1
|
Elfiky AM, Eid MM, El-Manawaty M, Elshahid ZA, Youssef EM, Mahmoud K. Production of novel theranostic nano-vector based on superparamagnetic iron oxide nanoparticles/miR-497 targeting colorectal cancer. Sci Rep 2025; 15:4247. [PMID: 39905036 DOI: 10.1038/s41598-025-88165-3] [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: 07/24/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
Colorectal cancer (CRC) is a serious public health concern worldwide. Immune checkpoint inhibition medication is likely to remain a crucial part of CRC clinical management. This study aims to create new super paramagnetic iron oxide nano-carrier (SPION) that can effectively transport miRNA to specific CRC cell lines. In addition, evaluate the efficiency of this nano-formulation as a therapeutic candidate for CRC. Bioinformatics tools were used to select a promising tumor suppressor miRNA (mir-497-5p). Green route, using Fusarium oxyporium fungal species, manipulated for the synthesis of SPION@Ag@Cs nanocomposite as a carrier of miR-497-5p. That specifically targets the suppression of PD1/PDL1 and CTLA4pathways for colorectal therapy. UV/visible and FTIR spectroscopy, Zeta potential and MTT were used to confirm the allocation of the miR-497 on SPION@Ag@Cs and its cytotoxicity against CRC cell lines. Immunofluorescence was employed to confirm transfection of cells with miR-497@NPs, and the down- regulation of CTLA4 in HT29, and Caco2 cell lines. On the other hand, PDL1 showed a significant increase in colorectal cell lines (HT-29 and Caco-2) in response to mir497-5p@Nano treatment. The data suggest that the mir-497 -loaded SPION@Ag@Cs nano-formulation could be a good candidate for the suppression of CTLA4in CRC human cell lines. Consequently, the targeting miR-497/CTLA4 axis is a potential immunotherapy treatment strategy for CRC.
Collapse
Affiliation(s)
- Asmaa M Elfiky
- Environmental and Occupational Medicine Department, Environment and Climate Change Research Institute, National Research Centre, Cairo, Egypt.
| | - May M Eid
- Physics Institute, National Research Center, Dokki, Cairo, Egypt
| | - May El-Manawaty
- Pharmaceutical Sciences Institute, Department of Pharmacognosy, National Research Centre, Cairo, Egypt
| | - Zeinab A Elshahid
- Chemistry of Natural and Microbial Products, Pharmaceutical Industry Research Institute, National Research Centre, Cairo, Egypt
| | | | - Khaled Mahmoud
- Pharmaceutical Sciences Institute, Department of Pharmacognosy, National Research Centre, Cairo, Egypt
| |
Collapse
|
2
|
Walmsley CS, Schoepflin Z, De Brabandt C, Rangachari D, Berwick S, Patell R. Hemophagocytic lymphohistiocytosis associated with immune checkpoint inhibitor use: A review of the current knowledge and future directions. Blood Cells Mol Dis 2025; 110:102896. [PMID: 39366077 DOI: 10.1016/j.bcmd.2024.102896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a severe and often lethal inflammatory syndrome characterized by excessive immune activation leading to fever, cytopenias, and multiorgan involvement. Immune checkpoint inhibitors (ICIs) are central to many contemporary cancer regimens, but their use is associated with immune-related adverse events. Here, we report a case of ICI-induced HLH successfully treated with single agent dexamethasone and provide a scoping review of the literature for cases of ICI-induced HLH with a focus on treatment strategies and outcomes. Using the Medline database, we searched for cases of ICI-associated HLH, with a total of 51 cases reported between 2017 and 2023. Our results underscore the severe nature of this disease, with a 13.7 % mortality rate across 51 case reports. Treatment strategies for ICI-induced HLH were variable: steroids alone (56.9 %), steroids with etoposide (17.6 %), steroids with tociluzumab (11.8 %), among other combinations. Our literature review indicates that steroids alone may be sufficient treatment in some cases of ICI-HLH, with comparable mortality with steroids alone (n = 29) (13.8 %) to that of cases treated with both steroids and immunomodulators (n = 15, 13.3 %). Moreover, all patients treated with steroids and tocilizumab survived (n = 6), suggesting that tocilizumab may be a reasonable next line of therapy when steroid monotherapy proves inadequate. We propose an outline for investigation and treatment of this rare complication of ICI use. Finally, we discuss possible future approaches to develop evidence-based strategies for the diagnosis and management of ICI-induced HLH including the importance of integrating the role of patient community involvement.
Collapse
Affiliation(s)
- Charlotte S Walmsley
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Zachary Schoepflin
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Charlotte De Brabandt
- Breast Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Patient Ambassador Program, American Cancer Society, Boston, MA, United States of America
| | - Deepa Rangachari
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Shana Berwick
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Rushad Patell
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America.
| |
Collapse
|
3
|
Murugan AK, Kannan S, Alzahrani AS. Immune checkpoint expression and therapeutic implications in IDH1-mutant and wild-type glioblastomas. Curr Probl Cancer 2025; 55:101182. [PMID: 39864140 DOI: 10.1016/j.currproblcancer.2025.101182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/17/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
Programmed cell death protein 1 (PDCD1) and cluster of differentiation 274 (CD274) expression is implicated in escaping tumors from immune surveillance. Immune checkpoint inhibitors show promise in cancer therapy, yet their efficacy in glioblastomas, particularly with IDH1 mutations, remains unclear. This study analyzed two independent NGS datasets (n = 577 and n = 153) from TCGA to investigate the expression of PDCD1 and CD274 in glioblastomas and their relationship with IDH1 mutations. We used cBioPortal for mutation analysis, RNA seq for expression analysis, miRDB and miRabel for differential expression of miRNAs, and Kaplan-Meier for survival prediction. We found that 5.4% of glioblastomas harbored IDH1 mutations, correlating with improved overall survival (OS) (p = 2.196e-3). Different glioblastoma cohorts showed a diverse IDH1 mutational prevalence (4-31%). Despite this, IDH1Mu was consistently associated with better OS (p = 8.235e-5). Notably, PDCD1 and CD274 were statistically significantly highly expressed in both IDH1Wt (p < 0.0001) and IDH1Mu tumors (p < 0.0001), with higher expression linked to poorer survival outcomes (PDCD1: p = 0.009; CD274: p = 0.02). Differential co-expression analyses revealed distinct gene and miRNA profiles for IDH1Wt and IDH1Mu glioblastomas, with specific upregulation of PTEN and downregulation of MUC16 in IDH1Wt, and upregulation of PIK3R1 in IDH1Mu. Additionally, PIK3R1 and ITGB2 emerged as critical druggable targets. Our findings indicate that PDCD1 and CD274 are highly expressed irrespective of IDH1 mutation statuses, suggesting that glioblastomas could benefit from immunotherapy. Moreover, IDH1Mu glioblastomas may require a combination of PI3K/AKT/mTOR inhibitors and immunotherapy due to PIK3R1 overexpression.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia
| |
Collapse
|
4
|
Zhao Z, Fetse J, Mamani UF, Guo Y, Li Y, Patel P, Liu Y, Lin CY, Li Y, Mustafa B, Cheng K. Development of a peptide-based tumor-activated checkpoint inhibitor for cancer immunotherapy. Acta Biomater 2025; 193:484-497. [PMID: 39716541 PMCID: PMC11788053 DOI: 10.1016/j.actbio.2024.12.051] [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: 08/26/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
Antibody-based checkpoint inhibitors have achieved great success in cancer immunotherapy, but their uncontrollable immune-related adverse events remain a major challenge. In this study, we developed a tumor-activated nanoparticle that is specifically active in tumors but not in normal tissues. We discovered a short anti-PD-L1 peptide that blocks the PD-1/PD-L1 interaction. The peptide was modified with a PEG chain through a novel matrix metalloproteinase-2 (MMP-2)-specific cleavage linker. The modified TR3 peptide self-assembles into a micelle-like nanoparticle (TR3-M-NP), which remains inactive and unable to block the PD-1/PD-L1 interaction in its native form. However, upon cleavage by MMP-2 in tumors, it releases the active peptide. The TR3-M-NP5k nanoparticle was specifically activated in tumors through enzyme-mediated cleavage, leading to the inhibition of tumor growth and extended survival compared to control groups. In summary, TR3-M-NP shows great potential as a tumor-responsive immunotherapy agent with reduced toxicities. STATEMENT OF SIGNIFICANCE: In this study, we developed a bioactive peptide-based checkpoint inhibitor that is active only in tumors and not in normal tissues, thereby potentially avoiding immune-related adverse effects. We discovered a short anti-PD-L1 peptide, TR3, that blocks the PD-1/PD-L1 interaction. We chemically modified the TR3 peptide to self-assemble into a micelle-like nanoparticle (TR3-M-NP), which itself cannot block the PD-1/PD-L1 interaction but releases the active TR3 peptide in tumors upon cleavage by MMP-2. In contrast, the nanoparticle is randomly degraded in normal tissues into peptides fragments that cannot block the PD-1/PD-L1 interaction. Upon intraperitoneal injection, TR3-M-NP5k was activated specifically in tumors through enzyme cleavage, leading to the inhibition of tumor growth and extended survival compared to the control groups. In summary, TR3-M-NP holds significant promise as a tumor-responsive immunotherapy agent with reduced toxicities. The bioactive platform has the potential to be used for other types of checkpoint inhibitor.
Collapse
Affiliation(s)
- Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - John Fetse
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Umar-Farouk Mamani
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yuhan Guo
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yuanke Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Pratikkumar Patel
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Chien-Yu Lin
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yongren Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Bahaa Mustafa
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
5
|
Irsara C, Weissenbacher A, Krendl FJ, Anliker M, Hofmann J, Hautz T, Schneeberger S, Griesmacher A, Loacker L. Expression of sPD-L1 levels in an ex vivo liver perfusion model. Clin Exp Immunol 2025; 219:uxae094. [PMID: 39435859 PMCID: PMC11773811 DOI: 10.1093/cei/uxae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/13/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
The programmed cell death protein 1 (PD-1) acts as a central inhibitory immune checkpoint receptor. The soluble form of its primary ligand, sPD-L1, was found to be elevated in the serum of patients with cancer, infectious diseases, and chronic inflammation. So far, the hepatic origin of sPD-L1 has received relatively little attention and is therefore the subject of this study in the context of normothermic machine perfusion (NMP) of liver grafts. sPD-L1 concentrations as well as several well-established clinically relevant laboratory parameters were determined in the perfusate of 16 donor liver grafts undergoing NMP up to 30 hours. sPD-L1 levels continuously increased during NMP and significantly correlated with markers of hepatic synthesis (cholinesterase), acute-phase proteins (von Willebrand factor, procalcitonin, antithrombin, interleukin-6, fibrinogen), and liver decay markers (gamma-glutamyltransferase, alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase). Perfusate leukocytes were in the lower reference range and decreased after 12 hours. Mean sPD-L1 levels in the perfusate correlated with donor levels of gamma-glutamyltransferase, alanine aminotransferase, creatinine, and blood urea nitrogen. Our study reveals a significant increase in the concentration of sPD-L1 following ischemia-reperfusion injury in a hepatic ex vivo model. sPD-L1 concentrations during NMP correlate with established acute-phase proteins and liver cell decay markers, suggesting that hepatic sPD-L1 synthesis or shedding increases during the acute phase and cell decay. Furthermore, sPD-L1 correlates with established liver function and synthesis parameters as well as with donor laboratory values and might therefore be a potential biomarker for the hepatic function of liver grafts.
