1
|
Choi YJ, Yang MK, Kim N, Khwarg SI, Choung H, Kim JE. Expression of nuclear receptors and glucose metabolic pathway proteins in sebaceous carcinoma: Androgen receptor and monocarboxylate transporter 1 have a key role in disease progression. Oncol Lett 2024; 28:593. [PMID: 39421321 PMCID: PMC11484244 DOI: 10.3892/ol.2024.14726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Standard systemic treatments are not consistently effective for treating unresectable or advanced sebaceous carcinoma (SC). The present study investigated the pathogenic roles of nuclear receptors (NRs), glucose metabolic dysregulation and immune checkpoint proteins in SC as prognostic markers or therapeutic targets. Patients with pathologically confirmed SC between January 2002 and December 2019 at three university hospitals in South Korea were included in the present study. Immunohistochemistry was performed on paraffin-embedded tumor tissues for glucocorticoid receptors (GR), androgen receptors (AR), estrogen receptors (ER), progesterone receptors (PR), glucose transporter 1 (GLUT1), monocarboxylate transporters (MCT1 and MCT4), CD147, phosphorylated adenosine monophosphate-activated protein kinase (pAMPK) and the immune checkpoint protein, programmed cell death-ligand 1 (PD-L1). The results were semi-quantitatively assessed and the associations of these proteins with various clinicopathological parameters were determined. A total of 39 cases of SC comprising 19 periocular and 20 extraocular tumors were enrolled. NRs were frequently detected in the tumor nuclei, with GR having the highest frequency (89.7%), followed by AR, ER (both 51.3%) and PR (41.0%). Regarding glucose metabolism, CD147, GLUT1 and MCT1 were highly expressed at 100, 89.7 and 87.2%, respectively, whereas MCT4 and pAMPK expression levels were relatively low at 38.5 and 35.9%, respectively. Membranous expression of PD-L1 was detected in five cases (12.8%), four of which were extraocular. In the multivariate analysis, advanced stage, low AR positivity and high MCT1 expression were independent poor prognostic factors for metastasis-free survival (all P<0.05). The present results suggested that hormonal and metabolic dysregulation may be associated with the pathogenesis of SC, and that AR and MCT1 in particular may serve as prognostic indicators and potential therapeutic targets. Additionally, ~10% of SC cases exhibited PD-L1 expression within the druggable range, and these patients are expected to benefit from treatment with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Youn Joo Choi
- Department of Ophthalmology, Kangdong Sacred Heart Hospital, Hallym University Medical Center, Seoul 05355, Republic of Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Min Kyu Yang
- Department of Ophthalmology, Asan Medical Center, Ulsan University College of Medicine, Seoul 05505, Republic of Korea
| | - Namju Kim
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Sang In Khwarg
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hokyung Choung
- Department of Ophthalmology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Republic of Korea
| | - Ji Eun Kim
- Department of Pathology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Republic of Korea
| |
Collapse
|
2
|
Bhujbal SP, Hah JM. An Innovative Approach to Address Neurodegenerative Diseases through Kinase-Targeted Therapies: Potential for Designing Covalent Inhibitors. Pharmaceuticals (Basel) 2023; 16:1295. [PMID: 37765103 PMCID: PMC10537995 DOI: 10.3390/ph16091295] [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: 07/28/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Owing to the dysregulation of protein kinase activity in various diseases such as cancer and autoimmune, cardiovascular, neurodegenerative, and inflammatory conditions, the protein kinase family has emerged as a crucial drug target in the 21st century. Notably, many kinases have been targeted to address cancer and neurodegenerative diseases using conventional ATP-mimicking kinase inhibitors. Likewise, irreversible covalent inhibitors have also been developed for different types of cancer. The application of covalent modification to target proteins has led to significant advancements in the treatment of cancer. However, while covalent drugs have significantly impacted medical treatment, their potential for neurodegenerative diseases remains largely unexplored. Neurodegenerative diseases present significant risks to brain function, leading to progressive deterioration in sensory, motor, and cognitive abilities. Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), and multiple sclerosis (MS) are among the various examples of such disorders. Numerous research groups have already reported insights through reviews and research articles on FDA-approved covalent inhibitors, revealing their mechanisms and the specific covalent warheads that preferentially interact with particular amino acid residues in intricate detail. Hence, in this review, we aim to provide a concise summary of these critical topics. This summary endeavors to guide medicinal chemists in their quest to design covalent inhibitors for protein kinases, specifically targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Swapnil P. Bhujbal
- College of Pharmacy, Hanyang University, Ansan 426-791, Republic of Korea;
| | - Jung-Mi Hah
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Republic of Korea
| |
Collapse
|
3
|
Zhang H, Zhou S, Plewka J, Wu C, Zhu M, Yu Q, Musielak B, Wang X, Awadasseid A, Magiera-Mularz K, Wu Y, Zhang W. Design, Synthesis, and Antitumor Activity Evaluation of 2-Arylmethoxy-4-(2,2'-dihalogen-substituted biphenyl-3-ylmethoxy) Benzylamine Derivatives as Potent PD-1/PD-L1 Inhibitors. J Med Chem 2023; 66:10579-10603. [PMID: 37496104 DOI: 10.1021/acs.jmedchem.3c00731] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Novel 2-arylmethoxy-4-(2,2'-dihalogen-substituted biphenyl-3-ylmethoxy) benzylamine derivatives were designed, synthesized, and evaluated in vitro and in vivo against cancers as PD-1/PD-L1 inhibitors. Through the computer-aided structural optimization and the homogeneous time-resolved fluorescence (HTRF) assay, compound A56 was found to most strongly block the PD-1/PD-L1 interaction with an IC50 value of 2.4 ± 0.8 nM and showed the most potent activity. 1H NMR titration results indicated that A56 can tightly bind to the PD-L1 protein with KD < 1 μM. The X-ray diffraction data for the cocrystal structure of the A56/PD-L1 complex (3.5 Å) deciphered a novel binding mode in detail, which can account for its most potent inhibitory activity. Cell-based assays further demonstrated the strong ability of A56 as an hPD-1/hPD-L1 blocker. Especially in an hPD-L1 MC38 humanized mouse model, A56 significantly inhibited tumor growth without obvious toxicity, with a TGI rate of 55.20% (50 mg/kg, i.g.). In conclusion, A56 is a promising clinical candidate worthy of further development.
Collapse
Affiliation(s)
- Hua Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shijia Zhou
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jacek Plewka
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Caiyun Wu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Mengyu Zhu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qimeng Yu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Bogdan Musielak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Xiao Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Katarzyna Magiera-Mularz
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
4
|
Lin J, Huang N, Li M, Zheng M, Wang Z, Zhang X, Gao H, Lao Y, Zhang J, Ding B. Dendritic Cell-Derived Exosomes Driven Drug Co-Delivery Biomimetic Nanosystem for Effective Combination of Malignant Melanoma Immunotherapy and Gene Therapy. Drug Des Devel Ther 2023; 17:2087-2106. [PMID: 37489176 PMCID: PMC10363389 DOI: 10.2147/dddt.s414758] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/04/2023] [Indexed: 07/26/2023] Open
Abstract
Purpose Malignant melanoma (MM), the most lethal skin cancer, is highly invasive and metastatic. These qualities are related to not only genetic mutations in MM itself but also the interaction of MM cells with the immune system and microenvironment. This study aimed to construct a combined immunotherapy and gene therapy drug delivery system for the effective treatment of MM. Methods Mature dendritic cell (mDC) exosomes (mDexos) with immune induction functions were used as carriers. BRAF siRNA (siBRAF) with the ability to silence mutated BRAF in MM was encapsulated in mDexos by electroporation to construct a biomimetic nanosystem for the codelivery of immunotherapy and gene therapy drugs (siBRAF-mDexos) to the MM microenvironment. Then, we investigated the nanosystem's serum stability and biocompatibility, uptake efficiency in mouse melanoma cells (B16-F10 cells), cytotoxicity against B16-F10 cells and inhibitory effect on BRAF expression. Furthermore, we evaluated its antimelanoma activity and safety in vivo. Results SiBRAF-mDexos were nanosized. Compared to siBRAF, siBRAF-mDexos displayed significantly increased serum stability, biocompatibility, uptake efficiency in B16-F10 cells, and cytotoxicity to B16-F10 melanoma cells; they also had a significantly greater inhibitory effect on BRAF expression and induced T-lymphocyte proliferation. Moreover, compared with siBRAF, siBRAF-mDexos showed significantly enhanced anti-MM activity and a high level of safety in vivo. Conclusion The study suggests that the siBRAF-mDexo biomimetic drug codelivery system can be used to effectively treat MM, which provides a new strategy for combined gene therapy and immunotherapy for MM.
Collapse
Affiliation(s)
- Jiecheng Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 214122, People’s Republic of China
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Na Huang
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Mingjuan Li
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Mengyuan Zheng
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Zhuoxiang Wang
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Xiaojuan Zhang
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Huan Gao
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Yunzhe Lao
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Jie Zhang
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| | - Baoyue Ding
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, 314001, People’s Republic of China
| |
Collapse
|
5
|
Li L, Li J. Dimerization of Transmembrane Proteins in Cancer Immunotherapy. MEMBRANES 2023; 13:393. [PMID: 37103820 PMCID: PMC10143916 DOI: 10.3390/membranes13040393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 06/19/2023]
Abstract
Transmembrane proteins (TMEMs) are integrated membrane proteins that span the entire lipid bilayer and are permanently anchored to it. TMEMs participate in various cellular processes. Some TMEMs usually exist and perform their physiological functions as dimers rather than monomers. TMEM dimerization is associated with various physiological functions, such as the regulation of enzyme activity, signal transduction, and cancer immunotherapy. In this review, we focus on the dimerization of transmembrane proteins in cancer immunotherapy. This review is divided into three parts. First, the structures and functions of several TMEMs related to tumor immunity are introduced. Second, the characteristics and functions of several typical TMEM dimerization processes are analyzed. Finally, the application of the regulation of TMEM dimerization in cancer immunotherapy is introduced.
Collapse
Affiliation(s)
- Lei Li
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jingying Li
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
6
|
PD-L1/PD-1 blockage enhanced the cytotoxicity of natural killer cell on the non-small cell lung cancer (NSCLC) by granzyme B secretion. Clin Transl Oncol 2023:10.1007/s12094-023-03120-w. [PMID: 36856921 DOI: 10.1007/s12094-023-03120-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023]
Abstract
OBJECTIVE To explore the role of PD-L1/PD-1 blockage in the cytotoxicity of natural killer cell in NSCLC. METHODS Two NSCLC cell lines, Calu-1 and H460, were tested for susceptibility to the cytolytic activity of freshly isolated healthy donor NK cells by a non-radioactive cellular cytotoxicity assay kit. Western blot analysis, FACS, ELISA and antibody blockage experiments were conducted to determine the mechanisms. NK cells isolated from NSCLC patients were also collected for functional assays. RESULTS Calu-1 and H460 cells were lysed by NK cells in a dose-dependent manner. H460 cells showed less susceptibility to NK cell-mediated lysis than Calu-1 cells at all ratios. The expression of PD-L1 on H460 cells was higher than that on Calu-1 cells, as determined by FACS and western blot analysis. The specific lysis of H460 cells by NK cells was enhanced when the PD-L1/PD-1 interaction was blocked by anti-PD-L1 antibody. This finding was also demonstrated in NK cells isolated from NSCLC patients. CONCLUSIONS The present study revealed that PD-L1/PD-1 blockage enhanced the cytotoxicity of natural killer cells in NSCLC via granzyme B secretion. This study will greatly facilitate the precise treatment of lung cancer through determination of PD-L1 expression in tumors.
