aspects of genetics, tumor cell biology, and immunology that are pertinent to the understanding of this multifaceted problem.
Collapse
|
23
|
Huaier extract synergizes with tamoxifen to induce autophagy and apoptosis in ER-positive breast cancer cells. Oncotarget 2018; 7:26003-15. [PMID: 27027343 PMCID: PMC5041960 DOI: 10.18632/oncotarget.8303] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/02/2016] [Indexed: 12/22/2022] Open
Abstract
Tamoxifen (TAM) is the most widely used endocrine therapy for estrogen receptor (ER)-positive breast cancer patients, but side effects and the gradual development of insensitivity limit its application. We investigated whether Huaier extract, a traditional Chinese medicine, in combination with TAM would improve treatment efficacy in ER-positive breast cancers. MTT, colony formation, and invasion and migration assays revealed that the combined treatment had stronger anticancer effects than either treatment alone. Huaier extract enhanced TAM-induced autophagy, apoptosis, and G0/G1 cell cycle arrest, as measured by acidic vesicular organelle (AVO) staining, TUNEL, flow cytometry, and western blot. Additionally, combined treatment inhibited tumorigenesis and metastasis by suppressing the AKT/mTOR signaling pathway. Huaier extract also enhanced the inhibitory effects of TAM on tumor growth in vivo in a xenograft mouse model. These results show that Huaier extract synergizes with TAM to induce autophagy and apoptosis in ER-positive breast cancer cells by suppressing the AKT/mTOR pathway.
Collapse
|
24
|
Cancer immune resistance: can theories converge? Emerg Top Life Sci 2017; 1:411-419. [PMID: 33525800 PMCID: PMC7289003 DOI: 10.1042/etls20170060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
Immune oncology (IO) is challenged to expand its usefulness to a broader range of cancers. A second generation of IO agents acting beyond the realm of Checkpoint Inhibitor Therapy (CIT) is sought with the intent of turning immune-resistant cancers into appealing IO targets. The published literature proposes a profusion of models to explain cancer immune resistance to CIT that largely outnumber the immune landscapes and corresponding resistance mechanisms. In spite of the complex and contradicting models suggested to explain refractoriness to CIT, the identification of prevailing mechanisms and their targeting may not be as daunting as it at first appears. Here, we suggest that cancer cells go through a conserved evolutionary bottleneck facing a Two-Option Choice to evade recognition by the immune competent host: they can either adopt a clean oncogenic process devoid of immunogenic stimuli (immune-silent tumors) or display an entropic biology prone to immune recognition (immune-active tumors) but resilient to rejection thanks to the recruitment of compensatory immune suppressive processes. Strategies aimed at enhancing the effectiveness of CIT will be different according to the immune landscape targeted.
Collapse
|
25
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
26
|
Chan WC, Knowlton GS, Bishop AC. Activation of Engineered Protein Tyrosine Phosphatases with the Biarsenical Compound AsCy3-EDT 2. Chembiochem 2017; 18:1950-1958. [PMID: 28745017 PMCID: PMC5923034 DOI: 10.1002/cbic.201700253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Indexed: 12/22/2022]
Abstract
Methods for activating signaling enzymes hold significant potential for the study of cellular signal transduction. Here we present a strategy for engineering chemically activatable protein tyrosine phosphatases (actPTPs). To generate actPTP1B, we introduced three cysteine point mutations in the enzyme's WPD loop. Biarsenical compounds were screened for the capability to bind actPTP1B's WPD loop and increase its phosphatase activity. We identified AsCy3-EDT2 as a robust activator that selectively targets actPTP1B in proteomic mixtures and intact cells. Introduction of the corresponding mutations in T-cell PTP also generates an enzyme (actTCPTP) that is strongly activated by AsCy3-EDT2 . Given the conservation of WPD-loop structure among the classical PTPs, our results potentially provide the groundwork of a widely generalizable approach for generating actPTPs as tools for elucidating PTP signaling roles as well as connections between dysregulated PTP activity and human disease.
