1
|
Huang D, Dai J, Yu H, Chen W. Immune dysregulation in endometrial tuberculosis: elevated HLA-G and IL-1Ra as key modulators. Front Cell Infect Microbiol 2025; 15:1548238. [PMID: 40375897 PMCID: PMC12078264 DOI: 10.3389/fcimb.2025.1548238] [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: 12/19/2024] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
Endometrial tuberculosis (ETB) is a reproductive system infection caused by Mycobacterium tuberculosis, primarily invading the endometrium through hematogenous dissemination. This study included 10 patients diagnosed with ETB and 10 patients with pulmonary tuberculosis (PTB) to analyze their clinical, pathological, and immunological characteristics. Anatomically, PTB presented the highest prevalence among tuberculosis cases. Compared to PTB imaging, CT scans of ETB showed less distinctive diagnostic features. Pathologically, abscess formation was more frequently observed in ETB patients than in PTB patients, suggesting a more intense local inflammatory response in ETB. However, there were no statistically significant differences in granulomatous lesions, caseous necrosis, coagulative necrosis, inflammatory necrosis, exudation, acute inflammation, or fibrous tissue hyperplasia between the two groups. Immunohistochemical analysis revealed higher infiltration of macrophages (CD68) in ETB lesions compared to PTB, whereas the counts of T cells (CD3+, CD4+, CD8+) and B cells (CD20) showed no significant differences. Notably, the expression levels of HLA-G and IP-10 were significantly elevated in the lesion areas of ETB compared to PTB. Similarly, the expression of HLA-G, IP-10, IL-1Ra, and IL-10 was significantly higher in the ETB group than in the PTB group. Furthermore, HLA-G and IL-1Ra expression levels were markedly elevated in ETB lesion areas compared to surrounding normal endometrial tissue. HLA-G plays a pivotal role in immune tolerance by modulating local immune responses, while IP-10 is involved in chronic inflammatory signaling. IL-1Ra and IL-10 are key regulators of endometrial immune homeostasis, counterbalancing inflammatory responses that could otherwise disrupt reproductive function. These immunoregulatory factors are crucial in maintaining immune tolerance within the endometrium and may influence immune responses associated with endometrial tuberculosis.
Collapse
Affiliation(s)
- Dan Huang
- Graduate School of Hebei North University, Zhangjiakou, China
- Department of Reproductive Medicine Clinic, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinlong Dai
- Department of Pathology, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Haotian Yu
- Department of Reproductive Medicine Clinic, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
de Oliveira IM, Chaves MM. The NLRP3 Inflammasome in inflammatory diseases: Cellular dynamics and role in granuloma formation. Cell Immunol 2025; 411-412:104961. [PMID: 40339528 DOI: 10.1016/j.cellimm.2025.104961] [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: 12/28/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
The innate immune system recognizes pathogen-associated molecular patterns (PAMPs) and damage associated molecular patterns (DAMPs) through pattern recognition receptors (PRRs). Inflammasomes, cytoplasmic protein complexes, are activated in response to PAMPs and DAMPs, leading to the release of inflammatory cytokines such as IL-1β and IL-18. NLRP3 inflammasome is one of the best characterized inflammasomes and recently its activation has been associated with granuloma formation, structures that aggregate immune cells in response to infections, such as those caused by bacteria, fungi and parasites, and autoinflammatory diseases, such as sarcoidosis. Activation of NLRP3 inflammasomes in macrophages induces the release of cytokines that recruit immune cells, such as monocytes and lymphocytes, to the site of infection. Neutrophils, monocytes, T and B lymphocytes are important in the formation and maintenance of granulomas. Although NLRP3 plays a key role in the immune response, cell recruitment and granuloma formation, many aspects of its function in different cell types remain to be elucidated. In this review, we aim to outline the NLRP3 inflammasome not only as a protein complex that aids innate immune cells in combating intracellular pathogens but also as a platform with broader implications in orchestrating immune responses. This underexplored aspect of the NLRP3 inflammasome presents a novel perspective on its involvement in immunity. Thus, we review the current understanding of the role of the NLRP3 inflammasome in immune cell infiltration and its significance in the organization and formation of granulomas in inflammatory diseases.
Collapse
Affiliation(s)
- Isadora M de Oliveira
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
3
|
Singh M, Sarhan MO, Damiba NNL, Singh AK, Villabona-Rueda A, Nino-Meza OJ, Chen X, Masias-Leon Y, Ruiz-Gonzalez CE, Ordonez AA, D'Alessio FR, Aboagye EO, Carroll LS, Jain SK. Proapoptotic Bcl-2 inhibitor as potential host directed therapy for pulmonary tuberculosis. Nat Commun 2025; 16:3003. [PMID: 40148277 PMCID: PMC11950383 DOI: 10.1038/s41467-025-58190-x] [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: 08/22/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Mycobacterium tuberculosis establishes within host cells by inducing anti-apoptotic Bcl-2 family proteins, triggering necrosis, inflammation, and fibrosis. Here, we demonstrate that navitoclax, an orally bioavailable, small-molecule Bcl-2 inhibitor, significantly improves pulmonary tuberculosis (TB) treatments as a host-directed therapy. Addition of navitoclax to standard TB treatments at human equipotent dosing in mouse models of TB, inhibits Bcl-2 expression, leading to improved bacterial clearance, reduced tissue necrosis, fibrosis and decreased extrapulmonary bacterial dissemination. Using immunohistochemistry and flow cytometry, we show that navitoclax induces apoptosis in several immune cells, including CD68+ and CD11b+ cells. Finally, positron emission tomography (PET) in live animals using clinically translatable biomarkers for apoptosis (18F-ICMT-11) and fibrosis (18F-FAPI-74), demonstrates that navitoclax significantly increases apoptosis and reduces fibrosis in pulmonary tissues, which are confirmed in postmortem analysis. Our studies suggest that proapoptotic drugs such as navitoclax can potentially improve pulmonary TB treatments, reduce lung damage / fibrosis and may be protective against post-TB lung disease.
Collapse
Affiliation(s)
- Medha Singh
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mona O Sarhan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nerketa N L Damiba
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alok K Singh
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | | | - Oscar J Nino-Meza
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xueyi Chen
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuderleys Masias-Leon
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carlos E Ruiz-Gonzalez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Franco R D'Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery & Cancer, Hammersmith Campus, Imperial College, London, UK
| | - Laurence S Carroll
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Krueger G, Faisal S, Dorhoi A. Microenvironments of tuberculous granuloma: advances and opportunities for therapy. Front Immunol 2025; 16:1575133. [PMID: 40196129 PMCID: PMC11973276 DOI: 10.3389/fimmu.2025.1575133] [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: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
The hallmark tissue lesions of tuberculosis (TB) are granulomas. These multicellular structures exhibit varying degrees of cellular complexity, are dynamic, and show considerable diversity within and between hosts. Categorization based on gross pathologic features, particularly caseation and necrosis, was historically coined prior to the identification of mycobacteria as the causative agent of TB. More recently, granuloma zonation based on immune cell composition, metabolite abundance, and physical characteristics has gained attention. With the advent of single-cell analyses, distinct microenvironments and cellular ecosystems within TB granulomas have been identified. We summarize the architecture of TB granulomas and highlight their cellular heterogeneity, including cell niches as well as physical factors such as oxygen gradients that modulate lesion fate. We discuss opportunities for therapy, highlighting new models and the power of in silico modeling to unravel granuloma features and trajectories. Understanding the relevance of the granuloma microenvironment to disease pathophysiology will facilitate the development of more effective interventions, such as host-directed therapies for TB.
Collapse
Affiliation(s)
- Gesa Krueger
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Shah Faisal
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
5
|
Senent Y, Remírez A, Repáraz D, Llopiz D, Celias DP, Sainz C, Entrialgo-Cadierno R, Suarez L, Rouzaut A, Alignani D, Tavira B, Lambris JD, Woodruff TM, de Andrea CE, Ruffell B, Sarobe P, Ajona D, Pio R. The C5a/C5aR1 Axis Promotes Migration of Tolerogenic Dendritic Cells to Lymph Nodes, Impairing the Anticancer Immune Response. Cancer Immunol Res 2025; 13:384-399. [PMID: 39666368 DOI: 10.1158/2326-6066.cir-24-0250] [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: 03/15/2024] [Revised: 07/30/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
The precise mechanisms by which the complement system contributes to the establishment of an immunosuppressive tumor microenvironment and promotes tumor progression remain unclear. In this study, we investigated the expression and function of complement C5a receptor 1 (C5aR1) in human and mouse cancer-associated dendritic cells (DC). First, we observed an overexpression of C5aR1 in tumor-infiltrating DCs, compared with DCs from the blood or spleen. C5aR1 expression was restricted to type 2 conventional DCs and monocyte-derived DCs, which displayed a tolerogenic phenotype capable of inhibiting T-cell activation and promoting tumor growth. C5aR1 engagement in DCs drove their migration from tumors to tumor-draining lymph nodes, where C5a levels were higher. We used this knowledge to optimize an anticancer therapy aimed at enhancing DC activity. In three syngeneic tumor models, C5aR1 inhibition significantly enhanced the efficacy of poly I:C, a Toll-like receptor 3 agonist, in combination with PD-1/PD-L1 blockade. The contribution of C5aR1 inhibition to the antitumor activity of the combination treatment relied on type 1 conventional DCs and antigen-specific CD8+ T cells, required lymphocyte egress from secondary lymphoid organs, and was associated with an increase in IFNγ signaling. In conclusion, our study highlights the importance of the C5a/C5aR1 axis in the biology of cancer-associated DCs and provides compelling evidence for the therapeutic potential of modulating the complement system to enhance DC-mediated immune responses against tumors.
Collapse
Affiliation(s)
- Yaiza Senent
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Ana Remírez
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - David Repáraz
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Program in Immunology and Immunotherapy, CCUN, Cima Universidad de Navarra, Pamplona, Spain
| | - Diana Llopiz
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Program in Immunology and Immunotherapy, CCUN, Cima Universidad de Navarra, Pamplona, Spain
| | - Daiana P Celias
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Cristina Sainz
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Rodrigo Entrialgo-Cadierno
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Lucia Suarez
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
| | - Ana Rouzaut
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Diego Alignani
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Cytometry Unit, Cima Universidad de Navarra, Pamplona, Spain
| | - Beatriz Tavira
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| | | | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Pablo Sarobe
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Program in Immunology and Immunotherapy, CCUN, Cima Universidad de Navarra, Pamplona, Spain
| | - Daniel Ajona
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Ruben Pio
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
6
|
Lawrence ALE, Tan S. Building Spatiotemporal Understanding of Mycobacterium tuberculosis-Host Interactions. ACS Infect Dis 2025; 11:277-286. [PMID: 39847659 PMCID: PMC11828672 DOI: 10.1021/acsinfecdis.4c00840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Heterogeneity during Mycobacterium tuberculosis (Mtb) infection greatly impacts disease outcome and complicates treatment. This heterogeneity encompasses many facets, spanning local differences in the host immune response to Mtb and the environment experienced by the bacterium, to nonuniformity in Mtb replication state. All of these facets are interlinked and each can affect Mtb susceptibility to antibiotic treatment. In-depth spatiotemporal understanding of Mtb-host interactions is thus critical to both fundamental comprehension of Mtb infection biology and for the development of effective therapeutic regimens. Such spatiotemporal understanding dictates the need for analysis at the single bacterium/cell level in the context of intact tissue architecture, which has been a significant technical challenge. Excitingly, innovations in spatial single cell methodology have opened the door to such studies, beginning to illuminate aspects ranging from intergranuloma differences in cellular composition and phenotype, to sublocation differences in Mtb physiology and replication state. In this perspective, we discuss recent studies that demonstrate the potential of these methodological advancements to reveal critical spatiotemporal insight into Mtb-host interactions, and highlight future avenues of research made possible by these advances.