Collapse
Affiliation(s)
- Christian Irsara
- Central Institute of Clinical and Chemical Laboratory Diagnostics, University Hospital of Innsbruck, Innsbruck, Austria
| | - Annemarie Weissenbacher
- Department of Visceral, Transplant and Thoracic Surgery, and organLife Laboratory, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Julius Krendl
- Department of Visceral, Transplant and Thoracic Surgery, and organLife Laboratory, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Anliker
- Central Institute of Clinical and Chemical Laboratory Diagnostics, University Hospital of Innsbruck, Innsbruck, Austria
| | - Julia Hofmann
- Department of Visceral, Transplant and Thoracic Surgery, and organLife Laboratory, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Theresa Hautz
- Department of Visceral, Transplant and Thoracic Surgery, and organLife Laboratory, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, and organLife Laboratory, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Griesmacher
- Central Institute of Clinical and Chemical Laboratory Diagnostics, University Hospital of Innsbruck, Innsbruck, Austria
| | - Lorin Loacker
- Central Institute of Clinical and Chemical Laboratory Diagnostics, University Hospital of Innsbruck, Innsbruck, Austria
| |
Collapse
|
6
|
Liu C, Yin Q, Wu Z, Li W, Huang J, Chen B, Yang Y, Zheng X, Zeng L, Wang J. Inflammation and Immune Escape in Ovarian Cancer: Pathways and Therapeutic Opportunities. J Inflamm Res 2025; 18:895-909. [PMID: 39867950 PMCID: PMC11762012 DOI: 10.2147/jir.s503479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecological malignancies, largely due to its late-stage diagnosis and high recurrence rates. Chronic inflammation is a critical driver of OC progression, contributing to immune evasion, tumor growth, and metastasis. Inflammatory cytokines, including IL-6, TNF-α, and IL-8, as well as key signaling pathways such as nuclear factor kappa B (NF-kB) and signal transducer and activator of transcription 3 (STAT3), are upregulated in OC, promoting a tumor-promoting environment. The tumor microenvironment (TME) is characterized by immune cells like tumor-associated macrophages (TAMs) and regulatory T cells (Tregs), which suppress anti-tumor immune responses, facilitating immune evasion. Furthermore, OC cells utilize immune checkpoint pathways, including PD-1/PD-L1, to inhibit cytotoxic T cell activity. Targeting these inflammatory and immune evasion mechanisms offers promising therapeutic strategies. COX-2 inhibitors, Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway blockers, and NF-kB inhibitors have shown potential in preclinical studies, while immune checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 have been explored with mixed results in OC. Additionally, emerging research on the microbiome and inflammation-related biomarkers, such as microRNAs (miRNAs) and exosomes, points to new opportunities for early detection and precision medicine. Future approaches to OC treatment must focus on personalized strategies that target the inflammatory TME, integrating anti-inflammatory therapies with immunotherapy to enhance patient outcomes. Continued research into the interplay between inflammation and immune evasion in OC is essential for developing effective, long-lasting treatments.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Qinan Yin
- Department of Radiation Oncology, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, People’s Republic of China
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Zhaoying Wu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Wenhui Li
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Jun Huang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Bo Chen
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yanjun Yang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Xuewei Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Li Zeng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Jingjing Wang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| |
Collapse
|
7
|
Manole S, Nguyen DH, Min JJ, Zhou S, Forbes N. Setting "cold" tumors on fire: Cancer therapy with live tumor-targeting bacteria. MED 2025; 6:100549. [PMID: 39689707 DOI: 10.1016/j.medj.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/18/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Immunotherapy with checkpoint blockade has shown remarkable efficacy in many patients with a variety of different types of cancer. However, the majority of patients with cancer have yet to benefit from this revolutionary therapy. Studies have shown that checkpoint blockade works best against immune-inflamed tumors characterized by the presence of tumor-infiltrating lymphocytes (TILs). In this review, we summarize studies using live tumor-targeting bacteria to treat cancer and describe various strategies to engineer the tumor-targeting bacteria for maximized immunoregulatory effects. We propose that tumor-localized infections by such engineered bacteria can create an immune microenvironment in favor of a more effective antitumor immunity with or without other therapies, such as immune checkpoint blockade (ICB). Finally, we will briefly outline some exemplary oncology clinical trials involving ICB plus live therapeutic bacteria, with a focus on their ability to modulate antitumor immune responses.
Collapse
Affiliation(s)
- Simin Manole
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeonnam 58128, South Korea.
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Neil Forbes
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA; Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA; Department of Microbiology, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
8
|
Song X, Chavez-Fuentes JC, Ma W, Fu W, Wang P, Yuan GC. sCCIgen: A high-fidelity spatially resolved transcriptomics data simulator for cell-cell interaction studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631830. [PMID: 39829773 PMCID: PMC11741276 DOI: 10.1101/2025.01.07.631830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Spatially resolved transcriptomics (SRT) provides an invaluable avenue for examining cell-cell interactions within native tissue environments. The development and evaluation of analytical tools for SRT data necessitate tools for generating synthetic datasets with known ground truth of cell-cell interaction induced features. To address this gap, we introduce sCCIgen, a novel real-data-based simulator tailored to generate high-fidelity SRT data with a focus on cell-cell interactions. sCCIgen preserves transcriptomic and spatial characteristics in SRT data, while comprehensively models various cell-cell interaction features, including cell colocalization, spatial dependence among gene expressions, and gene-gene interactions between nearby cells. We implemented sCCIgen as an interactive, easy-to-use, realistic, reproducible, and well-documented tool for studying cellular interactions and spatial biology.
Collapse
|
9
|
Peg V, Abengozar-Muela M, Acosta J, Andrés L, García-Rojo M, Hardisson D, Nicolau MJ, Ramos-Oliver I, Rodrigo M, Sánchez-Bernal ML, Sanz J, Garrote L, Ramírez I, Rojo F. New Approach in the Interpretation of Complex Triple-negative Breast Cancer Immunohistochemistry Specimens Processed With VENTANA PD-L1 (SP142) Assay. Appl Immunohistochem Mol Morphol 2025; 33:15-21. [PMID: 39636314 DOI: 10.1097/pai.0000000000001237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/13/2024] [Indexed: 12/07/2024]
Abstract
Triple-negative breast cancer (TNBC) is challenging to treat because of its lack of specific molecular targets. The IMMUNOPEG study aimed to evaluate a novel structured method for interpreting TNBC immunohistochemistry specimens processed with VENTANA PD-L1 (SP142) assay. The study involved 10 pathologists who evaluated 50 different immunohistochemistry specimens of TNBC with programmed death ligand 1 (PD-L1) expression considered challenging and that were previously evaluated by the scientific committee, using the NAVIFY Digital Pathology platform. Initially, the overall percent agreement (OPA) was 74%, with a negative percent agreement (NPA) of 68.2% for samples classified as negative, and a positive percent agreement (PPA) of 94.5% for positive samples. After training on the method, the OPA improved significantly to 81.6%, with the NPA increasing to 80.5% and the PPA decreasing to 85.5%. The mean percentage of the tumor area occupied by PD-L1-stained immune cells decreased from 2.5% to 1.6% post-training, approaching to the scientific committee's consensus of 1.029%. The study found that the pathologists' confidence in their assessments increased significantly when using the structured method, which was found to be easy to use by 9 out of 10 pathologists. All pathologists agreed that the structured method was useful for assessing PD-L1 expression. The study suggests that this method has potential value in interpreting challenging cases of PD-L1 immunohistochemistry (IHC) in TNBC. Further refinement and a training protocol may be necessary to enhance the method's efficiency. The potential for generalizing this structured method to other IHC procedures and pathologies warrants additional research.
Collapse
Affiliation(s)
- Vicente Peg
- Department of Pathology, Vall d'Hebron University Hospital
- Universidad Autónoma de Barcelona, Barcelona
| | | | - Jesús Acosta
- Department of Anatomic Pathology, Hospital General Universitario Santa Lucía, Cartagena
| | - Leire Andrés
- Department of Pathology, Hospital Universitario Cruces, Bilbao
| | - Marcial García-Rojo
- Department of Pathology, Hospital Universitario de Jerez, Jerez de la Frontera
| | | | - María Jesús Nicolau
- Department of Pathology, Hospital General Universitario de Castellón, Castellón
| | - Irma Ramos-Oliver
- Department of Pathology, Hospital Universitari Doctor Josep Trueta, Girona
| | | | | | - Julián Sanz
- Department of Pathology, Clínica Universidad de Navarra, Madrid
| | - Leia Garrote
- Medical Department, Roche Farma S.A., Madrid, Spain
| | | | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz, Madrid, Spain
| |
Collapse
|
10
|
Xu HZ, Chen FX, Li K, Zhang Q, Han N, Li TF, Xu YH, Chen Y, Chen X. Anti-lung cancer synergy of low-dose doxorubicin and PD-L1 blocker co-delivered via mild photothermia-responsive black phosphorus. Drug Deliv Transl Res 2025; 15:269-290. [PMID: 38597996 DOI: 10.1007/s13346-024-01595-w] [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] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
We have previously identified a latent interaction mechanism between non-small cell lung cancer cells (NSCLCC) and their associated macrophages (TAM) mediated by mutual paracrine activation of the HMGB1/RAGE/NF-κB signaling. Activation of this mechanism results in TAM stimulation and PD-L1 upregulation in the NSCLCC. In the present work, we found that free DOX at a low concentration that does not cause DNA damage could activate the HMGB1/RAGE/NF-κB/PD-L1 pathway byinducing oxidative stress. It was thus proposed that a combination of low-dose DOX and a PD-L1 blocker delivered in the NSCLC tumor would achieve synergistic TAM stimulation and thereby synergetic anti-tumor potency. To prove this idea, DOX and BMS-202 (a PD-L1 blocker) were loaded to black phosphorus (BP) nanoparticles after dosage titration to yield the BMS-202/DOX@BP composites that rapidly disintegrated and released drug cargo upon mild photothermal heating at 40 °C. In vitro experiments then demonstrated that low-dose DOX and BMS-202 delivered via BMS-202/DOX@BP under mild photothermia displayed enhanced tumor cell toxicity with a potent synergism only in the presence of TAM. This enhanced synergism was due to an anti-tumor M1-like TAM phenotype that was synergistically induced by low dose DOX plus BMS-202 only in the presence of the tumor cells, indicating the damaged tumor cells to be the cardinal contributor to the M1-like TAM stimulation. In vivo, BMS-202/DOX@BP under mild photothermia exhibited targeted delivery to NSCLC graft tumors in mice and synergistic anti-tumor efficacy of delivered DOX and BMS-202. In conclusion, low-dose DOX in combination with a PD-L1 blocker is an effective strategy to turn TAM against their host tumor cells exploiting the HMGB1/RAGE/NF-κB/PD-L1 pathway. The synergetic actions involved highlight the value of TAM and the significance of modulating tumor cell-TAM cross-talk in tumor therapy. Photothermia-responsive BP provides an efficient platform to translate this strategy into targeted, efficacious tumor therapy.
Collapse
Affiliation(s)
- Hua-Zhen Xu
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Fei-Xiang Chen
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Ke Li
- Center for Lab Teaching, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Quan Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Ning Han
- School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Hubei, 442000, China
| | - Tong-Fei Li
- School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Hubei, 442000, China
| | - Yong-Hong Xu
- Institute of Ophthalmological Research, Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yun Chen
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China.
| | - Xiao Chen
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430072, China.
| |
Collapse
|
11
|
Yue H, Li Y, Yang T, Wang Y, Bao Q, Xu Y, Liu X, Miao Y, Yang M, Mao C. Filamentous phages as tumour-targeting immunotherapeutic bionanofibres. NATURE NANOTECHNOLOGY 2025; 20:167-176. [PMID: 39468354 DOI: 10.1038/s41565-024-01800-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 09/04/2024] [Indexed: 10/30/2024]
Abstract
Programmed cell death-ligand 1 (PD-L1) blockers have advanced immunotherapy, but their lack of tumour homing capability represents a substantial challenge. Here we show that genetically engineered filamentous phages can be used as tumour-targeting immunotherapeutic agents that reduce the side effects caused by untargeted delivery of PD-L1 blockers. Specifically, we improved biopanning to discover a peptide binding the extracellular domain of PD-L1 and another targeting both melanoma tissues and cancer cells. The two peptides were genetically fused to the sidewall protein and tip protein of fd phages, respectively. The intravenously injected phages homed to tumours and bound PD-L1 on cancer cells, effectively blocking PD-1/PD-L1 recognition to trigger targeted immunotherapy without body weight loss, organ abnormalities and haematological aberrations. The phages, cost-effectively replicated by bacteria, are cancer-targeting immunotherapeutic nanofibres that can be flexibly designed to target different cancer types and immune checkpoints.
Collapse
Affiliation(s)
- Hui Yue
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Yan Li
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, P. R. China
| | - Tao Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Yecheng Wang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Qing Bao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Yajing Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Xiangyu Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Yao Miao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, P. R. China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China.
| |
Collapse
|
12
|
Hashim GM, Shahgolzari M, Hefferon K, Yavari A, Venkataraman S. Plant-Derived Anti-Cancer Therapeutics and Biopharmaceuticals. Bioengineering (Basel) 2024; 12:7. [PMID: 39851281 PMCID: PMC11759177 DOI: 10.3390/bioengineering12010007] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025] Open
Abstract
In spite of significant advancements in diagnosis and treatment, cancer remains one of the major threats to human health due to its ability to cause disease with high morbidity and mortality. A multifactorial and multitargeted approach is required towards intervention of the multitude of signaling pathways associated with carcinogenesis inclusive of angiogenesis and metastasis. In this context, plants provide an immense source of phytotherapeutics that show great promise as anticancer drugs. There is increasing epidemiological data indicating that diets rich in vegetables and fruits could decrease the risks of certain cancers. Several studies have proved that natural plant polyphenols, such as flavonoids, lignans, phenolic acids, alkaloids, phenylpropanoids, isoprenoids, terpenes, and stilbenes, could be used in anticancer prophylaxis and therapeutics by recruitment of mechanisms inclusive of antioxidant and anti-inflammatory activities and modulation of several molecular events associated with carcinogenesis. The current review discusses the anticancer activities of principal phytochemicals with focus on signaling circuits towards targeted cancer prophylaxis and therapy. Also addressed are plant-derived anti-cancer vaccines, nanoparticles, monoclonal antibodies, and immunotherapies. This review article brings to light the importance of plants and plant-based platforms as invaluable, low-cost sources of anti-cancer molecules of particular applicability in resource-poor developing countries.