Collapse
|
7
|
DiFrancesco M, Hofer J, Aradhya A, Rufinus J, Stoddart J, Finocchiaro S, Mani J, Tevis S, Visconti M, Walawender G, DiFlumeri J, Fattakhova E, Patil SP. Discovery of small-molecule PD-1/PD-L1 antagonists through combined virtual screening and experimental validation. Comput Biol Chem 2023; 102:107804. [PMID: 36610303 DOI: 10.1016/j.compbiolchem.2022.107804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 11/17/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Inhibition of the interaction between the PD-1 protein on activated lymphocytes and the PD-L1 protein on tumors represents a novel therapeutic approach for selective activation of the innate immune response against a variety of cancers. Therefore, the present study utilized a combined virtual and experimental screening approach to screen databases of both lead-like and larger molecules for identification of novel inhibitors of PD-1/PD-L1 interaction. First, high-throughput virtual screening of ∼3.7 million lead-like molecules using a rigid-receptor docking approach against both human PD-1 and PD-L1 proteins revealed possible small-molecule tractability of PD-1, but not PD-L1, binding interface. The subsequent work, therefore, involved screening of the National Cancer Institute (NCI) compound database against the PD-1 pocket. Several NCI compounds were identified with potential to bind to the PD-1 pocket and in turn inhibit the PD-1/PD-L1 interaction. The dynamic binding behavior of these molecules was further investigated using long 100 ns molecular dynamics (MD) stimulation revealing NSC631535 to be a potentially stable binder at PD-1 interface pocket. In support of these MD data, the experimental testing of NSC631535 exhibited 50% inhibition at ∼15 μM test concentration. The observed activity of this compound is promising as despite its relatively low molecular weight (415.5 g/mol) it is still capable of inhibiting the PD-1/PD-L1 interaction having a large interface area (∼1970 Å2). In summary, our integrated computational and experimental screening led to identification of a novel PD-1 antagonist that may serve as a starting point for further optimization into more potent small-molecule PD-1/PD-L1 inhibitors for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Jeremy Hofer
- Department of Computer Science, Widener University, Chester, PA, USA
| | - Abhay Aradhya
- Department of Computer Science, Widener University, Chester, PA, USA
| | - Jeffrey Rufinus
- Department of Computer Science, Widener University, Chester, PA, USA
| | - John Stoddart
- Department of Computer Science, Widener University, Chester, PA, USA
| | - Stephen Finocchiaro
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Jabari Mani
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Sean Tevis
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Michael Visconti
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Griffin Walawender
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Juliette DiFlumeri
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Elena Fattakhova
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Sachin P Patil
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA.
| |
Collapse
|
8
|
Brcic I, Kluba AM, Godschachner TM, Suppan C, Regitnig P, Dandachi N, Lax SF, Balić M. Tumor Microenvironment in Male Breast Carcinoma with Emphasis on Tumor Infiltrating Lymphocytes and PD-L1 Expression. Int J Mol Sci 2023; 24:818. [PMID: 36614261 PMCID: PMC9821263 DOI: 10.3390/ijms24010818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023] Open
Abstract
Male breast cancer (MBC) is rare and usually presents as a locally advanced disease. Stromal tumor-infiltrating lymphocytes (sTILs) are associated with a better response to neoadjuvant chemotherapy and improved prognosis in all molecular subtypes of female breast cancer, but their role in MBC is less clear. We studied sTILs and the expression of programmed cell death ligand 1 (PD-L1) and pan-TRK in MBC. We retrospectively studied 113 cases of MBC surgically treated between 1988 and 2015. The tumors were evaluated for histological type and grade, stage, intrinsic subtype and sTILs. We performed immunohistochemistry for PD-L1 (clone SP142) and pan-TRK (clone EPR17341) on tissue microarrays. Pan-TRK positive cases were further analyzed by next-generation sequencing. The median age was 69 years (range 60−77). Invasive carcinoma of no special type was found in 94.7% of cases, of which 53.1% were grade 2. Estrogen receptor was positive in 92% of the tumors, progesterone receptor in 85.8%, androgen receptor in 70.8%; 4.4% were human epidermal growth factor receptor 2 (HER2)-positive, and 55.8% HER2-low. 40.7% of tumors were luminal A and 51.3% luminal B, 4.4% HER2-enriched and 3.5% triple negative carcinoma. sTILs density was <50% in 96.4% of the tumors, >50% in 3.6% of the tumors. PD-L1 immune cell score >1% was found in 7.1% of the tumors (all of luminal subtype). A weak focal cytoplasmic pan-TRK staining was present in 8.8% but without NTRK fusion. Neither sTILs nor PD-L1 had statistically significant outcomes. Our findings suggest that a subset of MBC patients harbors an immunological environment characterized by increased sTILs with PD-L1 expression. These patients may potentially benefit from immune checkpoint inhibitor therapy. Frequent HER2-low may offer novel anti-HER2 treatment options.
Collapse
Affiliation(s)
- Iva Brcic
- Diagnostic and Research Institute of Pathology, Comprehensive Cancer Centre, Medical University of Graz, 8010 Graz, Austria
| | - Andrea Maria Kluba
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Theresa Marie Godschachner
- Diagnostic and Research Institute of Pathology, Comprehensive Cancer Centre, Medical University of Graz, 8010 Graz, Austria
| | - Christoph Suppan
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Peter Regitnig
- Diagnostic and Research Institute of Pathology, Comprehensive Cancer Centre, Medical University of Graz, 8010 Graz, Austria
| | - Nadia Dandachi
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Epigenetic and Genetic Cancer Biomarkers, Medical University of Graz, 8036 Graz, Austria
| | - Sigurd Friedwald Lax
- Diagnostic and Research Institute of Pathology, Comprehensive Cancer Centre, Medical University of Graz, 8010 Graz, Austria
- Department of Pathology, Hospital Graz II, Academic Teaching Hospital of the Medical University Graz, 8020 Graz, Austria
- School of Medicine, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Marija Balić
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- Unit for Translational Breast Cancer Research, Medical University of Graz, 8036 Graz, Austria
- Subcenter for Breast Care, Comprehensive Cancer Center, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
9
|
Oliushina EM, Zavalishina LE, Andreeva YY, Kuznetsova OA, Moskvina LV, Frank GA. [Predictive markers of immunotherapy in cervical cancer]. Arkh Patol 2023; 85:5-12. [PMID: 37814844 DOI: 10.17116/patol2023850515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
OBJECTIVE Study of PD-L1 expression in squamous and adenosquamous cell cervical cancer (CC) by immunohistochemical (IHC) method, assessment of the relationship between PD-L1 tumor status and its clinical and morphological characteristics, TILs, MSI/dMMR, and HPV tumor status. MATERIAL AND METHODS Surgical material was obtained from 41 patients with CC, on which the expression of PD-L1, proteins of the MMR system and p16 was studied by the IHC method, the TILs index was determined. RESULTS Positive PD-L1 status was found in 51.2% of the studied CC samples. In the study sample, the level of PD-L1 expression depended on the severity of lymphoid infiltration of the tumor (p=0.038), it was shown that a positive PD-L1 status of CC can be expected with a TILs value greater than or equal to 50%. The age of the patients, the histological variant of the tumor, the pT and pN stage, the presence of lymphovascular invasion, and the HPV status did not statistically significantly affect the level of PD-L1 expression, however, there was an association between the PD-L1 status and the grade of CC malignancy (p=0.027). The presence of the MSI/dMMR phenomenon was detected in a small percentage of carcinomas (4.9%), the PD-L1 status of these tumors was determined as positive. CONCLUSION A positive PD-L1 status is determined in a significant number of cases of CC, regardless of most of the studied clinical and morphological characteristics; there is a statistically significant relationship between PD-L1 expression and the degree of tumor differentiation and TILs. It has been shown that CC with the MSI/dMMR phenomenon is characterized by a positive PD-L1 status. The authors consider it necessary to study the expression of PD-L1 in patients with cervical carcinomas in order to determine the possibility of prescribing personalized therapy with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- E M Oliushina
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - L E Zavalishina
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - Yu Yu Andreeva
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - O A Kuznetsova
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - L V Moskvina
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - G A Frank
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
10
|
Torrente M, Blanco M, Franco F, Garitaonaindia Y, Calvo V, Collazo-Lorduy A, Gutiérrez L, Sánchez JC, González-del-Alba A, Hernández R, Méndez M, Cantos B, Núñez B, Sousa PAC, Provencio M. Assessing the risk of cardiovascular events in patients receiving immune checkpoint inhibitors. Front Cardiovasc Med 2022; 9:1062858. [PMID: 36531707 PMCID: PMC9751318 DOI: 10.3389/fcvm.2022.1062858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. However, despite their excellent therapeutic effect, these medications typically result in a broad spectrum of toxicity reactions. Immune-related cardiotoxicity is uncommon but can be potentially fatal, and its true incidence is underestimated in clinical trials. The aim of this study is to assess the incidence and identify risk factors for developing a cardiac event in patients treated with ICIs. METHODS We conducted a single-institution retrospective study, including patients treated with ICIs in our center. The main outcomes were cardiac events (CE) and cardiovascular death. RESULTS A total of 378 patients were analyzed. The incidence of CE was 16.7%, during a median follow-up of 50.5 months. The multivariable analysis showed that age, a history of arrhythmia or ischemic heart disease, and prior immune-related adverse events were significantly associated with CE. CONCLUSION CE during ICI treatment are more common than currently appreciated. A complete initial cardiovascular evaluation is recommended, especially in high-risk patients, being necessary a multidisciplinary approach of a specialized cardio-oncology team.
Collapse
Affiliation(s)
- María Torrente
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
- Faculty of Health Sciences, Francisco de Vitoria University, Madrid, Spain
| | - Mariola Blanco
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Fabio Franco
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Yago Garitaonaindia
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Virginia Calvo
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Ana Collazo-Lorduy
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Lourdes Gutiérrez
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Juan Cristóbal Sánchez
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | | | - Roberto Hernández
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Miriam Méndez
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Blanca Cantos
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Beatriz Núñez
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Pedro A. C. Sousa
- Department of Electrical Engineering, NOVA School of Science and Technology, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Mariano Provencio
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| |
Collapse
|
11
|
Chen J, Shen Z, Jiang X, Huang Z, Wu C, Jiang D, Yin L. Preclinical evaluation of IAP0971, a novel immunocytokine that binds specifically to PD1 and fuses IL15/IL15R α complex. Antib Ther 2022; 6:38-48. [PMID: 36683766 PMCID: PMC9847340 DOI: 10.1093/abt/tbac031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Background Currently, cytokine therapy for cancer has demonstrated efficacy in certain diseases but is generally accompanied by severe toxicity. The field of antibody-cytokine fusion proteins (immunocytokines) arose to target these effector molecules to the tumor microenvironment to expand the therapeutic window of cytokine therapy. Therefore, we have developed a novel immunocytokine that binds specifically to programmed death 1 (PD1) and fuses IL15/IL15Rα complex (referred to as IAP0971) for cancer immunotherapy. Methods We report here the making of IAP0971, a novel immunocytokine that binds specifically to PD1 and fuses IL15/IL15Rα complex, and preclinical characterization including pharmacology, pharmacodynamics, pharmacokinetics and toxicology, and discuss its potential as a novel agent for treating patients with advanced malignant tumors. Results IAP0971 bound to human IL2/15Rβ proteins specifically and blocked PD1/PDL1 signaling transduction pathway. IAP0971 promoted the proliferation of CD8 + T cells and natural killer T (NKT) cells, and further activated NK cells to kill tumor cells validated by in vitro assays. In an hPD1 knock-in mouse model, IAP0971 showed potent anti-tumor activity. Preclinical studies in non-human primates following single or repeated dosing of IAP0971 showed favorable pharmacokinetics and well-tolerated toxicology profile. Conclusion IAP0971 has demonstrated a favorable safety profile and potent anti-tumor activities in vivo. A Phase I/IIa clinical trial to evaluate the safety, tolerability and preliminary efficacy of IAP0971 in patients with advanced malignant tumors is on-going (NCT05396391).
