1
|
Dübbel L, Göken-Riebisch A, Koch KW. Intracellular and exosomal localization of the negative checkpoint regulator VISTA in immune cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119966. [PMID: 40262722 DOI: 10.1016/j.bbamcr.2025.119966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
Strategies in immunotherapy often target the immunosuppressive environment of tumor cells. One route of therapeutic interference could involve negative checkpoint regulators of which V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA) has raised more interest recently. The protein is expressed on the surface of tumor cells, T-lymphocytes, and antigen-presenting cells (APCs), but its intracellular expression pattern has not been investigated yet. We examined the intracellular distribution of VISTA and its possible role in translocation processes by immunofluorescence and Western blots. We analyzed the expression and localization of VISTA in murine bone marrow-derived macrophages (BMDMs), human monocyte-derived macrophages, and human T lymphocytes (Jurkat). We obtained different cell fractions and organelles of various cell types and analyzed for the presence of VISTA. Monitoring a VISTA-GFP fusion construct in transfected cell lines HL-60 and THP-1 confirmed VISTA localization in these cell lines. All used cell lines showed the colocalization of VISTA and several vesicle markers together with VISTA staining along microtubule fibers. Additionally, we found VISTA in secreted exosomes and have the first hints for nucleic expression in all tested cell lines. Therefore, the storage of VISTA in vesicles and its potential presence in nuclei resembles two other well-described checkpoint regulators, CTLA-4 and PD-L1, respectively. We conclude that VISTA storage in vesicles enables a fast response to immunogenic stimuli, which needs to be considered for inhibitory experiments. The localization of VISTA in exosomes suggests a signaling function to facilitate cell-cell communication. Furthermore, the VISTA expression in the nucleus proposes a transcriptional role.
Collapse
Affiliation(s)
- Lena Dübbel
- Division of Biochemistry, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany; University Clinic of Gynaecology and Obstetrics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany.
| | - Anna Göken-Riebisch
- University Clinic of Gynaecology and Obstetrics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Division of Biochemistry, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| |
Collapse
|
2
|
Day D, Ganju V, Chung K, Si L, Mao L, Aghmesheh M, Hoyer R, Brewin K, Zeng S, Zhang M, Lu Q, Jiang C, Ren F, Zhu Y, Guo J. First-in-human phase I study of EMB-02, a bispecific antibody targeting PD-1 and LAG-3 in patients with advanced solid tumors. Br J Cancer 2025:10.1038/s41416-025-02990-x. [PMID: 40234667 DOI: 10.1038/s41416-025-02990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND EMB-02 is a symmetric bispecific antibody targeting programmed cell death protein-1 and lymphocyte-activation gene 3 simultaneously. Here, we present the first-in-human study results of EMB-02 in patients with advanced solid tumors. METHODS Patients were treated with intravenous infusions of EMB-02 at doses of 6-900 mg. The primary objective was to evaluate the safety and tolerability and to determine the maximum tolerated dose and/or recommended phase II dose(s). Secondary objectives included characterizing the pharmacokinetic (PK) profile, assessing preliminary antitumor activity and the immunogenicity. RESULTS A total of 47 patients were enrolled. All grade and grade 3/4 treatment-emergent and treatment related adverse events occurred in 97.9%, 48.9%, 68.1% and 12.8% patients, respectively. The objective response rate (ORR) was 6.4% and clinical benefit rate at 24 weeks (CBR-24) was 25.5% in overall population. The CBR-24 was 33.3% in checkpoint inhibitor (CPI)-naïve patients, and 15% in CPI-treated. No clear relationship was observed between the efficacy and PD-L1, LAG-3, or MHC II expression level. Doses 360 mg or higher resulted in sustained saturation of PD-1 receptors on circulating CD3 + T cells. CONCLUSIONS EMB-02 demonstrated a favorable safety profile and early efficacy signals in multiple solid tumors, warranting further development. (NCT04618393).
Collapse
Affiliation(s)
- Daphne Day
- Medical Oncology Department, Monash Health-Monash MedicalCentre, Clayton, VIC, Australia
| | - Vinod Ganju
- Oncology Department, Peninsula And Southeast Oncology, Frankston, VIC, Australia
| | - Ki Chung
- Department of Medicine, Prisma Health Cancer Institute, Greenville, SC, USA
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China
| | - Lili Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China
| | - Morteza Aghmesheh
- Medical Oncology Department, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Robert Hoyer
- Medical Oncology Department, UCHealth Memorial Hospital Central, Colorado Springs, CO, USA
| | - Kim Brewin
- Medical Oncology Department, Monash Health-Monash MedicalCentre, Clayton, VIC, Australia
| | - Shuqi Zeng
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Mingfei Zhang
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Qiaoyang Lu
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Chengjun Jiang
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Fang Ren
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Yonghong Zhu
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China.
| |
Collapse
|
3
|
Papageorgiou GI, Skouteris N, Grenzelia M, Maragkoudakis E, Eleftheriou K. Is it time to revisit the significance of PD-L1 expression in assisting our treatment decisions? Immunotherapy 2025; 17:223-227. [PMID: 40116402 PMCID: PMC12013450 DOI: 10.1080/1750743x.2025.2483152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Affiliation(s)
| | | | - Maria Grenzelia
- Radiation Oncology Center, Iaso General Clinic, Marousi, Greece
| | | | | |
Collapse
|
4
|
Alghazali T, Saleh RO, Uthirapathy S, Ballal S, Abullais SS, Kalia R, Arya R, Sharma R, Kumar A, Abdulamer RS. Rheumatoid arthritis unmasked: the power of B cell depletion therapy. Mol Biol Rep 2025; 52:254. [PMID: 39976856 DOI: 10.1007/s11033-025-10366-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/12/2025] [Indexed: 05/10/2025]
Abstract
Rheumatoid arthritis (RA) is an enduring autoimmune illness characterized by persistent inflammation and joint damage. Recent advancements in B cell depletion therapies (BCDTs) have provided new avenues for managing RA. This review article delves into the pathophysiology of RA, highlighting the pivotal role of B cells in disease progression. We explore the mechanisms underlying B cell depletion, focusing on monoclonal antibodies such as rituximab as well as innovative approaches like chimeric antigen receptor (CAR) T cell therapies. An in-depth analysis of clinical studies reveals the efficacy and limitations of these therapies, including success rates, side effects, and cost implications for patients. Despite promising outcomes, the incomplete depletion of B cells and associated risks underscore the need for further research. This review aims to provide a comprehensive understanding of BCDTs in RA, shed light on their potential and challenges, and guide future therapeutic strategies.
Collapse
Affiliation(s)
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shahabe Saquib Abullais
- Department of Periodontics and Community Dental Science, College of Dentistry, King Khalid University, Abha, Saudi Arabia
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges- Jhanjeri, Ajitgarh, Punjab, India
| | - Rsk Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, India
| | - Abhinav Kumar
- Refrigeration and Air-condition Department, Technical Engineering College, The Islamic University, Najaf, Iraq
- Department of Mechanical Engineering, Karpagam Academy of Higher Education, 641021, Coimbatore, India
| | - Resan Shakir Abdulamer
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
5
|
Nisar MF, Yan T, Cai Y, Wan C. Immuno-oncological Challenges and Chemoresistance in Veterinary Medicine: Probiotics as a New Strategic Tool. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10468-8. [PMID: 39954194 DOI: 10.1007/s12602-025-10468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Cancer has the highest death rates due to increased immuno-oncological (IO) challenges and chemoresistance caused by gut dysbiosis, whereas administration of probiotics may reverse these responses against anticancer therapies. Recently, immunotherapeutics have extensively been focused for significant advancements in pharmacological drug discovery and clinical outcomes. Mammals have intestinal epithelial cells, mucosal immune cells, and indigenous gut microbiota which may reshape immunotherapeutics efficacy. These include use of T-cell immune checkpoint inhibitors (ICPI), genetically engineered T-cells, tumor vaccines, monoclonal antibodies (mAbs), and anti-B- and T-cell antibodies. Immunotherapeutics for cancer treatment became popular in both veterinary and human health care systems due to their strong inhibitory actions against PD-1 and CTLA-4 to check tumorigenesis. IO issues in animals also need special attention, where caninized mAbs targeting CD-20 and CD-52 have been clinically used in treating canine B-cell and T-cell lymphomas, respectively. Probiotics appeared as strong immunotherapeutics that might be shaping the epigenetics of the organisms specifically in animal breeding practices for desired features, but limited literature regarding the immunomodulatory effects in humans and animals is available. In addition, considering the important role of probiotics in humans and veterinary medicine, a new perspective on the probiotic-mediated modulation of ncRNAs (miRNAs, lncRNAs, circRNAs) is also highlighted and would be a new therapeutic tool. This review provides insight into the cellular processes and pharmacological activities for treating veterinary infectious diseases and covers small drug molecules as ncRNA-modulators in veterinary medicine.
Collapse
Affiliation(s)
- Muhammad Farrukh Nisar
- Ministry of Education and Jiangxi Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Jiangxi Agricultural University, Nanchang, 330045, China
- Jiangxi Key Laboratory for Post-harvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, China
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, Pakistan
| | - Tingdong Yan
- School of Pharmacy, Nantong University, Nantong, 226001, China.
| | - Yi Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chunpeng Wan
- Jiangxi Key Laboratory for Post-harvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
6
|
Filho GVDM, Costa GJ, Martins MR, Torres LC. Circulating levels of galectin-9 are a potential biomarker of survival in advanced non-small-cell lung cancer. J Surg Oncol 2024. [PMID: 39047056 DOI: 10.1002/jso.27758] [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: 03/27/2024] [Accepted: 05/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The immune system is recognized to have therapeutic potential to destroy cancer cells. Soluble T-cell immunoglobulin mucin domain-3 (sTIM-3) and its ligand galectin 9 (Gal-9) cause suppression of cytokine production, cell cycle arrest and cell death. sTIM-3 and Gal-9 levels may have prognostic implications in non-small-cell lung cancer (NSCLC) patients. METHODS This prospective cohort study was performed at Instituto de Medicina Integral Prof. Fernando Figueira, Recife, Pernambuco, Brazil. Fifty-eight patients were diagnosed with advanced NSCLC from January 2019 to January 2020. RESULTS The age median was of 64.0 years. Soluble galectin-9 (sGal-9) levels in the smokers compared to nonsmoker patients (p < 0.0001). By using the receiver operating characteristic curve, we found that a baseline of 1694 pg/mL (cutoff). sGAL9 with specificity (72.2%), sensitivity (83.2%) and area under the curve = 0.8497 (p < 0.0004). Until 18.2 months, 46.8% and 72.9% were alive in the sGAL9low and sGAL9high groups, respectively (log-rank test; p = 0.02). The median survival was 15.9 months for sGAL9low (≤1694 pg/mL). CONCLUSION This study indicated an association of tobacco with the release of circulating sGal-9 levels and the accuracy of sGal-9 as a potential biomarker predictive of survival time in advanced NSCLC patients. Furthermore, sGal-9 has may be a potential therapeutic target in the advanced NSCLC.