Collapse
Affiliation(s)
- Ghyda Murad Hashim
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Mehdi Shahgolzari
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan 65175-4171, Iran
| | - Kathleen Hefferon
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Afagh Yavari
- Department of Biology, Payame Noor University, Tehran P.O. Box 19395-3697, Iran
| | - Srividhya Venkataraman
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| |
Collapse
|
13
|
Dai Y, Ruan T, Yang W, Liu S, Chen J, Fang Y, Li Q. Efficacy and Safety of Paclitaxel-Based PD-1/PD-L1 Immunotherapies for Triple-Negative Breast Cancer: A Systematic Review and Network Meta-Analysis. Clin Med Insights Oncol 2024; 18:11795549241308072. [PMID: 39734512 PMCID: PMC11672372 DOI: 10.1177/11795549241308072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Background Triple negative breast cancer (TNBC) is a deadly subtype of breast cancer with limited treatment options. Currently, programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors have become the first choice for breast cancer immunotherapies. Despite paclitaxel being considered a cornerstone drug in breast cancer treatment, the effectiveness, safety, and optimal drug selection for its combination with PD-1/PD-L1 inhibitors remain uncertain. Methods We conducted a systematic review and network meta-analysis, performing a comprehensive literature search across PubMed, Embase, and the Cochrane Library from the inception of each database through May 18, 2024. Selected trials were those that assessed the efficacy and safety of paclitaxel-based PD-1/PD-L1 therapies for the treatment of TNBC. The primary endpoint assessed was overall survival (OS), while secondary outcomes included progression-free survival (PFS), adverse events (AEs), overall response rate (ORR), and Pathological complete response (pCR). This study is registered in PROSPERO under registration number CRD42023429651. Results A total of 8 RCTs meeting our eligibility criteria were included, involving 4626 patients who received either Paclitaxel (Paclitaxel-placebo/chemotherapy) or a combination of durvalumab, pembrolizumab, atezolizumab, toripalimab with paclitaxel. The pooled results demonstrated that Durvalumab combined with Paclitaxel significantly reduced the hazard ratio for OS (surface under the cumulative ranking [SUCRA]: 91.05%) and PFS compared with Paclitaxel alone (SUCRA: 83.52%). Additionally, Durvalumab plus Paclitaxel significantly improved the ORR compared with Paclitaxel (odds ratio [OR]: 2.30; 95% credible interval [CrI]: 1.10-5.20). For safety outcomes, Atezolizumab plus Paclitaxel showed a favorable profile in AEs, with no significant differences observed between groups. In the pCR study, Pembrolizumab plus Paclitaxel was the most effective treatment option (SUCRA: 81.85%). Conclusions When combined with paclitaxel, PD-1/PD-L1 inhibitors exhibit a favorable survival benefit. The combination of Durvalumab and paclitaxel represents the optimal treatment option. In the future, attention should be paid to the TNBC subtypes and drug dosage, as these factors may help to design personalized TNBC treatment programs.
Collapse
Affiliation(s)
| | - Tianyin Ruan
- Institute of Hepatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenhui Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shan Liu
- Center of Clinical Evaluation and Analysis, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jiahao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingying Fang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiushuang Li
- Center of Clinical Evaluation and Analysis, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
14
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [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: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
15
|
Yang D, Karanth SD, Yoon HS, Yang JJ, Lou X, Bian J, Zhang D, Guo Y, Yaghjyan L, Akinyemiju T, Rodriguez E, Mehta HJ, Braithwaite D. Disparities in Utilization of Immune Checkpoint Inhibitor Therapy Among Older Patients With Advanced Non-Small Cell Lung Cancer: A SEER-Medicare Analysis. JCO ONCOLOGY ADVANCES 2024; 1:e2400008. [PMID: 39758136 PMCID: PMC11698018 DOI: 10.1200/oa.24.00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE In the United States, there are disparities in access to care for patients with non-small cell lung cancer (NSCLC) on the basis of socioeconomic and racial/ethnic factors. This study investigates the association between race/ethnicity and the utilization of immune checkpoint inhibitor (ICI) therapy among older patients with advanced NSCLC (aNSCLC). METHODS This retrospective study used data from the SEER-Medicare-linked database. The cohort included patients (age 66 years or older) diagnosed with aNSCLC (stage III/IV) between March 2015 and December 2017, and they were followed through December 2019. Race/ethnicity was categorized as non-Hispanic (NH)-White, NH-Black, Hispanic, and Other. ICI therapy utilization was determined by identifying any usage of ICI agents (nivolumab, pembrolizumab, atezolizumab, durvalumab, ipilimumab, and cemiplimab-rwlc) from the Medicare database. Multivariable logistic regression models assessed the association between race/ethnicity and ICI therapy utilization (yes, no). Effect measure modification analyses were conducted by sex, socioeconomic status, and comorbidity. RESULTS The final sample included 26,836 patients; 76.2% were NH-White, 10.1% NH-Black, 5.7% Hispanic, and 8.0% Other. The overall ICI therapy utilization proportion was 17.8%, varying across ethnicities: NH-Black 14.1%, Hispanic 16.3%, NH-White 18.4%, and Other 18.5%. In comparison with NH-White patients, NH-Black patients were 15% less likely to receive ICI therapy (adjusted odds ratio, 0.85 [95% CI, 0.75 to 0.96]). Furthermore, the association between race/ethnicity and utilization of ICI therapy was modified by comorbidity status, sex, and socioeconomic status. CONCLUSION NH-Black patients with aNSCLC were less likely to receive ICI therapy than their NH-White counterparts. Our findings indicate the racial/ethnic disparities in ICI therapy utilization and call for further interventions to optimize access to care.
Collapse
Affiliation(s)
- Danting Yang
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
| | - Shama D. Karanth
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Hyung-Suk Yoon
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Jae Jeong Yang
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Xiwei Lou
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | - Jiang Bian
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | | | - Yi Guo
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | - Lusine Yaghjyan
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
| | - Tomi Akinyemiju
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC
- Duke Cancer Institute, School of Medicine, Duke University, Durham, NC
| | | | - Hiren J. Mehta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - Dejana Braithwaite
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
16
|
Vanders RL, Gomez HM, Daly K, Wark PA, Horvat JC, Hansbro PM. Immune checkpoints are suppressed during pregnancy following influenza A virus infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L890-L904. [PMID: 39254092 DOI: 10.1152/ajplung.00391.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Influenza A virus (IAV) infection is a major health risk during pregnancy. Although vaccination and antiviral agents are widely used and reduce IAV-induced symptoms, they are not sufficient to control IAV infections in pregnancy, especially during pandemics. Respiratory viruses like IAV exploit immune alterations that occur during pregnancy, including the upregulation of immune checkpoint proteins (ICPs) like programmed death ligand-1 (PDL1), programmed cell death receptor 1 (PD1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). We hypothesize that blocking expression of PDL1 on innate immune cells will improve maternal immunity following IAV infection. We used murine models of IAV infection during pregnancy with and without treatment with the immune checkpoint inhibitor (ICI), a-PDL1. Pregnant and nonpregnant mice were infected with mouse-adapted IAV (A/PR/8) and assessed at 3 days post infection (3 dpi). Lung cells were analyzed using flow cytometry. Lung mRNA expression of inflammatory and antiviral markers and histology was measured. Protein concentrations of inflammatory and antiviral markers, as well as viral titers were measured from lung bronchiolar lavage fluid (BALF). Lung function was also assessed. Following IAV infection, immune cells from pregnant mice had significant increases in the ICPs, PDL1, PD1, and CTLA4. a-PDL1 treatment effectively suppressed these ICPs and increased the activation marker, CD86. a-PDL1 treatment also reduced lung inflammatory cell infiltration and viral titers, increased antiviral responses, and improved lung function. Overall, IAV infection in pregnancy activates key inhibitory ICPs, leading to worsened disease outcomes. a-PDL1 treatment during IAV infection in pregnancy is an effective method to reduce ICP expression and improve overall immune cell responses.NEW & NOTEWORTHY Influenza infection worsens disease outcomes during pregnancy; however, treatment with anti-PDL1 can restore immune function during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Katie Daly
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Huelse JM, Bhasin SS, Jacobsen KM, Yim J, Thomas BE, Branella GM, Bakhtiari M, Chimenti ML, Baxter TA, Raikar SS, Wang X, Frye SV, Henry CJ, Earp HS, Bhasin M, DeRyckere D, Graham DK. MERTK inhibition selectively activates a DC - T-cell axis to provide anti-leukemia immunity. Leukemia 2024; 38:2685-2698. [PMID: 39322710 DOI: 10.1038/s41375-024-02408-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/27/2024]
Abstract
TAM-family tyrosine kinases (TYRO3, AXL and MERTK) are potential cancer therapeutic targets. In previous studies MERTK inhibition in the immune microenvironment was therapeutically effective in a B-cell acute leukemia (B-ALL) model. Here, we probed anti-leukemia immune mechanisms and evaluated roles for TYRO3 and AXL in the leukemia microenvironment. Host Mertk knock-out or MERTK inhibitor MRX-2843 increased CD8α+ dendritic cells (DCs) with enhanced antigen-presentation capacity in the leukemia microenvironment and inhibited leukemogenesis. High MERTK or low DC gene expression were associated with poor prognosis in pediatric ALL patients, indicating the clinical relevance of these findings. MRX-2843 increased CD8+ T-cell numbers and prevented induction of exhaustion markers, implicating a DC - T-cell axis. Indeed, combined depletion of CD8α+ DCs and CD8+ T-cells was required to abrogate anti-leukemia immunity in Mertk-/- mice. Tyro3-/- mice were also protected against B-ALL, implicating TYRO3 as an immunotherapeutic target. In contrast to Mertk-/- mice, Tyro3-/- did not increase CD8α+ DCs with enhanced antigen-presentation capacity and therapeutic activity was less dependent on DCs, indicating a different immune mechanism. Axl-/- did not impact leukemogenesis. These data demonstrate differential TAM kinase roles in the leukemia microenvironment and provide rationale for development of MERTK and/or TYRO3-targeted immunotherapies.
Collapse
Affiliation(s)
- Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Swati S Bhasin
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Kristen M Jacobsen
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Juhye Yim
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Beena E Thomas
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Gianna M Branella
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Mojtaba Bakhtiari
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Madison L Chimenti
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Travon A Baxter
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Sunil S Raikar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 27599, USA
| | - Stephen V Frye
- Center for Integrative Chemical Biology and Drug Discovery and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 27599, USA
| | - Curtis J Henry
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - H Shelton Earp
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 27599, USA
- Departments of Medicine and Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Manoj Bhasin
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
- Cancer Immunology Program, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, 30322, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
18
|
Janssen JC, van Dijk B, Hoeijmakers LL, Grünhagen DJ, Bramer WM, Verhoef C, de Gruijl TD, Blank CU, van der Veldt AAM. Local administration of immunotherapy for patients with skin cancer: A systematic review. Cancer Treat Rev 2024; 131:102848. [PMID: 39486396 DOI: 10.1016/j.ctrv.2024.102848] [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: 09/18/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024]
Abstract
Since the introduction of immune checkpoint inhibitors (ICIs) targeting PD-1 and CTLA-4 receptors, survival has improved significantly for patients with irresectable and metastatic skin cancer, including cutaneous squamous cell cancer and melanoma. However, systemic administration of these drugs is associated with immune related adverse events (irAEs), which can be severe, irreversible and even fatal. To reduce the risk of irAEs associated with systemic exposure to immunotherapeutic drugs, local administration of low doses could be considered. This systematic review provides an overview of early phase clinical trials with drugs that are currently under investigation for intratumoral administration in patients with melanoma and non-melanoma skin cancer.
Collapse
Affiliation(s)
- J C Janssen
- Department of Medical Oncology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Department of Surgical Oncology and Gastro Intestinal Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - B van Dijk
- Department of Medical Oncology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - L L Hoeijmakers
- Department of Medical Oncology, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D J Grünhagen
- Department of Surgical Oncology and Gastro Intestinal Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - W M Bramer
- Medical Library, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - C Verhoef
- Department of Surgical Oncology and Gastro Intestinal Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - T D de Gruijl
- Department of Immunology, Amsterdam UMC, University Medical Center, Amsterdam, the Netherlands
| | - C U Blank
- Department of Medical Oncology, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - A A M van der Veldt
- Department of Medical Oncology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
19
|
Shimada A, Ohnaka S, Nakashima M, Nagai A. Successful Resection of Small Intestinal Stenosis After Treatment With an Immune Checkpoint Inhibitor for Metastatic Lung Cancer in the Small Intestine: A Case Report. Cureus 2024; 16:e75909. [PMID: 39830565 PMCID: PMC11739535 DOI: 10.7759/cureus.75909] [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] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Gastrointestinal (GI) metastases from lung cancer are relatively rare, and their management strategies and outcomes in the era of immune checkpoint inhibitors are unknown. A 59-year-old man with lung cancer was hospitalized. He presented with vomiting due to a small intestinal metastasis. The metastatic lesion reduced in size with pembrolizumab treatment. However, the patient could not feed orally and required continuous central venous nutrition. Following the recurrence of a catheter-related bloodstream infection, the patient underwent small intestinal resection and gastrojejunal bypass surgery. Surgical specimens from the site of intestinal stenosis showed tumor disappearance and inflammatory cell invasion. GI stenosis due to lung cancer metastasis can persist even after effective treatment with immune checkpoint inhibitors. Therefore, surgery for GI metastases should be considered as necessary.
