1
|
Kallenberger EM, Khandelwal A, Nath P, Nguyen SA, DiGiovanni J, Nathan CA. FGFR2 in the Development and Progression of Cutaneous Squamous Cell Cancer. Mol Carcinog 2025; 64:5-13. [PMID: 39466044 DOI: 10.1002/mc.23835] [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: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is an increasingly common malignancy of the skin and the leading cause of death from skin cancer in adults over the age of 85. Fibroblast growth factor receptor 2 (FGFR2) has been identified as an important effector of signaling pathways that lead to the growth and development of cSCC. In recent years, there have been numerous studies evaluating the role FGFR2 plays in multiple cancers, its contribution to resistance to anticancer therapy, and new drugs that may be used to inhibit FGFR2. This review will provide an overview of our current understanding of FGFR2 and potential mechanisms in which we can target FGFR2 in cSCC. The goals of this review are the following: (1) to highlight our current knowledge of the role of FGFR2 in healthy skin and contrast this with its role in the development of cancer; (2) to further explain the specific molecular mechanisms that FGFR2 uses to promote tumorigenesis; (3) to describe how FGFR2 contributes to more invasive disease; (4) to describe its immunosuppressive effects in skin; and (5) to evaluate its effect on current anticancer therapy and discuss therapies on the horizon to target FGFR2 related malignancy.
Collapse
Affiliation(s)
- Ethan M Kallenberger
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alok Khandelwal
- Department of Oto/HNS, Health Sciences Center, Louisiana State University, Shreveport, Louisiana, USA
| | - Priyatosh Nath
- Department of Oto/HNS, Health Sciences Center, Louisiana State University, Shreveport, Louisiana, USA
| | - Shaun A Nguyen
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - John DiGiovanni
- Department of Pharmacology, University of Texas, Austin, Texas, USA
| | - Cherie-Ann Nathan
- Department of Oto/HNS, Health Sciences Center, Louisiana State University, Shreveport, Louisiana, USA
| |
Collapse
|
2
|
Cvammen W, Kemp MG. The REV-ERB antagonist SR8278 modulates keratinocyte viability in response to UVA and UVB radiation. Photochem Photobiol 2024; 100:1864-1873. [PMID: 38459721 DOI: 10.1111/php.13930] [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: 12/03/2023] [Revised: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/10/2024]
Abstract
The nucleotide excision repair (NER) system removes UV photoproducts from genomic DNA and is controlled by the circadian clock. Given that small-molecule compounds have been developed to target various clock proteins, we examined whether the cryptochrome inhibitor KS15 and REV-ERB antagonist SR8278 could modulate keratinocyte responses to UV radiation in vitro. We observed that though SR8278 promoted cell viability in UVB-irradiated cells, it had little effect on NER or on the expression of the clock-regulated NER factor XPA. Rather, we found that both KS15 and SR8278 absorb light within the UV spectrum to limit initial UV photoproduct formation in DNA. Moreover, SR8278 promoted UVB viability even in cells in which the core circadian clock protein BMAL1 was disrupted, which indicates that SR8278 is likely acting via other REV-ERB transcriptional targets. We further observed that SR8278 sensitized keratinocytes to light sources containing primarily UVA wavelengths of light likely due to the generation of toxic reactive oxygen species. Though other studies have demonstrated beneficial effects of SR8278 in other model systems, our results here suggest that SR8278 has limited utility for UV photoprotection in the skin and will likely cause phototoxicity in humans or mammals exposed to solar radiation.