Collapse
Affiliation(s)
- Guilherme Vieira de Mendonça Filho
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo City, São Paulo, Brazil
| | - Guilherme Jorge Costa
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo City, São Paulo, Brazil
| | - Mario R Martins
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Clinical Department, Hospital de Câncer de Pernambuco (HCP), Recife, Brazil
| | - Leuridan C Torres
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo City, São Paulo, Brazil
- Research Clinical Department, Hospital de Câncer de Pernambuco (HCP), Recife, Brazil
| |
Collapse
|
7
|
Pan S, Cai Q, Wei Y, Tang H, Zhang Y, Zhou W, Deng T, Mo W, Wang S, Wang C, Chen C. Increased co-expression of ICOS and PD-1 predicts poor overall survival in patients with acute myeloid leukemia. Immunobiology 2024; 229:152804. [PMID: 38615511 DOI: 10.1016/j.imbio.2024.152804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Inducible co-stimulatory factor (ICOS) has a dual role: activating cytotoxic T cells against tumors or exacerbating immunosuppression of regulatory T cells (Tregs) to participate in immune evasion. However, the correlation between ICOS and its co-expression with inhibitory immune checkpoints (IICs) and prognosis in acute myeloid leukemia (AML) is little known. METHODS The prognostic importance of ICOS and IICs in 62 bone marrow (BM) samples of de novo AML patients from our clinical center (GZFPH) was explored and then the RNA sequencing data of 155 AML patients from the Cancer Genome Atlas (TCGA) database was used for validation. RESULTS In both GZFPH and TCGA cohorts, high expression of ICOS was significantly associated with poor overall survival (OS) in patients with AML (P < 0.05). Importantly, co-expression of ICOS and PD-1, PD-L1, PD-L2, CTLA-4, and LAG-3 predicted poor OS in AML; among them, ICOS/PD-1 was the optimal combination of immune checkpoints (ICs). The co-expression of ICOS and PD-1 was correlated with poor OS in non-acute promyelocytic leukemia (non-APL) patients following chemotherapy. Additionally, ICOS/PD-1 was an independent OS-predicting factor (P < 0.05). Notably, a nomogram model was constructed by combining ICOS/PD-1, age, European Leukemia Net (ELN) risk stratification, and therapy to visually and personalized predict the 1-, 3-, and 5-year OS of patients with non-APL. CONCLUSION Increased expression of ICOS predicted poor outcomes, and ICOS/PD-1 was the optimal combination of ICs to predict outcomes in patients with AML, which might be a potential immune biomarker for designing novel AML therapy.
Collapse
Affiliation(s)
- Shiyi Pan
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Qinghua Cai
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Yiqiong Wei
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Haifeng Tang
- Department of Surgery, The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Wei Zhou
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Tingfen Deng
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| | - Caixia Wang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| | - Cunte Chen
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| |
Collapse
|
8
|
Sanchez‐Pupo RE, Finch GA, Johnston DE, Craig H, Abdo R, Barr K, Kerfoot S, Dagnino L, Penuela S. Global pannexin 1 deletion increases tumor-infiltrating lymphocytes in the BRAF/Pten mouse melanoma model. Mol Oncol 2024; 18:969-987. [PMID: 38327091 PMCID: PMC10994229 DOI: 10.1002/1878-0261.13596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/20/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
Immunotherapies for malignant melanoma seek to boost the anti-tumoral response of CD8+ T cells, but have a limited patient response rate, in part due to limited tumoral immune cell infiltration. Genetic or pharmacological inhibition of the pannexin 1 (PANX1) channel-forming protein is known to decrease melanoma cell tumorigenic properties in vitro and ex vivo. Here, we crossed Panx1 knockout (Panx1-/-) mice with the inducible melanoma model BrafCA, PtenloxP, Tyr::CreERT2 (BPC). We found that deleting the Panx1 gene in mice does not reduce BRAF(V600E)/Pten-driven primary tumor formation or improve survival. However, tumors in BPC-Panx1-/- mice exhibited a significant increase in the infiltration of CD8+ T lymphocytes, with no changes in the expression of early T-cell activation marker CD69, lymphocyte activation gene 3 protein (LAG-3) checkpoint receptor, or programmed cell death ligand-1 (PD-L1) in tumors when compared to the BPC-Panx1+/+ genotype. Our results suggest that, although Panx1 deletion does not overturn the aggressive BRAF/Pten-driven melanoma progression in vivo, it does increase the infiltration of effector immune T-cell populations in the tumor microenvironment. We propose that PANX1-targeted therapy could be explored as a strategy to increase tumor-infiltrating lymphocytes to boost anti-tumor immunity.
Collapse
Affiliation(s)
| | - Garth A. Finch
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | | | - Heather Craig
- Department of Microbiology and ImmunologyWestern UniversityLondonCanada
| | - Rober Abdo
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | - Kevin Barr
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | - Steven Kerfoot
- Department of Microbiology and ImmunologyWestern UniversityLondonCanada
| | - Lina Dagnino
- Department of Physiology and PharmacologyWestern UniversityLondonCanada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and DentistryWestern UniversityLondonCanada
| | - Silvia Penuela
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and DentistryWestern UniversityLondonCanada
| |
Collapse
|
9
|
Naini AA, Mayanti T, Maharani R, Harneti D, Nurlelasari, Farabi K, Fajriah S, Hilmayanti E, Kabayama K, Shimoyama A, Manabe Y, Fukase K, Jungsuttiwong S, Prescott TAK, Supratman U. Paraxylines A-G: Highly oxygenated preurianin-type limonoids with immunomodulatory TLR4 and cytotoxic activities from the stem bark of Dysoxylum parasiticum. PHYTOCHEMISTRY 2024; 220:114009. [PMID: 38342289 DOI: 10.1016/j.phytochem.2024.114009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Seven previously undescribed preurianin-type limonoids, namely paraxylines A-G, and three known analogs were isolated from stem bark of Dysoxylum parasiticum. The structures, including absolute configurations, were established through spectroscopic analyses, quantum chemical calculations using the density functional theory method, as well as the DP4+ algorithm. Paraxylines A-G were identified as the first preurianin-type with full substitution at C, D-rings, leading to the highly oxygenated seco-limonoids skeleton. The secreted alkaline phosphate assay against an engineered human and murine TLR4 of HEK-Blue cells was performed to evaluate the immune regulating effects. Among them, paraxyline B was found to be a remarkable TLR4 agonist whereas two analogs (toonapubesins A and B) were found to antagonise lipopolysaccharide stimulation of the TLR4 pathway. Paraxylines A and C-E acted either as agonists or antagonists depending on the origin of the TLR4 receptor (human or mouse). The effect of these selected compounds on the expression of pro-inflammatory cytokines TNF-α, IL-1α, IL-1β, and IL-6 of the NF-κB signaling pathway were examined in macrophage cell lines, revealing dose-dependent effects. Additionally, paraxylines A, C, D, and G also presented modest cytotoxic activity against MCF-7 and HeLa cell lines with IC50 values ranging from 23.1 to 43.5 μM.
Collapse
Affiliation(s)
- Al Arofatus Naini
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Tri Mayanti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Rani Maharani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Desi Harneti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Nurlelasari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Kindi Farabi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Sofa Fajriah
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center Complex - BRIN, Cibinong, 16911, Bogor, West Java, Indonesia
| | - Erina Hilmayanti
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Atsushi Shimoyama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Sirriporn Jungsuttiwong
- Department of Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | | | - Unang Supratman
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia.
| |
Collapse
|
10
|
Zhang M, Liu C, Li Y, Li H, Zhang W, Liu J, Wang L, Sun C. Galectin-9 in cancer therapy: from immune checkpoint ligand to promising therapeutic target. Front Cell Dev Biol 2024; 11:1332205. [PMID: 38264357 PMCID: PMC10803597 DOI: 10.3389/fcell.2023.1332205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Galectin-9 (Gal-9) is a vital member of the galectin family, functioning as a multi-subtype galactose lectin with diverse biological roles. Recent research has revealed that Gal-9's interaction with tumors is an independent factor that influences tumor progression. Furthermore, Gal-9 in the immune microenvironment cross-talks with tumor-associated immune cells, informing the clarification of Gal-9's identity as an immune checkpoint. A thorough investigation into Gal-9's role in various cancer types and its interaction with the immune microenvironment could yield novel strategies for subsequent targeted immunotherapy. This review focuses on the latest advances in understanding the direct and indirect cross-talk between Gal-9 and hematologic malignancies, in addition to solid tumors. In addition, we discuss the prospects of Gal-9 in tumor immunotherapy, including its cross-talk with the ligand TIM-3 and its potential in immune-combination therapy.
Collapse
Affiliation(s)
- Minpu Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Wenfeng Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jingyang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Liquan Wang
- Department of Thyroid and Breast Surgery, Weifang People’s Hospital, Weifang, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
11
|
Sun Y, Zhou X, Lucas E, Chen L, Zhang H, Chen H, Zhou F. Expression of B7-H3 and TIM-3 in gastric-type endocervical adenocarcinoma: prevalence, association with PD-L1 expression, and prognostic significance. J Pathol Clin Res 2024; 10:e345. [PMID: 37798754 PMCID: PMC10766062 DOI: 10.1002/cjp2.345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 10/07/2023]
Abstract
Gastric-type endocervical adenocarcinoma (GEA) is the second most common subtype of endocervical adenocarcinoma and has a poor prognosis. Anti-programmed death-1 and anti-programmed death-ligand 1 (PD-L1) inhibitors have emerged as a major treatment option for GEA; however, data on the expression of other immune checkpoints in GEA are limited. We analyzed the expression of T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and B7 homolog 3 protein (B7-H3) in 58 GEA and investigated their prognostic significance as well as association with PD-L1 expression and other known prognostic factors. Applying the tumor proportion score (TPS) with a cutoff of 1%, B7-H3 and TIM-3 were present in 48.3% and 17.2% of cases, respectively. Applying the combined positive score (CPS) with a cutoff of 1, TIM-3 expression was present in 70.7% of cases. Moreover, the expression of three checkpoints (B7-H3, TIM-3, and PD-L1) was incompletely overlapping. Patients with B7-H3 positive tumors (by TPS) or TIM-3 positive tumors (by TPS) had significantly worse recurrence-free survival (RFS) and overall survival (OS) (log-rank). Using CPS, patients with TIM-3 positive tumors showed significantly worse RFS (log-rank). Similarly, B7-H3 positivity (by TPS) and TIM-3 positivity (by TPS) were associated with worse RFS and OS in univariate analysis. TIM-3 positivity (by CPS) was associated with worse RFS in univariate analysis and the final Cox multivariate analysis. In conclusion, our results show that (1) B7-H3 and TIM-3 are frequently expressed in GEA and their expression overlaps incompletely with PD-L1; and (2) both B7-H3 and TIM-3 are independent negative prognostic markers in GEA.
Collapse
Affiliation(s)
- Yao Sun
- Department of PathologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
| | - Xin Zhou
- Department of PathologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
- Department of PathologyInternational Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Elena Lucas
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PathologyParkland HospitalDallasTXUSA
| | - Lili Chen
- Department of GynecologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
| | - Huijuan Zhang
- Department of PathologyInternational Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Hao Chen
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PathologyParkland HospitalDallasTXUSA
| | - Feng Zhou
- Department of PathologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
- Department of PathologyInternational Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
12
|
Ryapolova A, Minskaia E, Gasanov N, Moroz V, Krapivin B, Egorov AD, Laktyushkin V, Zhuravleva S, Nagornych M, Subcheva E, Malogolovkin A, Ivanov R, Karabelsky A. Development of Recombinant Oncolytic rVSV-mIL12-mGMCSF for Cancer Immunotherapy. Int J Mol Sci 2023; 25:211. [PMID: 38203382 PMCID: PMC10779112 DOI: 10.3390/ijms25010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Anti-cancer therapy based on oncolytic viruses (OVs) is a targeted approach that takes advantage of OVs' ability to selectively infect and replicate in tumor cells, activate the host immune response, and destroy malignant cells over healthy ones. Vesicular stomatitis virus (VSV) is known for its wide range of advantages: a lack of pre-existing immunity, a genome that is easily amenable to manipulation, and rapid growth to high titers in a broad range of cell lines, to name a few. VSV-induced tumor immunity can be enhanced by the delivery of immunostimulatory cytokines. The targeted cytokine delivery to tumors avoids the significant toxicity associated with systemic delivery while also boosting the immune response. To demonstrate this enhanced effect on both tumor growth and survival, a novel recombinant VSV (rVSV)-mIL12-mGMCSF, co-expressing mouse IL-12 (interleukin-12) and GM-CSF (granulocyte-macrophage colony-stimulating factor), was tested alongside rVSV-dM51-GFP (rVSV-GFP) that was injected intratumorally in a syngeneic in vivo C57BL/6 mouse model infused subcutaneously with B16-F10 melanoma cells. The pilot study tested the effect of two viral injections 4 days apart and demonstrated that treatment with the two rVSVs resulted in partial inhibition of tumor growth (TGII of around 40%) and an increased survival rate in animals from the treatment groups. The effect of the two VSVs on immune cell populations will be investigated in future in vivo studies with an optimized experimental design with multiple higher viral doses, as a lack of this information presents a limitation of this study.