Collapse
Affiliation(s)
- Anna-Lisa E Lawrence
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| |
Collapse
|
7
|
Garate-Soraluze E, Serrano-Mendioroz I, Fernández-Rubio L, De Andrea CE, Barrio-Alonso C, Herrero CDP, Teijeira A, Luri-Rey C, Claus C, Tanos T, Klein C, Umana P, Rullan A, Simón JA, Collantes M, Sánchez-Mateos P, Melero I, Rodriguez-Ruiz ME. 4-1BB agonist targeted to fibroblast activation protein α synergizes with radiotherapy to treat murine breast tumor models. J Immunother Cancer 2025; 13:e009852. [PMID: 39933836 PMCID: PMC11815443 DOI: 10.1136/jitc-2024-009852] [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: 06/11/2024] [Accepted: 11/01/2024] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Ionizing radiation (IR) is a double-edged sword for immunotherapy as it may have both immunosuppressive and immunostimulatory effects. The biological effects of IR on the tumor microenvironment (TME) are a key factor for this balance. Fibroblast activation protein (FAP) is expressed on the surface of cancer-associated fibroblasts (CAF) in many cancer types and its abundance is associated with the poor immune response to immune-checkpoint-blockade in patients. We hypothesized that IR increases FAP expression in CAFs, therefore the combination of IR with targeted immunomodulators such as an agonistic anti-FAP-4-1BBL fusion protein could enhance the immune-mediated antitumoral effects of these treatments. METHODS The murine transplantable TS/A tumor-cell-line co-engrafted with CAFs was used to investigate increases in FAP expression in tumors following irradiation using immunohistochemistry, real-time polymerase chain reaction (RT-PCR) and multiplex tissue immunofluorescence. One lesion of bilateral tumor-bearing mice was only locally irradiated or combined with weekly injections of the bispecific muFAP-4-1BBL fusion protein (a mouse surrogate for RG7826). Tumor sizes were followed over time and TME was assessed by flow cytometry. Selective monoclonal antibody (mAb)-mediated depletions of immune cell populations, neutralizing interferon alpha/beta receptor 1 (IFNAR-I) IFNAR and interferon (IFN)-γ mAbs and gene-modified mice (4-1BB-/-) were used to delineate the immune cell subsets and mechanisms required for efficacy. 67Ga labeled muFAP-4-1BBL tracked by SPECT-CT was used to study biodistribution. In human colorectal carcinoma samples, the inducibility of FAP expression following radiotherapy was explored by multiplex immunofluorescence. RESULTS Irradiation of TS/A+CAF tumors in mice showed an increase in FAP levels after local irradiation. A suboptimal radiotherapy regimen in combination with muFAP-4-1BBL attained primary tumor control and measurable abscopal effects. Immune TME landscape analyses showed post-treatment increased infiltration of activated immune cells associated with the combined radioimmunotherapy treatment. Efficacy depended on CD8+ T cells, type I IFN, IFN-γ and ability to express 4-1BB. Biodistribution studies of muFAP-4-1BBL indicated enriched tumor targeting to irradiated tumors. Human colorectal cancer samples pre and post irradiation showed enhanced FAP expression after radiotherapy. CONCLUSION Increased FAP expression in the TME as a result of radiotherapy can be exploited to target agonist 4-1BB immunotherapy to malignant tumor lesions using an FAP-4-1BBL antibody fusion protein.
Collapse
Affiliation(s)
- Eneko Garate-Soraluze
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Irantzu Serrano-Mendioroz
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Leticia Fernández-Rubio
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Carlos E De Andrea
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Spain
| | | | | | - Alvaro Teijeira
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Carlos Luri-Rey
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Christina Claus
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Tamara Tanos
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Pablo Umana
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Antonio Rullan
- Institute of Immunology and Transplantation, University College London, London, UK
| | - Jon Ander Simón
- Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - María Collantes
- Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Departments of Immunology-Immunotherapy and Radiation Oncology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Institute for Health Research (IDISNA), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Departments of Immunology-Immunotherapy and Radiation Oncology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Institute for Health Research (IDISNA), Pamplona, Spain
| |
Collapse
|
8
|
Datta M, Via LE, Dartois V, Xu L, Barry CE, Jain RK. Leveraging insights from cancer to improve tuberculosis therapy. Trends Mol Med 2025; 31:11-20. [PMID: 39142973 PMCID: PMC11717643 DOI: 10.1016/j.molmed.2024.07.011] [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: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024]
Abstract
Exploring and exploiting the microenvironmental similarities between pulmonary tuberculosis (TB) granulomas and malignant tumors has revealed new strategies for more efficacious host-directed therapies (HDTs). This opinion article discusses a paradigm shift in TB therapeutic development, drawing on critical insights from oncology. We summarize recent efforts to characterize and overcome key shared features between tumors and granulomas, including excessive fibrosis, abnormal angiogenesis, hypoxia and necrosis, and immunosuppression. We provide specific examples of cancer therapy application to TB to overcome these microenvironmental abnormalities, including matrix-targeting therapies, antiangiogenic agents, and immune-stimulatory drugs. Finally, we propose a new framework for combining HDTs with anti-TB agents to maximize therapeutic delivery and efficacy while reducing treatment dosages, duration, and harmful side effects to benefit TB patients.
Collapse
Affiliation(s)
- Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; Hackensack Meridian School of Medicine, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Lei Xu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
9
|
Li Q, Wang C, Gou J, Kitanovski S, Tang X, Cai Y, Zhang C, Zhang X, Zhang Z, Qiu Y, Zhao F, Lu M, He Y, Wang J, Lu H. Deciphering lung granulomas in HIV & TB co-infection: unveiling macrophages aggregation with IL6R/STAT3 activation. Emerg Microbes Infect 2024; 13:2366359. [PMID: 38855910 PMCID: PMC11188963 DOI: 10.1080/22221751.2024.2366359] [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: 03/31/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Tuberculosis (TB) remains a leading cause of mortality among individuals coinfected with HIV, characterized by progressive pulmonary inflammation. Despite TB's hallmark being focal granulomatous lung lesions, our understanding of the histopathological features and regulation of inflammation in HIV & TB coinfection remains incomplete. In this study, we aimed to elucidate these histopathological features through an immunohistochemistry analysis of HIV & TB co-infected and TB patients, revealing marked differences. Notably, HIV & TB granulomas exhibited aggregation of CD68 + macrophage (Mφ), while TB lesions predominantly featured aggregation of CD20+ B cells, highlighting distinct immune responses in coinfection. Spatial transcriptome profiling further elucidated CD68+ Mφ aggregation in HIV & TB, accompanied by activation of IL6 pathway, potentially exacerbating inflammation. Through multiplex immunostaining, we validated two granuloma types in HIV & TB versus three in TB, distinguished by cell architecture. Remarkably, in the two types of HIV & TB granulomas, CD68 + Mφ highly co-expressed IL6R/pSTAT3, contrasting TB granulomas' high IFNGRA/SOCS3 expression, indicating different signaling pathways at play. Thus, activation of IL6 pathway may intensify inflammation in HIV & TB-lungs, while SOCS3-enriched immune microenvironment suppresses IL6-induced over-inflammation in TB. These findings provide crucial insights into HIV & TB granuloma formation, shedding light on potential therapeutic targets, particularly for granulomatous pulmonary under HIV & TB co-infection. Our study emphasizes the importance of a comprehensive understanding of the immunopathogenesis of HIV & TB coinfection and suggests potential avenues for targeting IL6 signaling with SOCS3 activators or anti-IL6R agents to mitigate lung inflammation in HIV & TB coinfected individuals.
Collapse
MESH Headings
- Adult
- Female
- Humans
- Male
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- CD68 Molecule
- Coinfection/virology
- Coinfection/immunology
- Coinfection/microbiology
- Granuloma/immunology
- HIV Infections/complications
- HIV Infections/immunology
- Interleukin-6/metabolism
- Interleukin-6/genetics
- Lung/pathology
- Lung/immunology
- Macrophages/immunology
- Receptors, Interleukin-6/metabolism
- Receptors, Interleukin-6/genetics
- Signal Transduction
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Suppressor of Cytokine Signaling 3 Protein/metabolism
- Suppressor of Cytokine Signaling 3 Protein/genetics
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/complications
Collapse
Affiliation(s)
- Qian Li
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Cheng Wang
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Jizhou Gou
- Department of Pathology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Simo Kitanovski
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, Essen, Germany
| | - XiangYi Tang
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Yixuan Cai
- Clinical Research Center, The Fifth People’s Hospital of Wuxi, Jiangnan University, Wuxi, People’s Republic of China
| | - Chenxia Zhang
- Clinical Research Center, The Fifth People’s Hospital of Wuxi, Jiangnan University, Wuxi, People’s Republic of China
| | - Xiling Zhang
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Zhenfeng Zhang
- School of Public Health and Emergency Management, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Yuanwang Qiu
- Clinical Research Center, The Fifth People’s Hospital of Wuxi, Jiangnan University, Wuxi, People’s Republic of China
| | - Fang Zhao
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Mengji Lu
- Institute of Virology, Essen University Hospital, University of Duisburg-Essen, Essen, German
| | - Yun He
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Jun Wang
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, Essen, Germany
- Clinical Research Center, The Fifth People’s Hospital of Wuxi, Jiangnan University, Wuxi, People’s Republic of China
| | - Hongzhou Lu
- National Clinical Research Center for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China
| |
Collapse
|
10
|
Moura DS, Lopez-Marti JM, Benesova I, de Andrea C, di Lernia D, Lacerenza S, Mondaza-Hernandez JL, Martin-Ruiz M, Ramirez-Calvo M, Grignani G, Martinez-Trufero J, Redondo A, Valverde C, Stacchiotti S, Lopez-Pousa A, Lopez-Guerrero JA, Gutierrez A, Encinas-Tobajas V, Hindi N, Sangiolo D, Lopez-Martin JA, Strizova ZO, Martin-Broto J. Predictive and Dynamic Signature for Antiangiogenics in Combination with a PD1 Inhibitor in Soft-Tissue Sarcoma: Correlative Studies Linked to the IMMUNOSARC Trial. Clin Cancer Res 2024; 30:5192-5206. [PMID: 39283727 DOI: 10.1158/1078-0432.ccr-24-1782] [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: 06/06/2024] [Revised: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE The IMMUNOSARC trial combined an antiangiogenic agent (sunitinib) with a PD1 inhibitor (nivolumab) in advanced sarcomas. Here, we present the first correlative studies of the soft-tissue sarcoma cohort enrolled in this trial. EXPERIMENTAL DESIGN Formalin-fixed paraffin-embedded and peripheral blood samples were collected at baseline and week 13. Formalin-fixed paraffin-embedded samples were used for transcriptomics and multiplex immunofluorescence, whereas peripheral blood samples were used for multiplexed immunoassays. Flow cytometry and Luminex assays were performed to validate translational findings in tumor-isolated cells and peripheral blood mononuclear cells derived from patients. RESULTS The density of intratumoral CD8+ T cells, measured by multiplexed immunophenotyping, was significantly increased after treatment. This augment was accompanied by the dynamic significant increase in the gene expressions of CD86, CHI3L1, CXCL10, CXCL9, LAG3, and VCAM1 and the decrease in the expression levels of NR4A1. In peripheral blood, 12 proteins were significantly modulated by treatment at week 13. A score integrating the dynamic expression of the 7 genes and the 12 soluble factors separated 2 groups with distinct progression-free survival (PFS): 4.1 months [95% confidence interval, 3.5-not reached (NR)] versus 17 months (95% confidence interval, 12.0-NR), P = 0.014. This molecular score was predictive of PFS when applied to the normalized data determined in the baseline samples. CONCLUSIONS Treatment with sunitinib and nivolumab inflamed the sarcoma microenvironment, increasing CD8+ T-cell density and the expression of several genes/proteins with relevance in the response to PD1 inhibitors. A molecular signature identified two groups of patients with distinct PFS for the combination of antiangiogenics plus PD1 inhibitor therapy.