Collapse
Affiliation(s)
| | | | - Xiaoling Jiang
- SunHo (China) BioPharmaceutical Co., Ltd, No.5 Xingjian Road, Economic and Technological Development Zone, Nanjing 210008, Jiangsu Province, China
| | - Zhenzhen Huang
- SunHo (China) BioPharmaceutical Co., Ltd, No.5 Xingjian Road, Economic and Technological Development Zone, Nanjing 210008, Jiangsu Province, China
| | - Chongbing Wu
- SunHo (China) BioPharmaceutical Co., Ltd, No.5 Xingjian Road, Economic and Technological Development Zone, Nanjing 210008, Jiangsu Province, China
| | - Dongcheng Jiang
- SunHo (China) BioPharmaceutical Co., Ltd, No.5 Xingjian Road, Economic and Technological Development Zone, Nanjing 210008, Jiangsu Province, China
| | - Liusong Yin
- To whom correspondence should be addressed. Liusong Yin, No.5 Xingjian Road, Economic and Technological Development Zone, Nanjing 210008, Jiangsu Province, China. Tel: (+86) 18651612904; Fax: +86-25-85666030;
| |
Collapse
|
12
|
Chu J, Yeo MK, Lee SH, Lee MY, Chae SW, Kim HS, DO SI. Clinicopathological and Prognostic Significance of Programmed Death Ligand-1 SP142 Expression in 132 Patients With Triple-negative Breast Cancer. In Vivo 2022; 36:2890-2898. [PMID: 36309362 PMCID: PMC9677758 DOI: 10.21873/invivo.13030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND/AIM The prognostic value of programmed death ligand-1 (PD-L1) expression in triple-negative breast cancer (TNBC) has not been sufficiently investigated. In this study, we examined whether PD-L1 expression status is associated with clinicopathological features and outcomes of patients with TNBC. PATIENTS AND METHODS Immunostaining for PD-L1 SP142 was performed on tissue microarrays containing 132 TNBC samples. High PD-L1 expression was defined as ≥10% of the tumor area occupied by PD-L1-expressing cells. RESULTS Thirty-five (26.5%) patients showed high PD-L1 SP142 expression on immune cells (ICs). High IC PD-L1 expression was significantly correlated with smaller tumor size (p=0.030), absence of lymphovascular invasion (p=0.024), and fewer lymph node metastases (p=0.002). Multivariate survival analysis revealed that high IC PD-L1 expression independently predicted better disease-free survival (DFS) of TNBC patients. CONCLUSION High PD-L1 SP142 expression on ICs was significantly associated with favorable clinicopathological parameters and better outcomes in patients with TNBC. Our observations suggest that high IC PD-L1 expression can be used as an independent prognostic marker for predicting better DFS in patients with TNBC.
Collapse
Affiliation(s)
- Jinah Chu
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sang Hwa Lee
- Pathology Center, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Mi Yeon Lee
- Division of Biostatistics, Department of R&D Management, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seoung Wan Chae
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung-Im DO
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea;
| |
Collapse
|
13
|
The new progress in cancer immunotherapy. Clin Exp Med 2022:10.1007/s10238-022-00887-0. [DOI: 10.1007/s10238-022-00887-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022]
Abstract
AbstractThe cross talk between immune and non-immune cells in the tumor microenvironment leads to immunosuppression, which promotes tumor growth and survival. Immunotherapy is an advanced treatment that boosts humoral and cellular immunity rather than using chemotherapy or radiation-based strategy associated with non-specific targets and toxic effects on normal cells. Immune checkpoint inhibitors and T cell-based immunotherapy have already exhibited significant effects against solid tumors and leukemia. Tumor cells that escape immune surveillance create a major obstacle to acquiring an effective immune response in cancer patients. Tremendous progress had been made in recent years on a wide range of innate and adaptive immune checkpoints which play a significant role to prevent tumorigenesis, and might therefore be potential targets to suppress tumor cells growth. This review aimed to summarize the underlying molecular mechanisms of existing immunotherapy approaches including T cell and NK-derived immune checkpoint therapy, as well as other intrinsic and phagocytosis checkpoints. Together, these insights will pave the way for new innate and adaptive immunomodulatory targets for the development of highly effective new therapy in the future.
Collapse
|
14
|
Ahmed M, Ganesan A, Barakat K. Leveraging structural and 2D-QSAR to investigate the role of functional group substitutions, conserved surface residues and desolvation in triggering the small molecule-induced dimerization of hPD-L1. BMC Chem 2022; 16:49. [PMID: 35761353 PMCID: PMC9238240 DOI: 10.1186/s13065-022-00842-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 06/21/2022] [Indexed: 12/02/2022] Open
Abstract
Small molecules are rising as a new generation of immune checkpoints’ inhibitors, with compounds targeting the human Programmed death-ligand 1 (hPD-L1) protein are pioneering this area of research. Promising examples include the recently disclosed compounds from Bristol-Myers-Squibb (BMS). These molecules bind specifically to hPD-L1 through a unique mode of action. They induce dimerization between two hPD-L1 monomers through the hPD-1 binding interface in each monomer, thereby inhibiting the PD-1/PD-L1 axis. While the recently reported crystal structures of such small molecules bound to hPD-L1 reveal valuable insights regarding their molecular interactions, there is still limited information about the dynamics driving this unusual complex formation. The current study provides an in-depth computational structural analysis to study the interactions of five small molecule compounds in complex with hPD-L1. By employing a combination of molecular dynamic simulations, binding energy calculations and computational solvent mapping techniques, our analyses quantified the dynamic roles of different hydrophilic and lipophilic residues at the surface of hPD-L1 in mediating these interactions. Furthermore, ligand-based analyses, including Free-Wilson 2D-QSAR was conducted to quantify the impact of R-group substitutions at different sites of the phenoxy-methyl biphenyl core. Our results emphasize the importance of a terminal phenyl ring that must be present in any hPD-L1 small molecule inhibitor. This phenyl moiety overlaps with a very unfavorable hydration site, which can explain the ability of such small molecules to trigger hPD-L1 dimerization.
Collapse
Affiliation(s)
- Marawan Ahmed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Aravindhan Ganesan
- ArGan's Lab, School of Pharmacy, University of Waterloo, Kitchener, ON, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada. .,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
15
|
Ushio R, Murakami S, Saito H. Predictive Markers for Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. J Clin Med 2022; 11:1855. [PMID: 35407463 PMCID: PMC9000007 DOI: 10.3390/jcm11071855] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically improved the outcomes of non-small cell lung cancer patients and have increased the possibility of long-term survival. However, few patients benefit from ICIs, and no predictive biomarkers other than tumor programmed cell death ligand 1 (PD-L1) expression have been established. Hence, the identification of biomarkers is an urgent issue. This review outlines the current understanding of predictive markers for the efficacy of ICIs, including PD-L1, tumor mutation burden, DNA mismatch repair deficiency, microsatellite instability, CD8+ tumor-infiltrating lymphocytes, human leukocyte antigen class I, tumor/specific genotype, and blood biomarkers such as peripheral T-cell phenotype, neutrophil-to-lymphocyte ratio, interferon-gamma, and interleukin-8. A tremendous number of biomarkers are in development, but individual biomarkers are insufficient. Tissue biomarkers have issues in reproducibility and accuracy because of intratumoral heterogeneity and biopsy invasiveness. Furthermore, blood biomarkers have difficulty in reflecting the tumor microenvironment and therefore tend to be less predictive for the efficacy of ICIs than tissue samples. In addition to individual biomarkers, the development of composite markers, including novel technologies such as machine learning and high-throughput analysis, may make it easier to comprehensively analyze multiple biomarkers.
Collapse
Affiliation(s)
| | - Shuji Murakami
- Kanagawa Cancer Center, Department of Thoracic Oncology, 2-3-2 Nakao, Asahi, Yokohama 241-8515, Japan; (R.U.); (H.S.)
| | | |
Collapse
|
16
|
Ding J, Wang T, Chen Z, Lin Z, Chen X, He C. Enhanced antitumor chemo‐immunotherapy by local co‐delivery of chemotherapeutics, immune checkpoint blocking antibody and
IDO
inhibitor using an injectable polypeptide hydrogel. JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1002/pol.20210955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Junfeng Ding
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin P. R. China
- College of Applied Chemistry and Engineering University of Science and Technology of China Hefei Anhui P. R. China
| | - Tianran Wang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin P. R. China
- College of Applied Chemistry and Engineering University of Science and Technology of China Hefei Anhui P. R. China
| | - Zhixiong Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin P. R. China
- College of Applied Chemistry and Engineering University of Science and Technology of China Hefei Anhui P. R. China
| | - Zhiqiang Lin
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin P. R. China
- College of Applied Chemistry and Engineering University of Science and Technology of China Hefei Anhui P. R. China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin P. R. China
- College of Applied Chemistry and Engineering University of Science and Technology of China Hefei Anhui P. R. China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin P. R. China
- College of Applied Chemistry and Engineering University of Science and Technology of China Hefei Anhui P. R. China
| |
Collapse
|
17
|
Qu X, Sun D, Cheng Y, Wang J, Wang Y, Xu W, Qi G, Xu S. Electrostimulus-associated PD-L1 expression on cell membrane revealed by immune SERS nanoprobes. Analyst 2022; 147:527-533. [PMID: 35048911 DOI: 10.1039/d1an01930a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Programmed cell death ligand 1 (PD-L1) is considered a major immune checkpoint protein that mediates antitumor immune suppression and response. Effectively regulating PD-L1 expression and dynamic monitoring has become a significant challenge in immunotherapy. Herein, we adopted smart surface-enhanced Raman scattering (SERS) nanoprobes to discriminate and monitor the dynamic expression of PD-L1 under external electrostimulation (ES). The PD-L1 expression levels in three cell lines (MCF-7 cells, HeLa cells, and H8 cells) were assessed before and after ES. The results reveal that ES could effectively and rapidly mediate a transformation in the PD-L1 content (or activity) on the cell membrane. Moreover, the molecular profiles of the cell membrane before and after ES were revealed by using the label-free SERS method with the help of immune plasmonic nanoparticles. The cell membrane protein information presented identifiable conformation changes after ES, showing a significant inhibitory effect on the bridge of PD-L1 and its antibody. This study indicates that ES is superior to chemical drugs due to lesser side effects because ES-based regulation does not depend on intracellular signalling pathways. This strategy is versatile and robust for discriminating and monitoring PD-L1 on cell membranes, thus providing potential clinical application value to PD-L1-mediated systems. This study also offers a practical way to assess the molecular profiles of cell membrane proteins in the presence of an external stimulus, which may be applicable to many membrane protein-related studies.
Collapse
Affiliation(s)
- Xiaozhang Qu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China. .,The First Hospital of Jilin University, Changchun, 130031, P. R. China.,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Dan Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China. .,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yuqi Cheng
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China. .,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Jiafeng Wang
- Department of Endodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, P. R. China
| | - Yan Wang
- The Changchun Central Hospital, Changchun, 130061, P. R. China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China. .,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Guohua Qi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China. .,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
18
|
Qin J, Zhang M, Guan Y, Guo X, Li Z, Rankl C, Tang J. Imaging and quantifying analysis the binding behavior of PD-L1 at molecular resolution by atomic force microscopy. Anal Chim Acta 2022; 1191:339281. [PMID: 35033247 DOI: 10.1016/j.aca.2021.339281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/20/2021] [Accepted: 11/11/2021] [Indexed: 11/25/2022]
Abstract
Immunotherapy has emerged as an effective treatment modality for cancer. The interaction of programmed cell death ligand-1 (PD-L1) and programmed cell death protein-1 (PD-1) plays a key role in tumor-related immune escape and has become one of the most extensive targets for immunotherapy. Herein, we investigated the interaction of PD-L1 with its antibody and PD-1 using atomic force microscopy-based single molecule force spectroscopy for the first time. It was found that the PD-L1/anti-PD-L1 antibody complex was easier to dissociate than PD-L1/PD-1. The unbinding forces of specific interaction of PD-L1 on T24 cells with its antibody and PD-1 were quantitatively measured and similar to those on substrate. In addition, the location of PD-L1 on T24 cells was mapped at the single-molecule level by force-volume mapping. The force maps revealed that PD-L1 randomly distributed on T24 cells surface. The recognition events on cells obviously increased after INF-γ treatment, which proved that INF-γ up-regulated the expression of PD-L1 on T24 cells. These findings enrich our understanding of the molecular mechanisms by which PD-L1 interacts with its antibody and PD-1. It provides useful information for the physical factors that is needed to be considered in the design of inhibitors for tumor immunology.