Collapse
Affiliation(s)
- Anastasia Ryapolova
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Ekaterina Minskaia
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Nizami Gasanov
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Vasiliy Moroz
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Bogdan Krapivin
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Alexander D. Egorov
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Victor Laktyushkin
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Sofia Zhuravleva
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Maksim Nagornych
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Elena Subcheva
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Alexander Malogolovkin
- Department of Molecular Virology, First Moscow State Medical University (Sechenov University), 20 Pirogovskaya, 119991 Moscow, Russia;
| | - Roman Ivanov
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| | - Alexander Karabelsky
- Department of Gene Therapy, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (A.R.); (N.G.); (V.M.); (B.K.); (A.D.E.); (V.L.); (S.Z.); (M.N.); (E.S.); (R.I.); (A.K.)
| |
Collapse
|
13
|
Wang P, Zhang Q, Zhang H, Shao J, Zhang H, Wang Z. Molecular and clinical characterization of ICOS expression in breast cancer through large-scale transcriptome data. PLoS One 2023; 18:e0293469. [PMID: 38127899 PMCID: PMC10734928 DOI: 10.1371/journal.pone.0293469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/13/2023] [Indexed: 12/23/2023] Open
Abstract
ICOS (Inducible T Cell Costimulator), one of the co-stimulatory B7 superfamily members, was characterized as a co-stimulatory receptor for T-cell enhancement. However, the role of ICOS in breast cancer remains largely unknown. The present study systematically investigated the expression pattern and its relation to clinical characteristics and immunotherapy by integrating multiple clinical cohorts and large-scale gene expression data. This study included 2994 breast tumor samples with transcriptome data and matched clinical data. To make our findings more reliable, we set the TCGA cohort as the discovery set and the METABRIC cohort as the validation set. The expression of ICOS in breast cancer is strongly associated with major clinical and molecular characteristics. There is an association between higher ICOS expression and malignant subtypes and grades of tumors. In addition, gene ontology analysis based on genes significantly correlated with ICOS expression indicated that the expression of ICOS is mainly associated with immune responses and inflammation. We also observed strong correlations between ICOS and other promising immune-checkpoint molecules, including PD1, PDL1, CTLA4, and IDO1. Furthermore, we found that ICOS expression is associated with the response to anti-PDL1 immunotherapy and may serve as a biomarker for immunotherapy prediction. Our results indicated higher ICOS expression is significantly associated with favorable survival in triple-negative breast cancer (TNBC) patients, but not for all subtypes of breast cancer patients. In summary, ICOS correlates with higher malignant breast cancers, and it contributes to the regulation of the immune microenvironment of breast tumors, making it a potential biomarker and immunotherapy target.
Collapse
Affiliation(s)
- Peng Wang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Qin Zhang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Hengle Zhang
- Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jianqiang Shao
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Hui Zhang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Zunyi Wang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| |
Collapse
|
14
|
Song CH, Jeong M, In H, Kim JH, Lin CW, Han KH. Trends in the Development of Antibody-Drug Conjugates for Cancer Therapy. Antibodies (Basel) 2023; 12:72. [PMID: 37987250 PMCID: PMC10660735 DOI: 10.3390/antib12040072] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
In cancer treatment, the first-generation, cytotoxic drugs, though effective against cancer cells, also harmed healthy ones. The second-generation targeted cancer cells precisely to inhibit their growth. Enter the third-generation, consisting of immuno-oncology drugs, designed to combat drug resistance and bolster the immune system's defenses. These advanced therapies operate by obstructing the uncontrolled growth and spread of cancer cells through the body, ultimately eliminating them effectively. Within the arsenal of cancer treatment, monoclonal antibodies offer several advantages, including inducing cancer cell apoptosis, precise targeting, prolonged presence in the body, and minimal side effects. A recent development in cancer therapy is Antibody-Drug Conjugates (ADCs), initially developed in the mid-20th century. The second generation of ADCs addressed this issue through innovative antibody modification techniques, such as DAR regulation, amino acid substitutions, incorporation of non-natural amino acids, and enzymatic drug attachment. Currently, a third generation of ADCs is in development. This study presents an overview of 12 available ADCs, reviews 71 recent research papers, and analyzes 128 clinical trial reports. The overarching objective is to gain insights into the prevailing trends in ADC research and development, with a particular focus on emerging frontiers like potential targets, linkers, and drug payloads within the realm of cancer treatment.
Collapse
Affiliation(s)
- Chi Hun Song
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Minchan Jeong
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Hyukmin In
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Ji Hoe Kim
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Chih-Wei Lin
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406, Taiwan;
| | - Kyung Ho Han
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| |
Collapse
|
15
|
Ma Z, Han H, Zhou Z, Wang S, Liang F, Wang L, Ji H, Yang Y, Chen J. Machine learning-based establishment and validation of age-related patterns for predicting prognosis in non-small cell lung cancer within the context of the tumor microenvironment. IUBMB Life 2023; 75:941-956. [PMID: 37548145 DOI: 10.1002/iub.2768] [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: 05/01/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023]
Abstract
Lung cancer (LC) is a leading cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) accounting for over 80% of cases. The impact of aging on clinical outcomes in NSCLC remains poorly understood, particularly with respect to the immune response. In this study, we explored the effects of aging on NSCLC using 307 genes associated with human aging from the Human Ageing Genomic Resources. We identified 53 aging-associated genes that significantly correlate with overall survival of NSCLC patients, including the clinically validated gene BUB1B. Furthermore, we developed an aging-associated enrichment score to categorize patients based on their aging subtypes and evaluated their prognostic and therapeutic response values in LC. Our analyses yielded two aging-associated subtypes with unique profiles in the tumor microenvironment, demonstrating varying responses to immunotherapy. Consensus clustering based on transcriptome profiles provided insights into the effects of aging on NSCLC and highlighted the potential of personalized therapeutic approaches tailored to aging subtypes. Our findings provide a new target and theoretical support for personalized therapeutic approaches in patients with NSCLC, offering insights into the potential impact of aging on cancer outcomes.
Collapse
Affiliation(s)
- Zeming Ma
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Haibo Han
- Department of Clinical Laboratory, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhiwei Zhou
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Shijie Wang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Fan Liang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
- Department of Clinical Laboratory, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Liang Wang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Hong Ji
- Department of Clinical Laboratory, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yue Yang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jinfeng Chen
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
16
|
Mohamed DI, Abo Nahas HH, Elshaer AM, El-Waseef DAEDA, El-Kharashi OA, Mohamed SMY, Sabry YG, Almaimani RA, Almasmoum HA, Altamimi AS, Ibrahim IAA, Alshawwa SZ, Jaremko M, Emwas AH, Saied EM. Unveiling the interplay between NSAID-induced dysbiosis and autoimmune liver disease in children: insights into the hidden gateway to autism spectrum disorders. Evidence from ex vivo, in vivo, and clinical studies. Front Cell Neurosci 2023; 17:1268126. [PMID: 38026692 PMCID: PMC10644687 DOI: 10.3389/fncel.2023.1268126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/28/2023] [Indexed: 12/01/2023] Open
Abstract
Autism spectrum disorders (ASD) represent a diverse group of neuropsychiatric conditions, and recent evidence has suggested a connection between ASD and microbial dysbiosis. Immune and gastrointestinal dysfunction are associated with dysbiosis, and there are indications that modulating the microbiota could improve ASD-related behaviors. Additionally, recent findings highlighted the significant impact of microbiota on the development of autoimmune liver diseases, and the occurrence of autoimmune liver disease in children with ASD is noteworthy. In the present study, we conducted both an in vivo study and a clinical study to explore the relationship between indomethacin-induced dysbiosis, autoimmune hepatitis (AIH), and the development of ASD. Our results revealed that indomethacin administration induced intestinal dysbiosis and bacterial translocation, confirmed by microbiological analysis showing positive bacterial translocation in blood cultures. Furthermore, indomethacin administration led to disturbed intestinal permeability, evidenced by the activation of the NLRP3 inflammasomes pathway and elevation of downstream biomarkers (TLR4, IL18, caspase 1). The histological analysis supported these findings, showing widened intestinal tight junctions, decreased mucosal thickness, inflammatory cell infiltrates, and collagen deposition. Additionally, the disturbance of intestinal permeability was associated with immune activation in liver tissue and the development of AIH, as indicated by altered liver function, elevated ASMA and ANA in serum, and histological markers of autoimmune hepatitis. These results indicate that NSAID-induced intestinal dysbiosis and AIH are robust triggers for ASD existence. These findings were further confirmed by conducting a clinical study that involved children with ASD, autoimmune hepatitis (AIH), and a history of NSAID intake. Children exposed to NSAIDs in early life and complicated by dysbiosis and AIH exhibited elevated serum levels of NLRP3, IL18, liver enzymes, ASMA, ANA, JAK1, and IL6. Further, the correlation analysis demonstrated a positive relationship between the measured parameters and the severity of ASD. Our findings suggest a potential link between NSAIDs, dysbiosis-induced AIH, and the development of ASD. The identified markers hold promise as indicators for early diagnosis and prognosis of ASD. This research highlights the importance of maintaining healthy gut microbiota and supports the necessity for further investigation into the role of dysbiosis and AIH in the etiology of ASD.