Collapse
Affiliation(s)
- Ayako Shimada
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Shinnosuke Ohnaka
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Masanao Nakashima
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Atsushi Nagai
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| |
Collapse
|
20
|
Maggi E, Munari E, Landolina N, Mariotti FR, Azzarone B, Moretta L. T cell landscape in the microenvironment of human solid tumors. Immunol Lett 2024; 270:106942. [PMID: 39486594 DOI: 10.1016/j.imlet.2024.106942] [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: 10/03/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
T cells are the main effectors involved in anti-tumor immunity, mediating most of the adaptive response towards cancer. After priming in lymph nodes, tumor antigens-specific naïve T lymphocytes proliferate and differentiate into effector CD4+ and CD8+ T cells that migrate from periphery into tumor sites aiming to eliminate cancer cells. Then while most effector T cells die, a small fraction persists and recirculates as long-lived memory T cells which generate enhanced immune responses when re-encountering the same antigen. A number of T (and non-T) cell subsets, stably resides in non-lymphoid peripheral tissues and may provide rapid immune response independently of T cells recruited from blood, against the reemergence of cancer cells. When tumor grows, however, tumor cells have evaded immune surveillance of effector cells (NK and CTL cells) which are exhausted, thus favoring the local expansion of T (and non-T) regulatory cells. In this review, the current knowledge of features of T cells present in the tumor microenvironment (TME) of solid adult and pediatric tumors, the mechanisms upregulating immune-checkpoint molecules and transcriptional and epigenetic landscapes leading to dysfunction and exhaustion of T effector cells are reviewed. The interaction of T cells with cancer- or TME non-neoplastic cells and their secreted molecules shape the T cell profile compromising the intrinsic plasticity of T cells and, therefore, favoring immune evasion. In this phase regulatory T cells contribute to maintain a high immunosuppressive TME thus facilitating tumor cell proliferation and metastatic spread. Despite the advancements of cancer immunotherapy, many tumors are unresponsive to immune checkpoint inhibitors, or therapeutical vaccines or CAR T cell-based adoptive therapy: some novel strategies to improve these T cell-based treatments are lastly proposed.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | | | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy.
| |
Collapse
|
21
|
Huang L, Xiao Y, Yang L, Ren S. The development for emerging biomarkers of lymphangioleiomyomatosis. Orphanet J Rare Dis 2024; 19:445. [PMID: 39614360 PMCID: PMC11605962 DOI: 10.1186/s13023-024-03455-9] [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: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, slowly progressing, low-grade metastatic tumor primarily affecting women. Currently, vascular endothelial growth factor-D (VEGF-D) is the only validated diagnostic biomarker, enabling diagnosis of LAM without the need for lung biopsy in appropriate clinical settings. However, VEGF-D concentrations are normal in about 30% of patients, rendering it insufficient for diagnosing all cases of LAM. There remains a need to identify more non-invasive, safe, sensitive, and specific biomarkers associated with LAM. Therefore, it is imperative to explore novel non-invasive, safe, and specific diagnostic methods for LAM. This article aims to review biomarkers associated with LAM, including potential biomarkers newly discovered or showing advancements in classical biomarkers widely used in LAM, and discuss their application in LAM diagnosis, assessment of disease severity, prediction of treatment response, and prognosis. (LAM) 、,。,-D (VEGF-D) , LAM。, 30% VEGF-D , LAM 。 LAM 、、。,、 LAM 。 LAM , LAM , LAM 、、。.
Collapse
Affiliation(s)
- Liting Huang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Ying Xiao
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Lulu Yang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Siying Ren
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, 410011, China.
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
22
|
Zhao Y, Zhang D, Meng B, Zhang Y, Ma S, Zeng J, Wang X, Peng T, Gong X, Zhai R, Dong L, Jiang Y, Dai X, Fang X, Jia W. Integrated proteomic and glycoproteomic analysis reveals heterogeneity and molecular signatures of brain metastases from lung adenocarcinomas. Cancer Lett 2024; 605:217262. [PMID: 39341452 DOI: 10.1016/j.canlet.2024.217262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
Brain metastasis is a major cause of poor prognosis and death in lung adenocarcinoma (LUAD); however, the understanding of therapeutic strategies and mechanisms for brain metastases from LUAD (BM-LUAD) remains notably limited, especially at the proteomics levels. To address this issue, we conducted integrated proteomic and glycoproteomic analyses on 49 BM-LUAD tumors, revealing two distinct subtypes of the disease: BM-S1 and BM-S2. Whole exome sequencing analysis revealed that somatic mutations in STK11 and KEAP1, as well as copy number deletions on chr19p13.3, such as STK11, UQCR11, and SLC25A23, were more frequently detected in BM-S2. In BM-S1 tumors, we observed significant infiltration of GFAP + astrocytes, as evidenced by elevated levels of GFAP, GABRA2, GABRG1 and GAP43 proteins and an enrichment of astrocytic signatures in both our proteomic data and external spatial transcriptomic data. Conversely, BM-S2 tumors demonstrated higher levels of PD-1 immune cell infiltration, supported by the upregulation of PD-1 and LAG-3 genes. These findings suggest distinct microenvironmental adaptations required by the different BM-LUAD subtypes. Additionally, we observed unique glycosylation patterns between the subtypes, with increased fucosylation in BM-S1 and enhanced sialylation in BM-S2, primarily affected by glycosylation enzymes such as FUT9, B4GALT1, and ST6GAL1. Specifically, in BM-S2, these sialylation modifications are predominantly localized to the lysosomes, underscoring the critical role of N-glycosylation in the tumor progression of BM-LUAD. Overall, our study not only provides a comprehensive multi-omic data resource but also offers valuable biological insights into BM-LUAD, highlighting potential mechanisms and therapeutic targets for further investigation.
Collapse
Affiliation(s)
- Yang Zhao
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Yong Zhang
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jiaming Zeng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Tao Peng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Rui Zhai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Lianhua Dong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - You Jiang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xinhua Dai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China.
| | - Xiang Fang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China.
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
23
|
Ilieva N, Pencheva M, Hadzhiev H, Tashkova D, Daskalova E, Georgiev P, Genova S. Impact of Neoadjuvant Therapy on PD-L1 Expression in Triple-Negative Breast Cancer and Correlation with Clinicopathological Factors. Diagnostics (Basel) 2024; 14:2672. [PMID: 39682581 DOI: 10.3390/diagnostics14232672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND This study aims to deliver more insights on the impact of neoadjuvant treatment on Pd-L1 expression and to evaluate its correlation with clinicopathological factors. METHODS We reviewed 88 TNBC cases for the period 2021-2023. Data on age, tumor size, stage, and treatment were collected. Histological slides were assessed for subtype, grade, and TILs. A total of 48 received neoadjuvant treatment. HER2 and Ki67 were evaluated via immunohistochemistry. PD-L1 expression was tested on primary and residual tumors. Statistical analysis was performed using IBM SPSS (p < 0.05). RESULTS In this study, PD-L1 positive expression was found in 44.3% of primary tumors, with 52.9% of initially positive cases losing expression post-treatment. TILs were significantly higher in PD-L1-positive tumors (mean 41.79% vs. 27.55%, p = 0.001). A notable correlation was found between PD-L1 expression and Ki-67 proliferation index, with PD-L1-positive tumors having a median Ki-67 of 64.49 compared to 52.86 in negative cases (p = 0.015). Neoadjuvant immunotherapy led to a lower mean residual cancer burden (0.95 vs. 2.55, p = 0.002) compared to chemotherapy alone. Higher Ki-67 levels (≥50%) were associated with better treatment outcomes, showing a mean RCB score of 1.60 versus 3.16 for lower levels (p = 0.022). HER2-negative cases had a higher prevalence of favorable pathological response (54.5%) compared to HER2-low tumors (25%, p = 0.048), because of the strong correlation to high proliferative index. CONCLUSIONS In conclusion, PD-L1 expression in TNBC shows significant discordance post-treatment, highlighting the need for routine testing and further research on predictive biomarkers.
Collapse
Affiliation(s)
- Nevena Ilieva
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Clinical Pathology Department, Complex Oncology Center Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Mina Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Hristo Hadzhiev
- First Oncological Department, Complex Oncology Center Plovdiv, Bul. Al. Stamboliyski 2A, 4000 Plovdiv, Bulgaria
| | - Desislava Tashkova
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Clinical Pathology Department, Complex Oncology Center Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Elena Daskalova
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Petar Georgiev
- Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Sylvia Genova
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| |
Collapse
|
24
|
Cui Y, Zhou X, Zhang J, Fang B, Ge J, Tang H, Liu B, He H, Xu F, Shang X. Exploiting potential molecular compounds for treating testicular seminoma by targeting immune related genes. Cell Commun Signal 2024; 22:560. [PMID: 39574183 PMCID: PMC11580341 DOI: 10.1186/s12964-024-01927-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/04/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND In cases of advanced seminoma, up to 30% of patients may manifest cisplatin resistance, necessitating aggressive salvage therapy, with a consequent 50% risk of mortality attributable to cancer. Nevertheless, beyond chemotherapy and radiotherapy, no further therapeutic modalities have been implemented for these patients. METHODS The study commenced with the identification of differentially expressed immune-related genes, which were subsequently subjected to clustering using WGCNA. Prognostic signature construction ensued through the execution of univariable Cox regression, lasso regression, and multivariable Cox regression analyses. To validate the prognostic signature, the TCGA-TGCT and GSE99420 cohorts were employed, with assessments conducted via PFS, C-index, DCA, and ROC analyses. Subsequent exploration of the immune landscape and potential immunotherapeutic applications was undertaken through Cibersort and TIDE analyses. Molecular docking and dynamics simulation techniques were then employed for screening potential molecular compounds. Validation of these findings was pursued through in vitro and vivo assays. RESULTS CTLA4, SNX17, and TMX1 were selected to construct the signature. Patients in the high-risk group exhibited diminished progression-free survival rates. The AUC for predicting survival at 1, 3, and 5 years was 0.802, 0.899, and 0.943, respectively, surpassing those of other risk factors, such as lymphovascular invasion and T stage. The C-index for the risk score was 0.838. Decision curve analysis (DCA) suggests that incorporating lymphovascular invasion and the risk score yields the most favorable decision-making outcomes for patients. Moreover, individuals classified as high-risk may derive greater benefit from immunotherapy. Molecular compounds including Rutin, ICG-001, and Doxorubicin can selectively target CTLA4, SNX17, and TMX1, respectively, thereby inhibiting the proliferation and invasive capabilities of seminoma tumor cells in vitro and vivo. CONCLUSION The signature initially constructed based on immune-related genes shows promise for predicting outcomes and assessing the efficacy of immunotherapy in seminoma patients. Rutin, ICG-001, and Doxorubicin have demonstrated potential to target these signature genes and inhibit tumor cell viability.
Collapse
Affiliation(s)
- Yankang Cui
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaodie Zhou
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jing Zhang
- Jiangsu Product Quality Testing & Inspection Institute, Nanjing, China
| | - Bo Fang
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jingping Ge
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hao Tang
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bianjiang Liu
- Department of Urology, The First Afliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haowei He
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Feng Xu
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xuejun Shang
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
25
|
Zhou Y, Pang L, Ding T, Chen K, Liu J, Wu M, Wang W, Man K. Precise In Situ Delivery of a Photo-Enhanceable Inflammasome-Activating Nanovaccine Activates Anticancer Immunity. Cancer Res 2024; 84:3834-3847. [PMID: 39288074 PMCID: PMC11565167 DOI: 10.1158/0008-5472.can-24-0220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/04/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
A variety of state-of-the-art nanovaccines (NV) combined with immunotherapies have recently been developed to treat malignant tumors, showing promising results. However, immunosuppression in the tumor microenvironment (TME) restrains cytotoxic T-cell infiltration and limits the efficacy of immunotherapies in solid tumors. Therefore, tactics for enhancing antigen cross-presentation and reshaping the TME need to be explored to enhance the activity of NVs. Here, we developed photo-enhanceable inflammasome-activating NVs (PIN) to achieve precise in situ delivery of a tumor antigen and a hydrophobic small molecule activating the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 inflammasome (NLRP3) pathway. Near-infrared light irradiation promoted PIN accumulation in tumor sites through photo-triggered charge reversal of the nanocarrier. Systematic PIN administration facilitated intratumoral NLRP3 inflammasome activation and antigen cross-presentation in antigen-presenting cells upon light irradiation at tumor sites. Furthermore, PIN treatment triggered immune responses by promoting the production of proinflammatory cytokines and activating antitumor immunity without significant systematic toxicity. Importantly, the PIN enhanced the efficacy of immune checkpoint blockade and supported the establishment of long-term immune memory in mouse models of melanoma and hepatocellular carcinoma. Collectively, this study reports a safe and efficient photoresponsive system for codelivery of antigens and immune modulators into tumor tissues, with promising therapeutic potential. Significance: The development of a photoresponsive nanovaccine with spatiotemporal controllability enables robust tumor microenvironment modulation and enhances the efficacy of immune checkpoint blockade, providing an effective immunotherapeutic strategy for cancer treatment. See related commentary by Zhen and Chen, p. 3709.