Collapse
Affiliation(s)
- Juan Qin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China; University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Miaomiao Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China; University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Yanxue Guan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China; University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Xinyue Guo
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China; University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Zongjia Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China; University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Christian Rankl
- Research Center for Non Destructive Testing GmbH, Science Park 2/2. OG, Altenberger Straße 69, A-4040, Linz, Austria
| | - Jilin Tang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China; University of Science and Technology of China, Hefei, 230026, P.R. China.
| |
Collapse
|
19
|
Ma C, Qiao S, Liu Z, Shan L, Liang C, Fan M, Sun F. A Novel Type of PD-L1 Inhibitor rU1 snRNPA From Human-Derived Protein Scaffolds Library. Front Oncol 2021; 11:781046. [PMID: 34912719 PMCID: PMC8666589 DOI: 10.3389/fonc.2021.781046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Three marketed anti-PD-L1 antibodies almost have severe immune-mediated side effects. The therapeutic effects of anti-PD-L1 chemical inhibitors are not satisfied in the clinical trials. Here we constructed human-derived protein scaffolds library and screened scaffolds with a shape complementary to the PD-1 binding domain of PD-L1. The RNA binding domain of U1 snRNPA was selected as one of potential binders because it had the most favorable binding energies with PD-L1 and conformed to pre-established biological criteria for the screening of candidates. The recombinant U1 snRNPA (rU1 snRNPA) in Escherichia coli exhibits anti-cancer activity in melanoma and breast cancer by reactivating tumor-suppressed T cells in vitro and anti-melanoma activity in vivo. Considering hydrophobic and electrostatic interactions, three residues were mutated on the interface of U1 snRNPA and PD-L1 complex, and the ranked variants by PatchDock and A32D showed an increased active phenotype. The screening of human-derived protein scaffolds may become the potential development of therapeutic agents.
Collapse
Affiliation(s)
- Chuang Ma
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Sennan Qiao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zhiyi Liu
- Institute of Frontier Medical Science, Jilin University, Changchun, China
| | - Liang Shan
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Chongyang Liang
- Institute of Frontier Medical Science, Jilin University, Changchun, China
| | - Meiling Fan
- Jilin Academy of Chinese Medicine Sciences, Changchun, China
| | - Fei Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
20
|
Zhong Y, Lai D, Zhang L, Lu W, Shang Y, Zhou H. The Effects of Moxibustion on PD-1/PD-L1-Related Molecular Expression and Inflammatory Cytokine Levels in RA Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6658946. [PMID: 39290616 PMCID: PMC11407876 DOI: 10.1155/2021/6658946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 05/03/2021] [Accepted: 11/27/2021] [Indexed: 09/19/2024]
Abstract
Objective Rheumatoid arthritis (RA) is an autoimmune disease that starts with inflammation of the synovium. The pain and joint dysfunction caused by RA urgently need an effective treatment to alleviate the inflammatory reaction and delay the progression of the disease. The pathological damage of RA is proposed to associate with the dysfunction of the programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) pathway. Moxibustion, as a main complementary therapy of traditional Chinese medicine (TCM), has been proved effective to reduce chronic inflammatory reaction on RA, but whether the anti-inflammatory effects are mediated by PD-1/PD-L1 pathway is still unclear. Therefore, moxibustion was conducted in the rats with RA to investigate its effect on PD-1/PD-L1. Methods The rats' right hind paws were injected with Freund's complete adjuvant (FCA) to establish the model of RA. Seven days after the injection of FCA, moxibustion therapy was performed on the acupoints of Shenshu (BL23) and Zusanli (ST36) once a day for three weeks. Then, ELISA and immunohistochemical methods were used to analyze the influence of moxibustion on the expression of PD-1/PD-L1. If the moxibustion had an effect on the expression of PD-1/PD-L1-related molecules, we would knock down PD-1 with adenovirus vector. After moxibustion therapy, ELISA and histological analysis were performed to observe the anti-inflammatory effect of moxibustion. Results The results demonstrated that moxibustion had an effect on the expression of PD-1-related molecules. The results of ELISA manifested that moxibustion decreased the level of IFN-γ and increased the level of IL-4 and IL-10. HE staining revealed that moxibustion alleviated the proliferation of synovial tissue. However, the anti-inflammatory effect and pathological improvement were weakened when PD-1 was blocked. Conclusions The results indicate that moxibustion affected the expression of PD-1/PD-L1-related molecules and can effectively treat RA damage. The anti-inflammatory effect of moxibustion was weakened when PD-1 was knocked down.
Collapse
Affiliation(s)
- Yumei Zhong
- Chengdu First People's Hospital, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610095, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Deli Lai
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- The Affiliated Hospital, School of Medicine, UESTC, Chengdu Women's and Children's Central Hospital, Chengdu 610014, China
| | - Linlin Zhang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Wenting Lu
- Chengdu First People's Hospital, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610095, China
| | - Yanan Shang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Haiyan Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
21
|
Li Y, Hu Y, Yang B, Jin C, Ren H, Wu J, Wang Z, Wei Y, Yang L, Hu Y. Immunotherapy-Related Cardiotoxicity Re-Emergence in Non-Small Cell Lung Cancer - A Case Report. Onco Targets Ther 2021; 14:5309-5314. [PMID: 34848973 PMCID: PMC8627268 DOI: 10.2147/ott.s333242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/29/2021] [Indexed: 01/22/2023] Open
Abstract
PD-1/PD-L1 inhibitors activate immunological response and have become one of the main modalities of cancer treatment. However, they may result in the immune-related adverse events (irAEs). Immune-related cardiotoxicity is relatively rare but may become fatal. We will present a case of a male patient who experienced immunotherapy-related cardiotoxicity one year after received pembrolizumab treatment. The patient had atypical symptom presentation initially, but his condition deteriorated worsened rapidly and he developed severe cardiac disease. The patient experienced significant relief after corticosteroid treatment. Unfortunately, he experienced a reoccurence of the severe adverse event when discontinuing the use of corticosteroids. Ultimately, larger doses and longer courses of corticosteroid treatment cured the heart damage. Fortunately, we observed that lesions were stable and maintained for a long time after cessation of using pembrolizumab for eight months.
Collapse
Affiliation(s)
- Yuanxiang Li
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Yang Hu
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Bin Yang
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Caibao Jin
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Hui Ren
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Jingyi Wu
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Zhijun Wang
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Youying Wei
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Ling Yang
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| | - Yanping Hu
- Department of Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, People's Republic of China
| |
Collapse
|
22
|
Wu X, Lan X, Hu W, Zhang W, Lai X, Xu S, Li J, Qiu W, Wang W, Xiao J, Wang F, Ding Y, Liang L. CMTM6 expression in M2 macrophages is a potential predictor of PD-1/PD-L1 inhibitor response in colorectal cancer. Cancer Immunol Immunother 2021; 70:3235-3248. [DOI: 33818637 10.1007/s00262-021-02931-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/27/2021] [Indexed: 05/20/2025]
Abstract
Abstract
Background
CMTM6 is a novel key regulator of PD-L1. High expression of both CMTM6 and PD-L1 may predict the benefit of PD-1 axis blockade in lung cancer. We aimed to investigate the expression pattern of CMTM6 between mismatch repair-defective (dMMR) and mismatch repair-proficient (pMMR) colorectal cancer (CRC) tissues and assess its correlation with the response to PD-1/PD-L1 pathway blockade.
Methods
Immunohistochemistry (IHC) was used to analyze CMTM6 and PD-L1 expression and immune cell density in dMMR/pMMR CRC. Quantitative multiplex immunofluorescence (IF) was performed to detect CMTM6, PD-L1, CD4, CD8, CD68 and CD163 expression in CRC patients treated with PD-1/PD-L1 inhibitors.
Result
IHC analysis showed that CMTM6 and PD-L1 were both expressed in tumor cells (TCs) and invasion front immune cells (ICs). CMTM6 and PD-L1 expression and CD4+, CD8+, CD68+ or CD163+ cell density were significantly higher in dMMR CRC patients than in pMMR CRC patients. CMTM6 expression was positively correlated with PD-L1 expression and CD163+ M2 macrophage density in dMMR CRC. IF analysis showed that the coexpression rate of CMTM6/PD-L1 and the expression rate of CMTM6 in CD8+ T cells and CD163+ M2 macrophages were significantly increased in the group that exhibited clinical benefit. CMTM6 expression in M2 macrophages was identified as the best biomarker for predicting the responsiveness to PD-1/PD-L1 inhibitors.
Conclusions
CMTM6 expression in M2 macrophages may predict the PD-1/PD-L1 inhibitor response rate in CRC patients more accurately than dMMR/microsatellite instability-high (MSI-H) status. It can also identify pMMR CRC patients who could benefit from PD-1/PD-L1 inhibitors.
Collapse
|
23
|
Immunotherapy of cancer tumors with inhibition of PD-1 membrane protein and its ligands interaction. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2021-6.4.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The identification of the PD-1 receptor by Tasuku Honjo and CTLA-4 by James Ellison marked the beginning of the study of new regulatory pathways activating the immune response. The term “immune checkpoints” was introduced to denote the system of inhibitory mechanisms that include these proteins. The review presents the literature data on the molecular characteristics of the membrane protein PD-1 (programmed cell death 1 receptor) and its role in the regulation of immunity. We consider the PD-1 pathways used of by tumor cells to escape the immune response. The discovery of immune checkpoints made it possible to develop a new type of targeting therapy for cancer. The review presents the results of clinical trials of drugs that block the interaction between the PD-1 and its ligands in various types of cancer. These drugs include nivolumab, pembrolizumab, and avelumab. Studies of these drugs efficacy in patients with various types of cancer localization were conducted within the CheckMate, KEYNOTE and JAVELIN Solid Tumor programs, with some research being in progress. We analyze the results of studying the clinical efficacy of the drugs in patients with melanoma, lung cancer, renal cell cancer, colorectal cancer, classical Hodgkin’s lymphoma, Merkel carcinoma and stomach cancer. Both positive and inconclusive results in the treatment of patients are noted. These data made it possible to identify promising directions for the use of the drugs in certain localizations of the malignant process, as well as to determine the dose and time of their use to obtain an objective positive response to treatment.
Collapse
|
24
|
Kalpakoff M, Hund S, Musser J, Roach C, Apostolaki A, Vilardo M, Peltz L, Watts B, LaPlaca C, Tabuena-Frolli S, DiMaio MA, Welcher R, Kulangara K. Intrapatient Tumor Heterogeneity in IHC Interpretation Using PD-L1 IHC 22C3 pharmDx. Appl Immunohistochem Mol Morphol 2021; 29:667-673. [PMID: 33973887 PMCID: PMC8505133 DOI: 10.1097/pai.0000000000000941] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/01/2021] [Indexed: 11/26/2022]
Abstract
Tumor heterogeneity may impact immunohistochemical (IHC) interpretation, thus potentially affecting decision making by treating oncologists for cancer patient management. Programmed cell death ligand-1 (PD-L1) IHC 22C3 pharmDx is a companion diagnostic used as an aid in identifying patient eligibility for treatment with pembrolizumab (KEYTRUDA). This study aims to investigate tumor heterogeneity impact on IHC staining when evaluating PD-L1 expression using PD-L1 IHC 22C3 pharmDx. The effect of tumor heterogeneity was evaluated based on the PD-L1 diagnostic status of PD-L1 IHC 22C3 pharmDx stained tumor tissue sections at relevant diagnostic cutoffs for non-small cell lung carcinoma, gastric or gastroesophageal junction adenocarcinoma, urothelial carcinoma, head and neck squamous cell carcinoma, esophageal cancer and triple negative breast cancer. Overall agreement for the PD-L1 diagnostic status was assessed for each tumor type within a given specimen block (Intra-Block), between specimen blocks from the same surgical resection (Intra-Case), and between intrapatient primary and metastatic specimens. Intrablock and intracase point estimates were above 75%, and primary versus metastatic point estimates were above 50%. The results suggest that PD-L1 expression is consistent across cut sections through a minimum of 150 µm within a tissue block and between blocks from the same surgical resection and is significantly maintained across primary and metastatic blocks from the same patient despite changes to the tissue microenvironment. These data provide confidence for histopathologists and oncologists that evaluation of PD-L1 expression at clinically relevant cutoffs is reproducible among different assessments (or samplings) of a single tumor specimen.
