1
|
Meng HJ, Mao Y, Zhao DQ, Shi SG. Prognostic value of the triglyceride-glucose index in advanced gastric cancer: A call for further exploration. World J Gastroenterol 2025; 31:104574. [PMID: 40309236 PMCID: PMC12038551 DOI: 10.3748/wjg.v31.i15.104574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, necessitating the identification of reliable prognostic indicators to enhance treatment outcomes. Recent research has highlighted the triglyceride-glucose (TyG) index as a potential surrogate marker for insulin resistance, which may significantly influence the prognosis of patients undergoing immunotherapy combined with chemotherapy. In this context, the study by Yao et al demonstrates that a high TyG index correlates with improved overall survival and progression-free survival in advanced GC patients receiving sintilimab and chemotherapy. Specifically, patients in the high TyG group had a significantly longer median progression-free survival of 9.8 months [95% confidence interval (CI): 9.2-10.9] compared to 8.0 months (95%CI: 7.5-8.5) in the low TyG group (hazard ratio = 0.58, 95%CI: 0.43-0.79, P < 0.001). Similarly, the median overall survival was significantly longer in the high TyG group at 23.1 months (95%CI: 21.2-NA) vs 16.5 months (95%CI: 13.9-18.3) in the low TyG group (hazard ratio = 0.30, 95%CI: 0.21-0.42, P < 0.001). These findings underscore the strong prognostic potential of the TyG index in guiding treatment strategies for advanced GC. These findings underscore the need for further investigation into the TyG index's role as a prognostic tool and its underlying mechanisms in influencing treatment efficacy. We advocate for additional multicenter studies to validate these results and explore the TyG index's applicability across diverse patient populations, ultimately aiming to refine treatment strategies and improve patient outcomes in advanced GC.
Collapse
Affiliation(s)
- Hong-Jie Meng
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Yi Mao
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - De-Qing Zhao
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Sheng-Guang Shi
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| |
Collapse
|
2
|
Kaur M, Rüger K, Chen EC, Rangaswamy US, Davison LM, Moreno Arteaga S, Smith I, Chu R, Chattopadhyay S, Rickert M, Force Aldred S, Harris KE, Trinklein ND, Clarke SC. Potency-optimized CD28-activating bispecific antibody for the targeted treatment of Nectin-4 positive cancers. J Immunother Cancer 2025; 13:e011323. [PMID: 40187750 PMCID: PMC11973789 DOI: 10.1136/jitc-2024-011323] [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: 12/12/2024] [Accepted: 03/16/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND T-cell costimulation is crucial for an effective and sustained antitumor immune response, and inadequate expression of costimulatory ligands within tumors can impair T-cell function. Bispecific antibodies (bsAbs) targeting a tumor-associated antigen and the T-cell costimulatory receptor CD28 represent a novel class of immune-stimulatory therapeutics designed to enhance antitumor immune responses by selectively delivering T-cell costimulation directly to the tumor microenvironment. This approach holds the potential to improve the survival, proliferation, and cytotoxic function of antitumor T cells while minimizing the risk of systemic immune activation. Urothelial cancer (UC) is associated with significant morbidity and mortality worldwide, particularly in advanced disease settings. Nectin-4, a membrane protein highly expressed in UC with limited expression in healthy tissues, presents a compelling target for therapeutic intervention. METHODS Using our proprietary high-throughput antibody discovery pipeline, we identified a panel of novel antibodies with a range of affinities for CD28 and Nectin-4 and successfully engineered them as bsAbs. We tested the T-cell costimulatory function of these molecules in vitro using primary human T cells and human cancer cell lines. Based on these results, we selected a clinical candidate which we assessed in a syngeneic mouse tumor model system and investigated tolerability and pharmacokinetics (PK) in non-human primates (NHP). RESULTS Our in vitro studies demonstrated that these bsAbs effectively enhance T-cell activation and cytotoxicity against Nectin-4 positive tumor cells in the presence of T-cell receptor engagement. The bsAb panel exhibited a range of potencies, enabling the selection of a clinical candidate, termed RNDO-564, that maximized antitumor efficacy as well as the likelihood of a broad therapeutic window. Tumor-bearing syngeneic mouse models confirmed the in vivo efficacy of RNDO-564, demonstrating significant tumor regression both as a single agent and in combination with an immune checkpoint inhibitor. We observed favorable PK and tolerability profiles in NHP assessments. CONCLUSIONS Our study reports the first CD28 bsAb targeting Nectin-4 and highlights the potential of CD28 × Nectin-4 bsAbs as a new immunotherapeutic modality. The findings support the clinical development of RNDO-564 in patients with locally advanced and metastatic UC and other Nectin-4 positive malignancies.