Collapse
Affiliation(s)
- Yang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Dr. Li Dak-Sum Research Centre, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li Pang
- Department of Pancreato-Biliary Surgery and Liver Transplantation Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine and HKU-SZH, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Tao Ding
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine and HKU-SZH, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Kang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Dr. Li Dak-Sum Research Centre, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jinzhao Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Dr. Li Dak-Sum Research Centre, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Meicen Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Dr. Li Dak-Sum Research Centre, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Dr. Li Dak-Sum Research Centre, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwan Man
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine and HKU-SZH, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Mureanu N, Bowman AM, Porter-Wright IA, Verma P, Efthymiou A, Nicolaides KH, Scotta C, Lombardi G, Tribe RM, Shangaris P. The Immunomodulatory Role of Regulatory T Cells in Preterm Birth and Associated Pregnancy Outcomes. Int J Mol Sci 2024; 25:11878. [PMID: 39595948 PMCID: PMC11593591 DOI: 10.3390/ijms252211878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Spontaneous preterm birth (sPTB), defined as live birth before 37 weeks of gestational age, is associated with immune dysregulation and pro-inflammatory conditions that profoundly impact newborn health. The question of immune integrity at the maternal-foetal interface is a focus of recent studies centring not only sPTB but the conditions often affiliated with this outcome. Regulatory T cells (Tregs) play a critical anti-inflammatory role in pregnancy, promoting foetal tolerance and placentation. Due to this gestational role, it is hypothesised that decreased or dysfunctional Tregs may be implicated in cases of sPTB. This review examines studies comparing Treg presence in healthy term pregnancies and those with sPTB-associated conditions. Conflicting findings across different conditions and within sPTB itself have been identified. However, notable findings from the research indicate increased proinflammatory cytokines in pregnancies suffering from premature rupture of membranes (pPROM), chorioamnionitis, infection, preeclampsia, and gestational diabetes (GDM). Additionally, reduced Treg levels were identified in preeclampsia, GDM, and pPROM as well as chorioamnionitis presenting with increased Treg dysfunctionality. Treg deficiencies may contribute to health issues in preterm newborns. Current sPTB treatments are limited, underscoring the potential of in utero therapies targeting inflammation, including T cell interventions. Future research aims to establish consensus on the role of Tregs in sPTB and associated conditions and advancing understanding of mechanisms leading to Treg deficiencies in adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Nicoleta Mureanu
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
- Faculty of Medicine, Department of Obstetrics and Gynaecology, Carol Davila University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
| | - Amanda M. Bowman
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Imogen A. Porter-Wright
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Priya Verma
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Athina Efthymiou
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
| | - Kypros H. Nicolaides
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
| | - Cristiano Scotta
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London UB8 3PH, UK
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Rachel M. Tribe
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
| | - Panicos Shangaris
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| |
Collapse
|
27
|
Geng R, Ren M, Ma Y, Su W. Mechanism of the KIAA1429/KLF1/PD-L1 Axis in Regulating Immune Escape in Non-small Cell Lung Cancer. Cell Biochem Biophys 2024:10.1007/s12013-024-01592-3. [PMID: 39499390 DOI: 10.1007/s12013-024-01592-3] [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: 10/07/2024] [Indexed: 11/07/2024]
Abstract
Non-small cell lung cancer (NSCLC), accounting for approximately 80% of lung cancer cases, remains the leading cause of cancer-related mortality. Immune evasion is a critical challenge in NSCLC, contributing to poor treatment outcomes. This study investigates the role of KIAA1429 in immune evasion, aiming to identify novel therapeutic targets and provide a theoretical basis for NSCLC treatment. NSCLC cell lines were cultured to assess the expression of KIAA1429, KLF transcription factor (KLF1), and programmed cell death ligand 1 (PD-L1). Co-culture experiments were conducted with peripheral blood mononuclear cells (PBMCs) to evaluate cytotoxicity, CD8+T cell proportions, and levels of interferon-gamma (IFN-γ)/interleukin (IL)-10/IL-2. Additionally, N6-methyladenosine (m6A) modification in NSCLC cells, m6A enrichment on KLF1, and KLF1 mRNA stability were analyzed. Results showed increased expression of KIAA1429 and KLF1 in NSCLC cells. Knockdown of KIAA1429 inhibited NSCLC cell proliferation, enhanced PBMC cytotoxicity and CD8+T cell activation, increased IFN-γ and IL-2 levels, and decreased IL-10 levels. Mechanistically, KIAA1429 stabilized KLF1 mRNA level through m6A modification, promoting both KLF1 and PD-L1 expression. Overexpression of KLF1 or PD-L1 reversed the immune-modulating effects of KIAA1429 knockdown. In conclusion, KIAA1429 facilitates immune evasion in NSCLC by stabilizing KLF1 mRNA and upregulating PD-L1 expression.
Collapse
Affiliation(s)
- Rui Geng
- Department of Research-oriented Ward, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi, China
| | - Mingmin Ren
- Department of Research-oriented Ward, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi, China
| | - Yuhui Ma
- Department of Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences; Tongji Shanxi Hospital, Tongji Medical College, Huazhong University Science of and Technology, Taiyuan, Shanxi, China
| | - Wen Su
- Department of Immunology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
28
|
Zhao Y, Yin X, Zhou M, Rao W, Ji X, Wang X, Xiao X, Hu S. Noninvasive Monitoring of Programmed Death-Ligand 2 Expression with Positron Emission Tomography using 68Ga-labeled Peptide Antagonist in Preclinical and Exploratory Human Studies. RESEARCH (WASHINGTON, D.C.) 2024; 7:0523. [PMID: 39494220 PMCID: PMC11528066 DOI: 10.34133/research.0523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
While the expression of programmed death ligand-1 (PD-L1) is associated with response to immune therapy, PD-L1-negative patients may still benefit from immune treatment. Programmed death ligand-2 (PD-L2), another crucial immune checkpoint molecule interacting with PD-1, correlates with the efficacy of various tumor immune therapies. This study investigates the expression of PD-L2 in non-small cell lung cancer (NSCLC) patients following anti-PD-1 therapy and its predictive value for clinical survival outcomes. Additionally, we explore the noninvasive, real-time, and dynamic quantitative analysis potential of PD-L2 positron emission tomography (PET) imaging in transplanted tumors. We utilized [68Ga]Ga-labeled peptide HN11-1 for PD-L2 PET imaging. The results indicate a higher response rate to anti-PD-1 therapy in patients positive for both PD-L1 and PD-L2, with PD-L2 status independently predicting progression-free survival (PFS) with pembrolizumab treatment. Furthermore, [68Ga]Ga-HN11-1 PET imaging demonstrates specificity in assessing PD-L2 status. Overall, we confirm the correlation between high PD-L2 expression and favorable PFS in NSCLC patients post anti-PD-1 therapy and highlight the promising potential of [68Ga]Ga-HN11-1 as a specific tracer for PD-L2 in preclinical and initial human trials.
Collapse
Affiliation(s)
- Yajie Zhao
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Wanqian Rao
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Xuan Ji
- Department of Periodontology,
Suzhou Stomatological Hospital, Suzhou, Jiangsu 215026, China
| | - Xiaobo Wang
- Department of Nuclear Medicine and State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital,
Fourth Military Medical University, Xi’an 710032, China
| | - XiaoXiong Xiao
- Department of Thoracic Surgery, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital,
Central South University, Changsha 410008, China
| |
Collapse
|
29
|
Sun X, Lyu J, Yang M, Lin Y, Wu K, Liu K, Li A, Shuai X, Cai K, Wang Z, Wang G, Zhang P, Yin Y, Tao K. Two-Year Outcomes and Biomarker Analysis of Locally Advanced Gastric and Gastroesophageal Junction Adenocarcinoma After Neoadjuvant Chemotherapy and Immunotherapy from the Phase II WuhanUHGI001 Trial. Ann Surg Oncol 2024; 31:8157-8169. [PMID: 39154154 DOI: 10.1245/s10434-024-16041-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND This study reports the 2-year outcomes and biomarker analysis results of patients with locally advanced gastric and gastroesophageal junction (G/GEJ) adenocarcinoma who received neoadjuvant chemotherapy and immunotherapy in a phase II WuhanUHGI001 trial. METHODS Eligible patients with cT3/4aN+M0 locally advanced G/GEJ adenocarcinoma were screened, enrolled, and treated with 3 cycles of neoadjuvant tislelizumab and SOX followed by D2 gastrectomy and another 5 cycles of postoperative adjuvant SOX. The primary endpoint was major pathological response. RESULTS Of the 49 included patients, 24 (49.0%) achieved major pathological response and 13 (26.5%) achieved pathological complete response. During a median follow-up of 26.8 months, the 2-year progression-free survival (PFS) and overall survival (OS) rates were 69.4% and 81.2%, respectively. Grade 3-4 adverse events occurred in six patients (12.2%) during the neoadjuvant period, eight patients (17.0%) during the postoperative period, and seven patients (15.2%) during the adjuvant period. Biomarker analysis revealed that the pathological complete response showed no association with 2-year PFS and OS. Major pathological response showed a potentially strong association with improved 2-year PFS and OS rates. In addition, preoperative circulating tumor cells combined with pathological responses are helpful in prognosis assessment. In addition, our results showed that T downstaging, lymphocyte-to-monocyte ratio, and CD3+ T cells were independent factors that affect PFS. The signet ring cell component (SRCC), T downstaging, and neutrophil-to-lymphocyte ratio were independent factors affecting OS. Prognostic nomograms of PFS and OS constructed based on the multivariate Cox regression results demonstrated suitable calibration and discrimination ability. CONCLUSIONS Neoadjuvant tislelizumab plus SOX exhibits promising efficacy and acceptable toxicity in patients with locally advanced G/GEJ adenocarcinoma. In addition, our study established a prognostic risk signature and nomograms based on clinicopathological characteristics, which can accurately predict patient outcomes and aid in personalized treatment planning.
Collapse
Affiliation(s)
- Xiong Sun
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jianbo Lyu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ming Yang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yao Lin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ke Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ke Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Anshu Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoming Shuai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
30
|
Pan X, Xie X, Chen X, Chen H. High-Resolution CT Patterns of Anti-PD1 Checkpoint Inhibitor-Related Pneumonitis in Patients With Lung Cancer. J Comput Assist Tomogr 2024; 48:930-935. [PMID: 39143662 DOI: 10.1097/rct.0000000000001643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
BACKGROUND Lung cancer has the highest morbidity and mortality in the world, and immunotherapies have been developed for this disease in recent years. However, activation of the immune system can cause immune-related adverse events (irAEs), and checkpoint inhibitor-related pneumonitis (CIP), can be the most severe and fatal. But few reports have systematically examined the spectrum of imaging findings of this condition. Therefore, the objective of this paper is to investigate the high-resolution computed tomography (HRCT) characteristics of CIP in patients with lung cancer. OBJECTIVE To investigate the HRCT characteristics of CIP in patients with lung cancer. METHODS HRCT patterns in 41 lung cancer patients who developed CIP after treatment with immune checkpoint inhibitors were retrospectively characterized by interstitial lung disease classification, and their severity was graded. Specific HRCT characteristics related to CIP were identified. RESULTS There are 4 types of immunotherapy-induce pneumonitis patterns (organizing pneumonia OP 19 cases, nonspecific interstitial pneumonia NSIP 8 cases, acute interstitial pneumonia AIP 7 cases, 7 cases of undetermined type) and image grade (13 cases of grade 1, 17 cases of grade 2, 11 cases of grade 3, 0 cases of grade 4) were identified. Spatial distribution characteristics of these lesions were noted (17 cases predominantly distributed in tumor-containing lobes, 6 cases predominantly distributed in non-tumor-containing lobes, and no specific predilection in 18 cases). Specific CT imaging features found in CIP included, in the order of prevalence, the following: ground glass opacities (38 cases), subpleural/vertical line (37 cases), interstitial thickening around the bronchovascular bundles (36 cases), reticulation (34 cases), fine reticular shadow (31 cases), consolidation (31 cases), small cystic shadow (24 cases, may not having honeycombing), small nodules (17 cases), bronchiectasis (15 cases), honeycombing (11 cases), mosaic sign (11 cases), and pleural effusion (18 cases). CONCLUSION HRCT of CIP predominantly manifests as ground glass opacities, reticulation, subpleural/vertical line, interstitial thickening around the bronchovascular bundle, and consolidation.