Collapse
Affiliation(s)
- William Cvammen
- Department of Pharmacology and Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
| | - Michael G Kemp
- Department of Pharmacology and Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
- Dayton Veterans Administration Medical Center, Dayton, Ohio, USA
| |
Collapse
|
3
|
Shafi H, Lora AJ, Donow HM, Dickinson SE, Wondrak GT, Chow HHS, Curiel-Lewandrowski C, Mansour HM. Comprehensive Advanced Physicochemical Characterization and In Vitro Human Cell Culture Assessment of BMS-202: A Novel Inhibitor of Programmed Cell Death Ligand. Pharmaceutics 2024; 16:1409. [PMID: 39598533 PMCID: PMC11597381 DOI: 10.3390/pharmaceutics16111409] [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: 09/24/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: BMS-202, is a potent small molecule with demonstrated antitumor activity. The study aimed to comprehensively characterize the physical and chemical properties of BMS-202 and evaluate its suitability for topical formulation, focusing on uniformity, stability and safety profiles. Methods: A range of analytical techniques were employed to characterize BMS-202. Scanning Electron Microscopy (SEM) was used to assess morphology, Differential Scanning Calorimetry (DSC) provided insights of thermal behavior, and Hot-Stage Microscopy (HSM) corroborated these thermal behaviors. Molecular fingerprinting was conducted using Raman spectroscopy and Fourier Transform Infrared (FTIR) spectroscopy, with chemical uniformity of the batch further validated by mapping through FTIR and Raman microscopies. The residual water content was measured using Karl Fisher Coulometric titration, and vapor sorption isotherms examined moisture uptake across varying relative humidity levels. In vitro safety assessments involved testing with skin epithelial cell lines, such as HaCaT and NHEK, and Transepithelial Electrical Resistance (TEER) to evaluate barrier integrity. Results: SEM revealed a distinctive needle-like morphology, while DSC indicated a sharp melting point at 110.90 ± 0.54 ℃ with a high enthalpy of 84.41 ± 0.38 J/g. HSM confirmed the crystalline-to-amorphous transition at the melting point. Raman and FTIR spectroscopy, alongside chemical imaging, confirmed chemical uniformity as well as validated the batch consistency. A residual water content of 2.76 ± 1.37 % (w/w) and minimal moisture uptake across relative humidity levels demonstrated its low hygroscopicity and suitability for topical formulations. Cytotoxicity testing showed dose-dependent reduction in skin epithelial cell viability at high concentrations (100 µM and 500 µM), with lower doses (0.1 µM to 10 µM) demonstrating acceptable safety. TEER studies indicated that BMS-202 does not disrupt the HaCaT cell barrier function. Conclusions: The findings from this study establish that BMS-202 has promising physicochemical and in vitro characteristics at therapeutic concentrations for topical applications, providing a foundation for future formulation development focused on skin-related cancers or localized immune modulation.
Collapse
Affiliation(s)
- Hasham Shafi
- Florida International University Center for Translational Science, Port St. Lucie, FL 34987, USA
| | - Andrea J. Lora
- Florida International University Center for Translational Science, Port St. Lucie, FL 34987, USA
| | - Haley M. Donow
- Florida International University Center for Translational Science, Port St. Lucie, FL 34987, USA
| | - Sally E. Dickinson
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA (G.T.W.)
- Department of Pharmacology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Georg T. Wondrak
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA (G.T.W.)
- Department of Pharmacology and Toxicology, The University of Arizona College of Pharmacy, Tucson, AZ 85721, USA
| | - H.-H. Sherry Chow
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA (G.T.W.)
- Division of Hematology and Oncology, Department of Medicine, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Clara Curiel-Lewandrowski
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA (G.T.W.)