Collapse
Affiliation(s)
- Manpreet Kaur
- Preclinical Biology, Rondo Therapeutics Inc, Hayward, California, USA
| | - Katja Rüger
- Protein Science, Rondo Therapeutics Inc, Hayward, California, USA
| | - Elaine C Chen
- Translation Science, Rondo Therapeutics Inc, Hayward, California, USA
| | | | - Laura M Davison
- Discovery Biology, Rondo Therapeutics Inc, Hayward, California, USA
| | | | - Imani Smith
- Protein Science, Rondo Therapeutics Inc, Hayward, California, USA
| | - Ruth Chu
- Preclinical Biology, Rondo Therapeutics Inc, Hayward, California, USA
| | | | | | | | | | | | - Starlynn C Clarke
- Preclinical Biology, Rondo Therapeutics Inc, Hayward, California, USA
| |
Collapse
|
3
|
Vickram S, Infant SS, Manikandan S, Jenila Rani D, Mathan Muthu CM, Chopra H. Immune biomarkers and predictive signatures in gastric cancer: Optimizing immunotherapy responses. Pathol Res Pract 2025; 265:155743. [PMID: 39616978 DOI: 10.1016/j.prp.2024.155743] [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: 09/20/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
Gastric cancer is a malignant disease with a poor prognosis and few therapeutic options once it has advanced. Immunotherapy using ICIs has emerged as a viable therapeutic method; nevertheless, reliable immunological biomarkers are required to identify who may benefit from these therapies. It focuses on key immune biomarkers and predictive signatures in gastric cancer, such as PD-L1 expression, microsatellite instability (MSI), tumor mutational burden (TMB), and Epstein-Barr virus (EBV) status, to optimize gastric cancer patients' immunotherapy responses. PD-L1 expression is a popular biomarker for ICI effectiveness. Tumors with high MSI-H and TMB are the most susceptible to ICIs because they are highly immunogenic. EBV-positive stomach tumors are highly immunogenic, and immunotherapy has a high response rate. Combining composite biomarker panels with multi-omics-based techniques improved patient selection accuracy. In recent years, machine learning models have been integrated into next-generation sequencing. Dynamic, real-time-monitorable biomarkers for real-time immune response monitoring are also being considered. Thus, enhancing biomarker-driven immunotherapy is critical for improving clinical outcomes with gastric cancer. There is still more work to be done in this field, and verifying developing biomarkers will be an important component in the future of customized cancer therapy.
Collapse
Affiliation(s)
- Sundaram Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India.
| | - Shofia Saghya Infant
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - S Manikandan
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - D Jenila Rani
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - C M Mathan Muthu
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Hitesh Chopra
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
4
|
Drum DL, Jallorina AG, Wan LS, Chang VT, Lee-Wong MF. Non-Genetic Biomarkers in Merkel Cell Carcinoma: Prognostic Implications and Predictive Utility for Response to Anti-PD-(L)1 Immune Checkpoint Inhibitors. Exp Dermatol 2025; 34:e70030. [PMID: 39791602 DOI: 10.1111/exd.70030] [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: 05/31/2024] [Revised: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Merkel cell carcinoma (MCC) is a skin cancer that arises due to either Merkel cell polyomavirus infection (MCPyV) or ultraviolet (UV) radiation exposure, presenting primarily in the head and neck region of fair-skinned males. The recent success of PD-(L)1 immune checkpoint inhibitors (ICIs) in locally advanced/metastatic MCC, with an objective response rate (ORR) around 50% and improved survival, as a first-line treatment has moved ICIs to the forefront of therapy for MCC and generated interest in identifying biomarkers to predict clinical response. The MCC tumour microenvironment (TME) contains various components of the adaptive and innate immune system. These components can contribute to tumour immune escape through immunosuppression by preventing entrance of other immune cells or by aiding in the cytotoxic clearance of tumour cells. We aim to combine information from studies of baseline and on-treatment monitoring of the TME to help predict the success of ICIs in MCC. This review enhances the understanding of how CD8 T cells, γδ T cells and macrophages may impact predictions of response rates to ICIs in MCC patients. These immune cells are non-genetic biomarkers that can also be used to determine prognosis in MCC treatment.