Collapse
Affiliation(s)
- Wai Cheung Chan
- Amherst College, Department of Chemistry, Amherst, Massachusetts 01002
| | | | - Anthony C. Bishop
- Amherst College, Department of Chemistry, Amherst, Massachusetts 01002
| |
Collapse
|
27
|
Clawson GA, Matters GL, Xin P, McGovern C, Wafula E, dePamphilis C, Meckley M, Wong J, Stewart L, D’Jamoos C, Altman N, Imamura Kawasawa Y, Du Z, Honaas L, Abraham T. "Stealth dissemination" of macrophage-tumor cell fusions cultured from blood of patients with pancreatic ductal adenocarcinoma. PLoS One 2017; 12:e0184451. [PMID: 28957348 PMCID: PMC5619717 DOI: 10.1371/journal.pone.0184451] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022] Open
Abstract
Here we describe isolation and characterization of macrophage-tumor cell fusions (MTFs) from the blood of pancreatic ductal adenocarcinoma (PDAC) patients. The MTFs were generally aneuploidy, and immunophenotypic characterizations showed that the MTFs express markers characteristic of PDAC and stem cells, as well as M2-polarized macrophages. Single cell RNASeq analyses showed that the MTFs express many transcripts implicated in cancer progression, LINE1 retrotransposons, and very high levels of several long non-coding transcripts involved in metastasis (such as MALAT1). When cultured MTFs were transplanted orthotopically into mouse pancreas, they grew as obvious well-differentiated islands of cells, but they also disseminated widely throughout multiple tissues in "stealth" fashion. They were found distributed throughout multiple organs at 4, 8, or 12 weeks after transplantation (including liver, spleen, lung), occurring as single cells or small groups of cells, without formation of obvious tumors or any apparent progression over the 4 to 12 week period. We suggest that MTFs form continually during PDAC development, and that they disseminate early in cancer progression, forming "niches" at distant sites for subsequent colonization by metastasis-initiating cells.
Collapse
Affiliation(s)
- Gary A. Clawson
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Gail L. Matters
- Department of Biochemistry & Molecular Biology, HMC, PSU, Hershey, PA, United States of America
| | - Ping Xin
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Christopher McGovern
- Department of Biochemistry & Molecular Biology, HMC, PSU, Hershey, PA, United States of America
| | - Eric Wafula
- Department of Biology, Eberly College, University Park (UP), Pennsylvania State University, University Park, PA, United States of America
| | - Claude dePamphilis
- Department of Biology, Eberly College, University Park (UP), Pennsylvania State University, University Park, PA, United States of America
| | - Morgan Meckley
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Joyce Wong
- Department of Surgery, HMC, PSU, Hershey, PA, United States of America
| | - Luke Stewart
- Applications Support, Fluidigm Corporation, South San Francisco, CA, United States of America
| | - Christopher D’Jamoos
- Applications Support, Fluidigm Corporation, South San Francisco, CA, United States of America
| | - Naomi Altman
- Department of Statistics, Eberly College, UP, PSU, University Park, PA, United States of America
| | - Yuka Imamura Kawasawa
- Department of Pharmacology and Biochemistry & Molecular Biology, Institute for Personalized Medicine, HMC, PSU, Hershey, PA, United States of America
| | - Zhen Du
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Loren Honaas
- Department of Biology, Eberly College, University Park (UP), Pennsylvania State University, University Park, PA, United States of America
| | - Thomas Abraham
- Department of Neural & Behavioral Sciences and Microscopy Imaging Facility, HMC, PSU, Hershey, PA, United States of America
| |
Collapse
|
28
|
Targeted disruption of TC-PTP in the proliferative compartment augments STAT3 and AKT signaling and skin tumor development. Sci Rep 2017; 7:45077. [PMID: 28322331 PMCID: PMC5359614 DOI: 10.1038/srep45077] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/17/2017] [Indexed: 12/30/2022] Open
Abstract
Tyrosine phosphorylation is a vital mechanism that contributes to skin carcinogenesis. It is regulated by the counter-activities of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Here, we report the critical role of T-cell protein tyrosine phosphatase (TC-PTP), encoded by Ptpn2, in chemically-induced skin carcinogenesis via the negative regulation of STAT3 and AKT signaling. Using epidermal specific TC-PTP knockout (K14Cre.Ptpn2fl/fl) mice, we demonstrate loss of TC-PTP led to a desensitization to tumor initiator 7,12-dimethylbenz[a]anthracene (DMBA)-induced apoptosis both in vivo epidermis and in vitro keratinocytes. TC-PTP deficiency also resulted in a significant increase in epidermal thickness and hyperproliferation following exposure to the tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA). Western blot analysis showed that both phosphorylated STAT3 and phosphorylated AKT expressions were significantly increased in epidermis of TC-PTP-deficient mice compared to control mice following TPA treatment. Inhibition of STAT3 or AKT reversed the effects of TC-PTP deficiency on apoptosis and proliferation. Finally, TC-PTP knockout mice showed a shortened latency of tumorigenesis and significantly increased numbers of tumors during two-stage skin carcinogenesis. Our findings reveal that TC-PTP has potential as a novel target for the prevention of skin cancer through its role in the regulation of STAT3 and AKT signaling.