- Division of Dermatology, Department of Medicine, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
- BIO5 Institute, University of Arizona, Tucson, AZ 85724, USA
| | - Heidi M. Mansour
- Florida International University Center for Translational Science, Port St. Lucie, FL 34987, USA
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33174, USA
- Department of Biomedical Engineering, College of Engineering and Computing, Florida International University, Miami, FL 33174, USA
| |
Collapse
|
4
|
Salminen A. The role of the immunosuppressive PD-1/PD-L1 checkpoint pathway in the aging process and age-related diseases. J Mol Med (Berl) 2024; 102:733-750. [PMID: 38600305 PMCID: PMC11106179 DOI: 10.1007/s00109-024-02444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024]
Abstract
The accumulation of senescent cells within tissues is a hallmark of the aging process. Senescent cells are also commonly present in many age-related diseases and in the cancer microenvironment. The escape of abnormal cells from immune surveillance indicates that there is some defect in the function of cytotoxic immune cells, e.g., CD8+ T cells and natural killer (NK) cells. Recent studies have revealed that the expression of programmed death-ligand 1 (PD-L1) protein is abundantly increased in senescent cells. An increase in the amount of PD-L1 protein protects senescent cells from clearance by the PD-1 checkpoint receptor in cytotoxic immune cells. In fact, the activation of the PD-1 receptor suppresses the cytotoxic properties of CD8+ T and NK cells, promoting a state of immunosenescence. The inhibitory PD-1/PD-L1 checkpoint pathway acts in cooperation with immunosuppressive cells; for example, activation of PD-1 receptor can enhance the differentiation of regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC), and M2 macrophages, whereas the cytokines secreted by immunosuppressive cells stimulate the expression of the immunosuppressive PD-L1 protein. Interestingly, many signaling pathways known to promote cellular senescence and the aging process are crucial stimulators of the expression of PD-L1 protein, e.g., epigenetic regulation, inflammatory mediators, mTOR-related signaling, cGAS-STING pathway, and AhR signaling. It seems that the inhibitory PD-1/PD-L1 immune checkpoint axis has a crucial role in the accumulation of senescent cells and thus it promotes the aging process in tissues. Thus, the blockade of the PD-1/PD-L1 checkpoint signaling might be a potential anti-aging senolytic therapy. KEY MESSAGES: Senescent cells accumulate within tissues during aging and age-related diseases. Senescent cells are able to escape immune surveillance by cytotoxic immune cells. Expression of programmed death-ligand 1 (PD-L1) markedly increases in senescent cells. Age-related signaling stimulates the expression of PD-L1 protein in senescent cells. Inhibitory PD-1/PD-L1 checkpoint pathway suppresses clearance of senescent cells.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
5
|
Wu G, Deng W, Chen HY, Cho HJ, Kim J. Galectin 7 leads to a relative reduction in CD4+ T cells, mediated by PD-1. Sci Rep 2024; 14:6625. [PMID: 38503797 PMCID: PMC10951237 DOI: 10.1038/s41598-024-57162-3] [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: 12/14/2023] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
The role of glycan-binding proteins as an activator of immune regulatory receptors has gained attention recently. We report that galectin 7 reduced CD4+ T cell percentage in both in vitro culture and mouse tumor models. Immunohistochemical staining of esophageal cancer patient samples showed a lower percentage of CD4+ cells in the galectin 7 high area. The lack of CD4+ T cell depletion by galectin 7 in PD-1 knockout mice supports the role of PD-1 in mediating the effects of galectin 7. The binding assays demonstrate that galectin 7 binds to the N-glycosylation of PD-1 on N74 and N116 sites and leads to the recruitment of SHP-2. NFAT suppressive activity of galectin 7 was abrogated upon overexpression of the dominant negative SHP-2 mutant or inhibition of PD-1 by siRNA. Glycosylation of PD-1 has been reported to play a critical role in surface expression, stability, and interaction with its ligand PD-L1. This report further expands the significance of PD-1 glycosylation and suggests that galectin 7, a glycan-binding protein, interacts with the immune regulatory receptor PD-1 through glycosylation recognition.
Collapse
Affiliation(s)
- Guojin Wu
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9072, USA
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hsin-Yi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9072, USA
| | - Hye-Jeong Cho
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9072, USA
| | - Jaehyup Kim
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9072, USA.
| |
Collapse
|
6
|
Truong K, Chamberlin CV, Kim J, Carlino MS, Araujo RR. Immune checkpoint inhibitor-induced lichenoid drug eruption-sparing scar burns. Australas J Dermatol 2024; 65:163-166. [PMID: 38009870 DOI: 10.1111/ajd.14192] [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: 08/05/2023] [Revised: 10/31/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
The authors present a striking case of a patient experiencing a lichenoid drug eruption secondary to immunotherapy, curiously sparing scarred skin from past burns. We observed vastly higher amounts of inflammatory lymphoid cells staining for PD-1; 70% in skin with a lichenoid drug reaction and 50% in scarred skin. The lack of a lichenoid reaction at sites of scarred skin may indicate that a basement membrane component may be causative for a lichenoid drug eruption.