Collapse
|
25
|
Mechanistic Insights of Anti-Immune Evasion by Nobiletin through Regulating miR-197/STAT3/PD-L1 Signaling in Non-Small Cell Lung Cancer (NSCLC) Cells. Int J Mol Sci 2021; 22:ijms22189843. [PMID: 34576006 PMCID: PMC8468939 DOI: 10.3390/ijms22189843] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 01/02/2023] Open
Abstract
Tumor immune escape is a common process in the tumorigenesis of non-small cell lung cancer (NSCLC) cells where programmed death ligand-1 (PD-L1) expression, playing a vital role in immunosuppression activity. Additionally, epidermal growth factor receptor (EGFR) phosphorylation activates Janus kinase-2 (JAK2) and signal transduction, thus activating transcription 3 (STAT3) to results in the regulation of PD-L1 expression. Chemotherapy with commercially available drugs against NSCLC has struggled in the prospect of adverse effects. Nobiletin is a natural flavonoid isolated from the citrus peel that exhibits anti-cancer activity. Here, we demonstrated the role of nobiletin in evasion of immunosuppression in NSCLC cells by Western blotting and real-time polymerase chain reaction methods for molecular signaling analysis supported by gene silencing and specific inhibitors. From the results, we found that nobiletin inhibited PD-L1 expression through EGFR/JAK2/STAT3 signaling. We also demonstrated that nobiletin exhibited p53-independent PD-L1 suppression, and that miR-197 regulates the expression of STAT3 and PD-L1, thereby enhancing anti-tumor immunity. Further, we evaluated the combination ability of nobiletin with an anti-PD-1 monoclonal antibody in NSCLC co-culture with peripheral blood mononuclear cells. Similarly, we found that nobiletin assisted the induction of PD-1/PD-L1 blockade, which is a key factor for the immune escape mechanism. Altogether, we propose nobiletin as a modulator of tumor microenvironment for cancer immunotherapy.
Collapse
|
26
|
Nie D, Xue Y, Fang Q, Cheng J, Li B, Wang D, Li C, Gui S, Zhang Y, Zhao P. Immune Checkpoints: Therapeutic Targets for Pituitary Tumors. DISEASE MARKERS 2021; 2021:5300381. [PMID: 34447484 PMCID: PMC8384513 DOI: 10.1155/2021/5300381] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/06/2021] [Indexed: 11/18/2022]
Abstract
Pituitary tumors are the third most common intracranial tumors in adults. Treatment of refractory pituitary tumors is known to be difficult due to limited treatment options. As a promising therapeutic method, tumor immunotherapy has been applied in the treatment of many tumors, including pituitary tumors. Immune checkpoint blocking is one of the effective strategies to activate antitumor immunity. Immune checkpoints prevent tissue damage by regulating the immune response of peripheral tissues and participate in the maintenance of a normal immune environment. In the presence of a tumor, inhibition of T cell activity by tumor cells binding to immune checkpoints and their ligands is an important mechanism for tumor cells to escape immune injury. In this review, we summarize the latest findings of immune checkpoints and their potential as immunotherapeutic targets for pituitary tumors.
Collapse
Affiliation(s)
- Ding Nie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yimeng Xue
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qiuyue Fang
- Beijing Neurosurgical Institute, Beijing, China
| | | | - Bin Li
- Beijing Neurosurgical Institute, Beijing, China
| | - Dawei Wang
- Beijing Neurosurgical Institute, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | | | - Peng Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Discovery of phenyl-linked symmetric small molecules as inhibitors of the programmed cell death-1/programmed cell death-ligand 1 interaction. Eur J Med Chem 2021; 223:113637. [PMID: 34147746 DOI: 10.1016/j.ejmech.2021.113637] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/01/2021] [Accepted: 06/05/2021] [Indexed: 11/21/2022]
Abstract
Programmed cell death-1/programmed cell death ligand 1 (PD-1/PD-L1) is one of the most promising targets in the field of immune checkpoint blockade therapy. Beginning with our exploration of linkers and structure-activity relationship research, we found that the aromatic ring could replace the linker and aryl group to maintain the satisfactory activity of classic triaryl scaffold inhibitor. Based on previous studies, we designed and synthesized a series of C2-symmetric phenyl-linked compounds, and further tail optimization afforded the inhibitors, which displayed promising inhibitory activity against the PD-1/PD-L1 interaction with IC50 value at the single nanomolar range (C13-C15). Further cell-based PD-1/PD-L1 blockade bioassays indicated that these C2-symmetric molecules could significantly inhibit the PD-1/PD-L1 interaction at the cellular level and restore T cells' immune function at the safety concentrations. The discovery of these phenyl-linked symmetric small molecules showed the potential of simplified-linker and C2-symmetric strategy and provided a basis for developing symmetric small molecule inhibitors of PD-1/PD-L1 interaction. Moreover, C13 and C15 performed stable binding modes to PD-L1 dimeric after computational docking and dynamic simulation, which may serve as a good starting point for further development.
Collapse
|
28
|
Mehdizadeh S, Bayatipoor H, Pashangzadeh S, Jafarpour R, Shojaei Z, Motallebnezhad M. Immune checkpoints and cancer development: Therapeutic implications and future directions. Pathol Res Pract 2021; 223:153485. [PMID: 34022684 DOI: 10.1016/j.prp.2021.153485] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 02/08/2023]
Abstract
Over the past few decades, different inhibitory receptors have been identified, which have played prominent roles in reducing anti-tumor immune responses. The role of immune checkpoint inhibitors in cancer was revealed by critical blockade of the cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death protein-1 (PD-1) checkpoints. Immune checkpoint inhibitors, including anti-PD-1 (nivolumab and pembrolizumab), anti-PD-L1 (Atezolizumab, avelumab, and duravulumab), and anti-CTLA-4 (ipilimumab, tremelimumab), are currently FDA-approved treatment options for a broad range of cancer types. However, regarding immunotherapy advances in recent years, most studies have been focused on finding the antibodies against other inhibitory immune checkpoints in the tumor microenvironment such as lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin, and mucin domain 3 (TIM-3), B7-homolog 3 (B7-H3), V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA), diacylglycerol kinase-α (DGK-α), T cell immunoglobulin and ITIM domain (TIGIT), and B and T lymphocyte attenuator (BTLA). This immune checkpoint exerts differential inhibitory impacts on various types of lymphocytes. The suppression of immune responses demonstrates a surprising synergy with PD-1. Therefore, most antibodies against these immune checkpoints are undertaking clinical trials for cancer immunotherapy of advanced solid tumors and hematologic malignancies. In this review, we will summarize recent findings of immune checkpoint and the role of monoclonal antibodies in cancer immunotherapy targeting these receptors.
Collapse
Affiliation(s)
- Saber Mehdizadeh
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hashem Bayatipoor
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Salar Pashangzadeh
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Roghayeh Jafarpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Shojaei
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Motallebnezhad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Kim HR, Tagirasa R, Yoo E. Covalent Small Molecule Immunomodulators Targeting the Protease Active Site. J Med Chem 2021; 64:5291-5322. [PMID: 33904753 DOI: 10.1021/acs.jmedchem.1c00172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells of the immune system utilize multiple proteases to regulate cell functions and orchestrate innate and adaptive immune responses. Dysregulated protease activities are implicated in many immune-related disorders; thus, protease inhibitors have been actively investigated for pharmaceutical development. Although historically considered challenging with concerns about toxicity, compounds that covalently modify the protease active site represent an important class of agents, emerging not only as chemical probes but also as approved drugs. Here, we provide an overview of technologies useful for the study of proteases with the focus on recent advances in chemoproteomic methods and screening platforms. By highlighting covalent inhibitors that have been designed to target immunomodulatory proteases, we identify opportunities for the development of small molecule immunomodulators.
Collapse
Affiliation(s)
- Hong-Rae Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
30
|
PD-L1 signaling on human memory CD4+ T cells induces a regulatory phenotype. PLoS Biol 2021; 19:e3001199. [PMID: 33901179 PMCID: PMC8101994 DOI: 10.1371/journal.pbio.3001199] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 05/06/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) is expressed on T cells upon T cell receptor (TCR) stimulation. PD-1 ligand 1 (PD-L1) is expressed in most tumor environments, and its binding to PD-1 on T cells drives them to apoptosis or into a regulatory phenotype. The fact that PD-L1 itself is also expressed on T cells upon activation has been largely neglected. Here, we demonstrate that PD-L1 ligation on human CD25-depleted CD4+ T cells, combined with CD3/TCR stimulation, induces their conversion into highly suppressive T cells. Furthermore, this effect was most prominent in memory (CD45RA−CD45RO+) T cells. PD-L1 engagement on T cells resulted in reduced ERK phosphorylation and decreased AKT/mTOR/S6 signaling. Importantly, T cells from rheumatoid arthritis patients exhibited high basal levels of phosphorylated ERK and following PD-L1 cross-linking both ERK signaling and the AKT/mTOR/S6 pathway failed to be down modulated, making them refractory to the acquisition of a regulatory phenotype. Altogether, our results suggest that PD-L1 signaling on memory T cells could play an important role in resolving inflammatory responses; maintaining a tolerogenic environment and its failure could contribute to ongoing autoimmunity. This study shows that programmed death cell receptor ligand 1 (PD-L1) signaling in memory CD4+ T cells from healthy individuals induces a regulatory phenotype; this mechanism seems to be defective in equivalent T cells from rheumatoid arthritis patients and could be in part responsible for the pathology.
Collapse
|
31
|
Li L, Miao Q, Meng F, Li B, Xue T, Fang T, Zhang Z, Zhang J, Ye X, Kang Y, Zhang X, Chen Q, Liang X, Chen H, Zhang X. Genetic engineering cellular vesicles expressing CD64 as checkpoint antibody carrier for cancer immunotherapy. Am J Cancer Res 2021; 11:6033-6043. [PMID: 33897897 PMCID: PMC8058713 DOI: 10.7150/thno.48868] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint blockade therapies, especially those targeting the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) have achieved impressive clinical responses in multiple types of cancers. To optimize the therapeutic effect of the checkpoint antibodies, many strategies including targeting delivery, controlled release, and cellular synthesis have been developed. However, within these strategies, antibodies were attached to drug carriers by chemical bonding, which may affect the steric configuration and function of the antibodies. Herein, we prepared cluster of differentiation 64 (CD64), a natural catcher of the fragment crystalline (Fc) of monomeric immunoglobulin G (IgG), and over-expressed it on the cell membrane nanovesicles (NVs) as PD-L1 antibody delivery vehicle (CD64-NVs-aPD-L1), which was employed to disrupt the PD-1/PD-L1 immunosuppressive signal axis for boosting T cell dependent tumor elimination. Meanwhile, chemical immunomodulatory drug cyclophosphamide (CP) was also encapsulated in the vesicle (CD64-NVs-aPD-L1-CP), to simultaneously restrain the regulatory T cells (Tregs) and invigorate Ki67+CD8+ T cells, then further enhance their anti-tumor ability. Methods: The cell membrane NVs overexpressing CD64 were incubated with PD-L1 antibody and chemotherapeutic agent CP to prepare CD64-NVs-aPD-L1-CP. Results: The CD64-NVs-aPD-L1-CP could simultaneously interrupt the immunosuppressive effect of PD-L1 and decrease the inhibition of Tregs, leading to tumor growth suppression and survival time extension. Conclusion: CD64-NVs are charismatic carriers to achieve both checkpoint blockade and immunomodulatory drugs for combined cancer immunotherapy.