Collapse
|
29
|
Wang Y, Lin G. TP53INP1 3'-UTR functions as a ceRNA in repressing the metastasis of glioma cells by regulating miRNA activity. Biotechnol Lett 2016; 38:1699-707. [PMID: 27341836 DOI: 10.1007/s10529-016-2159-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/15/2016] [Indexed: 12/30/2022]
Abstract
OBJECTIVES To explore the effects of the competitive endogenous RNA (ceRNA) network between TP53INP1 and E-cadherin on the invasion and migration of glioma. RESULTS TP53INP1 and E-cadherin mRNA and protein were significantly overexpressed in normal brain tissues compared with glioma tissue specimens and correlated with the grades of glioma negatively. The expression of TP53INP1 and E-cadherin were correlated positively. Patients with higher TP53INP1 or E-cadherin expression had longer overall survival. Moreover, TP53INP1 3'-UTR inhibited glioma cell proliferation, invasion and proliferation; Furthermore, the 3'-UTRs of TP53INP1 and E-cadherin harboured seven identical miRNAs binding sites, and TP53INP1 3'-UTR could increase the expression of E-cadherin and decrease the expression of vimentin thus repressing the epithelial-mesenchymal transition (EMT). However, the coding sequence of TP53INP1 could not increase the expression of E-cadherin and the inhibitory effect on EMT of TP53INP1 3'-UTR was reversed by the siRNA against Dicer. CONCLUSIONS TP53INP1 3'-UTR could inhibit the EMT, thus hindering the migration and invasion of glioma via acting as a ceRNA for E-cadherin.
Collapse
Affiliation(s)
- Yi Wang
- Department of Neurosurgery, The Cangzhou Central Hospital, Cangzhou, 061001, China.
| | - Guihua Lin
- Department of Blood Transfusion, The Affiliated Ningde Municipal Hospital, Fujian Medical University, Ningde, 352000, China
| |
Collapse
|
30
|
Inoue K, Fry EA. Aberrant Splicing of Estrogen Receptor, HER2, and CD44 Genes in Breast Cancer. GENETICS & EPIGENETICS 2015; 7:19-32. [PMID: 26692764 PMCID: PMC4669075 DOI: 10.4137/geg.s35500] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/01/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022]
Abstract
Breast cancer (BC) is the most common cause of cancer-related death among women under the age of 50 years. Established biomarkers, such as hormone receptors (estrogen receptor [ER]/progesterone receptor) and human epidermal growth factor receptor 2 (HER2), play significant roles in the selection of patients for endocrine and trastuzumab therapies. However, the initial treatment response is often followed by tumor relapse with intrinsic resistance to the first-line therapy, so it has been expected to identify novel molecular markers to improve the survival and quality of life of patients. Alternative splicing of pre-messenger RNAs is a ubiquitous and flexible mechanism for the control of gene expression in mammalian cells. It provides cells with the opportunity to create protein isoforms with different, even opposing, functions from a single genomic locus. Aberrant alternative splicing is very common in cancer where emerging tumor cells take advantage of this flexibility to produce proteins that promote cell growth and survival. While a number of splicing alterations have been reported in human cancers, we focus on aberrant splicing of ER, HER2, and CD44 genes from the viewpoint of BC development. ERα36, a splice variant from the ER1 locus, governs nongenomic membrane signaling pathways triggered by estrogen and confers 4-hydroxytamoxifen resistance in BC therapy. The alternative spliced isoform of HER2 lacking exon 20 (Δ16HER2) has been reported in human BC; this isoform is associated with transforming ability than the wild-type HER2 and recapitulates the phenotypes of endocrine therapy-resistant BC. Although both CD44 splice isoforms (CD44s, CD44v) play essential roles in BC development, CD44v is more associated with those with favorable prognosis, such as luminal A subtype, while CD44s is linked to those with poor prognosis, such as HER2 or basal cell subtypes that are often metastatic. Hence, the detection of splice variants from these loci will provide keys to understand the pathogenesis, predict the prognosis, and choose specific therapies for BC.
Collapse
Affiliation(s)
- Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Elizabeth A. Fry
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| |
Collapse
|