Collapse
Affiliation(s)
- Kelvin Truong
- Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | | | - Jennifer Kim
- Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research (ICPMR) Westmead Hospital, NSW Health Pathology, Westmead, New South Wales, Australia
| | - Matteo S Carlino
- Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, New South Wales, Australia
- Melanoma Institute Australia, Sydney, New South Wales, Australia
| | - Raquel Ruiz Araujo
- Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
7
|
Dickinson SE, Vaishampayan P, Jandova J, Ai Y(E, Kirschnerova V, Zhang T, Calvert V, Petricoin E, Chow HHS, Hu C, Roe D, Bode A, Curiel-Lewandrowski C, Wondrak GT. Inhibition of UV-Induced Stress Signaling and Inflammatory Responses in SKH-1 Mouse Skin by Topical Small-Molecule PD-L1 Blockade. JID INNOVATIONS 2024; 4:100255. [PMID: 38328594 PMCID: PMC10847774 DOI: 10.1016/j.xjidi.2023.100255] [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] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/24/2023] [Accepted: 12/05/2023] [Indexed: 02/09/2024] Open
Abstract
The immune checkpoint ligand PD-L1 has emerged as a molecular target for skin cancer therapy and might also hold promise for preventive intervention targeting solar UV light-induced skin damage. In this study, we have explored the role of PD-L1 in acute keratinocytic photodamage testing the effects of small-molecule pharmacological inhibition. Epidermal PD-L1 upregulation in response to chronic photodamage was established using immunohistochemical and proteomic analyses of a human skin cohort, consistent with earlier observations that PD-L1 is upregulated in cutaneous squamous cell carcinoma. Topical application of the small-molecule PD-L1 inhibitor BMS-202 significantly attenuated UV-induced activator protein-1 transcriptional activity in SKH-1 bioluminescent reporter mouse skin, also confirmed in human HaCaT reporter keratinocytes. RT-qPCR analysis revealed that BMS-202 antagonized UV induction of inflammatory gene expression. Likewise, UV-induced cleavage of procaspase-3, a hallmark of acute skin photodamage, was attenuated by topical BMS-202. NanoString nCounter transcriptomic analysis confirmed downregulation of cutaneous innate immunity- and inflammation-related responses, together with upregulation of immune response pathway gene expression. Further mechanistic analysis confirmed that BMS-202 antagonizes UV-induced PD-L1 expression both at the mRNA and protein levels in SKH-1 epidermis. These data suggest that topical pharmacological PD-L1 antagonism using BMS-202 shows promise for skin protection against photodamage.
Collapse
Affiliation(s)
- Sally E. Dickinson
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology, College of Medicine Tucson, The University of Arizona, Tucson, Arizona, USA
- Skin Cancer Institute, University of Arizona, Tucson, Arizona, USA
| | - Prajakta Vaishampayan
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
| | - Jana Jandova
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, USA
| | - Yuchen (Ella) Ai
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
| | - Viktoria Kirschnerova
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
| | - Tianshun Zhang
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Valerie Calvert
- Center for Applied Proteomics and Molecular Medicine, College of Medicine, George Mason University, Fairfax, Virginia, USA
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine, College of Medicine, George Mason University, Fairfax, Virginia, USA
| | - H-H. Sherry Chow
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Department of Molecular & Cellular Biology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Chengcheng Hu
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Department of Epidemiology and Biostatistics, Mel and Enid Zukerman College of Public Health, The University of Arizona, Tucson, Arizona, USA
| | - Denise Roe
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Department of Epidemiology and Biostatistics, Mel and Enid Zukerman College of Public Health, The University of Arizona, Tucson, Arizona, USA
| | - Ann Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Clara Curiel-Lewandrowski
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Skin Cancer Institute, University of Arizona, Tucson, Arizona, USA
- Division of Dermatology, Department of Medicine, College of Medicine Tucson, The University of Arizona, Tucson, Arizona, USA
| | - Georg T. Wondrak
- The University of Arizona Cancer Center, The University of Arizona, Tucson, Arizona, USA
- Skin Cancer Institute, University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
8
|