Collapse
Affiliation(s)
- David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Jesus M Lopez-Marti
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Carlos de Andrea
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Davide di Lernia
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Serena Lacerenza
- Institute of Biomedicine of Seville (IBiS; US, CSIC, HUVR), Seville, Spain
| | | | - Marta Martin-Ruiz
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | | | - Giovanni Grignani
- Medical Oncology Unit, Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Andres Redondo
- Department of Medical Oncology, University Hospital La Paz, Madrid, Spain
| | - Claudia Valverde
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - José A Lopez-Guerrero
- Fundación Institute Valenciano of Oncology, Valencia, Spain
- Joint Cancer Research Unit IVO-CIPF, Valencia, Spain
- Department of Pathology, Medical School of the Catholic University of Valencia, Valencia, Spain
| | - Antonio Gutierrez
- Department of Hematology, University Hospital Son Espases, Mallorca, Spain
| | | | - Nadia Hindi
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
- Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy
| | - Jose A Lopez-Martin
- Department of Medical Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Zuzana Ozaniak Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Javier Martin-Broto
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
- Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| |
Collapse
|
11
|
Eguren-Santamaria I, Fernández de Piérola E, Camps G, Martín-Muñoz P, Campos M, Cuculescu D, Aguilera-Buenosvinos I, Rodríguez López I, Salido-Vallejo R, Alexandru R, De Andrea CE, Álvarez-Gigli L, Berraondo P, Melero I, Sanmamed MF. MHC class I and II-deficient humanized mice are suitable tools to test the long-term antitumor efficacy of immune checkpoint inhibitors and T-cell engagers. J Immunother Cancer 2024; 12:e008516. [PMID: 39244214 PMCID: PMC11381650 DOI: 10.1136/jitc-2023-008516] [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: 08/09/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Immunodeficient mice engrafted with peripheral blood mononuclear cells (PBMCs) are models to study new cancer immunotherapy agents. However, this approach is associated with xenograft-versus-host disease (xGVHD), which starts early after PBMC transfer and limits the duration and interpretation of experiments. Here, we explore different approaches to overcome xGVHD and better support the development of cancer immunotherapies. METHODS Immunodeficient NOD-scid IL2Rgnull (NSG) mice were intravenously transferred with human PBMCs and subcutaneously co-engrafted with HT29 human colon carcinoma cells. Diverse strategies to reduce xGVHD while preserving the antitumor activity of human immune cells were evaluated: (1) ex vivo immune graft modification by depleting CD4+ T cells pre-transfer using magnetic beads, (2) post-transplantation cyclophosphamide administration to eliminate proliferating xenoreactive T-cell clones and (3) using major histocompatibility complex (MHC) class I and II-deficient NSG mice: (Kb Db)null (IA)null (MHC-dKO NSG). Body weight and plasma murine alanine aminotransferase levels were measured as indicators of xGVHD and tumor size was measured every 2-3 days to monitor antitumor activity. The antitumor effects and pharmacodynamics of nivolumab plus ipilimumab and an anti-epithelial cell adhesion molecule (EpCAM)/CD3 T-cell engager (αEpCAM/CD3 bispecific antibody (BsAb)) were evaluated in the model. RESULTS CD4+ T-cell depletion attenuates xGVHD but also abrogates the antitumor activity. Cyclophosphamide limits the antitumor response and does not substantially prevent xGVHD. In contrast, xGVHD was significantly attenuated in MHC-dKO NSG recipients, while the antitumor effect of human PBMCs was preserved. Furthermore, the administration of nivolumab plus ipilimumab caused exacerbated xGVHD in conventional NSG mice, thereby precluding the observation of their antitumor effects. Severe xGVHD did not occur in MHC-dKO NSG mice thus enabling the study of complete and durable tumor rejections. Similarly, NSG mice treated with an αEpCAM/CD3 BsAb showed complete tumor regressions, but died due to xGVHD. In contrast, MHC-dKO NSG mice on treatment with the αEpCAM/CD3 BsAb achieved complete tumor responses without severe xGVHD. A significant proportion of mice rendered tumor-free showed tumor rejection on rechallenge with HT29 cells without further treatment. Finally, tumor-infiltrating CD8+ T-cell number increase, activation and CD137 upregulation were observed on αEpCAM/CD3 BsAb treatment. CONCLUSION Humanized MHC-dKO immunodeficient mice allow and refine the preclinical testing of immunotherapy agents for which experimentation is precluded in conventional immunodeficient mice due to severe xGVHD.
Collapse
Affiliation(s)
- Iñaki Eguren-Santamaria
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Medical Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
| | - Eva Fernández de Piérola
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
| | - Gracián Camps
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
| | - Paula Martín-Muñoz
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
| | | | - Doina Cuculescu
- Clinical Trial Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Inmaculada Rodríguez López
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
| | | | - Raluca Alexandru
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Carlos E De Andrea
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | | | - Pedro Berraondo
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| | - Miguel F Sanmamed
- Immunology and Immunotherapy, Centro de Investigación Médica Aplicada, Pamplona, Spain
- Medical Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Paplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| |
Collapse
|
12
|
Jain S, Singh M, Sarhan M, Damiba N, Singh A, Villabona-Rueda A, Meza ON, Chen X, Ordonez A, D'Alessio F, Aboagye E, Carroll L. Proapoptotic Bcl-2 inhibitor as host directed therapy for pulmonary tuberculosis. RESEARCH SQUARE 2024:rs.3.rs-4926508. [PMID: 39281866 PMCID: PMC11398574 DOI: 10.21203/rs.3.rs-4926508/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Mycobacterium tuberculosis establishes within host cells by inducing anti-apoptotic Bcl-2 family proteins, triggering necrosis, inflammation, and fibrosis. Here, we demonstrate that navitoclax, an orally bioavailable, small-molecule Bcl-2 inhibitor, significantly improves pulmonary tuberculosis (TB) treatments as a host-directed therapy. Addition of navitoclax to standard TB treatments at human equipotent dosing in mouse models of TB, inhibits Bcl-2 expression, leading to improved bacterial clearance, reduced tissue damage / fibrosis and decreased extrapulmonary bacterial dissemination. Using immunohistochemistry and flow cytometry, we show that navitoclax induces apoptosis in several immune cells, including CD68 + and CD11b + cells. Finally, positron emission tomography (PET) in live animals using novel, clinically translatable biomarkers for apoptosis (18F-ICMT-11) and fibrosis (18F-FAPI-74) demonstrates that navitoclax significantly increases apoptosis and reduces fibrosis in pulmonary tissues, which are confirmed using post-mortem studies. Our studies suggest that proapoptotic drugs such as navitoclax can improve pulmonary TB treatments, and should be evaluated in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Alok Singh
- Johns Hopkins University School of Medicine
| | | | | | - Xueyi Chen
- Johns Hopkins University School of Medicine
| | | | | | | | | |
Collapse
|
13
|
Tomás-Velázquez A, López-Gutiérrez JC, de Andrea C, Reyes-Múgica M, Salgado CM, Redondo P. Alpelisib decreases nevocytes of congenital melanocytic nevi. J Eur Acad Dermatol Venereol 2024; 38:1147-1151. [PMID: 38071601 DOI: 10.1111/jdv.19736] [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: 08/04/2023] [Accepted: 10/26/2023] [Indexed: 05/26/2024]
Abstract
BACKGROUND Multiple, large or giant congenital melanocytic nevi (CMN) are uncommon and affected patients can show progressive growth and thickening, associate neurocutaneous melanocytosis or develop melanoma. Current treatment modalities are mostly complex surgeries that frequently do not solve the disease and its risks completely. Thus, investigation on new treatment options for CMN and its complications must continue. MAPK pathway inhibitors are being investigated, also targeting PI3K-AKT. Omipalisib (PI3K inhibitor, with no indications approved yet) has been studied for CMN in vitro and in mice with promising results. However, alpelisib, a PI3K inhibitor approved with an adequate safety profile for patients with severe manifestations of PROS (PIK3CA-Related Overgrowth Spectrum), had not yet been tested for CMN. OBJECTIVE To evaluate the effect of alpelisib in nevocytes of congenital melanocytic nevi. METHODS Nevomelanocytic tissue samples of 10 patients were collected prospectively and, following a previously reported preclinical ex vivo model, explants were placed in organotypic culture for 5 days, with or without alpelisib. Consecutively, tissue sections were stained and using scanned images with Qupath and ImageJ softwares, representative regions from the dermis were analysed (using Wilcoxon test and Spearman's correlation). RESULTS When comparing alpelisib-treated explants with respect to control explants, we found a decrease in cell density (p = 0.0273), in density of SOX10+-cells (p = 0.0391) and also in the % of S-100+ area (p = 0.0078), in alpelisib samples. The three markers showed a positive correlation (p < 0.05). CONCLUSIONS This study provides first-time evidence that alpelisib induces nevocyte reduction in CMN from patient-derived explants, probably inducted by autophagy. Alpelisib is an approved drug with an adequate safety profile used in another mosaicism affecting PI3K (PROS). Further studies are needed to evaluate its efficacy in treating CMN and potentially, their complications, either with local or systemic administration, alone or in combination.