Collapse
Affiliation(s)
- Xiaohuan Pan
- From the Department of Radiology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohong Xie
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaojuan Chen
- From the Department of Radiology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huai Chen
- Department of Radiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Abdallah FM, Ghoneim AI, Abd-Alhaseeb MM, Abdel-Raheem IT, Helmy MW. Unveiling the antitumor synergy between pazopanib and metformin on lung cancer through suppressing p-Akt/ NF-κB/ STAT3/ PD-L1 signal pathway. Biomed Pharmacother 2024; 180:117468. [PMID: 39332188 DOI: 10.1016/j.biopha.2024.117468] [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: 07/06/2024] [Revised: 09/01/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Pazopanib, an inhibitor of the VEGF receptor tyrosine kinase, has demonstrated significant antitumor effects in lung cancer. However, its application as a standard treatment for this type of cancer is limited by its drug resistance and toxicity. Metformin has the potential to combat lung cancer by modifying the tumor's immune microenvironment. In this study, we investigated the potential antitumor effects and the associated underlying molecular mechanisms of the combination of pazopanib and metformin in lung cancer. In vitro studies were conducted using the A549 and H460 lung cancer cell lines, whereas urethane-induced lung cancer-bearing mice were used for in vivo assessments. The urethane-induced mice received oral administration of pazopanib (50 mg/kg) and/or metformin (250 mg/kg) for a duration of 21 days. The results indicated that the MTT assay demonstrated a combined cytotoxic effect of the pazopanib/metformin combination in H460 and A549 cells, as evidenced by CI and DRI analyses. The observed increase in annexin V levels and the corresponding increase in Caspase-3 activity strongly suggest that this combination induced apoptosis. Furthermore, the pazopanib/metformin combination significantly inhibited the p-Akt/NF-κB/IL-6/STAT3, HIF1α/VEGF, and TLR2/TGF-β/PD-L1 pathways while also increasing CD8 expression in vivo. Immunohistochemical analysis revealed that these antitumor mechanisms were manifested by the suppression of the proliferation marker Ki67. In conclusion, these findings revealed that metformin augments the antitumor efficacy of pazopanib in lung cancer by simultaneously targeting proliferative, angiogenic, and immunogenic signaling pathways, metformin enhances the antitumor effectiveness of pazopanib in lung cancer, making it a promising therapeutic option for lung cancer.
Collapse
Affiliation(s)
- Fatma M Abdallah
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt; Faculty of Health Sciences Technology, Borg Al Arab Technological University, New Borg El Arab, Egypt.
| | - Asser I Ghoneim
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt.
| | - Mohammad M Abd-Alhaseeb
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt; Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.
| | - Ihab T Abdel-Raheem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt.
| | - Maged W Helmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt.
| |
Collapse
|
32
|
Hinojosa-Gonzalez DE, Saffati G, Salgado-Garza G, Patel S, Kronstedt S, Jones JA, Taylor JM, Yen AE, Slawin JR. Novel therapeutic regimens in previously untreated metastatic urothelial carcinoma: A systematic review and bayesian network meta-analysis. Urol Oncol 2024; 42:361-369. [PMID: 39112104 DOI: 10.1016/j.urolonc.2024.07.006] [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: 04/26/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 09/07/2024]
Abstract
Metastatic urothelial carcinoma (muC) has historically had few effective therapeutic options. Recently, immune checkpoint inhibitors (ICIs), were introduced as therapeutic options for cisplatin-ineligible patients, however, direct head-to-head trials comparing these treatments are lacking. To address this gap, this study employs a Bayesian framework to indirectly compare the performance of ICIs as first-line agents for muC. A systematic review was performed to identify randomized controlled trials evaluating different ICI for mUC. Data was inputted into Review Manager 5.4 for pairwise meta-analysis. Data was then used to build a network in R Studio. These networks were used to model 200,000 Markov Chains via MonteCarlo sampling. The results are expressed as hazard ratios (HR) with 95% credible intervals (CrI). Six studies with 5,449 patients were included, 3,255 received ICI monotherapy or combination. Moreover, a total of 3,006 had PD-L1 positive tumors and 2,362 were PD-L1 negative. Median overall survival (OS) ranged from 12.1 to 31.5 months across the studies, with the combination of enfortumab vedotin and pembrolizumab demonstrating the most substantial reduction in the risk of death (HR 0.47 [95% CrI: 0.38, 0.58]), followed by avelumab monotherapy (HR 0.69 [95% CrI: 0.56, 0.86]). The limitations of this network meta-analysis include variability in study follow-up duration, lack of standardized methods for assessing PD-L1 positivity, and potential bias introduced by control arms with poorer survival outcomes across included trials. The enfortumab vedotin/pembrolizumab combination significantly improved survival and response rates. Avelumab showed notable single-agent activity. These findings provide a valuable framework to guide clinical decision-making and highlight priority areas for future research, including biomarker refinement and novel combination strategies to enhance antitumor immunity in this challenging malignancy.
Collapse
Affiliation(s)
| | - Gal Saffati
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | | | - Sagar Patel
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | - Shane Kronstedt
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | - Jeffrey A Jones
- Scott Department of Urology, Baylor College of Medicine, Houston, TX; Michael E. DeBakey VA Medical Center, Houston, TX
| | - Jennifer M Taylor
- Scott Department of Urology, Baylor College of Medicine, Houston, TX; Michael E. DeBakey VA Medical Center, Houston, TX
| | - Aihua E Yen
- Bladder Cancer Center, Daniel L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Jeremy R Slawin
- Scott Department of Urology, Baylor College of Medicine, Houston, TX; Michael E. DeBakey VA Medical Center, Houston, TX
| |
Collapse
|
33
|
Jalil A, Donate MM, Mattei J. Exploring resistance to immune checkpoint inhibitors and targeted therapies in melanoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:42. [PMID: 39534873 PMCID: PMC11555183 DOI: 10.20517/cdr.2024.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Melanoma is the most aggressive form of skin cancer, characterized by a poor prognosis, and its incidence has risen rapidly over the past 30 years. Recent therapies, notably immunotherapy and targeted therapy, have significantly improved the outcome of patients with metastatic melanoma. Previously dismal five-year survival rates of below 5% have shifted to over 50% of patients surviving the five-year mark, marking a significant shift in the landscape of melanoma treatment and survival. Unfortunately, about 50% of patients either do not respond to therapy or experience early or late relapses following an initial response. The underlying mechanisms for primary and secondary resistance to targeted therapies or immunotherapy and relapse patterns remain not fully identified. However, several molecular pathways and genetic factors have been associated with melanoma resistance to these treatments. Understanding these mechanisms paves the way for creating novel treatments that can address resistance and ultimately enhance patient outcomes in melanoma. This review explores the mechanisms behind immunotherapy and targeted therapy resistance in melanoma patients. Additionally, it describes the treatment strategies to overcome resistance, which have improved patients' outcomes in clinical trials and practice.
Collapse
Affiliation(s)
- Anum Jalil
- Department of Medicine, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| | - Melissa M Donate
- Long School of Medicine, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| | - Jane Mattei
- Department of Hematology Oncology, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| |
Collapse
|
34
|
Chick RC, Pawlik TM. Updates in Immunotherapy for Pancreatic Cancer. J Clin Med 2024; 13:6419. [PMID: 39518557 PMCID: PMC11546190 DOI: 10.3390/jcm13216419] [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: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with limited effective therapeutic options. Due to a variety of cancer cell-intrinsic factors, including KRAS mutations, chemokine production, and other mechanisms that elicit a dysregulated host immune response, PDAC is often characterized by poor immune infiltration and an immune-privileged fibrotic stroma. As understanding of the tumor microenvironment (TME) evolves, novel therapies are being developed to target immunosuppressive mechanisms. Immune checkpoint inhibitors have limited efficacy when used alone or with radiation. Combinations of immune therapies, along with chemotherapy or chemoradiation, have demonstrated promise in preclinical and early clinical trials. Despite dismal response rates for immunotherapy for metastatic PDAC, response rates with neoadjuvant immunotherapy are somewhat encouraging, suggesting that incorporation of immunotherapy in the treatment of PDAC should be earlier in the disease course. Precision therapy for PDAC may be informed by advances in transcriptomic sequencing that can identify immunophenotypes, allowing for more appropriate treatment selection for each individual patient. Personalized and antigen-specific therapies are an increasing topic of interest, including adjuvant immunotherapy using personalized mRNA vaccines to prevent recurrence. Further development of personalized immune therapies will need to balance precision with generalizability and cost.
Collapse
Affiliation(s)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| |
Collapse
|
35
|
Cen S, Yuan M, Sun Q, Hou G, Ying J, Xu Q, Zheng Y, Dong Y, Pan H, Han W. Efficacy and safety of dual blockade of HER2 and PD-1 in patients with HER2-positive gastric cancer: a retrospective, multicentre study. Sci Rep 2024; 14:25030. [PMID: 39443515 PMCID: PMC11500361 DOI: 10.1038/s41598-024-76296-y] [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: 06/12/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is one of the most important pathological characteristics of gastric cancer. The positive rate of HER2 expression in patients with gastric cancer is approximately 20%. The phase III Keynote-811 study revealed that anti-HER2 and anti-PD-1 therapy combined with chemotherapy could significantly improve the objective response rate as first-line treatment in patients with HER2-positive advanced gastric cancer. In the present study, we aimed to evaluate the efficacy of combination therapy with trastuzumab and PD-1 inhibitors in patients with advanced HER2-positive gastric cancer in a real-world setting. Seventy-two HER2-positive gastric cancer patients from three hospitals in China were retrospectively reviewed. These patients were treated with trastuzumab plus one anti-PD-1 agent with or without chemotherapy. The overall response rate, progression-free survival and overall survival were assessed according to the Response Evaluation Criteria in Solid Tumours (RECIST 1.1). From January 2018 to October 2021, 72 patients with HER2-positive gastric cancer received trastuzumab and a PD-1 inhibitor with or without chemotherapy as neoadjuvant chemotherapy, first-line therapy, second-line therapy or salvage therapy. The ORR was 54.2% for all patients and 79.4% for previously untreated patients. The median PFS and median OS were 10 months (95% CI: 8-13 months) and 26.1 months (95% CI: 18.5-NA months), respectively, for all patients. Grade 3 adverse effects occurred in approximately 25% of patients. Immune-related adverse effects occurred in approximately 12.5% of patients. Trastuzumab and PD-1 inhibitor combination therapy with or without chemotherapy achieved satisfactory survival outcomes in patients with HER2-positive gastric cancer with acceptable safety.
Collapse
Affiliation(s)
- Shuyi Cen
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China
| | - Meiqin Yuan
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Qunan Sun
- Department of Medical Oncology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, PRC, China
| | - Guilan Hou
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Qi Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Yu Zheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China
| | - Ying Dong
- Department of Medical Oncology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, PRC, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China.
| | - Weidong Han
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China.
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China.
| |
Collapse
|
36
|
Tirone B, Scarabosio A, Surico PL, Parodi PC, D’Esposito F, Avitabile A, Foti C, Gagliano C, Zeppieri M. Targeted Drug Delivery in Periorbital Non-Melanocytic Skin Malignancies. Bioengineering (Basel) 2024; 11:1029. [PMID: 39451404 PMCID: PMC11504966 DOI: 10.3390/bioengineering11101029] [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: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Targeted drug delivery has emerged as a transformative approach in the treatment of periorbital skin malignancies, offering the potential for enhanced efficacy and reduced side effects compared to traditional therapies. This review provides a comprehensive overview of targeted therapies in the context of periorbital malignancies, including basal cell carcinoma, squamous cell carcinoma, sebaceous gland carcinoma, and Merkel cell carcinoma. It explores the mechanisms of action for various targeted therapies, such as monoclonal antibodies, small molecule inhibitors, and immunotherapies, and their applications in treating these malignancies. Additionally, this review addresses the management of ocular and periocular side effects associated with these therapies, emphasizing the importance of a multidisciplinary approach to minimize impact and ensure patient adherence. By integrating current findings and discussing emerging trends, this review aims to highlight the advancements in targeted drug delivery and its potential to improve treatment outcomes and quality of life for patients with periorbital skin malignancies.
Collapse
Affiliation(s)
- Benedetta Tirone
- Dermatology and Venerology Section, Department of Precision and Regenerative Medicine and Ionan Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Scarabosio
- Clinic of Plastic and Reconstructive Surgery, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Pier Luigi Surico
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy
| | - Pier Camillo Parodi
- Clinic of Plastic and Reconstructive Surgery, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy
| | - Fabiana D’Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, 153-173 Marylebone Rd, London NW15QH, UK
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Alessandro Avitabile
- Eye Clinic Catania San Marco Hospital, Viale Carlo Azeglio Ciampi, 95121 Catania, Italy
| | - Caterina Foti
- Dermatology and Venerology Section, Department of Precision and Regenerative Medicine and Ionan Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Caterina Gagliano
- Mediterranean Foundation “G.B. Morgagni”, 95125 Catania, Italy
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| |
Collapse
|
37
|
Song L, Liang Y, Li Y, Guo T, Li H, Liang S. Development of an LC-TOF/MS Method to Quantify Camrelizumab in Human Serum. Molecules 2024; 29:4862. [PMID: 39459229 PMCID: PMC11510712 DOI: 10.3390/molecules29204862] [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: 07/07/2024] [Revised: 09/28/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
With the advantages of a high specificity, a long half-life, and a high safety, the use of antibody biologic drugs, including camrelizumab, has been rapidly increasing in clinical practice. Camrelizumab, an immune checkpoint inhibitor and humanized monoclonal antibody, is used to treat several advanced solid cancers. Measuring its concentration supports personalized dosage adjustments, influences treatment decisions for patients, strengthens the control of disease activity through therapeutic drug monitoring, and helps evaluate and prevent drug interactions in combination therapy. Because antibodies are present in complex biological matrices, quantifying monoclonal antibody drugs is challenging, and must rely on precise, selective, and reliable analytical methods. In this study, a quadrupole time-of-flight mass spectrometry TripleTOF 6600+ (AB SCIEX, Framingham, MA, USA) system equipped with a Turbo V ion source was used for the qualitative analysis of monoclonal antibodies using the data-dependent acquisition (IDA) MS/MS mode, followed by quantitative analysis using a targeted MRMHR workflow. This method showed a good linear relationship within the range of 4-160 μg/mL, with a correlation coefficient of R2 ≥ 0.996. It demonstrated an acceptable accuracy (88.95-101.18%) and precision (≤15%). Furthermore, the lower limit of quantification was found to be 4 μg/mL, with the lowest detection limit of 0.3217 μg/mL, indicating that this method is rapid, accurate, and reliable for the quantitative analysis of camrelizumab in human serum.