Romashin D, Rusanov A, Arzumanian V, Varshaver A, Poverennaya E, Vakhrushev I, Netrusov A, Luzgina N. Exploring the Functions of Mutant p53 through TP53 Knockout in HaCaT Keratinocytes. Curr Issues Mol Biol 2024; 46:1451-1466. [PMID: 38392212 PMCID: PMC10887868 DOI: 10.3390/cimb46020094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Approximately 50% of tumors carry mutations in TP53; thus, evaluation of the features of mutant p53 is crucial to understanding the mechanisms underlying cell transformation and tumor progression. HaCaT keratinocytes represent a valuable model for research in this area since they are considered normal, although they bear two gain-of-function mutations in TP53. In the present study, transcriptomic and proteomic profiling were employed to examine the functions of mutant p53 and to investigate the impact of its complete abolishment. Our findings indicate that CRISPR-mediated TP53 knockout results in significant changes at the transcriptomic and proteomic levels. The knockout of TP53 significantly increased the migration rate and altered the expression of genes associated with invasion, migration, and EMT but suppressed the epidermal differentiation program. These outcomes suggest that, despite being dysfunctional, p53 may still possess oncosuppressive functions. However, despite being considered normal keratinocytes, HaCaT cells exhibit oncogenic properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alexander Netrusov
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- Faculty of Biology and Biotechnology, HSE University, Moscow 101000, Russia
| | | |
Collapse
|
9
|
Han Z, Wang N, Qiao Q, He X, Wang N. Association of PD-L1 Expression with Clinicopathologic Characters in Gastric Cancer: A Comprehensive Meta-analysis. Curr Med Chem 2024; 31:3198-3216. [PMID: 37921182 DOI: 10.2174/0109298673263784230922060257] [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: 05/24/2023] [Revised: 07/18/2023] [Accepted: 08/16/2023] [Indexed: 11/04/2023]
Abstract
PURPOSE The expression level of programmed death ligand-1(PD-L1) in patients with gastric cancer is the key to determining the use of immune drugs. The relationship between PD-L1 expression level and clinical characteristics is worth exploring. METHODS By setting the search terms correlated to PD-L1 and gastric cancer, a nearly comprehensive search was carried out in four major databases, and the deadline for searching was September 1, 2022. The retrieved documents were further screened by strict inclusion and exclusion criteria after removing the duplication. Next, the quality of the included studies was evaluated with the Newcastle-Ottawa Scale (NOS) scale. Finally, the STATA15.1 software was used to process data and draw plots, and the odds ratios (ORs) were adopted to assess the pooled effect size. RESULTS A total of 85 works of literature were included in this study through screening strictly, and detailed data were extracted after evaluating the quality of the literature. The process of analysis was conducted in the whole population, Asia-Africa population, European and American population, and Asian population with CPS≥1, amd all found that the expression of PD-L1 in gastric cancer was correlated with age, tumor size, EBV infection, Her-2 expression and microsatellite status. However, the subgroup of the region also found some differences in Asian and Western regions, which was interesting and worth studying further. The included research of this study did not have significant publish bias. CONCLUSION After careful analysis, this study found that age (>60 years), tumor size (>5cm), EBV infection (+), Her-2 expression (+), microsatellite status (MSI), and mismatch repair status (dMMR) were risk factors for positive expression of PD-L1 in gastric cancer.
Collapse
Affiliation(s)
- Zhuo Han
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Nan Wang
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Nan Wang
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| |
Collapse
|
10
|
Vaishampayan P, Curiel-Lewandrowski C, Dickinson SE. Review: PD-L1 as an emerging target in the treatment and prevention of keratinocytic skin cancer. Mol Carcinog 2023; 62:52-61. [PMID: 36121318 PMCID: PMC9771956 DOI: 10.1002/mc.23464] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 02/03/2023]
Abstract
Recent advances in the understanding and targeting of immune checkpoints have led to great progress in immune therapies against many forms of cancer. While many types of immune checkpoints are currently targeted in the clinic, this review will focus on recent research implicating the programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) axis as an emerging focus for the treatment of keratinocytic tumors. PD-L1 is of particular interest in nonmelanoma skin cancer (NMSC), as it is not only upregulated in these tumors but is stimulated by environmental ultraviolet exposure. This response may also make PD-L1 an excellent target for photochemoprevention using topically applied small molecule inhibitors. Here, we summarize recent investigations on PD-L1 expression and clinically relevant immune checkpoint inhibitor treatment in cutaneous squamous cell carcinoma, basal cell carcinoma, and head and neck squamous cell carcinoma, as well as small molecule agents targeting PD-L1 that may be useful for clinical development aiming at treatment or prevention of NMSC.