Collapse
Affiliation(s)
| | | | - Carlos de Andrea
- Department of Pathology, University Clinic of Navarra, Pamplona, Spain
| | - Miguel Reyes-Múgica
- Department of Pathology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Claudia M Salgado
- Department of Pathology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pedro Redondo
- Department of Dermatology, University Clinic of Navarra, Madrid, Spain
| |
Collapse
|
14
|
Carow B, Muliadi V, Skålén K, Yokota C, Kathamuthu GR, Setiabudiawan TP, Lange C, Scheu K, Gaede KI, Goldmann T, Pandita A, Masood KI, Pervez S, Grunewald J, Hasan Z, Levin M, Rottenberg ME. Immune mapping of human tuberculosis and sarcoidosis lung granulomas. Front Immunol 2024; 14:1332733. [PMID: 38385142 PMCID: PMC10879604 DOI: 10.3389/fimmu.2023.1332733] [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: 11/03/2023] [Accepted: 12/18/2023] [Indexed: 02/23/2024] Open
Abstract
Tuberculosis (TB) and sarcoidosis are both granulomatous diseases. Here, we compared the immunological microenvironments of granulomas from TB and sarcoidosis patients using in situ sequencing (ISS) transcriptomic analysis and multiplexed immunolabeling of tissue sections. TB lesions consisted of large necrotic and cellular granulomas, whereas "multifocal" granulomas with macrophages or epitheloid cell core and a T-cell rim were observed in sarcoidosis samples. The necrotic core in TB lesions was surrounded by macrophages and encircled by a dense T-cell layer. Within the T-cell layer, compact B-cell aggregates were observed in most TB samples. These B-cell clusters were vascularized and could contain defined B-/T-cell and macrophage-rich areas. The ISS of 40-60 immune transcripts revealed the enriched expression of transcripts involved in homing or migration to lymph nodes, which formed networks at single-cell distances in lymphoid areas of the TB lesions. Instead, myeloid-annotated regions were enriched in CD68, CD14, ITGAM, ITGAX, and CD4 mRNA. CXCL8 and IL1B mRNA were observed in granulocytic areas in which M. tuberculosis was also detected. In line with ISS data indicating tertiary lymphoid structures, immune labeling of TB sections expressed markers of high endothelial venules, follicular dendritic cells, follicular helper T cells, and lymph-node homing receptors on T cells. Neither ISS nor immunolabeling showed evidence of tertiary lymphoid aggregates in sarcoidosis samples. Together, our finding suggests that despite their heterogeneity, the formation of tertiary immune structures is a common feature in granulomas from TB patients.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Victoria Muliadi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Skålén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chika Yokota
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Gokul Raj Kathamuthu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Christoph Lange
- Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Katrin Scheu
- Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Karoline I Gaede
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Borstel, Germany
- BioMaterialBank North, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Torsten Goldmann
- Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Borstel, Germany
| | - Ankur Pandita
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kiran Iqbal Masood
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - Shahid Pervez
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - Johan Grunewald
- Respiratory Medicine Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Zahra Hasan
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - Max Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Pavlova EN, Lepekha LN, Rybalkina EY, Tarasov RV, Sychevskaya KA, Voronezhskaya EE, Masyutin AG, Ergeshov AE, Erokhina MV. High and Low Levels of ABCB1 Expression Are Associated with Two Distinct Gene Signatures in Lung Tissue of Pulmonary TB Patients with High Inflammation Activity. Int J Mol Sci 2023; 24:14839. [PMID: 37834286 PMCID: PMC10573207 DOI: 10.3390/ijms241914839] [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: 08/24/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
P-glycoprotein (encoded by the ABCB1 gene) has a dual role in regulating inflammation and reducing chemotherapy efficacy in various diseases, but there are few studies focused on pulmonary TB patients. In this study, our objective was to identify a list of genes that correlate with high and low levels of ABCB1 gene expression in the lungs of pulmonary TB patients with different activity of chronic granulomatous inflammation. We compared gene expression in two groups of samples (with moderate and high activity of tuberculomas) to identify their characteristic gene signatures. Gene expression levels were determined using quantitative PCR in samples of perifocal area of granulomas, which were obtained from 65 patients after surgical intervention. Subsequently, two distinct gene signatures associated with high inflammation activity were identified. The first signature demonstrated increased expression of HIF1a, TGM2, IL6, SOCS3, and STAT3, which correlated with high ABCB1 expression. The second signature was characterized by high expression of TNFa and CD163 and low expression of ABCB1. These results provide insight into various inflammatory mechanisms and association with P-gp gene expression in lung tissue of pulmonary TB patients and will be useful in the development of a host-directed therapy approach to improving the effectiveness of anti-TB treatment.
Collapse
Affiliation(s)
- Ekaterina N. Pavlova
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Larisa N. Lepekha
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
| | - Ekaterina Yu. Rybalkina
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
- FSBI N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Ruslan V. Tarasov
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
| | - Ksenia A. Sychevskaya
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
- FSBI N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Elena E. Voronezhskaya
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alexander G. Masyutin
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Atadzhan E. Ergeshov
- Director of the Institute, Central Tuberculosis Research Institute, 2 Yauzskaya Alleya, 107564 Moscow, Russia;
| | - Maria V. Erokhina
- Central Tuberculosis Research Institute, 107564 Moscow, Russia; (E.N.P.); (A.G.M.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
16
|
Shi H, Yang L, Zhang F, Zhou Y, Zhou Y. Diagnostic Value of CD25, CD69, and CD134 on Tuberculosis-Specific Antigen-Stimulated CD4+ T Cells for Tuberculous Pleurisy. J Immunol Res 2023; 2023:5309816. [PMID: 37809012 PMCID: PMC10551431 DOI: 10.1155/2023/5309816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 10/10/2023] Open
Abstract
Rapid and accurate methods for the diagnosis of tuberculous pleurisy (TP) are urgently needed. Activation markers of tuberculosis (TB)-reactive T cells are considered promising for the diagnosis of active TB (ATB). Different activation indexes may play different roles in the progression of TB, but there are few reports on T cell activation indicators, except for HLA-DR. Hence, we evaluated the expression of early (CD25 and CD69) and late (CD134) activation markers on TB antigen-stimulated CD4+ T cells in populations with different TB infection status and investigated their diagnostic value for ATB, particularly, for TP. Moreover, we compared the differences in the diagnostic efficacy among the indexes from peripheral blood (PB) and pleural fluid (PF) for TP. The expression of each activation marker was significantly increased in TB-infected populations (patients with ATB and latent TB infection vs. healthy individuals; patients with TP vs. non-TP) and was significantly higher in the PF than in the PB of patients with TP. The diagnostic performance of the coexpressed activation markers was superior to that of single expression markers in the differential diagnosis of ATB and non-TB, with CD25+CD134+ showing the best diagnostic efficiency (AUC: 0.93, 95% CI, 0.87-0.99; sensitivity: 86.7%, 95% CI, 72.5%-94.5%; and specificity: 94.0%, 95% CI, 82.5%-98.4%). Except for TB-IGRA, the activation indexes were more accurate than conventional laboratory methods for ATB diagnosis. In addition, the expression of CD25+CD134+ in PB and PF was the best values for differential diagnosis of TP and NTP, with AUCs of 0.87 (95% CI, 0.77-0.96) and 0.95 (95% CI, 0.90-1.00), respectively. Our study provides information on the diagnostic value of different activation markers for TB and shows that the expression of CD25+CD134+ on CD4+ T cells in PF can serve as a potential marker for TP diagnosis.
Collapse
Affiliation(s)
- Hanlu Shi
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liping Yang
- The Quzhou Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Fujie Zhang
- Qian Xi Nan Hospital of Traditional Chinese Medicine, Qian Xi Nan Buyei and Miao Autonomous Prefecture, Guizhou 562499, China
| | - Yu Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
- Key Laboratory of Biomarkers and in vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang 310063, China
| | - Yonglie Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
| |
Collapse
|
17
|
Sawyer AJ, Patrick E, Edwards J, Wilmott JS, Fielder T, Yang Q, Barber DL, Ernst JD, Britton WJ, Palendira U, Chen X, Feng CG. Spatial mapping reveals granuloma diversity and histopathological superstructure in human tuberculosis. J Exp Med 2023; 220:e20221392. [PMID: 36920308 PMCID: PMC10035589 DOI: 10.1084/jem.20221392] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/07/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
The hallmark of tuberculosis (TB) is the formation of immune cell-enriched aggregates called granulomas. While granulomas are pathologically diverse, their tissue-wide heterogeneity has not been spatially resolved at the single-cell level in human tissues. By spatially mapping individual immune cells in every lesion across entire tissue sections, we report that in addition to necrotizing granulomas, the human TB lung contains abundant non-necrotizing leukocyte aggregates surrounding areas of necrotizing tissue. These cellular lesions were more diverse in composition than necrotizing lesions and could be stratified into four general classes based on cellular composition and spatial distribution of B cells and macrophages. The cellular composition of non-necrotizing structures also correlates with their proximity to necrotizing lesions, indicating these are foci of distinct immune reactions adjacent to necrotizing granulomas. Together, we show that during TB, diseased lung tissue develops a histopathological superstructure comprising at least four different types of non-necrotizing cellular aggregates organized as satellites of necrotizing granulomas.