Collapse
Affiliation(s)
- Li Song
- College of Chemistry and Materials Science, Hebei University, Baoding 071000, China
| | - Yan Liang
- College of Pharmacy, Hebei Medical University, Shijiazhuang 050000, China
| | - Yilin Li
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang 050000, China
| | - Tingting Guo
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang 050000, China
| | - Hui Li
- Hebei Institute of Drug and Medical Device Inspection, Shijiazhuang 050000, China
| | - Shuxuan Liang
- College of Chemistry and Materials Science, Hebei University, Baoding 071000, China
| |
Collapse
|
38
|
Wang X, Ma S, Twardowski P, Lau C, Chan YS, Wong K, Xiao S, Wang J, Wu X, Frankel P, Wilson TG, Synold TW, Presant C, Dorff T, Yu J, Sadava D, Chen S. Reduction of myeloid-derived suppressor cells in prostate cancer murine models and patients following white button mushroom treatment. Clin Transl Med 2024; 14:e70048. [PMID: 39390760 PMCID: PMC11467013 DOI: 10.1002/ctm2.70048] [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: 06/17/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND In a previously reported Phase I trial, we observed therapy-associated declines in circulating myeloid-derived suppressor cells (MDSCs) with the administration of white button mushroom (WBM) tablets in prostate cancer (PCa) patients. These observations led us to hypothesise that WBM could mitigate PCa progression by suppressing MDSCs. METHODS We performed bidirectional translational research to examine the immunomodulatory effects of WBM consumption in both syngeneic murine PCa models and patients with PCa participating in an ongoing randomised Phase II trial (NCT04519879). RESULTS In murine models, WBM treatment significantly suppressed tumour growth with a reduction in both the number and function of MDSCs, which in turn promoted antitumour immune responses mediated by T cells and natural killer (NK) cells. In patients, after consumption of WBM tablets for 3 months, we observed a decline in circulating polymorphonuclear MDSCs (PMN-MDSCs), along with an increase in cytotoxic CD8+ T and NK cells. Furthermore, single immune cell profiling of peripheral blood from WBM-treated patients showed suppressed STAT3/IRF1 and TGFβ signalling in circulating PMN-MDSCs. Subclusters of PMN-MDSCs presented transcriptional profiles associated with responsiveness to fungi, neutrophil chemotaxis, leukocyte aggregation, and regulation of inflammatory response. Finally, in mouse models of PCa, we found that WBM consumption enhanced the anticancer activity of anti-PD-1 antibodies, indicating that WBM may be used as an adjuvant therapy with immune checkpoint inhibitors. CONCLUSION Our results from PCa murine models and patients provide mechanistic insights into the immunomodulatory effects of WBM and provide a scientific foundation for WBM as a nutraceutical intervention to delay or prevent PCa progression. HIGHLIGHTS White button mushroom (WBM) treatment resulted in a reduction in pro-tumoural MDSCs, notably polymorphonuclear MDSCs (PMN-MDSCs), along with activation of anti-tumoural T and NK cells. Human single immune cell gene expression profiling shed light on the molecular alterations induced by WBM, specifically on PMN-MDSCs. A proof-of-concept study combining WBM with PD-1 blockade in murine models revealed an additive effect on tumour regression and survival outcomes, highlighting the clinical relevance of WBM in cancer management.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Przemyslaw Twardowski
- Department of Urology and Urologic OncologyProvidence Saint John's Cancer InstituteSanta MonicaCaliforniaUSA
| | - Clayton Lau
- Department of SurgeryCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Yin S. Chan
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Kelly Wong
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Sai Xiao
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jinhui Wang
- Integrative Genomics CoreBeckman Research Institute, City of HopeMonroviaCaliforniaUSA
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Paul Frankel
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Timothy G. Wilson
- Department of Urology and Urologic OncologyProvidence Saint John's Cancer InstituteSanta MonicaCaliforniaUSA
| | - Timothy W Synold
- Department of Medical Oncology & Therapeutics ResearchCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Cary Presant
- Department of Medical Oncology & Therapeutics ResearchCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Tanya Dorff
- Department of Medical Oncology & Therapeutics ResearchCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - David Sadava
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Shiuan Chen
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| |
Collapse
|
39
|
Yadav R, Khatkar R, Yap KCH, Kang CYH, Lyu J, Singh RK, Mandal S, Mohanta A, Lam HY, Okina E, Kumar RR, Uttam V, Sharma U, Jain M, Prakash H, Tuli HS, Kumar AP, Jain A. The miRNA and PD-1/PD-L1 signaling axis: an arsenal of immunotherapeutic targets against lung cancer. Cell Death Discov 2024; 10:414. [PMID: 39343796 PMCID: PMC11439964 DOI: 10.1038/s41420-024-02182-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Lung cancer is a severe challenge to the health care system with intrinsic resistance to first and second-line chemo/radiotherapies. In view of the sterile environment of lung cancer, several immunotherapeutic drugs including nivolumab, pembrolizumab, atezolizumab, and durvalumab are currently being used in clinics globally with the intention of releasing exhausted T-cells back against refractory tumor cells. Immunotherapies have a limited response rate and may cause immune-related adverse events (irAEs) in some patients. Hence, a deeper understanding of regulating immune checkpoint interactions could significantly enhance lung cancer treatments. In this review, we explore the role of miRNAs in modulating immunogenic responses against tumors. We discuss various aspects of how manipulating these checkpoints can bias the immune system's response against lung cancer. Specifically, we examine how altering the miRNA profile can impact the activity of various immune checkpoint inhibitors, focusing on the PD-1/PD-L1 pathway within the complex landscape of lung cancer. We believe that a clear understanding of the host's miRNA profile can influence the efficacy of checkpoint inhibitors and significantly contribute to existing immunotherapies for lung cancer patients. Additionally, we discuss ongoing clinical trials involving immunotherapeutic drugs, both as standalone treatments and in combination with other therapies, intending to advance the development of immunotherapy for lung cancer.
Collapse
Affiliation(s)
- Ritu Yadav
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Rinku Khatkar
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Kenneth C-H Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chloe Yun-Hui Kang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juncheng Lyu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rahul Kumar Singh
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Surojit Mandal
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Adrija Mohanta
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rajiv Ranjan Kumar
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Vivek Uttam
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Uttam Sharma
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Manju Jain
- Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| | | | | | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Aklank Jain
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
40
|
He X, Gao Y, Deng Y, He J, Nolte I, Murua Escobar H, Yu F. The Comparative Oncology of Canine Malignant Melanoma in Targeted Therapy: A Systematic Review of In Vitro Experiments and Animal Model Reports. Int J Mol Sci 2024; 25:10387. [PMID: 39408717 PMCID: PMC11476434 DOI: 10.3390/ijms251910387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 10/20/2024] Open
Abstract
Canine malignant melanoma (CMM) is highly aggressive and mostly located in the oral cavity. CMM is the predominant type of canine oral malignancy and shows striking homologies with human mucosal melanoma. In comparative oncology, canine oral melanomas (COMs), as spontaneous tumor models, have the potential to acquire a unique value as a translational model of rare human melanoma subtypes. This review aims to provide a comprehensive summary of targeted therapies for canine malignant melanoma and to enrich the field of comparative oncology. Following the PRISMA guidelines, a comprehensive literature search was conducted across databases for studies from 1976 to April 2024. Studies were selected based on their relevance to targeted treatments. A total of 30 studies met the inclusion criteria. Based on the treatment approaches, the studies were further categorized into immunotherapies, small molecule signaling inhibitors, indirect kinase inhibitors, and other alternative strategies. Some treatments have been shown to result in stable disease or partial response, accounting for 29% (monoclonal antibody) and 76.5% (micro-RNA therapies) in clinical trials. Moreover, in vitro experiments of small molecule inhibitors, including cell signaling inhibitors and indirect kinase inhibitors, have shown the potential to be an effective treatment option for the development of therapeutic strategies in canine malignant melanoma. The observed response in in vitro experiments of CMM (particularly the oral and certain cutaneous subtypes) to drugs used in the treatment of human melanoma underlines the resemblance to human melanoma, therefore supporting the notion that CMM may be a valuable model for understanding rare human melanoma subtypes and exploring potential therapeutic avenues in preclinical trials. Finally, this literature review serves as a valuable resource for the development of therapeutic strategies for CMM and highlights the potential for translating these findings to human cancer treatment.
Collapse
Affiliation(s)
- Xiaohui He
- Department of Small Animal Medicine, College of Veterinary Medicine, China Agriculture University, Beijing 100193, China
| | - Yu Gao
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Yuqing Deng
- Department of Small Animal Medicine, College of Veterinary Medicine, China Agriculture University, Beijing 100193, China
| | - Junying He
- Department of Small Animal Medicine, College of Veterinary Medicine, China Agriculture University, Beijing 100193, China
| | - Ingo Nolte
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Feng Yu
- Department of Small Animal Medicine, College of Veterinary Medicine, China Agriculture University, Beijing 100193, China
| |
Collapse
|
41
|
Ryba-Stanisławowska M. Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00992-0. [PMID: 39325360 DOI: 10.1007/s13402-024-00992-0] [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: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Monika Ryba-Stanisławowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland.
| |
Collapse
|
42
|
Tang S, Lyles KV, Wang Y, Fan D, Luo M. Enhancing the Efficacy of Breast Cancer Immunotherapy Using a Smac-Armed Oncolytic Virus. Cancers (Basel) 2024; 16:3248. [PMID: 39409870 PMCID: PMC11476252 DOI: 10.3390/cancers16193248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
It has been shown that the response rate of TNBC is dependent on the level of PD-L1 and the tumor microenvironment (TME). Approaches that alter the TME can improve the efficacy of ICIs. Background: We have engineered a Smac-armed oncolytic virus by inserting a Smac transgene into the genome of a vesicular stomatitis virus to generate VSV-S. Our previous study shows that the anticancer efficacy of VSV-S is more potent than that of wild-typed VSV in a subcutaneous TNBC mouse model. VSV-S treatment reverts the immunosuppressive TME by reducing MDSCs and TAMs, while increasing infiltration of neutrophils and CD8+ T cells. Methods: VSV-S was used to treat TNBC in an orthotopic mouse model, and in a combination therapy with an anti-PD-1 antibody to treat metastatic TNBC in a mouse model. Changes in the TME were evaluated. Results: In this current study, we show that neoadjuvant VSV-S treatment of primary orthotopic TNBC tumors in mice drastically lowered lung metastasis after surgical removal of the primary tumor, and significantly increased the survival rate. The mechanism of action and changes to the TME were delineated, among which one significant marker is the elevation of PD-L1 expression in tumors. In the TNBC lung metastasis mouse model, pulmonary treatment with VSV-S greatly enhanced the efficacy of ICI treatment. Conclusions: Our results suggest that the combination of oncolytic virus and ICI therapies has the potential to substantially improve the outcome of TNBC treatment.
Collapse
Affiliation(s)
- Sijia Tang
- Institute of Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA;
| | - Kristin V. Lyles
- Department of Chemistry, Georgia State University, Atlanta, GA 30302, USA;
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 20209, USA; (Y.W.); (D.F.)
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 20209, USA; (Y.W.); (D.F.)
| | - Ming Luo
- Department of Chemistry, Georgia State University, Atlanta, GA 30302, USA;
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA
| |
Collapse
|
43
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
44
|
Yin Z(S, Wang Z. Strategies for engineering oncolytic viruses to enhance cancer immunotherapy. Front Pharmacol 2024; 15:1450203. [PMID: 39309012 PMCID: PMC11413971 DOI: 10.3389/fphar.2024.1450203] [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: 06/17/2024] [Accepted: 07/30/2024] [Indexed: 09/25/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the predominant form of lung cancer and is characterized by rapid metastasis and high mortality, presenting a challenge for early-stage treatment modalities. The heterogeneity of NSCLC's tumor microenvironment (TME) significantly influences the efficacy of anti-PD-1 immune checkpoint inhibitors (ICIs) therapy, leading to varied patient responses. This review characterized different strains of oncolytic viruses in NSCLC and the different gene edits in pre-existing oncolytic viruses. This study also aimed to provide strategies to enhance anti-PD-1 therapy in NSCLC by engineering oncolytic viruses (OVs). This study offers insights into the genomic adaptations necessary for OVs targeting NSCLC, identify genetic determinants of anti-PD-1 response variability, and propose genomic edits to bolster therapy effectiveness. The primary goal of this study is to present a theoretically designed OV with a detailed genomic framework capable of enhancing the response to anti-PD-1 therapy, thereby advancing the field of cancer immunotherapy.