Collapse
Affiliation(s)
| | - Clara Curiel-Lewandrowski
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
- Division of Dermatology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Sally E Dickinson
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| |
Collapse
|
11
|
Cvammen W, Kemp MG. Flavonoid Nobiletin Exhibits Differential Effects on Cell Viability in Keratinocytes Exposed to UVA versus UVB Radiation. Photochem Photobiol 2022; 98:1372-1378. [PMID: 35348223 PMCID: PMC9790495 DOI: 10.1111/php.13625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
The polymethoxylated flavonoid nobiletin has been shown to suppress inflammatory responses to UVB radiation and to enhance circadian rhythms. Because expression of the core nucleotide excision repair (NER) factor XPA and the rate of removal of UV photoproducts from DNA are regulated by the circadian clock, we investigated whether the beneficial effects of nobiletin in UVB-exposed cells could be due in part to enhanced NER. Although nobiletin limited UVB-irradiated human keratinocytes from undergoing cell death, we found that this enhanced survival was not associated with increased NER or XPA expression. Instead, nobiletin reduced initial UV photoproduct formation and promoted a G1 cell cycle arrest. We then examined the implications of this findings for exposures to solar radiation through use of a solar simulated light (SSL) source that contains primarily UVA radiation. In striking contrast to the results obtained with UVB radiation, nobiletin instead sensitized keratinocytes to both the SSL and a more defined UVA radiation source. This enhanced cell death was correlated with a photochemical change in nobiletin absorption spectrum and the production of reactive oxygen species. We conclude that nobiletin is unlikely to be a useful compound for protecting keratinocytes against the harmful effects of solar UV radiation.
Collapse
Affiliation(s)
- William Cvammen
- Department of Pharmacology and ToxicologyWright State University Boonshoft School of MedicineDaytonOH
| | - Michael G. Kemp
- Department of Pharmacology and ToxicologyWright State University Boonshoft School of MedicineDaytonOH,Dayton Veterans Administration Medical CenterDaytonOH
| |
Collapse
|
12
|
Liotta LA, Pappalardo PA, Carpino A, Haymond A, Howard M, Espina V, Wulfkuhle J, Petricoin E. Laser Capture Proteomics: spatial tissue molecular profiling from the bench to personalized medicine. Expert Rev Proteomics 2021; 18:845-861. [PMID: 34607525 PMCID: PMC10720974 DOI: 10.1080/14789450.2021.1984886] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Laser Capture Microdissection (LCM) uses a laser to isolate, or capture, specific cells of interest in a complex heterogeneous tissue section, under direct microscopic visualization. Recently, there has been a surge of publications using LCM for tissue spatial molecular profiling relevant to a wide range of research topics. AREAS COVERED We summarize the many advances in tissue Laser Capture Proteomics (LCP) using mass spectrometry for discovery, and protein arrays for signal pathway network mapping. This review emphasizes: a) transition of LCM phosphoproteomics from the lab to the clinic for individualized cancer therapy, and b) the emerging frontier of LCM single cell molecular analysis combining proteomics with genomic, and transcriptomic analysis. The search strategy was based on the combination of MeSH terms with expert refinement. EXPERT OPINION LCM is complemented by a rich set of instruments, methodology protocols, and analytical A.I. (artificial intelligence) software for basic and translational research. Resolution is advancing to the tissue single cell level. A vision for the future evolution of LCM is presented. Emerging LCM technology is combining digital and AI guided remote imaging with automation, and telepathology, to a achieve multi-omic profiling that was not previously possible.
Collapse
Affiliation(s)
- Lance A. Liotta
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Philip A. Pappalardo
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Alan Carpino
- Fluidigm Corporation, South San Francisco, CA, USA
| | - Amanda Haymond
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Marissa Howard
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Virginia Espina
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Julie Wulfkuhle
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Emanuel Petricoin
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| |
Collapse
|