Collapse
Affiliation(s)
- Andrew J. Sawyer
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Ellis Patrick
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, Australia
- Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, Australia
| | - Jarem Edwards
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - James S. Wilmott
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Timothy Fielder
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Qianting Yang
- Guangdong Key Lab for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen, Third People’s Hospital, Shenzhen, Shenzhen, China
| | - Daniel L. Barber
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joel D. Ernst
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Warwick J. Britton
- Centenary Institute, The University of Sydney, Sydney, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Umaimainthan Palendira
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen UniversitySchool of Medicine, Shenzhen, China
| | - Carl G. Feng
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Ochoa MC, Sanchez-Gregorio S, de Andrea CE, Garasa S, Alvarez M, Olivera I, Glez-Vaz J, Luri-Rey C, Etxeberria I, Cirella A, Azpilikueta A, Berraondo P, Argemi J, Sangro B, Teijeira A, Melero I. Synergistic effects of combined immunotherapy strategies in a model of multifocal hepatocellular carcinoma. Cell Rep Med 2023; 4:101009. [PMID: 37040772 PMCID: PMC10140615 DOI: 10.1016/j.xcrm.2023.101009] [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: 09/16/2022] [Revised: 10/20/2022] [Accepted: 03/20/2023] [Indexed: 04/13/2023]
Abstract
Immune checkpoint-inhibitor combinations are the best therapeutic option for advanced hepatocellular carcinoma (HCC) patients, but improvements in efficacy are needed to improve response rates. We develop a multifocal HCC model to test immunotherapies by introducing c-myc using hydrodynamic gene transfer along with CRISPR-Cas9-mediated disruption of p53 in mouse hepatocytes. Additionally, induced co-expression of luciferase, EGFP, and the melanosomal antigen gp100 facilitates studies on the underlying immunological mechanisms. We show that treatment of the mice with a combination of anti-CTLA-4 + anti-PD1 mAbs results in partial clearance of the tumor with an improvement in survival. However, the addition of either recombinant IL-2 or an anti-CD137 mAb markedly improves both outcomes in these mice. Combining tumor-specific adoptive T cell therapy to the aCTLA-4/aPD1/rIL2 or aCTLA-4/aPD1/aCD137 regimens enhances efficacy in a synergistic manner. As shown by multiplex tissue immunofluorescence and intravital microscopy, combined immunotherapy treatments enhance T cell infiltration and the intratumoral performance of T lymphocytes.
Collapse
Affiliation(s)
- Maria Carmen Ochoa
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sandra Sanchez-Gregorio
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain; Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Saray Garasa
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Maite Alvarez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Iñaki Etxeberria
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Josepmaria Argemi
- Liver Unit and HPB Oncology Area, Clínica Universidad de Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBEREHD), Madrid, Spain
| | - Bruno Sangro
- Liver Unit and HPB Oncology Area, Clínica Universidad de Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBEREHD), Madrid, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Staal AHJ, Cortenbach KRG, Gorris MAJ, van der Woude LL, Srinivas M, Heijmen RH, Geuzebroek GSC, Grewal N, Hebeda KM, de Vries IJM, DeRuiter MC, van Kimmenade RRJ. Adventitial adaptive immune cells are associated with ascending aortic dilatation in patients with a bicuspid aortic valve. Front Cardiovasc Med 2023; 10:1127685. [PMID: 37057097 PMCID: PMC10086356 DOI: 10.3389/fcvm.2023.1127685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/14/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundBicuspid aortic valve (BAV) is associated with ascending aorta aneurysms and dissections. Presently, genetic factors and pathological flow patterns are considered responsible for aneurysm formation in BAV while the exact role of inflammatory processes remains unknown.MethodsIn order to objectify inflammation, we employ a highly sensitive, quantitative immunohistochemistry approach. Whole slides of dissected, dilated and non-dilated ascending aortas from BAV patients were quantitatively analyzed.ResultsDilated aortas show a 4-fold increase of lymphocytes and a 25-fold increase in B lymphocytes in the adventitia compared to non-dilated aortas. Tertiary lymphoid structures with B cell follicles and helper T cell expansion were identified in dilated and dissected aortas. Dilated aortas were associated with an increase in M1-like macrophages in the aorta media, in contrast the number of M2-like macrophages did not change significantly.ConclusionThis study finds unexpected large numbers of immune cells in dilating aortas of BAV patients. These findings raise the question whether immune cells in BAV aortopathy are innocent bystanders or contribute to the deterioration of the aortic wall.
Collapse
Affiliation(s)
- Alexander H. J. Staal
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kimberley R. G. Cortenbach
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lieke L. van der Woude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
| | - Robin H. Heijmen
- Department of Cardiothoracic Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Nimrat Grewal
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marco C. DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Roland R. J. van Kimmenade
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Roland R. J. van Kimmenade,
| |
Collapse
|
20
|
Jiménez-Sánchez D, López-Janeiro Á, Villalba-Esparza M, Ariz M, Kadioglu E, Masetto I, Goubert V, Lozano MD, Melero I, Hardisson D, Ortiz-de-Solórzano C, de Andrea CE. Weakly supervised deep learning to predict recurrence in low-grade endometrial cancer from multiplexed immunofluorescence images. NPJ Digit Med 2023; 6:48. [PMID: 36959234 PMCID: PMC10036616 DOI: 10.1038/s41746-023-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/10/2023] [Indexed: 03/25/2023] Open
Abstract
Predicting recurrence in low-grade, early-stage endometrial cancer (EC) is both challenging and clinically relevant. We present a weakly-supervised deep learning framework, NaroNet, that can learn, without manual expert annotation, the complex tumor-immune interrelations at three levels: local phenotypes, cellular neighborhoods, and tissue areas. It uses multiplexed immunofluorescence for the simultaneous visualization and quantification of CD68 + macrophages, CD8 + T cells, FOXP3 + regulatory T cells, PD-L1/PD-1 protein expression, and tumor cells. We used 489 tumor cores from 250 patients to train a multilevel deep-learning model to predict tumor recurrence. Using a tenfold cross-validation strategy, our model achieved an area under the curve of 0.90 with a 95% confidence interval of 0.83-0.95. Our model predictions resulted in concordance for 96,8% of cases (κ = 0.88). This method could accurately assess the risk of recurrence in EC, outperforming current prognostic factors, including molecular subtyping.
Collapse
Affiliation(s)
- Daniel Jiménez-Sánchez
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Álvaro López-Janeiro
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - María Villalba-Esparza
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Mikel Ariz
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Ece Kadioglu
- Lunaphore Technologies SA, Tolochenaz, Switzerland
| | | | | | - Maria D Lozano
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Ortiz-de-Solórzano
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
| | - Carlos E de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain.
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain.
| |
Collapse
|
21
|
Cirella A, Bolaños E, Di Trani CA, de Andrea CE, Sánchez-Gregorio S, Etxeberria I, Gonzalez-Gomariz J, Olivera I, Brocco D, Glez-Vaz J, Luri-Rey C, Azpilikueta A, Rodríguez I, Fernandez-Sendín M, Egea J, Eguren I, Sanmamed MF, Palencia B, Teijeira A, Berraondo P, Melero I. Intratumoral Gene Transfer of mRNAs Encoding IL12 in Combination with Decoy-Resistant IL18 Improves Local and Systemic Antitumor Immunity. Cancer Immunol Res 2023; 11:184-198. [PMID: 36478221 DOI: 10.1158/2326-6066.cir-22-0373] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/18/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
IL12-based local gene therapy of cancer constitutes an active area of clinical research using plasmids, mRNAs, and viral vectors. To improve antitumor effects, we have experimentally tested the combination of mRNA constructs encoding IL12 and IL18. Moreover, we have used a form of IL18 [decoy-resistant IL18 (DR-18)] which has preserved bioactivity but does not bind to the IL18 binding protein decoy receptor. Both cytokines dramatically synergize to induce IFNγ release from mouse splenocytes, and, if systemically cotransferred to the liver, they mediate lethal toxicity. However, if given intratumorally to B16OVA tumor-bearing mice, the combination attains efficacy against the directly treated tumor and moderate tumor-delaying activity on distant noninjected lesions. Cotreatment was conducive to the presence of more activated CD8+ T cells in the treated and noninjected tumors. In keeping with these findings, the efficacy of treatment was contingent on the integrity of CD8+ T cells and cDC1 dendritic cells in the treated mice. Furthermore, efficacy of IL12 plus DR-18 local mRNA coinjection against distant concomitant tumors could be enhanced upon combination with anti-PD-1 mAb systemic treatment, thus defining a feasible synergistic immunotherapy strategy.
Collapse
Affiliation(s)
- Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos E de Andrea
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Pathology, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sandra Sánchez-Gregorio
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Iñaki Etxeberria
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Jose Gonzalez-Gomariz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Davide Brocco
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Inmaculada Rodríguez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendín
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Josune Egea
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Iñaki Eguren
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Oncology, Clinica Universidad de Navarra, Madrid, Spain
| | - Belen Palencia
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Oncology, Clinica Universidad de Navarra, Madrid, Spain.,Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
22
|
Marafioti T, Lozano MD, de Andrea CE. Characterization of the immune infiltrate in mouse tissue by multiplex immunofluorescence. Methods Cell Biol 2023; 174:43-53. [PMID: 36710050 DOI: 10.1016/bs.mcb.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Multiplexed immunofluorescence imaging of formalin-fixed, paraffin-embedded (FFPE) specimens mounted on glass slides allow the identification of multiple cell phenotypes while retaining spatial and morphological context. Multiplex immunofluorescence protocols have already been developed and validated for mouse tissues. Immunophenotyping analysis reliably depicts the immune landscape of cancer tissues that has been demonstrated to influence cancer development and progression as well as to have an impact on therapy responsiveness and resistance. Here, we describe a method for multiplexed fluorescence image analysis, enabling analysis of mouse cancer morphology and cell phenotypes in FFPE sections.
Collapse
Affiliation(s)
- Teresa Marafioti
- Department of Histopathology, University College London, London, United Kingdom; Department of Cellular Pathology, University College Hospital, London, United Kingdom
| | - Maria D Lozano
- Department of Pathology, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain; Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Department of Pathology, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain; Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain.
| |
Collapse
|
23
|
Schmitt F, Lozano MD. Molecular/biomarker testing in lung cytology: A practical approach. Diagn Cytopathol 2023; 51:59-67. [PMID: 36098379 DOI: 10.1002/dc.25054] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/13/2022]
Abstract
The increasing comprehension of molecular mechanisms underlying lung cancer and the discovery of targetable genomic alterations has dramatically change the pathological approach to lung cancer, especially non-small cell lung cancer (NSCLC). This unstoppable knowledge has taken pathologists to the leading front on lung cancer management. This is especially relevant in the world of cytopathology where "doing more with less" is a daily challenge. Nowadays with a growing number of predictive biomarkers needed to manage patients with NSCLC, there has been a paradigm shift in care and handling of diagnostic samples. One of the main emphasis and interest relies on the utilization of cytologic samples and small biopsies for not only diagnostic purposes but also for ancillary testing. Moreover, lung cytopathology is in continuous evolutions with implementation of new diagnostic techniques, new tools, and facing new challenges. The goal of this paper will be to provide the reader with the necessary concepts than can be used to exploit the cytological samples in order to use these samples for comprehensive diagnosis and relevant ancillary testing purposes.