Collapse
Affiliation(s)
- Doaa I. Mohamed
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Asmaa M. Elshaer
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Omnyah A. El-Kharashi
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Soha M. Y. Mohamed
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yasmine Gamal Sabry
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Riyad A. Almaimani
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hussain A. Almasmoum
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulmalik S. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Samar Z. Alshawwa
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative and Red Sea Research Center, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Abdul-Hamid Emwas
- Advanced Nanofabrication Imaging and Characterization Center, King Abdullah University of Science and Technology, Core Labs, Thuwal, Saudi Arabia
| | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| |
Collapse
|
17
|
Guo X, Yu S, Ren X, Li L. Immune checkpoints represent a promising breakthrough in targeted therapy and prognosis of myelodysplastic syndrome. Heliyon 2023; 9:e19222. [PMID: 37810157 PMCID: PMC10558320 DOI: 10.1016/j.heliyon.2023.e19222] [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: 11/11/2022] [Revised: 06/27/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a hematological malignancy of undetermined etiology, possibly linked to chromosomal structural alterations, genetic mutations, presentation and carcinogenicity of variant antigens on cell surface, and the generation of pro-inflammatory microenvironment in the bone marrow. Current drugs are unable to cure this disease, and therefore, decreasing the survival and proliferation of malignant cells to delay disease progression and extend the survival time of patients becomes the primary approach to management. In recent years, the immune system has received increasing attention for its potential role in the occurrence and development of MDS, leading to the emergence of immunoregulation as a viable treatment option. The current review provides a brief overview of pathogenesis of MDS and current treatment principles. In the meantime, the significance of immune proteins in treatment and prognosis of MDS is also discussed.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Shunjie Yu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Xiaotong Ren
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| |
Collapse
|
18
|
Herzog TJ, Hays JL, Barlin JN, Buscema J, Cloven NG, Kong LR, Tyagi NK, Lanneau GS, Long BJ, Marsh RL, Seward SM, Starks DC, Welch S, Moore KN, Konstantinopoulos PA, Gilbert L, Monk BJ, O'Malley DM, Chen X, Dalal R, Coleman RL, Sehouli J. ARTISTRY-7: phase III trial of nemvaleukin alfa plus pembrolizumab vs chemotherapy for platinum-resistant ovarian cancer. Future Oncol 2023; 19:1577-1591. [PMID: 37334673 DOI: 10.2217/fon-2023-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023] Open
Abstract
Standard single-agent nonplatinum chemotherapy provides only modest benefit in a small proportion of patients with platinum-resistant/-refractory ovarian cancer, with objective response rates of 6-20% and progression-free survival of ≈3-4 months. Nemvaleukin alfa (nemvaleukin, ALKS 4230) is a novel cytokine designed to capture and expand the therapeutic potential of high-dose interleukin-2 (IL-2) while mitigating its associated toxicity issues. Nemvaleukin preferentially activates cytotoxic CD8+ T cells and natural killer cells with minimal, non-dose-dependent effects on CD4+ regulatory T cells. The global, randomized, open-label, phase III ARTISTRY-7 trial will compare efficacy and safety of nemvaleukin plus pembrolizumab with chemotherapy in patients with platinum-resistant ovarian cancer. The primary end point is investigator-assessed progression-free survival. Clinical Trial Registration: GOG-3063; ENGOT-OV68; NCT05092360 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Thomas J Herzog
- University of Cincinnati Cancer Center, UC College of Medicine, Cincinnati, OH 45267, USA
| | - John L Hays
- Wexner Medical Center & James Cancer Hospital, Ohio State University, Columbus, OH 43210, USA
| | | | | | - Noelle G Cloven
- Texas Oncology - Fort Worth Cancer Center, Fort Worth, TX 76104, USA
| | - Lynn R Kong
- Ventura County Hematology Oncology Specialists, Oxnard, CA 93930, USA
| | | | | | - Beverly J Long
- Sarasota Memorial Healthcare System, Sarasota, FL 34239, USA
| | | | | | | | - Stephen Welch
- London Health Sciences Centre, London, ON, N6A 5A5, Canada
| | - Kathleen N Moore
- College of Medicine, University of Oklahoma, Oklahoma City, OK 73117, USA
| | | | - Lucy Gilbert
- McGill University Health Centre, Women's Health Research Unit, Montréal, QC, H3H 2R9, Canada
| | - Bradley J Monk
- HonorHealth Research Institute, University of Arizona College of Medicine, Creighton University School of Medicine, Phoenix, AZ 85012, USA
| | - David M O'Malley
- Wexner Medical Center & James Cancer Hospital, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | - Jalid Sehouli
- Charité Universitaetsmedizin Berlin Charité Campus Virchow-Klinikum, Berlin, 11017, Germany
| |
Collapse
|
19
|
Ibrahim R, Saleh K, Chahine C, Khoury R, Khalife N, Cesne AL. LAG-3 Inhibitors: Novel Immune Checkpoint Inhibitors Changing the Landscape of Immunotherapy. Biomedicines 2023; 11:1878. [PMID: 37509517 PMCID: PMC10377063 DOI: 10.3390/biomedicines11071878] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
One of the most important steps forward in the management of cancer was the discovery of immunotherapy. It has become an essential pillar in the treatment paradigm of cancer patients. Unfortunately, despite the various options presented with immune checkpoint inhibitors (ICIs), the benefit is still limited to select patients and the vast majority of these patients gain either minimal benefit or eventually progress, leaving an unmet need for the development of novel therapeutic agents and strategies. Lymphocyte activation gene-3 (LAG-3), an immune checkpoint receptor protein, is a molecule found on the surface of activated T-cells. It plays a major role in negatively regulating T-cell function thereby providing tumors with an immune escape in the tumor microenvironment (TME). Given its importance in regulating the immune system, LAG-3 has been considered as a promising target in oncology and precision medicine. To date, two LAG-3-directed agents (eftilagimod alpha and relatlimab) have been approved in combination with programmed death-1 (PD-1) inhibitors in the setting of advanced solid tumors. In this review, we discuss the structure of LAG-3, its mechanism of action, and its interaction with its ligands. We also shed light on the emerging treatments targeting LAG-3 for the treatment of solid tumors.
Collapse
Affiliation(s)
- Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Nadine Khalife
- Department of head and neck Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| |
Collapse
|
20
|
Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023; 8:156. [PMID: 37041165 PMCID: PMC10090134 DOI: 10.1038/s41392-023-01407-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023] Open
Abstract
Oncolytic viruses (OVs) have attracted growing awareness in the twenty-first century, as they are generally considered to have direct oncolysis and cancer immune effects. With the progress in genetic engineering technology, OVs have been adopted as versatile platforms for developing novel antitumor strategies, used alone or in combination with other therapies. Recent studies have yielded eye-catching results that delineate the promising clinical outcomes that OVs would bring about in the future. In this review, we summarized the basic principles of OVs in terms of their classifications, as well as the recent advances in OV-modification strategies based on their characteristics, biofunctions, and cancer hallmarks. Candidate OVs are expected to be designed as "qualified soldiers" first by improving target fidelity and safety, and then equipped with "cold weapons" for a proper cytocidal effect, "hot weapons" capable of activating cancer immunotherapy, or "auxiliary weapons" by harnessing tactics such as anti-angiogenesis, reversed metabolic reprogramming and decomposing extracellular matrix around tumors. Combinations with other cancer therapeutic agents have also been elaborated to show encouraging antitumor effects. Robust results from clinical trials using OV as a treatment congruously suggested its significance in future application directions and challenges in developing OVs as novel weapons for tactical decisions in cancer treatment.
Collapse
Affiliation(s)
- Danni Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Tomela K, Pietrzak B, Galus Ł, Mackiewicz J, Schmidt M, Mackiewicz AA, Kaczmarek M. Myeloid-Derived Suppressor Cells (MDSC) in Melanoma Patients Treated with Anti-PD-1 Immunotherapy. Cells 2023; 12:cells12050789. [PMID: 36899926 PMCID: PMC10000540 DOI: 10.3390/cells12050789] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a subset of immature myeloid cells with suppressive activity well described in the context of cancer. They inhibit anti-tumour immunity, promote metastasis formation and can lead to immune therapy resistance. In a retrospective study, blood probes of 46 advanced melanoma patients were analysed before the first administration of anti-PD-1 immunotherapy and in the third month of treatment for MDSC, immature monocytic (ImMC), monocytic MDSC (MoMDSC) and granulocytic MDSC (GrMDSC) by multi-channel flow cytometry. Cell frequencies were correlated with response to immunotherapy, progression-free survival (PFS) and lactate dehydrogenase (LDH) serum level. Responders to anti-PD-1 therapy had higher MoMDSC levels (4.1 ± 1.2%) compared to non-responders (3.0 ± 1.2%) (p = 0.0333) before the first administration of anti-PD-1. No significant changes in MDSCs frequencies were observed in the groups of patients before and in the third month of therapy. The cut-off values of MDSCs, MoMDSCs, GrMDSCs and ImMCs for favourable 2- and 3-year PFS were established. Elevated LDH level is a negative prognostic factor of response to the treatment and is related to an elevated ratio of GrMDSCs and ImMCs level compared to patients' LDH level below the cut-off. Our data may provide a new perspective for more careful consideration of MDSCs, and specially MoMDSCs, as a tool for monitoring the immune status of melanoma patients. Changes in MDSC levels may have a potential prognostic value, however a correlation with other parameters must be established.
Collapse
Affiliation(s)
- Katarzyna Tomela
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Correspondence:
| | - Bernadeta Pietrzak
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland
| | - Łukasz Galus
- Department of Medical and Experimental Oncology, Institute of Oncology, University of Medical Sciences, 60-355 Poznan, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Institute of Oncology, University of Medical Sciences, 60-355 Poznan, Poland
| | - Marcin Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland
| | - Andrzej Adam Mackiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
22
|
Zhang Y, Yang R, Xu C, Zhang Y, Deng M, Wu D, Tang F, Liu X, Han Y, Zhan Y, Miao J. Risk stratification and molecular heterogeneity of endometrial cancer and expression profile of TIM-3: A retrospective cohort study. Gynecol Oncol 2023; 170:210-220. [PMID: 36709662 DOI: 10.1016/j.ygyno.2023.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/07/2023] [Accepted: 01/18/2023] [Indexed: 01/29/2023]
Abstract
OBJECTIVE The present study aimed to implement ProMisE classification and risk grouping on a retrospective cohort of 628 patients with endometrial cancer (EC) and determine the molecular heterogeneity across subtypes and subgroups, as well as to investigate the potential beneficiary for TIM-3 checkpoint inhibition in ECs. METHODS Protein expressions of p53, MMR, TIM-3 and CD8 were measured by immunohistochemistry, and massively parallel sequencing was conducted for 128 cancer-related genes. Patients were categorized into four ProMisE subtypes: MMR-deficient (MMRd), POLE-ultramutated (POLEmut), p53-wild type (p53wt), and p53-abnormal (p53abn), and were subjected to risk classification. RESULTS 43 (6.9%) patients belonged to POLEmut, 118 (18.8%) to MMRd, 69 (11%) to p53abn, and 398 (63.3%) to p53wt. Compared to the 2016 stratification system, the 2021 ESGO/ESTRO/ESP risk stratification integrated with molecular classification revealed that 11 patients (11/628, 1.8%) were upgraded due to the p53abn signature, whereas 23 patients (23/628, 3.7%) were downgraded due to the POLEmut signature. JAK1 and RAD50 mutations showed higher frequencies in patients with aggressive phenotypes. RAD51B mutation was significantly related to poor RFS of the p53wt subtype but not of the other three molecular subgroups. TIM-3 expression was detected in 30.9% immune cells (ICs) and 29.0% tumor cells (TCs) in ECs, respectively. It was frequently expressed in POLEmut and MMRd ECs as compared to that in the other two molecular subtypes in TCs and ICs. CONCLUSIONS Our study revealed the molecular heterogeneity across subtypes and subgroups. The new risk stratification system changed the risk grouping of some patients due to the integration of molecular features. RAD51B mutation can further stratify the recurrence risk in the p53wt subtype. Patients with MMRd or POLEmut may benefit most from immunotherapy against TIM-3.
Collapse
Affiliation(s)
- Yubo Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Ruiye Yang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chunyu Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanqin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Mengqi Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Di Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fan Tang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xinyu Liu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yiding Han
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yang Zhan
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jinwei Miao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China.
| |
Collapse
|
23
|
A systematic review and meta-analysis of immune checkpoint therapy in relapsed or refractory non-Hodgkin lymphoma; a friend or foe? Transl Oncol 2023; 30:101636. [PMID: 36773442 PMCID: PMC9941575 DOI: 10.1016/j.tranon.2023.101636] [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: 11/27/2022] [Revised: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Over the last decades, a revolution has occurred in oncology with the development of immune checkpoint inhibitors (ICIs). Following tremendous successes in solid tumors, interest has risen to explore these inhibitors in hematologic malignancies; while Hodgkin's lymphoma (HL) has shown overwhelming achievements, available data on different types of non-Hodgkin's lymphoma (NHL) vary considerably. To the best of our knowledge, no meta-analysis has assessed the efficacy and safety of ICI therapy in relapsed or refractory NHL patients. Meta-analysis of the included studies (n = 29) indicated PD-1 may probably be the more attractive ICI target rather than PD-L1 and CTLA-4 in NHL patients. Also, there is a plausible correlation between NHL subtypes and response to ICI therapy. While MF, ENKTL, RT, and PMBCL showed promising responses to ICI monotherapy, neither FL nor DLBCL had satisfactory responses; further necessitating novel strategies such as the application of ICIs in combination with other treatment strategies. Notably, among different combinations, BTK inhibitors showed an obvious improvement as compared to ICI monotherapy in both FL and DLBCL, however, the best results were obtained when ICI was combined with anti-CD20 monoclonal antibodies. Finally, while most NHL patients who received ICI treatment have experienced mild AEs, larger trials with long-term follow-up are required to confirm the safety, as well as the efficacy, of ICI therapy in NHL patients.