Collapse
|
32
|
Wu X, Lan X, Hu W, Zhang W, Lai X, Xu S, Li J, Qiu W, Wang W, Xiao J, Wang F, Ding Y, Liang L. CMTM6 expression in M2 macrophages is a potential predictor of PD-1/PD-L1 inhibitor response in colorectal cancer. Cancer Immunol Immunother 2021; 70:3235-3248. [PMID: 33818637 PMCID: PMC8505364 DOI: 10.1007/s00262-021-02931-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Background CMTM6 is a novel key regulator of PD-L1. High expression of both CMTM6 and PD-L1 may predict the benefit of PD-1 axis blockade in lung cancer. We aimed to investigate the expression pattern of CMTM6 between mismatch repair-defective (dMMR) and mismatch repair-proficient (pMMR) colorectal cancer (CRC) tissues and assess its correlation with the response to PD-1/PD-L1 pathway blockade.
Methods Immunohistochemistry (IHC) was used to analyze CMTM6 and PD-L1 expression and immune cell density in dMMR/pMMR CRC. Quantitative multiplex immunofluorescence (IF) was performed to detect CMTM6, PD-L1, CD4, CD8, CD68 and CD163 expression in CRC patients treated with PD-1/PD-L1 inhibitors. Result IHC analysis showed that CMTM6 and PD-L1 were both expressed in tumor cells (TCs) and invasion front immune cells (ICs). CMTM6 and PD-L1 expression and CD4+, CD8+, CD68+ or CD163+ cell density were significantly higher in dMMR CRC patients than in pMMR CRC patients. CMTM6 expression was positively correlated with PD-L1 expression and CD163+ M2 macrophage density in dMMR CRC. IF analysis showed that the coexpression rate of CMTM6/PD-L1 and the expression rate of CMTM6 in CD8+ T cells and CD163+ M2 macrophages were significantly increased in the group that exhibited clinical benefit. CMTM6 expression in M2 macrophages was identified as the best biomarker for predicting the responsiveness to PD-1/PD-L1 inhibitors. Conclusions CMTM6 expression in M2 macrophages may predict the PD-1/PD-L1 inhibitor response rate in CRC patients more accurately than dMMR/microsatellite instability-high (MSI-H) status. It can also identify pMMR CRC patients who could benefit from PD-1/PD-L1 inhibitors. Supplementary Information The online version contains supplementary material available at 10.1007/s00262-021-02931-6.
Collapse
Affiliation(s)
- Xuehui Wu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Department of Pathology, The People's Hospital of China Three Gorges University, Yichang, 443000, Hubei Province, People's Republic of China
| | - Xiaoliang Lan
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Wanming Hu
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, People's Republic of China
| | - Wanning Zhang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Xiangmeng Lai
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Shaowan Xu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Jiaoying Li
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong Province, People's Republic of China
| | - Weihao Qiu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Wei Wang
- General Hospital of Southern Theater Command, People's Liberation Army of China, Guangzhou, 510010, Guangdong Province, People's Republic of China
| | - Jianbiao Xiao
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Feifei Wang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China.
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong Province, People's Republic of China.
| |
Collapse
|
33
|
Poto R, Marone G, Pirozzi F, Galdiero MR, Cuomo A, Formisano L, Bianco R, Della Corte CM, Morgillo F, Napolitano S, Troiani T, Tocchetti CG, Mercurio V, Varricchi G. How can we manage the cardiac toxicity of immune checkpoint inhibitors? Expert Opin Drug Saf 2021; 20:685-694. [PMID: 33749484 DOI: 10.1080/14740338.2021.1906860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Cancer immunotherapies with monoclonal antibodies (mAbs) against immune checkpoints (i.e. CTLA-4 and PD-1/PD-L1) have revolutionized antineoplastic treatments. Immune checkpoint inhibitors (ICIs) approved for cancer immunotherapy are mAbs anti-CTLA-4 (ipilimumab), anti-PD-1 (nivolumab, pembrolizumab, and cemiplimab), and anti-PD-L1 (atezolizumab, avelumab, and durvalumab). Treatment with ICIs can be associated with immune-related adverse events (irAEs), including an increased risk of developing myocarditis. These findings are compatible with the observation that, CTLA-4, PD-1, and PD-L1 pathways play a central role in the modulation of autoimmunity.Areas covered: In this paper, we start from examining the pathogenesis of cardiovascular adverse events from ICIs, and then we focus on risk factors and strategies to prevent and manage this cardiotoxicity.Expert opinion: There is a growing need for a multidisciplinary approach of ICI-associated cardiotoxicity, involving oncologists, cardiologists, and immunologists. Prevention and effective management of ICIs cardiotoxicity starts with an in-depth screening and surveillance strategies of high-risk patients, in order to improve early detection and appropriate management in a personalized approach.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giancarlo Marone
- Department of Public Health, Section of Hygiene, University of Naples Federico II, Naples, Italy.,Monaldi Hospital Pharmacy, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | | | - Floriana Morgillo
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy.,Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
34
|
Xiong W, Qi L, Jiang N, Zhao Q, Chen L, Jiang X, Li Y, Zhou Z, Shen J. Metformin Liposome-Mediated PD-L1 Downregulation for Amplifying the Photodynamic Immunotherapy Efficacy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:8026-8041. [PMID: 33577301 DOI: 10.1021/acsami.0c21743] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Photodynamic therapy (PDT) is a promising strategy for cancer treatment. It can not only generate reactive oxygen species (ROS) to cause the chemical damage of tumor cells in the presence of enough oxygen but also promote the antitumor immunity of T cells through enhancing the production of interferon γ (IFN-γ). However, one phenomenon is ignored so far that the enhanced production of IFN-γ caused by PDT may significantly increase the expression of programmed death-ligand 1 (PD-L1) on the tumor cell membrane and thus could inhibit the immune killing effects of T cells. Herein, we report the construction of a composite by loading metformin (Met) and IR775 into a clinically usable liposome as a two-in-one nanoplatform (IR775@Met@Lip) to solve this problem. The IR775@Met@Lip could reverse tumor hypoxia to enhance ROS production to elicit more chemical damage. Besides, the overexpression of PD-L1 by PDT was also effectively down-regulated. These therapeutic benefits including decreased PD-L1 expression, alleviated T cell exhaustion, and reversed tumor hypoxia successfully suppressed both the primary and abscopal tumor growth in bladder and colon cancers, respectively. Combining with its excellent biocompatibility, our results indicate that this IR775@Met@Lip system has great potential to become a highly effective cancer therapy modality.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lin Qi
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ning Jiang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Qi Zhao
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Lingxiao Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuan Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zaigang Zhou
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jianliang Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| |
Collapse
|
35
|
Ning B, Ren X, Hagiwara K, Takeoka S, Ito Y, Miyatake H. Development of a Non-IgG PD-1/PD-L1 Inhibitor by in Silico Mutagenesis and an In-Cell Protein-Protein Interaction Assay. ACS Chem Biol 2021; 16:316-323. [PMID: 33448787 DOI: 10.1021/acschembio.0c00817] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inhibiting the programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) axis by monoclonal antibodies (mAbs) is a successful cancer immunotherapy. However, mAb-based drugs have various disadvantages including high production costs and large molecular sizes, which motivated us to develop a smaller alternative drug. Since PD-L1 binds PD-1 with moderate affinity, a higher affinity PD-1 variant should serve as a competitive inhibitor of the wild-type PD-1/PD-L1 interaction. In this report, we conducted in silico point mutagenesis of PD-1 to identify potent PD-1 variants with a higher affinity toward PD-L1 and refined the in silico results using a luciferase-based in-cell protein-protein interaction (PPI) assay. As a result, a PD-1 variant was developed that had two mutated amino acids (T76Y, A132V), termed 2-PD-1. 2-PD-1 could bind with PD-L1 at a dissociation constant of 12.74 nM. Moreover, 2-PD-1 successfully inhibited the PD-1/PD-L1 interaction with a half maximal inhibitory concentration of 19.15 nM and reactivated the T cell with a half maximal effective concentration of 136.1 nM. These results show that in silico mutagenesis combined with an in-cell PPI assay verification strategy successfully prepared a non-IgG inhibitor of the PD-1/PD-L1 interaction.
Collapse
Affiliation(s)
- Boyang Ning
- Nano Medical Engineering Laboratory, RIKEN Cluster of Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Xueli Ren
- Nano Medical Engineering Laboratory, RIKEN Cluster of Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kyoji Hagiwara
- Nano Medical Engineering Laboratory, RIKEN Cluster of Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shinji Takeoka
- Nano Medical Engineering Laboratory, RIKEN Cluster of Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster of Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hideyuki Miyatake
- Nano Medical Engineering Laboratory, RIKEN Cluster of Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama city, Saitama 338-8570, Japan
| |
Collapse
|
36
|
Lim J, Lee J, Jung S, Kim WJ. Phenylboronic-acid-based nanocomplex as a feasible delivery platform of immune checkpoint inhibitor for potent cancer immunotherapy. J Control Release 2021; 330:1168-1177. [DOI: 10.1016/j.jconrel.2020.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022]
|
37
|
Jin KT, Chen B, Liu YY, Lan HUR, Yan JP. Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer. Cancer Cell Int 2021; 21:83. [PMID: 33522929 PMCID: PMC7851946 DOI: 10.1186/s12935-021-01763-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer deaths worldwide. Besides common therapeutic approaches, such as surgery, chemotherapy, and radiotherapy, novel therapeutic approaches, including immunotherapy, have been an advent in CRC treatment. The immunotherapy approaches try to elicit patients` immune responses against tumor cells to eradicate the tumor. Monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells are two branches of cancer immunotherapy. MAbs demonstrate the great ability to completely recognize cancer cell-surface receptors and blockade proliferative or inhibitory pathways. On the other hand, T cell activation by genetically engineered CAR receptor via the TCR/CD3 and costimulatory domains can induce potent immune responses against specific tumor-associated antigens (TAAs). Both of these approaches have beneficial anti-tumor effects on CRC. Herein, we review the different mAbs against various pathways and their applications in clinical trials, the different types of CAR-T cells, various specific CAR-T cells against TAAs, and their clinical use in CRC treatment.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - Bo Chen
- Department of Neurology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Yu-Yao Liu
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - H Uan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - Jie-Ping Yan
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, 310014, China.
| |
Collapse
|
38
|
Diagnosis and Differential Diagnosis of Neurological Adverse Events during Immune Checkpoint Inhibitor Therapy. JOURNAL OF ONCOLOGY 2021; 2020:8865054. [PMID: 33381178 PMCID: PMC7749779 DOI: 10.1155/2020/8865054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/13/2020] [Accepted: 11/21/2020] [Indexed: 01/05/2023]
Abstract
Therapy with immune checkpoint inhibitors (ICIs) has improved overall survival and cancer-related morbidity of cancer treatment even in cancer entities with poor prognosis. Since the approval of the first ICI, ipilimumab, for treatment of advanced melanoma by the Food and Drug Administration (FDA) in 2011, the spectrum of indications and approved ICIs has grown, rapidly. Up to now, seven different ICIs for more than 20 indications are available. However, their mechanisms of action can lead to immune-related adverse events (irAEs). In particular, neurological irAEs are clinically relevant. Although they are rare, an early and accurate diagnosis is challenging and neurological disease course and sequelae are potentially fatal. Between 08/2017 and 03/2020, 31 patients received ICI treatment at Hannover Medical School and presented with neurological adverse events (N-irAEs). Treated malignancies were metastatic melanoma, bronchial carcinoma, and urothelial cell carcinoma. All patients received comprehensive neurological diagnostics including clinical examination and magnetic resonance imaging (MRI). Cerebrospinal fluid (CSF) analysis was obtained in 21 patients and electroneurography was performed in 22 patients. Although N-irAEs were suspected in all 31 patients, 11 patients had other conditions leading to neurological symptoms including tumor metastases in seven patients and hemorrhagic or ischemic stroke in four patients. In the following, these patients are referred to as the differential diagnosis (DD) group. Patients with N-irAEs suffered from immune mediated neuropathy (9/20), myositis and/or myasthenic syndrome (6/20), or encephalitis/cerebellitis (5/20). Except for cell count, CSF results did not differ between the N-irAEs and the DD group. Symptoms related to N-irAEs are rather unspecific potentially mimicking other tumor-related symptoms such as metastases. Patients with malignancy are predominantly not treated by neurologists. Because of the complexity of neurological symptoms, detailed neurological investigations in specialized institutions are necessary in patients with new neurological symptoms and need to be critically discussed with treating oncologists.