Collapse
Affiliation(s)
- Fernando Schmitt
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Cintesis@RISE, Health Research Network, Porto, Portugal.,IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Maria D Lozano
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
24
|
López‐Janeiro Á, Villalba‐Esparza M, Brizzi ME, Jiménez‐Sánchez D, Ruz‐Caracuel I, Kadioglu E, Masetto I, Goubert V, Garcia‐Ros D, Melero I, Peláez‐García A, Hardisson D, de Andrea CE. The association between the tumor immune microenvironments and clinical outcome in low-grade, early-stage endometrial cancer patients. J Pathol 2022; 258:426-436. [PMID: 36169332 PMCID: PMC9828119 DOI: 10.1002/path.6012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/04/2022] [Accepted: 09/21/2022] [Indexed: 01/19/2023]
Abstract
Endometrial tumors show substantial heterogeneity in their immune microenvironment. This heterogeneity could be used to improve the accuracy of current outcome prediction tools. We assessed the immune microenvironment of 235 patients diagnosed with low-grade, early-stage endometrial cancer. Multiplex quantitative immunofluorescence was carried out to measure CD8, CD68, FOXP3, PD-1, and PD-L1 markers, as well as cytokeratin (CK), on tissue microarrays. Clustering results revealed five robust immune response patterns, each associated with specific immune populations, cell phenotypes, and cell spatial clustering. Most samples (69%) belonged to the immune-desert subtype, characterized by low immune cell densities. Tumor-infiltrating lymphocyte (TIL)-rich samples (4%) displayed high CD8+ T-cell infiltration, as well as a high percentage of CD8/PD-1+ cells. Immune-exclusion samples (19%) displayed the lowest CD8+ infiltration combined with high PD-L1 expression levels in CK+ tumor cells. In addition, they demonstrated high tumor cell spatial clustering as well as increased spatial proximity of CD8+ /PD-1+ and CK/PD-L1+ cells. FOXP3 and macrophage-rich phenotypes (3% and 4% of total samples) displayed relatively high levels of FOXP3+ regulatory T-cells and CD68+ macrophages, respectively. These phenotypes correlated with clinical outcomes, with immune-exclusion tumors showing an association with tumor relapse. When compared with prediction models built using routine pathological variables, models optimized with immune variables showed increased outcome prediction capacity (AUC = 0.89 versus 0.78) and stratification potential. The improved prediction capacity was independent of mismatch repair protein status and adjuvant radiotherapy treatment. Further, immunofluorescence results could be partially recapitulated using single-marker immunohistochemistry (IHC) performed on whole tissue sections. TIL-rich tumors demonstrated increased CD8+ T-cells by IHC, while immune-exclusion tumors displayed a lack of CD8+ T-cells and frequent expression of PD-L1 in tumor cells. Our results demonstrate the capability of the immune microenvironment to improve standard prediction tools in low-grade, early-stage endometrial carcinomas. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Álvaro López‐Janeiro
- Department of PathologyHospital Universitario La Paz, IdiPAZMadridSpain,Present address:
Department of Pathology, Clínica Universidad de NavarraUniversity of NavarraPamplonaSpain
| | - María Villalba‐Esparza
- Department of Pathology, Clínica Universidad de NavarraUniversity of NavarraPamplonaSpain,Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| | | | - Daniel Jiménez‐Sánchez
- Department of Pathology, Clínica Universidad de NavarraUniversity of NavarraPamplonaSpain
| | | | | | | | | | - David Garcia‐Ros
- Department of Pathology, Clínica Universidad de NavarraUniversity of NavarraPamplonaSpain
| | - Ignacio Melero
- Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC)Instituto de Salud Carlos IIIMadridSpain,Department of Immunology and ImmunotherapyClínica Universidad de NavarraPamplonaSpain,Program of Immunology and ImmunotherapyCIMA Universidad de NavarraPamplonaSpain
| | - Alberto Peláez‐García
- Molecular Pathology and Therapeutic Targets GroupLa Paz University Hospital (IdiPAZ)MadridSpain
| | - David Hardisson
- Department of PathologyHospital Universitario La Paz, IdiPAZMadridSpain,Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC)Instituto de Salud Carlos IIIMadridSpain,Molecular Pathology and Therapeutic Targets GroupLa Paz University Hospital (IdiPAZ)MadridSpain,Faculty of MedicineUniversidad Autónoma de MadridMadridSpain
| | - Carlos E de Andrea
- Department of Pathology, Clínica Universidad de NavarraUniversity of NavarraPamplonaSpain,Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
25
|
Sonzini G, Granados-Aparici S, Sanegre S, Diaz-Lagares A, Diaz-Martin J, de Andrea C, Eritja N, Bao-Caamano A, Costa-Fraga N, García-Ros D, Salguero-Aranda C, Davidson B, López-López R, Melero I, Navarro S, Ramon y Cajal S, de Alava E, Matias-Guiu X, Noguera R. Integrating digital pathology with transcriptomic and epigenomic tools for predicting metastatic uterine tumor aggressiveness. Front Cell Dev Biol 2022; 10:1052098. [PMID: 36467415 PMCID: PMC9716026 DOI: 10.3389/fcell.2022.1052098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/04/2022] [Indexed: 10/13/2024] Open
Abstract
The incidence of new cancer cases is expected to increase significantly in the future, posing a worldwide problem. In this regard, precision oncology and its diagnostic tools are essential for developing personalized cancer treatments. Digital pathology (DP) is a particularly key strategy to study the interactions of tumor cells and the tumor microenvironment (TME), which play a crucial role in tumor initiation, progression and metastasis. The purpose of this study was to integrate data on the digital patterns of reticulin fiber scaffolding and the immune cell infiltrate, transcriptomic and epigenetic profiles in aggressive uterine adenocarcinoma (uADC), uterine leiomyosarcoma (uLMS) and their respective lung metastases, with the aim of obtaining key TME biomarkers that can help improve metastatic prediction and shed light on potential therapeutic targets. Automatized algorithms were used to analyze reticulin fiber architecture and immune infiltration in colocalized regions of interest (ROIs) of 133 invasive tumor front (ITF), 89 tumor niches and 70 target tissues in a total of six paired samples of uADC and nine of uLMS. Microdissected tissue from the ITF was employed for transcriptomic and epigenetic studies in primary and metastatic tumors. Reticulin fiber scaffolding was characterized by a large and loose reticular fiber network in uADC, while dense bundles were found in uLMS. Notably, more similarities between reticulin fibers were observed in paired uLMS then paired uADCs. Transcriptomic and multiplex immunofluorescence-based immune profiling showed a higher abundance of T and B cells in primary tumor and in metastatic uADC than uLMS. Moreover, the epigenetic signature of paired samples in uADCs showed more differences than paired samples in uLMS. Some epigenetic variation was also found between the ITF of metastatic uADC and uLMS. Altogether, our data suggest a correlation between morphological and molecular changes at the ITF and the degree of aggressiveness. The use of DP tools for characterizing reticulin scaffolding and immune cell infiltration at the ITF in paired samples together with information provided by omics analyses in a large cohort will hopefully help validate novel biomarkers of tumor aggressiveness, develop new drugs and improve patient quality of life in a much more efficient way.
Collapse
Affiliation(s)
- Giorgia Sonzini
- Department of Pathology, Medical School, University of Valencia-INCLIVA, Valencia, Spain
| | - Sofia Granados-Aparici
- Department of Pathology, Medical School, University of Valencia-INCLIVA, Valencia, Spain
| | - Sabina Sanegre
- Department of Pathology, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Cancer CIBER (CIBERONC), Madrid, Spain
| | - Angel Diaz-Lagares
- Cancer CIBER (CIBERONC), Madrid, Spain
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - Juan Diaz-Martin
- Cancer CIBER (CIBERONC), Madrid, Spain
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla, Seville, Spain
| | - Carlos de Andrea
- Cancer CIBER (CIBERONC), Madrid, Spain
- Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Núria Eritja
- Cancer CIBER (CIBERONC), Madrid, Spain
- Institut de Recerca Biomèdica de LLeida (IRBLLEIDA), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Department of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, University of Lleida - University of Barcelona, Barcelona, Spain
| | - Aida Bao-Caamano
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
- Universidad de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Nicolás Costa-Fraga
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
- Universidad de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - David García-Ros
- Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Carmen Salguero-Aranda
- Cancer CIBER (CIBERONC), Madrid, Spain
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla, Seville, Spain
| | - Ben Davidson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Rafael López-López
- Cancer CIBER (CIBERONC), Madrid, Spain
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Ignacio Melero
- Cancer CIBER (CIBERONC), Madrid, Spain
- Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Samuel Navarro
- Department of Pathology, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Cancer CIBER (CIBERONC), Madrid, Spain
| | - Santiago Ramon y Cajal
- Cancer CIBER (CIBERONC), Madrid, Spain
- Department of Pathology, Vall d'Hebron University Hospital, Autonoma University of Barcelona, Barcelona, Spain
| | - Enrique de Alava
- Cancer CIBER (CIBERONC), Madrid, Spain
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla, Seville, Spain
| | - Xavier Matias-Guiu
- Cancer CIBER (CIBERONC), Madrid, Spain
- Institut de Recerca Biomèdica de LLeida (IRBLLEIDA), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Department of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, University of Lleida - University of Barcelona, Barcelona, Spain
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Cancer CIBER (CIBERONC), Madrid, Spain
| |
Collapse
|
26
|
Melero I, Villalba-Esparza M, Recalde-Zamacona B, Jiménez-Sánchez D, Teijeira Á, Argueta A, García-Tobar L, Álvarez-Gigli L, Sainz C, Garcia-Ros D, Toledo E, Abengozar-Muela M, Fernández-Alonso M, Rodríguez-Mateos M, Reina G, Carmona-Torre F, Quiroga JA, Del Pozo JL, Cross A, López-Janeiro Á, Hardisson D, Echeveste JI, Lozano MD, Ho LP, Klenerman P, Issa F, Landecho MF, de Andrea CE. Neutrophil Extracellular Traps, Local IL-8 Expression, and Cytotoxic T-Lymphocyte Response in the Lungs of Patients With Fatal COVID-19. Chest 2022; 162:1006-1016. [PMID: 35714708 PMCID: PMC9197577 DOI: 10.1016/j.chest.2022.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Excessive inflammation is pathogenic in the pneumonitis associated with severe COVID-19. Neutrophils are among the most abundantly present leukocytes in the inflammatory infiltrates and may form neutrophil extracellular traps (NETs) under the local influence of cytokines. NETs constitute a defense mechanism against bacteria, but have also been shown to mediate tissue damage in a number of diseases. RESEARCH QUESTION Could NETs and their tissue-damaging properties inherent to neutrophil-associated functions play a role in the respiratory failure seen in patients with severe COVID-19, and how does this relate to the SARS-CoV-2 viral loads, IL-8 (CXCL8) chemokine expression, and cytotoxic T-lymphocyte infiltrates? STUDY DESIGN AND METHODS Sixteen lung biopsy samples obtained immediately after death were analyzed methodically as exploratory and validation cohorts. NETs were analyzed quantitatively by multiplexed immunofluorescence and were correlated with local levels of IL-8 messenger RNA (mRNA) and the density of CD8+ T-cell infiltration. SARS-CoV-2 presence in tissue was quantified by reverse-transcriptase polymerase chain reaction and immunohistochemistry analysis. RESULTS NETs were found in the lung interstitium and surrounding the bronchiolar epithelium with interindividual and spatial heterogeneity. NET density did not correlate with SARS-CoV-2 tissue viral load. NETs were associated with local IL-8 mRNA levels. NETs were also detected in pulmonary thrombi and in only one of eight liver tissues. NET focal presence correlated negatively with CD8+ T-cell infiltration in the lungs. INTERPRETATION Abundant neutrophils undergoing NETosis are found in the lungs of patients with fatal COVID-19, but no correlation was found with viral loads. The strong association between NETs and IL-8 points to this chemokine as a potentially causative factor. The function of cytotoxic T-lymphocytes in the immune responses against SARS-CoV-2 may be interfered with by the presence of NETs.