Collapse
|
24
|
From the Catastrophic Objective Irreproducibility of Cancer Research and Unavoidable Failures of Molecular Targeted Therapies to the Sparkling Hope of Supramolecular Targeted Strategies. Int J Mol Sci 2023; 24:ijms24032796. [PMID: 36769134 PMCID: PMC9917659 DOI: 10.3390/ijms24032796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
The unprecedented non-reproducibility of the results published in the field of cancer research has recently come under the spotlight. In this short review, we try to highlight some general principles in the organization and evolution of cancerous tumors, which objectively lead to their enormous variability and, consequently, the irreproducibility of the results of their investigation. This heterogeneity is also extremely unfavorable for the effective use of molecularly targeted medicine. Against the seemingly comprehensive background of this heterogeneity, we single out two supramolecular characteristics common to all tumors: the clustered nature of tumor interactions with their microenvironment and the formation of biomolecular condensates with tumor-specific distinctive features. We suggest that these features can form the basis of strategies for tumor-specific supramolecular targeted therapies.
Collapse
|
25
|
Tang G, Liu J, Qi L, Li Y. The evolving role of checkpoint inhibitors in the treatment of urothelial carcinoma. Br J Clin Pharmacol 2023; 89:93-113. [PMID: 35997657 DOI: 10.1111/bcp.15504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Abstract
The most prevalent pathological subtype of bladder and upper urinary tract malignancy is urothelial carcinoma (UC). Traditional therapies mainly include surgical resection, chemotherapy and radiotherapy. Checkpoint inhibitors, which are monoclonal antibodies developed to specifically target immune checkpoint molecules, have recently emerged as potential treatment options for UC patients, especially those targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1). However, anti-PD-1/PD-L1 therapy does not work for a considerable number of UC patients. Current antitumour immunotherapy research hotspots include seeking biomarkers that might predict therapeutic effects and exploring novel immune checkpoint molecules crucial for the antitumour immune response. Hence, we will recapitulate the latest preclinical and clinical trials of 5 PD-1/PD-L1 inhibitors, 1 cytotoxic T-lymphocyte-associated protein 4 inhibitor and combination therapies for UC treatment, including combined immunotherapy and immunotherapy with chemotherapy or radiotherapy. We will also summarize other potential immune checkpoint molecules found in ongoing UC studies. Moreover, we will highlight the role of biomarkers linked with the oncological efficacy of anti-PD-1/PD-L1 immunotherapy and address the mechanisms of immunotherapy drug resistance in UC, with the hope of providing more systematic guidance for its application and development.
Collapse
Affiliation(s)
- Guyu Tang
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Jing Liu
- Department of Oncology, Xiangya Hospital of Central South University, Changsha, China
| | - Lin Qi
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Yuan Li
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
26
|
Ji C, He Y, Wang Y. Identification of necroptosis subtypes and development of necroptosis-related risk score model for in ovarian cancer. Front Genet 2022; 13:1043870. [PMID: 36568363 PMCID: PMC9773578 DOI: 10.3389/fgene.2022.1043870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Background: ith the ongoing development of targeted therapy, non-apoptotic cell death, including necroptosis, has become a popular topic in the field of prevention and treatment. The purpose of this study was to explore the effect of necroptosis-related genes (NRGs) on the classification of ovarian cancer (OV) subtypes and to develop a necroptosis-related risk score (NRRS) classification system. Methods: 74 NRGs were obtained from the published studies, and univariate COX regression analysis was carried out between them and OV survival. Consensus clustering analysis was performed on OV samples according to the expression of NRGs related to prognosis. Furthermore, the NRRS model was developed by combining Weighted Gene Co-Expression Network Analysis (WGCNA) with least absolute shrinkage and selection operator (Lasso)-penalized Cox regression and multivariate Cox regression analysis. And the decision tree model was constructed based on the principle of random forest screening factors principle. Results: According to the post-related NRGs, OV was divided into two necroptosis subtypes. Compared with Cluster 1 (C1), the overall survival (OS) of Cluster 2 (C2) was significantly shorter, stromal score and immune score, the infiltration level of tumor associated immune cells and the expression of 20 immune checkpoints were significantly higher. WGCNA identified the blue module most related to necroptosis subtype, and 12 genes in the module were used to construct NRRS. NRRS was an independent prognostic variable of OV. The OS of samples with lower NRRS was significantly longer, and tumor mutation burden and homologous recombination defect were more obvious. Conclusion: This study showed that necroptosis plays an important role in the classification, prognosis, immune infiltration and biological characteristics of OV subtypes. The evaluation of tumor necroptosis may provide a new perspective for OV treatment.
Collapse
|
27
|
Chen H, Molberg K, Carrick K, Niu S, Rivera Colon G, Gwin K, Lewis C, Zheng W, Castrillon DH, Lucas E. Prevalence and prognostic significance of PD-L1, TIM-3 and B7-H3 expression in endometrial serous carcinoma. Mod Pathol 2022; 35:1955-1965. [PMID: 35804040 DOI: 10.1038/s41379-022-01131-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/24/2022]
Abstract
Endometrial serous carcinoma (ESC) is an aggressive type of endometrial carcinoma with a poor prognosis. Immune checkpoint blockade has evolved as a novel treatment option for endometrial cancers; however, data on expression of immune checkpoints that may be potential targets for immunotherapy in ESC are limited. We analyzed the prevalence and prognostic significance of PD-L1, TIM-3 and B7-H3 immune checkpoints in 99 ESC and evaluated their correlation with CD8 + tumor infiltrating lymphocytes. Applying the tumor proportion score (TPS) with a cutoff of 1%, PD-L1, TIM-3 and B7-H3 expression was present in 17%, 10% and 93% of cases, respectively. Applying the combined positive score (CPS) with a cutoff of 1, PD-L1, TIM-3 and B7-H3 expression was present in 63%, 67% and 94% of cases, respectively. Expression of these markers was largely independent of one another. PD-L1 correlated with higher CD8 + T-cell density when evaluated by either TPS (p = 0.02) or CPS (p < 0.0001). TIM-3 correlated with CD8 + T-cell density when evaluated by CPS (p < 0.0001). PD-L1 positivity was associated with improved overall survival (p = 0.038) when applying CPS. No association between PD-L1 expression and survival was found using TPS, and there was no association between TIM-3 or B7-H3 positivity and survival by either TPS or CPS. Using TPS, PD-L1 correlated with a higher tumor stage but not with survival, whereas the converse was true when PD-L1 was evaluated by CPS, suggesting that PD-L1 expression in immune cells correlates with prognosis and is independent of tumor stage. In conclusion, PD-L1, TIM-3 and B7-H3 may be potential therapeutic targets in selected patients with ESC. Further investigation of their roles as predictive biomarkers is needed.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA
| | - Kyle Molberg
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA
| | - Kelley Carrick
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA
| | - Shuang Niu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA
| | - Glorimar Rivera Colon
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA
| | - Katja Gwin
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Wenxin Zheng
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA.,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Obstetrics and Gynecology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Diego H Castrillon
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA.,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Obstetrics and Gynecology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Elena Lucas
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Pathology, Parkland Hospital, Dallas, TX, 75235, USA. .,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
28
|
Identification of a New Prediction Model for Bladder Cancer Related to Immune Functions and Chemotherapy Using Gene Sets of Biological Processes. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4740686. [DOI: 10.1155/2022/4740686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022]
Abstract
Background. Biological processes serve crucial functions in the initiation and development of cancer. Therefore, we constructed and validated a model for bladder cancer (BLCA) with good predictive power for immunity, prognosis, and therapy. Methods. Using the expression of the gene sets based on biological processes, BLCA patients were divided into three clusters by consensus cluster analysis. By performing LASSO regression analysis twice, key genes were selected, and the biological processes-related genes’ (BPRG) score was calculated. Differences in immune infiltration, tumor microenvironment, tumor mutation burden, immunotherapy, and sensitivity towards chemotherapy were analyzed between two groups divided by BPRG score. Results. Good accuracy was observed for the three clusters. They showed different prognoses and levels of immune cell infiltration. The selected key genes were mainly enriched in immune-related pathways. The high-BPRG score group was related to poor prognosis, higher immune cell infiltration, interstitial scores, and increased tumor mutation. Moreover, the effects of immunotherapy were good, and those of chemotherapy were poor. Conclusion. Overall, key genes may be involved in various complex immune regulation processes. Therefore, the quantification and verification of the BPRG score are expected to facilitate the understanding of the immunosuppressive microenvironment in BLCA and guide the choice of chemotherapeutic drugs and immunotherapeutic regimens and help predict the prognoses of patients with BLCA.
Collapse
|
29
|
Zhou R, Peng N, Li W. Identification of ISCA1 as novel immunological and prognostic biomarker for bladder cancer. Front Immunol 2022; 13:975503. [PMID: 36072584 PMCID: PMC9442282 DOI: 10.3389/fimmu.2022.975503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/05/2022] [Indexed: 12/09/2022] Open
Abstract
BackgroundIron-sulfur cluster assembly 1 (ISCA1) has a significant effect on respiratory complexes and energy metabolism. Although there is some evidence that ISCA1 gene expression impacts energy metabolism and consequently has a role in tumorigenesis and cancer metastasis in different types of malignancies, no systematic pan-cancer study of the ISCA1 has been conducted. As a result, we sought to investigate ISCA1’s predictive value in 33 cancer types as well as its possible immunological function.MethodsWe included the pan-cancer expression profile dataset and clinical data from the public database. Firstly, the single-sample Gene Set Enrichment Analysis (ssGSEa) approach was employed for analyzing the immune link in pan-cancer, while the limma package was utilized for analyzing the differential expression in cancer species. Subsequently, ciberport, MCP-counter, TIMER2, quanTIseq, and xCELL were employed for analyzing bladder cancer (BLCA)’s immune infiltration. Least absolute shrinkage and selection operator (Lasso) were employed for choosing the best gene to develop the immune risk scoring model.ResultsISCA1 gene expression was positively related to four immune signatures (chemokine, immunostimulator, MHC, and receptor) in BLCA. Samples of BLCA were sorted into two groups by the best cut-off of ISCA1 expression degree. The group with a high level of ISCA1 expression had a higher risk, suggesting that the ISCA1 gene was a risk factor in BLCA, and its high expression resulted in a poorer prognosis. Additionally, it was noted that ISCA1 was positively linked with these immune checkpoints. Moreover, there was a considerable positive link between ISCA1 and different immune properties in subgroups with different immune checkpoint inhibiting responses. Finally, an immune risk scoring model was made and it showed a better score in comparison to that of TIDE.ConclusionISCA1 can be a prognostic marker for a variety of cancers, particularly BLCA. Its high level of expression has a deleterious impact on the prognosis of BLCA patients. This strongly shows that ISCA1 is a significant prognostic factor for BLCA and that it could be used as a new prognostic detection target and treatment approach.