Collapse
|
39
|
Esen E, Sergin I, Jesudason R, Himmels P, Webster JD, Zhang H, Xu M, Piskol R, McNamara E, Gould S, Capietto AH, Delamarre L, Walsh K, Ye W. MAP4K4 negatively regulates CD8 T cell-mediated antitumor and antiviral immunity. Sci Immunol 2020; 5:5/45/eaay2245. [PMID: 32220977 DOI: 10.1126/sciimmunol.aay2245] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/01/2019] [Accepted: 02/27/2020] [Indexed: 12/28/2022]
Abstract
During cytotoxic T cell activation, lymphocyte function-associated antigen-1 (LFA-1) engages its ligands on antigen-presenting cells (APCs) or target cells to enhance T cell priming or lytic activity. Inhibiting LFA-1 dampens T cell-dependent symptoms in inflammation, autoimmune diseases, and graft-versus-host disease. However, the therapeutic potential of augmenting LFA-1 function is less explored. Here, we show that genetic deletion or inhibition of mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) enhances LFA-1 activation on CD8 T cells and improves their adherence to APCs or LFA-1 ligand. In addition, loss of Map4k4 increases CD8 T cell priming, which culminates in enhanced antigen-dependent activation, proliferation, cytokine production, and cytotoxic activity, resulting in impaired tumor growth and improved response to viral infection. LFA-1 inhibition reverses these phenotypes. The ERM (ezrin, radixin, and moesin) proteins reportedly regulate T cell-APC conjugation, but the molecular regulator and effector of ERM proteins in T cells have not been defined. In this study, we demonstrate that the ERM proteins serve as mediators between MAP4K4 and LFA-1. Last, systematic analyses of many organs revealed that inducible whole-body deletion of Map4k4 in adult animals is tolerated under homeostatic conditions. Our results uncover MAP4K4 as a potential target to augment antitumor and antiviral immunity.
Collapse
Affiliation(s)
- Emel Esen
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Ismail Sergin
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Rajiv Jesudason
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Patricia Himmels
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Joshua D Webster
- Department of Research Pathology, Genentech, South San Francisco, CA, USA
| | - Hua Zhang
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Min Xu
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Robert Piskol
- Department of Bioinformatics, Genentech, South San Francisco, CA, USA
| | - Erin McNamara
- Department of Translational Oncology, Genentech, South San Francisco, CA, USA
| | - Stephen Gould
- Department of Translational Oncology, Genentech, South San Francisco, CA, USA
| | | | - Lélia Delamarre
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Kevin Walsh
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA.
| | - Weilan Ye
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
40
|
Guo J, Luo L, Wang Z, Hu N, Wang W, Xie F, Liang E, Yan X, Xiao J, Li S. Design, Synthesis, and Biological Evaluation of Linear Aliphatic Amine-Linked Triaryl Derivatives as Potent Small-Molecule Inhibitors of the Programmed Cell Death-1/Programmed Cell Death-Ligand 1 Interaction with Promising Antitumor Effects In Vivo. J Med Chem 2020; 63:13825-13850. [DOI: 10.1021/acs.jmedchem.0c01329] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jialin Guo
- National Engineering Research Center for the Emergency Drug, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and ToxicologyBeijing 100850, China
| | - Zhihong Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and ToxicologyBeijing 100850, China
| | - Naijing Hu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and ToxicologyBeijing 100850, China
| | - Wei Wang
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Erguang Liang
- National Engineering Research Center for the Emergency Drug, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xinlin Yan
- National Engineering Research Center for the Emergency Drug, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Junhai Xiao
- National Engineering Research Center for the Emergency Drug, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Song Li
- National Engineering Research Center for the Emergency Drug, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
41
|
Yun J, Yu G, Hu P, Chao Y, Li X, Chen X, Wei Q, Wang J. PD-1 expression is elevated in monocytes from hepatocellular carcinoma patients and contributes to CD8 T cell suppression. Immunol Res 2020; 68:436-444. [PMID: 32975728 DOI: 10.1007/s12026-020-09155-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022]
Abstract
It is recently shown that PD-1 expression by immune cells of the myeloid lineage contributes to the suppression of antitumor immunity. The expression of PD-1 by antigen-presenting cells in hepatocellular carcinoma (HCC), a malignancy with high intratumoral PD-L1 expression, remained understudied. Here, we found that circulating monocytes from HBV-associated HCC patients upregulated PD-1 in a severity-dependent manner. Monocyte stimulation using IFN-γ or high levels of IL-10 could slightly increase PD-1 expression, while LPS could markedly increase PD-1 expression. Interestingly, LPS in combination with IL-4 or IL-10 presented stronger stimulation of PD-1 expression than LPS in combination with IFN-γ or LPS alone. At resting state, PD-1+ monocytes presented comparable MHC-I and IL-12 expression and higher MHC-II, CD86, iNOS, arginase I, and IL-10 expression than PD-1- monocytes. Upon LPS stimulation, PD-1+ monocytes presented lower iNOS and higher arginase I and IL-10 expression than PD-1- monocytes, indicating an M2-polarization bias in PD-1+ monocytes. CD8 T cells following coculture with PD-1+ monocytes presented lower IFN-γ and lower TNF-α expression, together with lower proliferation capacity, than CD8 T cells following coculture with PD-1- monocytes, suggesting that PD-1+ monocytes were less capable of supporting CD8 T cell activation. Overall, these data indicated that PD-1 expression was elevated in monocytes from hepatocellular carcinoma patients. In addition, PD-1+ monocytes presented a preference toward M2 polarization and had a deficiency in supporting CD8 T cells.
Collapse
Affiliation(s)
- Jin Yun
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Genhua Yu
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pingping Hu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.,Department of Digital Medical Research Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yang Chao
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xingyu Li
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiaobo Chen
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qichun Wei
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Junfeng Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China. .,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China. .,Department of Digital Medical Research Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| |
Collapse
|
42
|
Oner G, Altintas S, Canturk Z, Tjalma W, Van Berckelaer C, Broeckx G, Zwaenepoel K, Tholhuijsen M, Verhoeven Y, Berneman Z, Peeters M, Pauwels P, van Dam PA. The immunologic aspects in hormone receptor positive breast cancer. Cancer Treat Res Commun 2020; 25:100207. [PMID: 32896829 DOI: 10.1016/j.ctarc.2020.100207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/04/2020] [Accepted: 07/05/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Although hormone receptor positive/HER2-negative (HR +/HER2-) breast cancer is the most diagnosed breast cancer type, the immunologic aspects HR positive breast cancer (BC) has been neglected until recently. The purpose of this paper is to review the current knowledge of the immune environment in HR positive BC and the potential use of immunotherapy in these patients. METHOD A computer-based literature research was carried out using PubMed, American Society of Clinical Oncology Annual Meeting (ASCO) and San Antonio Breast Cancer Symposium (SABCS). RESULTS The tumour microenvironment (TME), with infiltrating immune cells, plays an important role in HR positive BC. However, the effects of these immune cells are different in the luminal cancers compared to the other breast cancer types. Even though PD-1 and PD-L1 are less expressed in HR positive BC, pathological complete response (pCR) was more often seen after PD-1 inhibitor treatment in patients with an increased expression. The studies support the assertion that endocrine therapy has immunomodulatory effect. CONCLUSION The reviewed literature indicates that immune cells play an important role in HR positive BC. Considerably more research is needed to determine the real effect of the TME in this patient group.
Collapse
Affiliation(s)
- Gizem Oner
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Department of General Surgery, Kocaeli University, Kocaeli, Turkey.
| | - Sevilay Altintas
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Zafer Canturk
- Department of General Surgery, Kocaeli University, Kocaeli, Turkey
| | - Wiebren Tjalma
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | | | - Glenn Broeckx
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Karen Zwaenepoel
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Maria Tholhuijsen
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Yannick Verhoeven
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Zwi Berneman
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Department of Hematology, Antwerp University, Edegem, Belgium
| | - Marc Peeters
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Peter A van Dam
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
43
|
Wang Z, Huang L, Yan Y, El‐Zohry AM, Toffoletti A, Zhao J, Barbon A, Dick B, Mohammed OF, Han G. Elucidation of the Intersystem Crossing Mechanism in a Helical BODIPY for Low-Dose Photodynamic Therapy. Angew Chem Int Ed Engl 2020; 59:16114-16121. [PMID: 32449273 PMCID: PMC7540422 DOI: 10.1002/anie.202005269] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Indexed: 12/22/2022]
Abstract
Intersystem crossing (ISC) of triplet photosensitizers is a vital process for fundamental photochemistry and photodynamic therapy (PDT). Herein, we report the co-existence of efficient ISC and long triplet excited lifetime in a heavy atom-free bodipy helicene molecule. Via theoretical computation and time-resolved EPR spectroscopy, we confirmed that the ISC of the bodipy results from its twisted molecular structure and reduced symmetry. The twisted bodipy shows intense long wavelength absorption (ϵ=1.76×105 m-1 cm-1 at 630 nm), satisfactory triplet quantum yield (ΦT =52 %), and long-lived triplet state (τT =492 μs), leading to unprecedented performance as a triplet photosensitizer for PDT. Moreover, nanoparticles constructed with such helical bodipy show efficient PDT-mediated antitumor immunity amplification with an ultra-low dose (0.25 μg kg-1 ), which is several hundred times lower than that of the existing PDT reagents.
Collapse
Affiliation(s)
- Zhijia Wang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of Technology2 Ling Gong RoadDalian116024China
| | - Ling Huang
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts01605USA
| | - Yuxin Yan
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of Technology2 Ling Gong RoadDalian116024China
| | - Ahmed M. El‐Zohry
- Division of Physical Sciences and EngineeringKing Abdullah University of Science and Technology (KAUST)Thuwal23955-6900Kingdom of Saudi Arabia
| | - Antonio Toffoletti
- Dipartimento di Scienze ChimicheUniversità degli Studi di PadovaVia Marzolo 135121PadovaItaly
| | - Jianzhang Zhao
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of Technology2 Ling Gong RoadDalian116024China
| | - Antonio Barbon
- Dipartimento di Scienze ChimicheUniversità degli Studi di PadovaVia Marzolo 135121PadovaItaly
| | - Bernhard Dick
- Lehrstuhl für Physikalische ChemieInstitut für Physikalische und Theoretische ChemieUniversität RegensburgUniversitätsstr. 3193053RegensburgGermany
| | - Omar F. Mohammed
- Division of Physical Sciences and EngineeringKing Abdullah University of Science and Technology (KAUST)Thuwal23955-6900Kingdom of Saudi Arabia
| | - Gang Han
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts01605USA
| |
Collapse
|
44
|
Pascual T, Cejalvo JM, Oliveira M, Vidal M, Vega E, Ganau S, Julve A, Zamora E, Miranda I, Delgado A, Bermejo B, la Cruz-Merino LD, Juan M, Ferrero-Cafiero JM, Canes J, Gonzalez X, Villagrasa P, Prat A. SOLTI-1503 PROMETEO TRIAL: combination of talimogene laherparepvec with atezolizumab in early breast cancer. Future Oncol 2020; 16:1801-1813. [PMID: 32633563 DOI: 10.2217/fon-2020-0246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
New treatment strategies such as immune checkpoint inhibitors and oncolytic viruses are opening new possibilities in cancer therapy. Preliminary results in melanoma and other tumors showed that the combination of talimogene laherparepvec with an anti-PD-1/PD-L1 or anti-CTLA4 has greater efficacy than either therapy alone, without additional safety concerns beyond those expected for each agent. The presence of residual cancer after neoadjuvant chemotherapy in early breast cancer patients is an unmet medical need. SOLTI-1503 PROMETEO is a window of opportunity trial, which evaluates the combination of talimogene laherparepvec in combination with atezolizumab in women with operable HER2-negative breast cancer who present residual disease after neoadjuvant chemotherapy. The primary end point is the rate of residual cancer burden 0/1. Clinical Trial Registration: NCT03802604.