Collapse
Affiliation(s)
- Ignacio Melero
- Division of Immunology and Immunotherapy, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - María Villalba-Esparza
- Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | - Álvaro Teijeira
- Division of Immunology and Immunotherapy, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Alan Argueta
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Laura García-Tobar
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Cristina Sainz
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - David Garcia-Ros
- Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Estefanía Toledo
- Department of Preventive Medicine and Public Health, IdiSNA, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición, Institute of Health Carlos III, Madrid, Spain
| | | | - Mirian Fernández-Alonso
- Navarra Institute for Health Research, Pamplona, Spain; Department of Microbiology and Infectious Diseases, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Rodríguez-Mateos
- Department of Microbiology and Infectious Diseases, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Reina
- Navarra Institute for Health Research, Pamplona, Spain; Department of Microbiology and Infectious Diseases, Clínica Universidad de Navarra, Pamplona, Spain
| | - Francisco Carmona-Torre
- Navarra Institute for Health Research, Pamplona, Spain; Department of Microbiology and Infectious Diseases, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Jose L Del Pozo
- Navarra Institute for Health Research, Pamplona, Spain; Department of Microbiology and Infectious Diseases, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amy Cross
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, England
| | | | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - José I Echeveste
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain; Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Maria D Lozano
- Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain; Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Ling-Pei Ho
- MRC Human Immunology Unit, University of Oxford, Oxford, England
| | - Paul Klenerman
- Nuffield Department of Clinical Medicine, Peter Medawar Building for Pathogen Research, Oxford, England
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, England
| | - Manuel F Landecho
- Department of Internal Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain; Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain.
| |
Collapse
|
27
|
High IGKC-Expressing Intratumoral Plasma Cells Predict Response to Immune Checkpoint Blockade. Int J Mol Sci 2022; 23:ijms23169124. [PMID: 36012390 PMCID: PMC9408876 DOI: 10.3390/ijms23169124] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/06/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
Resistance to Immune Checkpoint Blockade (ICB) constitutes the current limiting factor for the optimal implementation of this novel therapy, which otherwise demonstrates durable responses with acceptable toxicity scores. This limitation is exacerbated by a lack of robust biomarkers. In this study, we have dissected the basal TME composition at the gene expression and cellular levels that predict response to Nivolumab and prognosis. BCR, TCR and HLA profiling were employed for further characterization of the molecular variables associated with response. The findings were validated using a single-cell RNA-seq data of metastatic melanoma patients treated with ICB, and by multispectral immunofluorescence. Finally, machine learning was employed to construct a prediction algorithm that was validated across eight metastatic melanoma cohorts treated with ICB. Using this strategy, we have unmasked a major role played by basal intratumoral Plasma cells expressing high levels of IGKC in efficacy. IGKC, differentially expressed in good responders, was also identified within the Top response-related BCR clonotypes, together with IGK variants. These results were validated at gene, cellular and protein levels; CD138+ Plasma-like and Plasma cells were more abundant in good responders and correlated with the same RNA-seq-defined fraction. Finally, we generated a 15-gene prediction model that outperformed the current reference score in eight ICB-treated metastatic melanoma cohorts. The evidenced major contribution of basal intratumoral IGKC and Plasma cells in good response and outcome in ICB in metastatic melanoma is a groundbreaking finding in the field beyond the role of T lymphocytes.
Collapse
|
28
|
Laspidea V, Puigdelloses M, Labiano S, Marrodán L, Garcia-Moure M, Zalacain M, Gonzalez-Huarriz M, Martínez-Vélez N, Ausejo-Mauleon I, de la Nava D, Herrador-Cañete G, Marco-Sanz J, Guruceaga E, de Andrea CE, Villalba M, Becher O, Squatrito M, Matía V, Gállego Pérez-Larraya J, Patiño-García A, Gupta S, Gomez-Manzano C, Fueyo J, Alonso MM. Exploiting 4-1BB immune checkpoint to enhance the efficacy of oncolytic virotherapy for diffuse intrinsic pontine gliomas. JCI Insight 2022; 7:154812. [PMID: 35393952 PMCID: PMC9057625 DOI: 10.1172/jci.insight.154812] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/25/2022] [Indexed: 12/28/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors, and patient survival has not changed despite many therapeutic efforts, emphasizing the urgent need for effective treatments. Here, we evaluated the anti-DIPG effect of the oncolytic adenovirus Delta-24-ACT, which was engineered to express the costimulatory ligand 4-1BBL to potentiate the antitumor immune response of the virus. Delta-24-ACT induced the expression of functional 4-1BBL on the membranes of infected DIPG cells, which enhanced the costimulation of CD8+ T lymphocytes. In vivo, Delta-24-ACT treatment of murine DIPG orthotopic tumors significantly improved the survival of treated mice, leading to long-term survivors that developed immunological memory against these tumors. In addition, Delta-24-ACT was safe and caused no local or systemic toxicity. Mechanistic studies showed that Delta-24-ACT modulated the tumor-immune content, not only increasing the number, but also improving the functionality of immune cells. All of these data highlight the safety and potential therapeutic benefit of Delta-24-ACT the treatment of patients with DIPG.
Collapse
Affiliation(s)
- Virginia Laspidea
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Montserrat Puigdelloses
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Sara Labiano
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Lucía Marrodán
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Marc Garcia-Moure
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Marta Zalacain
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Naiara Martínez-Vélez
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Iker Ausejo-Mauleon
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Daniel de la Nava
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Guillermo Herrador-Cañete
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Gene Therapy and Regulation of Gene Expression Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
| | - Javier Marco-Sanz
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Elisabeth Guruceaga
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Bioinformatics Platform, El Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Department of Pathology, Navarra University Clinic, Pamplona, Spain
| | - María Villalba
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Department of Pathology, Navarra University Clinic, Pamplona, Spain
| | - Oren Becher
- Department of Pediatrics.,Department of Biochemistry and Molecular Genetics, and.,Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA.,Division of Hematology Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Massimo Squatrito
- Seve Ballesteros Foundation Brain Tumor Group, Molecular Oncology Programme, Spanish National Cancer Research Center, Madrid, Spain
| | - Verónica Matía
- Seve Ballesteros Foundation Brain Tumor Group, Molecular Oncology Programme, Spanish National Cancer Research Center, Madrid, Spain
| | - Jaime Gállego Pérez-Larraya
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Neurology, Navarra University Clinic, Pamplona, Spain
| | - Ana Patiño-García
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Sumit Gupta
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marta M Alonso
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| |
Collapse
|
29
|
Baykan AH, Sayiner HS, Aydin E, Koc M, Inan I, Erturk SM. Extrapulmonary tuberculosıs: an old but resurgent problem. Insights Imaging 2022; 13:39. [PMID: 35254534 PMCID: PMC8901940 DOI: 10.1186/s13244-022-01172-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) primarily affects the lungs, but some of its most devastating clinical consequences arise because of its ability to spread from the lungs to other organs. Extrapulmonary TB (EPTB) constitutes 15-20% of all TB cases. Imaging findings are not always specific and can mimic many diseases; therefore, EPTB should be considered in the differential diagnosis, particularly in patients with immune system disorders (AIDS, patients receiving chemotherapy, etc.) and those in other high-risk groups including people with diabetes. The bacterium's passage to the regional lymph nodes is essential for developing a protective T-cell-mediated immune response, but the bacterium can spread hematologically and via the lymphatic system, leading to extrapulmonary involvement. Diagnosis of EPTB in high-risk patients is made based on suspected clinical and radiological findings, but further positive culture and histopathological confirmation may be required in some instances. Radiological evaluations are critical for diagnosis and crucial in planning the treatment and follow-up. This paper aims to review the typical and atypical imaging features and the differential diagnosis of EPTB.
Collapse
Affiliation(s)
- Ali H Baykan
- Department of Radiology, Faculty of Medicine, Adiyaman University, Yunus Emre Mahallesi 1164 Sokak No:13, 02200, Merkez, Adıyaman, Turkey.
| | - Hakan S Sayiner
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey
| | - Elcin Aydin
- Department of Radiology, Izmir Tepecik Training and Research Hospital, Izmir, Turkey
| | - Mustafa Koc
- Department of Radiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Ibrahim Inan
- Department of Radiology,, King's College Hospital London, Dubai, United Arab Emirates
| | - Sukru M Erturk
- Department of Radiology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
30
|
Ge G, Jiang H, Xiong J, Zhang W, Shi Y, Tao C, Wang H. Progress of the Art of Macrophage Polarization and Different Subtypes in Mycobacterial Infection. Front Immunol 2021; 12:752657. [PMID: 34899703 PMCID: PMC8660122 DOI: 10.3389/fimmu.2021.752657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Mycobacteriosis, mostly resulting from Mycobacterium tuberculosis (MTb), nontuberculous mycobacteria (NTM), and Mycobacterium leprae (M. leprae), is the long-standing granulomatous disease that ravages several organs including skin, lung, and peripheral nerves, and it has a spectrum of clinical-pathologic features based on the interaction of bacilli and host immune response. Histiocytes in infectious granulomas mainly consist of infected and uninfected macrophages (Mφs), multinucleated giant cells (MGCs), epithelioid cells (ECs), and foam cells (FCs), which are commonly discovered in lesions in patients with mycobacteriosis. Granuloma Mφ polarization or reprogramming is the crucial appearance of the host immune response to pathogen aggression, which gets a command of endocellular microbe persistence. Herein, we recapitulate the current gaps and challenges during Mφ polarization and the different subpopulations of mycobacteriosis.