Collapse
Affiliation(s)
- Renlong Zhou
- Department of Blood Transfusion, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Naixiong Peng
- Department of Urology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Wei Li
- Department of Urology, Shenzhen Longhua District Central Hospital, Shenzhen, China
- *Correspondence: Wei Li,
| |
Collapse
|
30
|
Magnetic Nanochain-Based Smart Drug Delivery System with Remote Tunable Drug Release by a Magnetic Field. BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00072-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
31
|
Gianni C, Palleschi M, Schepisi G, Casadei C, Bleve S, Merloni F, Sirico M, Sarti S, Cecconetto L, Di Menna G, Schettini F, De Giorgi U. Circulating inflammatory cells in patients with metastatic breast cancer: Implications for treatment. Front Oncol 2022; 12:882896. [PMID: 36003772 PMCID: PMC9393759 DOI: 10.3389/fonc.2022.882896] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022] Open
Abstract
Adaptive and innate immune cells play a crucial role as regulators of cancer development. Inflammatory cells in blood flow seem to be involved in pro-tumor activities and contribute to breast cancer progression. Circulating lymphocyte ratios such as the platelet-lymphocytes ratio (PLR), the monocyte-lymphocyte ratio (MLR) and the neutrophil-lymphocyte ratio (NLR) are new reproducible, routinely feasible and cheap biomarkers of immune response. These indexes have been correlated to prognosis in many solid tumors and there is growing evidence on their clinical applicability as independent prognostic markers also for breast cancer. In this review we give an overview of the possible value of lymphocytic indexes in advanced breast cancer prognosis and prediction of outcome. Furthermore, targeting the immune system appear to be a promising therapeutic strategy for breast cancer, especially macrophage-targeted therapies. Herein we present an overview of the ongoing clinical trials testing systemic inflammatory cells as therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- *Correspondence: Caterina Gianni,
| | - Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bleve
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Filippo Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Samanta Sarti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lorenzo Cecconetto
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giandomenico Di Menna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Francesco Schettini
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
32
|
Huo C, Luo Z, Ning X, Kang X, Yan Q, Guo Y, Li G, Wang Z, Li Y, Qian S. 4,6-Disubstituted-1H-Indazole-4-Amine derivatives with immune-chemotherapy effect and in vivo antitumor activity. Eur J Med Chem 2022; 241:114625. [DOI: 10.1016/j.ejmech.2022.114625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/04/2022]
|
33
|
A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
34
|
Cao ZX, Weng X, Huang JS, Long X. Receptor–ligand pair typing and prognostic risk model for papillary thyroid carcinoma based on single-cell sequencing. Front Immunol 2022; 13:902550. [PMID: 35935973 PMCID: PMC9354623 DOI: 10.3389/fimmu.2022.902550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
The papillary thyroid carcinoma (PTC) microenvironment consists of various cancer and surrounding cells, and the communication between them is mainly performed through ligand–receptor (LR) interactions. Single-cell RNA sequencing (scRNA-seq) has been performed to investigate the role of intercellular communication networks in tumor progression. In addition, scRNA-seq can accurately identify the characteristics of immune cell subsets, which is of great significance for predicting the efficacy of immunotherapy. In this study, the cell–cell communication network was analyzed through LR pairs, and a new PTC molecular phenotype was developed based on LR pairs. Furthermore, a risk model was established to predict patient response to PD-1 blockade immunotherapy. The scRNA-seq dataset was obtained from GSE184362, and the bulk tumor RNA-seq dataset was obtained from The Cancer Genome Atlas. CellPhoneDB was used for cellular communication analysis. LR pair correlations were calculated and used to identify molecular subtypes, and the least absolute shrinkage and selection operator (Lasso) Cox regression was used to develop a risk model based on LR pairs. The IMvigor210 and GSE78220 cohorts were used as external validations for the LR.score to predict responses to PD-L1 blockade therapy. A total of 149 LR pairs with significant expression and prognostic correlation were included, and three PTC molecular subtypes were obtained from those with significant prognostic differences. Then, five LR pairs were selected to construct the risk scoring model, a reliable and independent prognostic factor in the training set, test set, and whole dataset. Furthermore, two external validation sets confirmed the predictive efficacy of the LR.score for response to PD-1 blockade therapy.
Collapse
Affiliation(s)
- Zhe Xu Cao
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xin Weng
- Hunan Sixth Engineering Company Construction Hospital, Changsha, China
| | - Jiang Sheng Huang
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xia Long
- Hospital Office, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xia Long,
| |
Collapse
|
35
|
Zhang Y, Yang R, Xu C, Zhang Y, Deng M, Wu D, Tang F, Liu X, Han Y, Zhan Y, Miao J. Analysis of the immune checkpoint lymphocyte activation gene-3 (LAG-3) in endometrial cancer: An emerging target for immunotherapy. Pathol Res Pract 2022; 236:153990. [PMID: 35749914 DOI: 10.1016/j.prp.2022.153990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Lymphocyte activation gene-3 (LAG-3) is a novel molecule that participates in the immune escape of tumor cells and is a target for immunotherapy. However, the expression of LAG-3 in patients with endometrial cancer (EC) has not been comprehensively characterized. OBJECTIVES We elucidated the expression of LAG-3 and investigated its correlation with clinicopathological parameters, ProMisE subtypes, CD8+ T-cell infiltration and relapse-free survival (RFS) in a retrospective cohort of 421 patients with endometrial cancer. METHODS Next-generation sequencing of the polymerase epsilon (POLE) and immunohistochemistry of mismatch repair (MMR)-related protein (MLH1, PMS2, MSH2, and MSH6), p53, CD8 and LAG-3 protein in whole sections were performed. RESULTS Positive LAG-3 was detected in tumor cells (TCs) and immune cells (ICs) in 31.6% (133/421) and 24.0% (101/421) of the patients, respectively. LAG-3 positivity in ICs was more common in high-grade, high-intermediate risk, high-risk, and advanced/metastatic subgroups and was relevant to lymphovascular space invasion, while that in TCs was more common in older individuals (≥54 years). LAG-3 expression was more prevalent in POLE ultramutated (POLEmut) and MMR-deficient (MMRd) EC than in p53-abnormal (p53abn) and p53-wild (p53wt) EC in TCs (34.4 % and 66.3% in POLEmut and MMRd versus 28.6% and 19.5% in p53abn and p53wt, P < 0.001) and ICs (78.1 % and 65.1% in POLEmut and MMRd versus 2.9% and 5.2% in p53abn and p53wt, P < 0.001). Positive expression of LAG-3 in TCs and ICs was associated with high levels of tumor-associated CD8+ T-cell immune infiltration. Additionally, LAG-3 positivity in TCs was related to improved RFS. CONCLUSIONS This study suggests that immunotherapy targeting LAG-3 may play a role in EC patients with POLEmut or MMRd molecular markers. Positive LAG-3 expression in TCs may be a predictor of improved RFS.
Collapse
Affiliation(s)
- Yubo Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Ruiye Yang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chunyu Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanqin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Mengqi Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Di Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fan Tang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xinyu Liu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yiding Han
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yang Zhan
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jinwei Miao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China.
| |
Collapse
|
36
|
Goldman JW, Piha-Paul SA, Curti B, Pedersen KS, Bauer TM, Groenland SL, Carvajal RD, Chhaya V, Kirby G, McGlinchey K, Hammond SA, Streicher KL, Townsley D, Chae YK, Voortman J, Marabelle A, Powderly J. Safety and tolerability of MEDI0562, an OX40 agonist monoclonal antibody, in combination with durvalumab or tremelimumab in adult patients with advanced solid tumors. Clin Cancer Res 2022; 28:3709-3719. [PMID: 35699623 DOI: 10.1158/1078-0432.ccr-21-3016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/03/2021] [Accepted: 06/10/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Combination therapies targeting immunological checkpoints have shown promise in treating multiple tumor types. We report safety and tolerability of MEDI0562, a humanized IgG1K OX40 monoclonal antibody, in combination with durvalumab (anti-PD-L1), or tremelimumab (anti-CTLA-4), in adult patients with previously treated advanced solid tumors. EXPERIMENTAL DESIGN In this phase 1, multicenter, open-label study, patients received escalating doses of MEDI0562 (2.25, 7.5, or 22.5 mg) every two weeks (Q2W) in combination with durvalumab (1500 mg) or tremelimumab (75 or 225 mg) Q4W, intravenously, until unacceptable toxicity or progressive disease. Tumor assessments were performed Q8W. The primary objective was to evaluate safety and tolerability. RESULTS Among the 27 and 31 patients who received MEDI0562 + durvalumab or MEDI0562 + tremelimumab, 74.1% and 67.7% reported a treatment-related adverse event (AE), and 22.2% and 19.4% experienced a treatment‑emergent AE that led to discontinuation, respectively. The maximum tolerated dose of MEDI0562 + durvalumab was 7.5 mg MEDI0562 + 1500 mg durvalumab; the maximum administered dose of MEDI0562 + tremelimumab was 22.5 mg MEDI0562 + 225 mg tremelimumab. Three patients in the MEDI0562 + durvalumab arm had a partial response. The mean percentage of Ki67+CD4+ and Ki67+CD8+ memory T cells increased by >100% following the first dose of MEDI0562 + durvalumab or tremelimumab in all dose cohorts. A decrease in OX40+FOXP3 T regulatory cells was observed in a subset of patients with available paired biopsies. CONCLUSIONS Following dose escalation, moderate toxicity was observed in both treatment arms, with no clear efficacy signals demonstrated.
Collapse
Affiliation(s)
| | - Sarina A Piha-Paul
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brendan Curti
- Providence Cancer Center and Earle A. Chiles Research Institute, Portland, OR, United States
| | | | - Todd M Bauer
- Sarah Cannon Research Institute / Tennessee Oncology, PLLC., Nashville, TN, United States
| | | | | | - Vaishali Chhaya
- AstraZeneca (United States), Gaithersburg, MD, United States
| | - Gray Kirby
- AstraZeneca (United States), Gaithersburg, MD, United States
| | - Kelly McGlinchey
- AstraZeneca (United Kingdom), Gaithersburg, Maryland, United States
| | | | | | | | - Young Kwang Chae
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jens Voortman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - John Powderly
- Carolina BioOncology Institute, Huntersville, NC, United States
| |
Collapse
|
37
|
Zhang Z, Yang A, Chaurasiya S, Park AK, Lu J, Kim SI, Warner SG, Yuan YC, Liu Z, Han H, Von Hoff D, Fong Y, Woo Y. CF33-hNIS-antiPDL1 virus primes pancreatic ductal adenocarcinoma for enhanced anti-PD-L1 therapy. Cancer Gene Ther 2022; 29:722-733. [PMID: 34108669 PMCID: PMC8896143 DOI: 10.1038/s41417-021-00350-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Immunotherapeutic strategies that combine oncolytic virus (OV) and immune checkpoint inhibitors have the potential to overcome treatment resistance in pancreatic ductal adenocarcinoma (PDAC), one of the least immunogenic solid tumors. Oncolytic viral chimera, CF33-hNIS-antiPDL1 genetically modified to express anti-human PD-L1 antibody and CF33-hNIS-Δ without the anti-PD-L1 gene, were used to investigate the immunogenic effects of OVs and virus-delivered anti-PD-L1 in PDAC in vitro. Western blot, flow cytometry, and immunofluorescence microscopy were used to evaluate the effects of CF33-hNIS-Δ and IFNγ on PD-L1 upregulation in AsPC-1 and BxPC-3 cells, and CF33-hNIS-antiPDL1 production of anti-PD-L1 and surface PD-L1 blockade of AsPC-1 and BxPC-3 with or without cocultured activated T cells. The cytosolic and cell surface levels of PD-L1 in PDAC cell lines varied; only BxPC-3 showed high cell surface expression. Treatment of these cells with CF33-hNIS-Δ and IFNγ significantly upregulated PD-L1 expression and translocation of PD-L1 from the cytosol onto the cell surface. Following coculture of activated T cells and BxPC-3 with CF33-hNIS-antiPDL1, the cell surface PD-L1 blockade on BxPC-3 cells by virus-delivered anti-PD-L1 antibody increased granzyme B release and prevented virus-induced decrease of perforin release from activated CD8+ T cells. Our results suggest that CF33-IOVs can prime immune checkpoint inhibition of PDAC and enhance antitumor immune killing.
Collapse
Affiliation(s)
- Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Annie Yang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Anthony K Park
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Susanne G Warner
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Yate-Ching Yuan
- Division of Translational Bioinformatics, Center for Informatics, City of Hope National Medical Center, Duarte, CA, USA
| | - Zheng Liu
- Division of Translational Bioinformatics, Center for Informatics, City of Hope National Medical Center, Duarte, CA, USA
| | - Haiyong Han
- The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Daniel Von Hoff
- The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA.