Collapse
Affiliation(s)
- David L Drum
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Anika G Jallorina
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Leo S Wan
- Department of Medicine, West Virginia School of Osteopathic Medicine, Lewisburg, West Virginia, USA
| | - Victor T Chang
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey School of Medicine, Newark, New Jersey, USA
- Section of Hematology/Oncology, Veterans Administration New Jersey Health Care System, East Orange, New Jersey, USA
| | - Mary F Lee-Wong
- Department of Medicine and Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Allergy and Immunology, Maimonides Medical Center, Brooklyn, New York, USA
| |
Collapse
|
5
|
Xia M, Han Y, Sun L, Li D, Zhu C, Li D. The role of neutrophils in osteosarcoma: insights from laboratory to clinic. Front Immunol 2024; 15:1490712. [PMID: 39582869 PMCID: PMC11582048 DOI: 10.3389/fimmu.2024.1490712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024] Open
Abstract
Osteosarcoma, a highly aggressive malignant bone tumor, is significantly influenced by the intricate interactions within its tumor microenvironment (TME), particularly involving neutrophils. This review delineates the multifaceted roles of neutrophils, including tumor-associated neutrophils (TANs) and neutrophil extracellular traps (NETs), in osteosarcoma's pathogenesis. TANs exhibit both pro- and anti-tumor phenotypes, modulating tumor growth and immune evasion, while NETs facilitate tumor cell adhesion, migration, and immunosuppression. Clinically, neutrophil-related markers such as the neutrophil-to-lymphocyte ratio (NLR) predict patient outcomes, highlighting the potential for neutrophil-targeted therapies. Unraveling these complex interactions is crucial for developing novel treatment strategies that harness the TME to improve osteosarcoma management.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongsong Li
- Department of Orthopedics, The First Hospital of Jilin University,
Changchun, Jilin, China
| |
Collapse
|
6
|
Kudo-Saito C, Imazeki H, Ozawa H, Kawakubo H, Hirano H, Boku N, Kato K, Shoji H. Targeting SNCA in the treatment of malignant ascites in gastrointestinal cancer. Transl Oncol 2024; 48:102075. [PMID: 39098214 PMCID: PMC11345905 DOI: 10.1016/j.tranon.2024.102075] [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: 05/13/2024] [Revised: 07/09/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
Peritoneal tumor dissemination and subsequent malignant tumor ascites (MTA) occur unexpectedly and repeatedly in patients with gastrointestinal (GI) cancers, and worsen quality of life and prognosis of the patients. Various treatments have been clinically developed for these patients, while most of the MTA cases are refractory to the treatments. Thus, effective treatments are urgently needed to improve the clinical outcomes. In this study, we identified α-synuclein (SNCA) as an immunological determinant of MTA progression in GI cancer through translational research using mouse tumor models and clinical specimens collected from gastric cancer patients. We found that the SNCA+ subsets were significantly increased in CD3+ T cells, CD56+ NK cells, and CD11b+ myeloid cells within MTA and peripheral blood cells (PBCs) of MTA cases, albeit almost absent in PBCs of healthy donors, and spleen of naive mice. Of note, the SNCA+ T-cell subset was rarely seen in patients that intraperitoneal lavage fluid without tumor cells was collected before surgery as a tumor-free control, suggesting a possible cancer-induced product, especially within the peritoneal cavity. In vivo treatment with anti-SNCA blocking mAb significantly induced anti-tumor effects in mouse MTA models, and synergistically improved anti-PD1 therapeutic efficacy, providing a significantly better prognosis. These suggest that SNCA is involved in severe immunosuppression in the MTA cases, and that blocking SNCA is effective in dramatically improving the immune status in the hosts. Targeting SNCA will be a promising strategy to improve clinical outcomes in the treatment of GI cancer patients, especially with MTA.