Collapse
Affiliation(s)
- Tomas Pascual
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Juan M Cejalvo
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Breast Cancer Biology Research Group, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Mafalda Oliveira
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Vidal
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Estela Vega
- Medical Oncology Department, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Sergi Ganau
- Radiology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Ana Julve
- Radiology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Esther Zamora
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ignacio Miranda
- Radiology Department, Breast Imaging Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ana Delgado
- Radiology Department, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Breast Cancer Biology Research Group, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Luis de la Cruz-Merino
- Medical Oncology Department, Hospital Universitario Virgen Macarena. Sevilla, Spain
- Medicine Department, Universidad de Sevilla, Sevilla, Spain
| | - Manel Juan
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Biomedicine Department, University of Barcelona, Barcelona, Spain
- Immunogenetics of the Autoinflammatory Response, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Jordi Canes
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Xavier Gonzalez
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department Hospital Universitari General de Catalunya, Sant Cugat del Vallès, Spain
| | | | - Aleix Prat
- Scientific Department, SOLTI Breast Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| |
Collapse
|
45
|
Keikha M, Ghazvini K, Eslami M, Yousefi B, Casseb J, Yousefi M, Karbalaei M. Molecular targeting of PD-1 signaling pathway as a novel therapeutic approach in HTLV-1 infection. Microb Pathog 2020; 144:104198. [PMID: 32283259 DOI: 10.1016/j.micpath.2020.104198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 02/08/2023]
Abstract
HTLV-1, the first human oncogenic retrovirus, is a type C retrovirus that belongs to the Deltaretrovirus genus. The HTLV-1 genome has 8.5 kbp length, and consists of major genes such as gag, pol, pro, env, and pX region. This retrovirus is considered as one of the most deadly infectious agent for peripheral-blood mononuclear cells (PBMC). The infection of HTLV-1 can lead to dangerous complications, such as infective dermatitis (ID), uveitis, arthritis, lymphadenitis, arthropathies, Sjögren's Syndrome (SS), and particularly HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) or Adult T-Cell Leukemia Lymphoma (ATLL). At the moment, Zidovudine (AZT) plus IFN-α is the only treatment available for HTLV-1 infections. Based on scientific studies, alongside the therapeutic regimens, intrinsic mechanisms also play a determinant role in reducing the signs of disease. Programmed cell death-1 (PD-1) signaling pathway, one of the most important checkpoints, has recently received interest, such as the development of a novel generation of anti-tumors. In the present study, we discuss the role of PD-1 signaling pathway in HTLV-1 infection as well as its application as a novel approach for treatment of HTLV-1 infections.
Collapse
Affiliation(s)
- Masoud Keikha
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Jorge Casseb
- Institute of Tropical Medicine of São Paulo/Laboratory of Dermatology and Immunodeficiencies, Department of Dermatology, University of São Paulo Medical School, São Paulo, SP, 01246-100, Brazil
| | - Masoud Yousefi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohsen Karbalaei
- Department of Microbiology and Virology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
46
|
Wang Z, Huang L, Yan Y, El‐Zohry AM, Toffoletti A, Zhao J, Barbon A, Dick B, Mohammed OF, Han G. Elucidation of the Intersystem Crossing Mechanism in a Helical BODIPY for Low‐Dose Photodynamic Therapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005269] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Zhijia Wang
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology 2 Ling Gong Road Dalian 116024 China
| | - Ling Huang
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester Massachusetts 01605 USA
| | - Yuxin Yan
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology 2 Ling Gong Road Dalian 116024 China
| | - Ahmed M. El‐Zohry
- Division of Physical Sciences and Engineering King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Kingdom of Saudi Arabia
| | - Antonio Toffoletti
- Dipartimento di Scienze Chimiche Università degli Studi di Padova Via Marzolo 1 35121 Padova Italy
| | - Jianzhang Zhao
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology 2 Ling Gong Road Dalian 116024 China
| | - Antonio Barbon
- Dipartimento di Scienze Chimiche Università degli Studi di Padova Via Marzolo 1 35121 Padova Italy
| | - Bernhard Dick
- Lehrstuhl für Physikalische Chemie Institut für Physikalische und Theoretische Chemie Universität Regensburg Universitätsstr. 31 93053 Regensburg Germany
| | - Omar F. Mohammed
- Division of Physical Sciences and Engineering King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Kingdom of Saudi Arabia
| | - Gang Han
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester Massachusetts 01605 USA
| |
Collapse
|
47
|
Jain S, Kumar S. Cancer immunotherapy: dawn of the death of cancer? Int Rev Immunol 2020; 39:1-18. [PMID: 32530336 DOI: 10.1080/08830185.2020.1775827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 05/11/2020] [Accepted: 05/16/2020] [Indexed: 02/08/2023]
Abstract
Cancer is one of the proficient evaders of the immune system which claims millions of lives every year. Developing therapeutics against cancer is extremely challenging as cancer involves aberrations in self, most of which are not detected by the immune system. Conventional therapeutics like chemotherapy, radiotherapy are not only toxic but they significantly lower the quality of life. Immunotherapy, which gained momentum in the 20th century, is emerging as one of the alternatives to the conventional therapies and is relatively less harmful but more costly. This review explores the modern advances in an array of such therapies and try to compare them along with a limited analysis of concerns associated with them.
Collapse
Affiliation(s)
- Sidhant Jain
- Department of Zoology, University of Delhi, Delhi, India
| | - Sahil Kumar
- Department of Pharmacology, Maulana Azad Medical College and Lok Nayak Hospital, New Delhi, India
| |
Collapse
|
48
|
Hendrix A, Yeo AE, Lejeune S, Seront E. Rare case of life-threatening thrombocytopenia occurring after radiotherapy in a patient treated with immune checkpoint inhibitor. BMJ Case Rep 2020; 13:13/6/e235249. [PMID: 32522726 DOI: 10.1136/bcr-2020-235249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) improve significantly outcome of patients with advanced renal cancer. Although immune-related adverse events involve frequently skin, digestive tract, lung, liver and endocrine organs, haematological toxicities are rare. We describe the case of a patient with metastatic renal cancer who was treated with nivolumab. Eight courses of nivolumab were administered without any toxicity; brain metastases were then diagnosed and treated with stereotactic radiotherapy. As the extra-cranial disease was stable, the ninth course of nivolumab was administered 5 days after the end of radiotherapy. One week later, he presented with rectal and nasal bleeding in a context of severe thrombocytopenia (1000/mm3). High dose of steroids and intravenous immunoglobulin reversed slowly the thrombocytopenia. This case highlights the possibility of life-threatening thrombocytopenia with ICIs. Interestingly, the close time relation with radiotherapy highlights a potential interaction, warranting a close follow-up of patients in this situation.
Collapse
Affiliation(s)
- Aurore Hendrix
- Medical Oncology, Hopital de Jolimont, Haine Saint Paul, Belgium
| | | | - Sarah Lejeune
- Medical Oncology, Hopital de Jolimont, Haine Saint Paul, Belgium
| | - Emmanuel Seront
- Medical Oncology, Hopital de Jolimont, Haine Saint Paul, Belgium
| |
Collapse
|
49
|
Wang J, Huang F, Jiang C, Chi P. Silencing Signal Transducer and Activator of Transcription 3 (STAT3) and Use of Anti-Programmed Cell Death-Ligand 1 (PD-L1) Antibody Induces Immune Response and Anti-Tumor Activity. Med Sci Monit 2020; 26:e915854. [PMID: 32343679 PMCID: PMC7201895 DOI: 10.12659/msm.915854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The treatment of cancer is still unable to meet the needs of patients and remains a huge challenge. This study investigated the immune response and anti-cancer effect of silencing STAT3 combined with the use of anti-PD-L1 antibody. MATERIAL AND METHODS Transfected CT26.WT cells were used to subcutaneously inoculate C57B/L6 mice, which were subsequently injected with anti-PD-L1 antibody. Treated mice were examined for tumor formation and inflammation using HE staining. Tumors were investigated for apoptosis using the TUNEL assay. The expression of STAT3, PD-L1, and C-met was studied immunohistochemistrially and by using PCR and Western blot analysis. RESULTS Four weeks after inoculation, tumors were observed in the inoculated mice. HE staining showed obvious inflammation in mice injected with cells that were silenced for STAT3 and injected with PD-L1 antibody. TUNEL assay showed low level of apoptosis in mice injected with cells silenced for STAT3 or injected with PD-L1 antibody, and higher level of apoptosis following combined treatment of STAT3 silencing and PD-L1 antibody injection. Immunohistochemistry, PCR, and Western blot analyses revealed that the expression of C-met, PD-L1, and STAT3 was significantly reduced in tumors following the combined treatment. Compared with treatment of STAT3 silencing or PD-L1 antibody injection, the combined treatment enhanced apoptosis. CONCLUSIONS Silencing STAT3 and PD-L1 antibody injection in combination increased apoptosis in tumor cells and thus offers better anti-cancer activity.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of General Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China (mainland)
| | - Fakun Huang
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China (mainland)
| | - Caiyun Jiang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China (mainland)
| | - Pan Chi
- Department of General Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China (mainland)
| |
Collapse
|
50
|
Kang DY, Sp N, Jo ES, Rugamba A, Hong DY, Lee HG, Yoo JS, Liu Q, Jang KJ, Yang YM. The Inhibitory Mechanisms of Tumor PD-L1 Expression by Natural Bioactive Gallic Acid in Non-Small-Cell Lung Cancer (NSCLC) Cells. Cancers (Basel) 2020; 12:E727. [PMID: 32204508 PMCID: PMC7140102 DOI: 10.3390/cancers12030727] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most common lung cancer subtype and accounts for more than 80% of all lung cancer cases. Epidermal growth factor receptor (EGFR) phosphorylation by binding growth factors such as EGF activates downstream prooncogenic signaling pathways including KRAS-ERK, JAK-STAT, and PI3K-AKT. These pathways promote the tumor progression of NSCLC by inducing uncontrolled cell cycle, proliferation, migration, and programmed death-ligand 1 (PD-L1) expression. New cytotoxic drugs have facilitated considerable progress in NSCLC treatment, but side effects are still a significant cause of mortality. Gallic acid (3,4,5-trihydroxybenzoic acid; GA) is a phenolic natural compound, isolated from plant derivatives, that has been reported to show anticancer effects. We demonstrated the tumor-suppressive effect of GA, which induced the decrease of PD-L1 expression through binding to EGFR in NSCLC. This binding inhibited the phosphorylation of EGFR, subsequently inducing the inhibition of PI3K and AKT phosphorylation, which triggered the activation of p53. The p53-dependent upregulation of miR-34a induced PD-L1 downregulation. Further, we revealed the combination effect of GA and anti-PD-1 monoclonal antibody in an NSCLC-cell and peripheral blood mononuclear-cell coculture system. We propose a novel therapeutic application of GA for immunotherapy and chemotherapy in NSCLC.
Collapse
Affiliation(s)
- Dong Young Kang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (D.Y.K.); (N.S.); (E.S.J.); (A.R.)
| | - Nipin Sp
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (D.Y.K.); (N.S.); (E.S.J.); (A.R.)
| | - Eun Seong Jo
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (D.Y.K.); (N.S.); (E.S.J.); (A.R.)
| | - Alexis Rugamba
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (D.Y.K.); (N.S.); (E.S.J.); (A.R.)
| | - Dae Young Hong
- Department of Emergency Medicine, School of Medicine, Konkuk University, Seoul 05029, Korea;
| | - Hong Ghi Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Seoul 05029, Korea;
| | - Ji-Seung Yoo
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-0808, Japan;
| | - Qing Liu
- Jilin Green Food Engineering Research Institute, Changchun 130000, Jilin, China;
| | - Kyoung-Jin Jang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (D.Y.K.); (N.S.); (E.S.J.); (A.R.)
| | - Young Mok Yang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (D.Y.K.); (N.S.); (E.S.J.); (A.R.)
| |
Collapse
|