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
38
|
Dong S, Liu B, Hu S, Guo F, Zhong Y, Cai Q, Zhang S, Qian Y, Wang J, Zhou F. A novel oncolytic virus induces a regional cytokine storm and safely eliminates malignant ascites of colon cancer. Cancer Med 2022; 11:4297-4309. [PMID: 35510373 DOI: 10.1002/cam4.4772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/23/2022] [Accepted: 04/12/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Given malignant ascites with a terrible prognosis and a unique immune microenvironment, our purpose is to evaluate whether oncolytic herpes simplex virus type 2(OH2) is able to safely eliminate ascites of colon cancer and through which specific mechanism it exerts antitumor immunity. METHODS We established an ascites mice model through intraperitoneal injection of CT26 cells and obtained an appropriate dose range for in vivo tests. Efficacy and safety of OH2 were detected by weight of ascites, blood routine analysis, histopathological examination, and the survival time of mice. The specific mechanism underlying antitumor immunity was analyzed by cytometric bead array, flow cytometry, and single-cell RNA sequencing. Furthermore, anti-interleukin (IL)-6R antibody tocilizumab was synchronously or sequentially delivered with OH2 to explore the role of the regional cytokine storm, mainly IL-6 hypersecretion. RESULTS OH2 was able to eliminate ascites and significantly prolong the survival of mice-bearing CT26 tumor cells by intraperitoneal injection, without obvious systemic damage to the main organs even though a regional cytokine storm. Hypersecretion of pro-inflammatory cytokines, mainly IL-6, and increased infiltration of CD4+ and CD8+ T cells were observed in ascites mice treated by OH2, compared with those treated by 5-fluorouracil or nonresponders. Furthermore, the initial-stage blocking of the IL-6 pathway was able to considerably suppress antitumor immune responses driven by OH2. Surprisingly, we discovered upregulations of the immune checkpoint genes such as Cd274 and Pdcd1 by single-cell RNA sequencing. CONCLUSIONS OH2 could safely eliminate malignant ascites of colon cancer and convert the cold immune microenvironment by inducing a remarkably regional cytokine storm in ascites, mainly IL-6, in the early stage of antitumor immune responses beyond directed oncolytic virotherapy.
Collapse
Affiliation(s)
- Shuang Dong
- Hubei Key Laboratory of Tumor Biological Behaviors, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.,Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Binlei Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
| | - Sheng Hu
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fang Guo
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Zhong
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Cai
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Siqi Zhang
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yu Qian
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Wang
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fuxiang Zhou
- Hubei Key Laboratory of Tumor Biological Behaviors, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
39
|
Minami Y, Takaki H, Yamakado K, Kudo M. How Compatible Are Immune Checkpoint Inhibitors and Thermal Ablation for Liver Metastases? Cancers (Basel) 2022; 14:cancers14092206. [PMID: 35565338 PMCID: PMC9103121 DOI: 10.3390/cancers14092206] [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: 03/10/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although immune checkpoint inhibitors (ICIs) have achieved great progression in cancer treatment, the efficacy of ICI monotherapy is still limited. Meanwhile, the negative efficacy of thermal ablation for liver metastases is the high rate of local tumor progression. Since thermal ablation-induced inflammation and increases in tumor antigens have been suggested to promote the cancer-immunity cycle, thermal ablation and ICI can boost the immune response against cancer cells as one of the positive synergy effects. The findings of preclinical and clinical research have provided supportive evidence for the combination of ICIs with thermal ablation reversing T-cell exhaustion and demonstrating synergy. However, the clinical feasibility of immune response activation by combination therapy with ICI monotherapy and thermal ablation appears to be limited, it may be not very common phenomena. Abstract Cancer immunotherapy, which reactivates the weakened immune cells of cancer patients, has achieved great success, and several immune checkpoint inhibitors (ICIs) are now available in clinical practice. Despite promising clinical outcomes, favorable responses are only observed in a fraction of patients, and resistance mechanisms, including the absence of tumor antigens, have been reported. Thermal ablation involves the induction of irreversible damage to cancer cells by localized heat and may result in the release of tumor antigens. The combination of immunotherapy and thermal ablation is an emerging therapeutic option with enhanced efficacy. Since thermal ablation-induced inflammation and increases in tumor antigens have been suggested to promote the cancer-immunity cycle, the combination of immuno-oncology (IO) therapy and thermal ablation may be mutually beneficial. In preclinical and clinical studies, the combination of ICI and thermal ablation significantly inhibited tumor growth, and synergistic antitumor effects appeared to prolong the survival of patients with secondary liver cancer. However, evidence for the efficacy of ICI monotherapy combined with thermal ablation is currently insufficient. Therefore, the clinical feasibility of immune response activation by ICI monotherapy combined with thermal ablation may be limited, and thermal ablation may be more compatible with dual ICIs (the IO–IO combination) to induce strong immune responses.
Collapse
Affiliation(s)
- Yasunori Minami
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, 377-2 Ohno-Higashi Osaka-Sayama, Osaka 589-8511, Japan;
- Correspondence: ; Tel.: +81-72-366-0221 (ext. 3525); Fax: +81-72-367-2880
| | - Haruyuki Takaki
- Department of Radiology, Hyogo College of Medicine, 1-1 Mukogawa Nishinomiya, Nishinomiya 663-8501, Japan; (H.T.); (K.Y.)
| | - Koichiro Yamakado
- Department of Radiology, Hyogo College of Medicine, 1-1 Mukogawa Nishinomiya, Nishinomiya 663-8501, Japan; (H.T.); (K.Y.)
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, 377-2 Ohno-Higashi Osaka-Sayama, Osaka 589-8511, Japan;
| |
Collapse
|
40
|
Correia C, Weiskittel TM, Ung CY, Villasboas Bisneto JC, Billadeau DD, Kaufmann SH, Li H. Uncovering Pharmacological Opportunities for Cancer Stem Cells-A Systems Biology View. Front Cell Dev Biol 2022; 10:752326. [PMID: 35359437 PMCID: PMC8962639 DOI: 10.3389/fcell.2022.752326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs) represent a small fraction of the total cancer cell population, yet they are thought to drive disease propagation, therapy resistance and relapse. Like healthy stem cells, CSCs possess the ability to self-renew and differentiate. These stemness phenotypes of CSCs rely on multiple molecular cues, including signaling pathways (for example, WNT, Notch and Hedgehog), cell surface molecules that interact with cellular niche components, and microenvironmental interactions with immune cells. Despite the importance of understanding CSC biology, our knowledge of how neighboring immune and tumor cell populations collectively shape CSC stemness is incomplete. Here, we provide a systems biology perspective on the crucial roles of cellular population identification and dissection of cell regulatory states. By reviewing state-of-the-art single-cell technologies, we show how innovative systems-based analysis enables a deeper understanding of the stemness of the tumor niche and the influence of intratumoral cancer cell and immune cell compositions. We also summarize strategies for refining CSC systems biology, and the potential role of this approach in the development of improved anticancer treatments. Because CSCs are amenable to cellular transitions, we envision how systems pharmacology can become a major engine for discovery of novel targets and drug candidates that can modulate state transitions for tumor cell reprogramming. Our aim is to provide deeper insights into cancer stemness from a systems perspective. We believe this approach has great potential to guide the development of more effective personalized cancer therapies that can prevent CSC-mediated relapse.
Collapse
Affiliation(s)
- Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Taylor M Weiskittel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | | | - Daniel D Billadeau
- Department of Immunology, Mayo Clinic, Rochester, MN, United States,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States,Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, United States,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States,*Correspondence: Hu Li,
| |
Collapse
|
41
|
Pandey PR, Young KH, Kumar D, Jain N. RNA-mediated immunotherapy regulating tumor immune microenvironment: next wave of cancer therapeutics. Mol Cancer 2022; 21:58. [PMID: 35189921 PMCID: PMC8860277 DOI: 10.1186/s12943-022-01528-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/31/2022] [Indexed: 12/16/2022] Open
Abstract
AbstractAccumulating research suggests that the tumor immune microenvironment (TIME) plays an essential role in regulation of tumor growth and metastasis. The cellular and molecular nature of the TIME influences cancer progression and metastasis by altering the ratio of immune- suppressive versus cytotoxic responses in the vicinity of the tumor. Targeting or activating the TIME components show a promising therapeutic avenue to combat cancer. The success of immunotherapy is both astounding and unsatisfactory in the clinic. Advancements in RNA-based technology have improved understanding of the complexity and diversity of the TIME and its effects on therapy. TIME-related RNA or RNA regulators could be promising targets for anticancer immunotherapy. In this review, we discuss the available RNA-based cancer immunotherapies targeting the TIME. More importantly, we summarize the potential of various RNA-based therapeutics clinically available for cancer treatment. RNA-dependent targeting of the TIME, as monotherapy or combined with other evolving therapeutics, might be beneficial for cancer patients’ treatment in the near future.
Collapse
|
42
|
Li J, Che L, Xu C, Lu D, Xu Y, Liu M, Chai W. XIST/miR-34a-5p/PDL1 axis regulated the development of lung cancer cells and the immune function of CD8+ T cells. J Recept Signal Transduct Res 2022; 42:469-478. [PMID: 35067156 DOI: 10.1080/10799893.2021.2019274] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Jing Li
- Respiratory Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Liyan Che
- Respiratory Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Chang Xu
- Emergency Dpartment, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Dongdong Lu
- Respiratory Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Yan Xu
- Respiratory Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Mengru Liu
- Respiratory Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Wenshu Chai
- Respiratory Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| |
Collapse
|
43
|
Yan L, Yang Y, Li YM, Fan JW, Wang XD, Bai YJ, Wang LL, Shi YY, Li Y. Soluble Tim-3/Gal-9 as predictors of adverse outcomes after kidney transplantation: a cohort study. Clin Biochem 2021; 102:19-25. [PMID: 34968481 DOI: 10.1016/j.clinbiochem.2021.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND In our previous study, serum soluble T-cell immunoglobulin and mucin structure-3 (stim-3) and galactosin-9 (sGal-9) were found to be associated with renal function after kidney transplantation. However, it is unclear whether these two indicators can predict adverse outcomes after transplantation. METHODS Ninety-one recipients of kidney transplantation were enrolled and divided into a stable group and an adverse outcome group (consisting of biopsy-proven rejection, graft loss, death and clinically diagnosed rejection). The expression levels of sTim-3 and sGal-9 before (pre-Tim-3 and pre-Gal-9) and one month after transplantation (post-Tim-3 and post-Gal-9) were measured by ELISA. RESULTS The level of pre-Tim-3 was significantly higher in the stable group than in the adverse outcome group [median (range), 2275 (840-4236) pg/mL vs. 1589 (353-3094) pg/mL, P=0.002]. The level of post-Gal-9 was significantly lower in the stable group than in the adverse outcome group [median (range), 4869 (1418-13080) pg/mL vs. 6852: (4128-10760) pg/mL, P=0.003]. The areas under the curve (AUCs) for pre-Tim-3 and post-Gal-9 were 0.737 (P=0.002) and 0.751 (P=0.003), respectively, better than AUC of post-eGFR (0.633) (P=0.071), according to the receiver operating characteristic (ROC) curve. Through Cox regression analysis, including pre-Tim-3, post-Gal-9, post-eGFR, sex, age, BMI of recipients and donors, pre-Tim-3 and post-Gal-9 were independent risk factors for adverse outcomes after kidney transplantation (P=0.016, P=0.033, respectively). CONCLUSION Serum sTim-3 and sGal-9 can predict adverse outcomes within two years after kidney transplantation.