Collapse
Affiliation(s)
- Chie Kudo-Saito
- Department of Immune Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Hiroshi Imazeki
- Department of Immune Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hiroki Ozawa
- Department of Immune Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hirofumi Kawakubo
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hidekazu Hirano
- Department of Medical Oncology and General Medicine, IMS Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan; Department of Medical Oncology and General Medicine, IMS Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Ken Kato
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hirokazu Shoji
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| |
Collapse
|
7
|
Pandey S, Cholak ME, Yadali R, Sosman JA, Tetreault MP, Fang D, Pollack SM, Gnjatic S, Obeng RC, Lyerly HK, Sonabend AM, Guevara-Patiño JA, Butterfield LH, Zhang B, Maecker HT, Le Poole IC. Immune Assessment Today: Optimizing and Standardizing Efforts to Monitor Immune Responses in Cancer and Beyond. Cancers (Basel) 2024; 16:475. [PMID: 38339227 PMCID: PMC10854499 DOI: 10.3390/cancers16030475] [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: 12/09/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
As part of a symposium, current and former directors of Immune Monitoring cores and investigative oncologists presented insights into the past, present and future of immune assessment. Dr. Gnjatic presented a classification of immune monitoring technologies ranging from universally applicable to experimental protocols, while emphasizing the need for assay harmonization. Dr. Obeng discussed physiologic differences among CD8 T cells that align with anti-tumor responses. Dr. Lyerly presented the Soldano Ferrone lecture, commemorating the passionate tumor immunologist who inspired many, and covered a timeline of monitoring technology development and its importance to immuno-oncology. Dr. Sonabend presented recent achievements in glioblastoma treatment, accentuating the range of monitoring techniques that allowed him to refine patient selection for clinical trials. Dr. Guevara-Patiño focused on hypoxia within the tumor environment and stressed that T cell viability is not to be confused with functionality. Dr. Butterfield accentuated monitoring of dendritic cell metabolic (dys)function as a determinant for tumor vaccine success. Lectures were interspersed with select abstract presentations. To summarize the concepts, Dr. Maecker from Stanford led an informative forum discussion, pointing towards the future of immune monitoring. Immune monitoring continues to be a guiding light towards effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Surya Pandey
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Meghan E. Cholak
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Rishita Yadali
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Jeffrey A. Sosman
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Marie-Pier Tetreault
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Deyu Fang
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Seth M. Pollack
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Sacha Gnjatic
- Human Immune Monitoring Center, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Rebecca C. Obeng
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - H. Kim Lyerly
- Center for Applied Therapeutics, Duke Cancer Center, Duke University, Durham, NC 27710, USA;
| | - Adam M. Sonabend
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | | | - Lisa H. Butterfield
- Merck Research Laboratories, Boston, MA 02115, USA;
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | - Bin Zhang
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Holden T. Maecker
- Human Immune Monitoring Center, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - I. Caroline Le Poole
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| |
Collapse
|
8
|
Nirala BK, Yamamichi T, Petrescu DI, Shafin TN, Yustein JT. Decoding the Impact of Tumor Microenvironment in Osteosarcoma Progression and Metastasis. Cancers (Basel) 2023; 15:5108. [PMID: 37894474 PMCID: PMC10605493 DOI: 10.3390/cancers15205108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Osteosarcoma (OS) is a heterogeneous, highly metastatic bone malignancy in children and adolescents. Despite advancements in multimodal treatment strategies, the prognosis for patients with metastatic or recurrent disease has not improved significantly in the last four decades. OS is a highly heterogeneous tumor; its genetic background and the mechanism of oncogenesis are not well defined. Unfortunately, no effective molecular targeted therapy is currently available for this disease. Understanding osteosarcoma's tumor microenvironment (TME) has recently gained much interest among scientists hoping to provide valuable insights into tumor heterogeneity, progression, metastasis, and the identification of novel therapeutic avenues. Here, we review the current understanding of the TME of OS, including different cellular and noncellular components, their crosstalk with OS tumor cells, and their involvement in tumor progression and metastasis. We also highlight past/current clinical trials targeting the TME of OS for effective therapies and potential future therapeutic strategies with negligible adverse effects.
Collapse
Affiliation(s)
| | | | | | | | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA 30322, USA; (B.K.N.); (T.Y.); (D.I.P.); (T.N.S.)
| |
Collapse
|