Collapse
Affiliation(s)
| | - Zhengfeng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
45
|
van Houtum EJ, Valk AH, Granado D, Lok J, van den Bogaard L, Remkes N, van Eck van der Sluijs J, Span PN, Cornelissen LA, Adema GJ. Siglec-7 and Siglec-9 expression in primary triple negative and oestrogen receptor positive breast cancer and in vitro signalling. Clin Transl Immunology 2024; 13:e1524. [PMID: 39246414 PMCID: PMC11378723 DOI: 10.1002/cti2.1524] [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: 05/27/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 09/10/2024] Open
Abstract
Objectives PD-1/PD-L1 immune checkpoint blockade can be an effective treatment for advanced breast cancer patients. However, patients with oestrogen receptor positive (ER+) tumors often display only low lymphocyte infiltration, while a large part of triple negative (TN) breast tumors does not generate an effective immunotherapy response. Therefore, new treatment strategies have to be developed. Here, we investigate Siglec-7 and Siglec-9 as novel ITIM-bearing inhibitory immune checkpoint receptors similar to PD-1, but expressed on a broader range of immune cells. Methods We assessed Siglec-7 and Siglec-9 (ligand) expression in TN and ER+ breast cancer tumors and their breast cancer cell line-induced signalling. Results We report that Siglec-7 and Siglec-9 are highly expressed in TN tumors, and to a low extent in ER+ tumors. Siglec-7 was observed on myeloid cells, T cells, and NK cells and Siglec-9 preferentially on myeloid cells. Expression of sialoglycans, including Siglec-7 and Siglec-9 ligands, was observed in both TN and ER+ breast cancer tissue sections. Expression levels of Siglec-7 and Siglec-9 ligands were higher on in vitro cultured TN cell lines than ER+ cell lines. Importantly, by applying chimeric Siglec-7 reporter cells, we showed the induction of Siglec-7 signalling by multiple TN cell lines, but only by one ER+ cell line. Moreover, Siglec-7 signalling is directly related to Siglec-7 ligand expression levels of breast cancer cell lines. Conclusion These data imply that immunotherapy targeting Siglec receptors may be particularly interesting for TN breast cancer patients not responding to current treatment strategies with tumors displaying high immune cell infiltration.
Collapse
Affiliation(s)
- Eline Jh van Houtum
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Anne Hc Valk
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Daniel Granado
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Jasper Lok
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Lune van den Bogaard
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Naomi Remkes
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Jesper van Eck van der Sluijs
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Paul N Span
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Lenneke Am Cornelissen
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Gosse J Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
46
|
Roozitalab G, Abedi B, Imani S, Farghadani R, Jabbarzadeh Kaboli P. Comprehensive assessment of TECENTRIQ® and OPDIVO®: analyzing immunotherapy indications withdrawn in triple-negative breast cancer and hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:889-918. [PMID: 38409546 DOI: 10.1007/s10555-024-10174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/05/2024] [Indexed: 02/28/2024]
Abstract
Atezolizumab (TECENTRIQ®) and nivolumab (OPDIVO®) are both immunotherapeutic indications targeting programmed cell death 1 ligand 1 (PD-L1) and programmed cell death 1 (PD-1), respectively. These inhibitors hold promise as therapies for triple-negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) and have demonstrated encouraging results in reducing the progression and spread of tumors. However, due to their adverse effects and low response rates, the US Food and Drug Administration (FDA) has withdrawn the approval of atezolizumab in TNBC and nivolumab in HCC treatment. The withdrawals of atezolizumab and nivolumab have raised concerns regarding their effectiveness and the ability to predict treatment responses. Therefore, the current study aims to investigate the immunotherapy withdrawal of PD-1/PD-L1 inhibitors, specifically atezolizumab for TNBC and nivolumab for HCC. This study will examine both the structural and clinical aspects. This review provides detailed insights into the structure of the PD-1 receptor and its ligands, the interactions between PD-1 and PD-L1, and their interactions with the withdrawn antibodies (atezolizumab and nivolumab) as well as PD-1 and PD-L1 modifications. In addition, this review further assesses these antibodies in the context of TNBC and HCC. It seeks to elucidate the factors that contribute to diverse responses to PD-1/PD-L1 therapy in different types of cancer and propose approaches for predicting responses, mitigating the potential risks linked to therapy withdrawals, and optimizing patient outcomes. By better understanding the mechanisms underlying responses to PD-1/PD-L1 therapy and developing strategies to predict these responses, it is possible to create more efficient treatments for TNBC and HCC.
Collapse
Affiliation(s)
- Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan.
| |
Collapse
|
47
|
Schumaier NP, Heidemann DG, Gupta C. Durvalumab-associated limbal stem cell deficiency and secondary corneal perforation. Am J Ophthalmol Case Rep 2024; 35:102074. [PMID: 38975031 PMCID: PMC11225003 DOI: 10.1016/j.ajoc.2024.102074] [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: 01/06/2024] [Revised: 03/18/2024] [Accepted: 04/29/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose We report a patient with bilateral limbal stem cell deficiency (LSCD) like clinical manifestations and secondary corneal perforation presumably induced by durvalumab following its use for the treatment of non-small cell lung carcinoma. Observations A 65-year-old male diagnosed with non-small cell lung carcinoma was treated with monthly durvalumab infusions. Two months after starting durvalumab, the patient was found to have bilateral severe keratoconjunctivitis and LSCD-like clinical findings. Despite topical management and oral prednisone for presumed ocular cicatricial pemphigoid, the patient continued to worsen clinically. The patient was transferred to our institution about one year later with persistent inflammation. The patient eventually developed a corneal perforation of the left eye, which required the application of cyanoacrylic tissue adhesive. Due to the lack of response to oral prednisone, durvalumab was discontinued with the approval of the patient's oncologist. Several months following the discontinuation of durvalumab, the conjunctival inflammation subsided, and corneal epithelial breakdown and ulceration resolved. Conclusions We report an association between durvalumab and the development of bilateral LSCD-like clinical findings with subsequent corneal perforation. We hope this case reinforces the importance of routine ophthalmologic follow-up after starting any cancer treatment, especially in patients with symptoms and signs suggesting ocular surface disease or inflammation.
Collapse
Affiliation(s)
- Nahrain Putris Schumaier
- Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
- Corewell Health William Beaumont University Hospital Eye Institute, Royal Oak, MI, USA
| | - David G. Heidemann
- Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
- Corewell Health William Beaumont University Hospital Eye Institute, Royal Oak, MI, USA
| | - Chirag Gupta
- Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
- Corewell Health William Beaumont University Hospital Eye Institute, Royal Oak, MI, USA
| |
Collapse
|
48
|
Fabi A, Cortesi L, Duranti S, Cordisco EL, Di Leone A, Terribile D, Paris I, de Belvis AG, Orlandi A, Marazzi F, Muratore M, Garganese G, Fuso P, Paoletti F, Dell'Aquila R, Minucci A, Scambia G, Franceschini G, Masetti R, Genuardi M. Multigenic panels in breast cancer: Clinical utility and management of patients with pathogenic variants other than BRCA1/2. Crit Rev Oncol Hematol 2024; 201:104431. [PMID: 38977141 DOI: 10.1016/j.critrevonc.2024.104431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Multigene panels can analyze high and moderate/intermediate penetrance genes that predispose to breast cancer (BC), providing an opportunity to identify at-risk individuals within affected families. However, considering the complexity of different pathogenic variants and correlated clinical manifestations, a multidisciplinary team is needed to effectively manage BC. A classification of pathogenic variants included in multigene panels was presented in this narrative review to evaluate their clinical utility in BC. Clinical management was discussed for each category and focused on BC, including available evidence regarding the multidisciplinary and integrated management of patients with BC. The integration of both genetic testing and counseling is required for customized decisions in therapeutic strategies and preventative initiatives, as well as for a defined multidisciplinary approach, considering the continuous evolution of guidelines and research in the field.
Collapse
Affiliation(s)
- Alessandra Fabi
- Precision Medicine Unit in Senology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Cortesi
- Department of Oncology and Haematology, Modena Hospital University, Modena Italy (Cortesi)
| | - Simona Duranti
- Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Emanuela Lucci Cordisco
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Genetics Unit, Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alba Di Leone
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Terribile
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ida Paris
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Giulio de Belvis
- Value Lab, Faculty of Economics, Università Cattolica del Sacro Cuore, Rome, Italy; Critical Pathways and Outcomes Evaluation Unit, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Armando Orlandi
- Unit of Oncology, Comprehensive Cancer Centre, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Marazzi
- UOC Oncological Radiotherapy, Department of Diagnostic Imaging, Radiation Oncology and Haematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Margherita Muratore
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori"
| | - Giorgia Garganese
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Section of Obstetrics and Gynecology, Department of Woman and Child Health and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paola Fuso
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Filippo Paoletti
- Critical Pathways and Outcomes Evaluation Unit, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Rossella Dell'Aquila
- Critical Pathways and Outcomes Evaluation Unit, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Angelo Minucci
- Genomics Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Gianluca Franceschini
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Riccardo Masetti
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Maurizio Genuardi
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Genetics Unit, Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
49
|
Xin Z, Qin L, Tang Y, Guo S, Li F, Fang Y, Li G, Yao Y, Zheng B, Zhang B, Wu D, Xiao J, Ni C, Wei Q, Zhang T. Immune mediated support of metastasis: Implication for bone invasion. Cancer Commun (Lond) 2024; 44:967-991. [PMID: 39003618 PMCID: PMC11492328 DOI: 10.1002/cac2.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024] Open
Abstract
Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.
Collapse
Affiliation(s)
- Zengfeng Xin
- Department of Orthopedic SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Luying Qin
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yang Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Siyu Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Fangfang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yuan Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Gege Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yihan Yao
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Binbin Zheng
- Department of Respiratory MedicineNingbo Hangzhou Bay HospitalNingboZhejiangP. R. China
| | - Bicheng Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Dang Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jie Xiao
- Department of Orthopedic SurgerySecond Affiliated Hospital (Jiande Branch)Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Chao Ni
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Breast SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qichun Wei
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Ting Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
50
|
Shen X, Xie J, Liu S, Cai Y, Yuan S, Uehara Y, Zhu D, Zheng M. Anoikis-related subtype and prognosis analyses based on bioinformatics, and an expression verification of ANGPTL4 based on experiments of lung adenocarcinoma. J Thorac Dis 2024; 16:5361-5378. [PMID: 39268091 PMCID: PMC11388259 DOI: 10.21037/jtd-24-1123] [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: 07/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024]
Abstract
Background Lung adenocarcinoma (LUAD) is one of the most common malignant tumors with high mortality. Anoikis resistance is an important mechanism of tumor cell proliferation and migration. Our research is devoted to exploring the role of anoikis in the diagnosis, classification, and prognosis of LUAD. Methods We downloaded the expression profile, mutation, and clinical data of LUAD from The Cancer Genome Atlas (TCGA) database. The "ConsensusClusterPlus" package was then used for the cluster analysis, and least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analyses were used to establish the prognostic model. We verified the reliability of the model using a Gene Expression Omnibus (GEO) data set. A gene set variation analysis (GSVA) was conducted to investigate the functional enrichment differences in the different clusters and risk groups. The CIBERSORT algorithm and a single-sample gene set enrichment analysis (ssGSEA) were used to analyze immune cell infiltration. The tumor mutation burden (TMB) and Tumor Immune Dysfunction and Exclusion (TIDE) scores were used to evaluate the patients' sensitivity to immunotherapy. Immunohistochemical staining of tissue microarrays was used to verify the correlation between ANGPTL4 expression and the clinicopathological characteristics and prognosis of LUAD patients. Results First, we screened 135 differentially expressed anoikis-related genes (ARGs) and 23 prognosis-related ARGs from TCGA-LUAD data set. Next, 494 LUAD samples were allocated to cluster A and cluster B based on the 23 prognosis-related ARGs. The Kaplan-Meier (K-M) analysis showed the overall survival (OS) of cluster B was better than that of cluster A. The clinicopathological characteristics and functional enrichment analyses revealed significant differences between clusters A and B. The tumor microenvironment (TME) analysis showed that cluster B had more immune cell infiltration and a higher TME score than cluster A. Subsequently, a LASSO Cox regression model of LUAD was constructed with ten ARGs. The K-M analysis showed that the low-risk patients had longer OS than the high-risk patients. The receiver operating characteristic curve, nomogram, and GEO data set verification results showed that the model had high accuracy and reliability. The level of immune cell infiltration and TME score were higher in the low-risk group than the high-risk group. The high-risk group had stronger sensitivity to immune checkpoint block therapy and weaker sensitivity to chemotherapy drugs than the low-risk group. ANGPTL4 expression was correlated with stage, tumor differentiation, tumor size, lymph node metastasis, and OS. Conclusions We discovered novel molecular subtypes and constructed a novel prognostic model of LUAD. Our findings provide important insights into subtype classification and the accurate survival prediction of LUAD. We also identified ANGPTL4 as a prognostic indicator of LUAD.
Collapse
Affiliation(s)
- Xiaojian Shen
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| | - Jing Xie
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| | - Shu Liu
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| | - Yun Cai
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| | - Shen Yuan
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| | - Yuji Uehara
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Honkomagoame, Tokyo, Japan
- Division of Cancer Evolution, National Cancer Center Japan Research Institute, Tokyo, Japan
| | - Dongbing Zhu
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| | - Miaosen Zheng
- Department of Pathology, The People's Hospital of Rugao, Rugao Hospital Affiliated to Nantong University, Rugao, China
| |
Collapse
|