Collapse
Affiliation(s)
- Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Yan Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Ya-Mei Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Ji-Wen Fan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Xian-Ding Wang
- Department of Nephrology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Yang-Juan Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Lan-Lan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Yun-Ying Shi
- Department of Nephrology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu, Sichuan, China.
| |
Collapse
|
44
|
Bailly C, Thuru X, Quesnel B. Modulation of the Gal-9/TIM-3 Immune Checkpoint with α-Lactose. Does Anomery of Lactose Matter? Cancers (Basel) 2021; 13:cancers13246365. [PMID: 34944985 PMCID: PMC8699133 DOI: 10.3390/cancers13246365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The disaccharide lactose is a common excipient in pharmaceutical products. In addition, the two anomers α- and β-lactose can exert immuno-modulatory effects. α-Lactose functions as a major regulator of the T-cell immunoglobulin mucin-3 (Tim-3)/Galectin-9 (Gal-9) immune checkpoint, through direct binding to the β-galactoside-binding lectin galectin-9. The blockade of TIM-3 with monoclonal antibodies or small molecules represents a promising approach to combat onco-hematological diseases, in particular myelodysplastic syndromes, and acute myeloid leukemia. Alternatively, the activity of the checkpoint can be modulated via targeting of Gal-9 with both α- and β-lactose. In fact, lactose is a quasi-pan-galectin ligand, capable of modulating the functions of most of the 16 galectin molecules. This review discusses the capacity of lactose and Gal-9 to modulate the TIM-3/Gal-9 and PD-1/PD-L1 immune checkpoints in oncology. The immuno-regulatory roles of lactose and Gal-9 are highlighted. Abstract The disaccharide lactose is an excipient commonly used in pharmaceutical products. The two anomers, α- and β-lactose (α-L/β-L), differ by the orientation of the C-1 hydroxyl group on the glucose unit. In aqueous solution, a mutarotation process leads to an equilibrium of about 40% α-L and 60% β-L at room temperature. Beyond a pharmaceutical excipient in solid products, α-L has immuno-modulatory effects and functions as a major regulator of TIM-3/Gal-9 immune checkpoint, through direct binding to the β-galactoside-binding lectin galectin-9. The blockade of the co-inhibitory checkpoint TIM-3 expressed on T cells with anti-TIM-3 antibodies represents a promising approach to combat different onco-hematological diseases, in particular myelodysplastic syndromes and acute myeloid leukemia. In parallel, the discovery and development of anti-TIM-3 small molecule ligands is emerging, including peptides, RNA aptamers and a few specifically designed heterocyclic molecules. An alternative option consists of targeting the different ligands of TIM-3, notably Gal-9 recognized by α-lactose. Modulation of the TIM-3/Gal-9 checkpoint can be achieved with both α- and β-lactose. Moreover, lactose is a quasi-pan-galectin ligand, capable of modulating the functions of most of the 16 galectin molecules. The present review provides a complete analysis of the pharmaceutical and galectin-related biological functions of (α/β)-lactose. A focus is made on the capacity of lactose and Gal-9 to modulate both the TIM-3/Gal-9 and PD-1/PD-L1 immune checkpoints in oncology. Modulation of the TIM-3/Gal-9 checkpoint is a promising approach for the treatment of cancers and the role of lactose in this context is discussed. The review highlights the immuno-regulatory functions of lactose, and the benefit of the molecule well beyond its use as a pharmaceutical excipient.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, 59290 Lille, France
- Correspondence:
| | - Xavier Thuru
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020—UMR1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France; (X.T.); (B.Q.)
| | - Bruno Quesnel
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020—UMR1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France; (X.T.); (B.Q.)
| |
Collapse
|
45
|
Immune Checkpoint Inhibitor Therapy for Bone Metastases: Specific Microenvironment and Current Situation. J Immunol Res 2021; 2021:8970173. [PMID: 34877360 PMCID: PMC8645368 DOI: 10.1155/2021/8970173] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/19/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022] Open
Abstract
The treatment of bone metastases is a thorny issue. Immunotherapy may be one of the few hopes for patients with unresectable bone metastases. Immune checkpoint inhibitors are the most commonly used immunotherapy drugs currently. In this review, the characteristics and interaction of bone metastases and their immune microenvironment were systematically discussed, and the relevant research progress of the immunological mechanism of tumor bone metastasis was reviewed. On this basis, we expounded the clinical application of immune checkpoint inhibitors for bone metastasis of common tumors, including non-small-cell lung cancer, renal cell carcinoma, prostate cancer, melanoma, and breast cancer. Then, the deficiencies and limitations in current researches were summarized. In-depth basic research on bone metastases and optimization of clinical treatment is needed.
Collapse
|
46
|
Aguirre LE, Komrokji R, Padron E. It is time to shift the treatment paradigm in myelodysplastic syndromes: A focus on novel developments and current investigational approaches exploring combinatorial therapy in high-risk MDS. Best Pract Res Clin Haematol 2021; 34:101325. [PMID: 34865697 DOI: 10.1016/j.beha.2021.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Higher risk myelodysplastic syndromes are defined as a subset of disease with higher risk of AML transformation and poor overall survival. For decades, therapeutic options for high-risk MDS have been limited to allogeneic stem cell transplant (the only option for cure but limited to only a handful of patients) or hypomethylating agents, with the goal to alter the natural history of disease, delay progression and improve survival, while addressing cytopenias, transfusion requirements and improving quality of life. Recent developments in DNA sequencing and other technologies have shed significant light into the pathogenesis of MDS and led to rational and targeted drug development across a variety of therapeutic vulnerabilities, including disruption of protein ubiquitination through NAE inhibition, selective modulation of macrophage activity and immune checkpoint inhibition through blockade of TIM-3. This review highlights some of the most promising agents in recent drug development and their therapeutic efficacy in the management of high-risk MDS, and further explores the rationale behind potential combinatorial approaches using an HMA backbone to synergistically improve treatment outcomes.
Collapse
Affiliation(s)
- Luis E Aguirre
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rami Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Eric Padron
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
47
|
Chen Y, Xia F, Jiang B, Wang W, Li X. Role of Immune Cell-Specific Hypermethylation Signatures in Classification and Risk Stratification of Breast Cancer. Front Med (Lausanne) 2021; 8:674338. [PMID: 34513864 PMCID: PMC8426625 DOI: 10.3389/fmed.2021.674338] [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: 03/01/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Epigenetic regulation, including DNA methylation, plays a major role in shaping the identity and function of immune cells. Innate and adaptive immune cells recruited into tumor tissues contribute to the formation of the tumor immune microenvironment (TIME), which is closely involved in tumor progression in breast cancer (BC). However, the specific methylation signatures of immune cells have not been thoroughly investigated yet. Additionally, it remains unknown whether immune cells-specific methylation signatures can identify subgroups and stratify the prognosis of BC patients. Methods: DNA methylation profiles of six immune cell types from eight datasets downloaded from the Gene Expression Omnibus were collected to identify immune cell-specific hypermethylation signatures (IC-SHMSs). Univariate and multivariate cox regression analyses were performed using BC data obtained from The Cancer Genome Atlas to identify the prognostic value of these IC-SHMSs. An unsupervised clustering analysis of the IC-SHMSs with prognostic value was performed to categorize BC patients into subgroups. Multiple Cox proportional hazard models were constructed to explore the role of IC-SHMSs and their relationship to clinical characteristics in the risk stratification of BC patients. Integrated discrimination improvement (IDI) was performed to determine whether the improvement of IC-SHMSs on clinical characteristics in risk stratification was statistically significant. Results: A total of 655 IC-SHMSs of six immune cell types were identified. Thirty of them had prognostic value, and 10 showed independent prognostic value. Four subgroups of BC patients, which showed significant heterogeneity in terms of survival prognosis and immune landscape, were identified. The model incorporating nine IC-SHMSs showed similar survival prediction accuracy as the clinical model incorporating age and TNM stage [3-year area under the curve (AUC): 0.793 vs. 0.785; 5-year AUC: 0.735 vs. 0.761]. Adding the IC-SHMSs to the clinical model significantly improved its prediction accuracy in risk stratification (3-year AUC: 0.897; 5-year AUC: 0.856). The results of IDI validated the statistical significance of the improvement (p < 0.05). Conclusions: Our study suggests that IC-SHMSs may serve as signatures of classification and risk stratification in BC. Our findings provide new insights into epigenetic signatures, which may help improve subgroup identification, risk stratification, and treatment management.
Collapse
Affiliation(s)
- Yong Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Fada Xia
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Jiang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wenlong Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Sun J, Tang Q, Zhang J, Chen G, Peng J, Chen L. Possible Immunotherapeutic Strategies Based on Carcinogen-Dependent Subgroup Classification for Oral Cancer. Front Mol Biosci 2021; 8:717038. [PMID: 34497832 PMCID: PMC8419237 DOI: 10.3389/fmolb.2021.717038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/23/2021] [Indexed: 11/13/2022] Open
Abstract
The oral cavity serves as an open local organ of the human body, exposed to multiple external factors from the outside environment. Coincidentally, initiation and development of oral cancer are attributed to many external factors, such as smoking and drinking, to a great extent. This phenomenon was partly explained by the genetic abnormalities traditionally induced by carcinogens. However, more and more attention has been attracted to the influence of carcinogens on the local immune status. On the other hand, immune heterogeneity of cancer patients is a huge obstacle for enhancing the clinical efficacy of tumor immunotherapy. Thus, in this review, we try to summarize the current opinions about variant genetic changes and multiple immune alterations induced by different oral cancer carcinogens and discuss the prospects of targeted immunotherapeutic strategies based on specific immune abnormalities caused by different carcinogens, as a predictive way to improve clinical outcomes of immunotherapy-treated oral cancer patients.
Collapse
Affiliation(s)
- Jiwei Sun
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Junyuan Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jinfeng Peng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
49
|
Bashash D, Zandi Z, Kashani B, Pourbagheri-Sigaroodi A, Salari S, Ghaffari SH. Resistance to immunotherapy in human malignancies: Mechanisms, research progresses, challenges, and opportunities. J Cell Physiol 2021; 237:346-372. [PMID: 34498289 DOI: 10.1002/jcp.30575] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 12/31/2022]
Abstract
Despite remarkable advances in different types of cancer therapies, an effective therapeutic strategy is still a major and significant challenge. One of the most promising approaches in this regard is immunotherapy, which takes advantage of the patients' immune system; however, the many mechanisms that cancerous cells harbor to extend their survival make it impossible to gain perfect eradication of tumors. The response rate to cancer immunotherapies, especially checkpoint inhibitors and adoptive T cell therapy, substantially differs in various cancer types with the highest rates in advanced melanoma and non-small cell lung cancer. Indeed, the lack of response in many tumors indicates primary resistance that can originate from either tumor cells (intrinsic) or tumor microenvironment (extrinsic). On the other hand, some tumors show an initial response to immunotherapy followed by relapse in few months (acquired resistance). Understanding the underlying molecular mechanisms of immunotherapy resistance makes it possible to develop effective strategies to overcome this hurdle and boost therapy outcomes. In this review, we take a look at immunotherapy strategies and go through a number of primary and acquired resistance mechanisms. Also, we present various ongoing methods to overcoming resistance and introduce some promising fields to improve the outcome of immunotherapy in patients affected with cancer.
Collapse
Affiliation(s)
- Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Salari
- Department of Medical Oncology, Hematology and Bone Marrow Transplantation, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Laird DW, Penuela S. Pannexin biology and emerging linkages to cancer. Trends Cancer 2021; 7:1119-1131. [PMID: 34389277 DOI: 10.1016/j.trecan.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022]
Abstract
Pannexins are a family of glycoproteins that comprises three members, PANX1, PANX2, and PANX3. The widely expressed and interrogated PANX1 forms heptameric membrane channels that primarily serve to connect the cytoplasm to the extracellular milieu by being selectively permeable to small signaling molecules when activated. Apart from notable exceptions, PANX1 in many tumor cells appears to facilitate tumor growth and metastasis, suggesting that pannexin-blocking therapeutics may have utility in cancer. Attenuation of PANX1 function must also consider the fact that PANX1 is found in stromal cells of the tumor microenvironment (TME), including immune cells. This review highlights the key discoveries of the past 5 years that suggest pannexins facilitate, or in some cases inhibit, tumor cell growth and metastasis via direct protein interactions and through the regulated efflux of signaling molecules.
Collapse
Affiliation(s)
- Dale W Laird
- Department of Anatomy and Cell Biology, and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Oncology, Divisions of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|