Collapse
Affiliation(s)
- Gai Ge
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Haiqin Jiang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jingshu Xiong
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wenyue Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ying Shi
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Chenyue Tao
- Imperial College London, London, United Kingdom
| | - Hongsheng Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,National Center for Sexually Transmitted Disease and Leprosy Control, China Centers for Disease Control and Prevention, Nanjing, China.,Centre for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Alvarez M, Molina C, De Andrea CE, Fernandez-Sendin M, Villalba M, Gonzalez-Gomariz J, Ochoa MC, Teijeira A, Glez-Vaz J, Aranda F, Sanmamed MF, Rodriguez-Ruiz ME, Fan X, Shen WH, Berraondo P, Quintero M, Melero I. Intratumoral co-injection of the poly I:C-derivative BO-112 and a STING agonist synergize to achieve local and distant anti-tumor efficacy. J Immunother Cancer 2021; 9:jitc-2021-002953. [PMID: 34824158 PMCID: PMC8627419 DOI: 10.1136/jitc-2021-002953] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND BO-112 is a nanoplexed form of polyinosinic:polycytidylic acid that acting on toll-like receptor 3 (TLR3), melanoma differentiation-associated protein 5 (MDA5) and protein kinase RNA-activated (PKR) elicits rejection of directly injected transplanted tumors, but has only modest efficacy against distant untreated tumors. Its clinical activity has also been documented in early phase clinical trials. The 5,6-dimethylxanthenone-4-acetic acid (DMXAA) stimulator of interferon genes (STING) agonist shows a comparable pattern of efficacy when used via intratumoral injections. METHODS Mice subcutaneously engrafted with bilateral MC38 and B16.OVA-derived tumors were treated with proinflammatory immunotherapy agents known to be active when intratumorally delivered. The combination of BO-112 and DMXAA was chosen given its excellent efficacy and the requirements for antitumor effects were studied on selective depletion of immune cell types and in gene-modified mouse strains lacking basic leucine zipper ATF-like transcription factor 3 (BATF3), interferon-α/β receptor (IFNAR) or STING. Spatial requirements for the injections were studied in mice bearing three tumor lesions. RESULTS BO-112 and DMXAA when co-injected in one of the lesions of mice bearing concomitant bilateral tumors frequently achieved complete local and distant antitumor efficacy. Synergistic effects were contingent on CD8 T cell lymphocytes and dependent on conventional type 1 dendritic cells, responsiveness to type I interferon (IFN) and STING function in the tumor-bearing host. Efficacy was preserved even if BO-112 and DMXAA were injected in separate lesions in a manner able to control another untreated third-party tumor. Efficacy could be further enhanced on concurrent PD-1 blockade. CONCLUSION Clinically feasible co-injections of BO-112 and a STING agonist attain synergistic efficacy able to eradicate distant untreated tumor lesions.
Collapse
Affiliation(s)
- Maite Alvarez
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain .,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Carmen Molina
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Carlos E De Andrea
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain.,Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maria Villalba
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain.,Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Jose Gonzalez-Gomariz
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain
| | - Maria Carmen Ochoa
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Alvaro Teijeira
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Javier Glez-Vaz
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Fernando Aranda
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Miguel F Sanmamed
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain.,Immunology and Oncology, Clinica Universidad de Navarra, Pamplona, Spain
| | | | - Xinyi Fan
- Radiation Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Wen H Shen
- Radiation Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Pedro Berraondo
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain
| | | | - Ignacio Melero
- Immunology and Immunotherapy, Center for Applied Medical Research (CIMA). University of Navarra, Pamplona, Spain .,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain.,Immunology and Oncology, Clinica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
32
|
de Andrea CE, Ochoa MC, Villalba-Esparza M, Teijeira Á, Schalper KA, Abengozar-Muela M, Eguren-Santamaría I, Sainz C, Sánchez-Gregorio S, Garasa S, Ariz M, Ortiz-de-Solorzano C, Rodriguez-Ruiz ME, Perez-Gracia JL, Lozano MD, Echeveste JI, Sanmamed MF, Melero I. Heterogenous presence of neutrophil extracellular traps in human solid tumours is partially dependent on IL-8. J Pathol 2021; 255:190-201. [PMID: 34184758 DOI: 10.1002/path.5753] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/17/2021] [Accepted: 06/23/2021] [Indexed: 01/21/2023]
Abstract
Neutrophil extracellular traps (NETs) are webs of extracellular nuclear DNA extruded by dying neutrophils infiltrating tissue. NETs constitute a defence mechanism to entrap and kill fungi and bacteria. Tumours induce the formation of NETs to the advantage of the malignancy via a variety of mechanisms shown in mouse models. Here, we investigated the presence of NETs in a variety of human solid tumours and their association with IL-8 (CXCL8) protein expression and CD8+ T-cell density in the tumour microenvironment. Multiplex immunofluorescence panels were developed to identify NETs in human cancer tissues by co-staining with the granulocyte marker CD15, the neutrophil marker myeloperoxidase and citrullinated histone H3 (H3Cit), as well as IL-8 protein and CD8+ T cells. Three ELISA methods to detect and quantify circulating NETs in serum were optimised and utilised. Whole tumour sections and tissue microarrays from patients with non-small cell lung cancer (NSCLC; n = 14), bladder cancer (n = 14), melanoma (n = 11), breast cancer (n = 31), colorectal cancer (n = 20) and mesothelioma (n = 61) were studied. Also, serum samples collected retrospectively from patients with metastatic melanoma (n = 12) and NSCLC (n = 34) were ELISA assayed to quantify circulating NETs and IL-8. NETs were detected in six different human cancer types with wide individual variation in terms of tissue density and distribution. At least in NSCLC, bladder cancer and metastatic melanoma, NET density positively correlated with IL-8 protein expression and inversely correlated with CD8+ T-cell densities. In a series of serum samples from melanoma and NSCLC patients, a positive correlation between circulating NETs and IL-8 was found. In conclusion, NETs are detectable in formalin-fixed human biopsy samples from solid tumours and in the circulation of cancer patients with a considerable degree of individual variation. NETs show a positive association with IL-8 and a trend towards a negative association with CD8+ tumour-infiltrating lymphocytes. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Carlos E de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - María Carmen Ochoa
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - María Villalba-Esparza
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Álvaro Teijeira
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Marta Abengozar-Muela
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Iñaki Eguren-Santamaría
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Cristina Sainz
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sandra Sánchez-Gregorio
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Saray Garasa
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Mikel Ariz
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Carlos Ortiz-de-Solorzano
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - María E Rodriguez-Ruiz
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - María D Lozano
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - José I Echeveste
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Miguel F Sanmamed
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| |
Collapse
|
33
|
Sanegre S, Eritja N, de Andrea C, Diaz-Martin J, Diaz-Lagares Á, Jácome MA, Salguero-Aranda C, García Ros D, Davidson B, Lopez R, Melero I, Navarro S, Ramon Y Cajal S, de Alava E, Matias-Guiu X, Noguera R. Characterizing the Invasive Tumor Front of Aggressive Uterine Adenocarcinoma and Leiomyosarcoma. Front Cell Dev Biol 2021; 9:670185. [PMID: 34150764 PMCID: PMC8209546 DOI: 10.3389/fcell.2021.670185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
The invasive tumor front (the tumor–host interface) is vitally important in malignant cell progression and metastasis. Tumor cell interactions with resident and infiltrating host cells and with the surrounding extracellular matrix and secreted factors ultimately determine the fate of the tumor. Herein we focus on the invasive tumor front, making an in-depth characterization of reticular fiber scaffolding, infiltrating immune cells, gene expression, and epigenetic profiles of classified aggressive primary uterine adenocarcinomas (24 patients) and leiomyosarcomas (11 patients). Sections of formalin-fixed samples before and after microdissection were scanned and studied. Reticular fiber architecture and immune cell infiltration were analyzed by automatized algorithms in colocalized regions of interest. Despite morphometric resemblance between reticular fibers and high presence of macrophages, we found some variance in other immune cell populations and distinctive gene expression and cell adhesion-related methylation signatures. Although no evident overall differences in immune response were detected at the gene expression and methylation level, impaired antimicrobial humoral response might be involved in uterine leiomyosarcoma spread. Similarities found at the invasive tumor front of uterine adenocarcinomas and leiomyosarcomas could facilitate the use of common biomarkers and therapies. Furthermore, molecular and architectural characterization of the invasive front of uterine malignancies may provide additional prognostic information beyond established prognostic factors.
Collapse
Affiliation(s)
- Sabina Sanegre
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, School of Medical, University of Valencia-INCLIVA, Valencia, Spain
| | - Núria Eritja
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institut de Recerca Biomèdica de LLeida (IRBLLEIDA), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Department of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, University of Lleida - University of Barcelona, Barcelona, Spain
| | - Carlos de Andrea
- Cancer CIBER (CIBERONC), Madrid, Spain.,Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Juan Diaz-Martin
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institute of Biomedicine of Sevilla, Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Ángel Diaz-Lagares
- Cancer CIBER (CIBERONC), Madrid, Spain.,Cancer Epigenomics, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - María Amalia Jácome
- Department of Mathematics, MODES Group, CITIC, Faculty of Science, Universidade da Coruña, A Coruña, Spain
| | - Carmen Salguero-Aranda
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institute of Biomedicine of Sevilla, Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - David García Ros
- Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Ben Davidson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Rafel Lopez
- Cancer CIBER (CIBERONC), Madrid, Spain.,Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.,Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Ignacio Melero
- Cancer CIBER (CIBERONC), Madrid, Spain.,Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Samuel Navarro
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, School of Medical, University of Valencia-INCLIVA, Valencia, Spain
| | - Santiago Ramon Y Cajal
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Enrique de Alava
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institute of Biomedicine of Sevilla, Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Xavier Matias-Guiu
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institut de Recerca Biomèdica de LLeida (IRBLLEIDA), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Department of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, University of Lleida - University of Barcelona, Barcelona, Spain
| | - Rosa Noguera
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, School of Medical, University of Valencia-INCLIVA, Valencia, Spain
| |
Collapse
|
34
|
Tejerina E, Garca Tobar L, Echeveste JI, de Andrea CE, Vigliar E, Lozano MD. PD-L1 in Cytological Samples: A Review and a Practical Approach. Front Med (Lausanne) 2021; 8:668612. [PMID: 34026795 PMCID: PMC8139418 DOI: 10.3389/fmed.2021.668612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 12/25/2022] Open
Abstract
With a growing number of predictive biomarkers needed to manage patients with non-small cell lung cancer (NSCLC), there has been a paradigm shift in care and handling of diagnostic samples. Among the various testing methods, immunohistochemistry (IHC) is the most cost- effective and widely available. Furthermore, over the past decade immunotherapy has emerged as one of the most promising cancer treatments. In this scenario IHC is the most used testing method available for PDL-1/PD1 immunotherapy. Several monoclonal antibodies targeting programmed death 1 (PD-1)/programmed death ligand-1 (PD-L1) pathways have been integrated into standard-of-care treatments of a wide range of cancer types, once provided evidence of PD-L1 expression in tumor cells by immunohistochemistry (IHC). Since currently available PD-L1 assays have been developed on formalin-fixed paraffin embedded (FFPE) histological specimens, a growing body of research is being dedicated to confirm the feasibility of applying PDL-1 assays also to cytological samples. Albeit promising results have been reported, several important issues still need to be addressed. Among these are the type of cytological samples, pre-analytical issues, cyto-histological correlation, and inter-observer agreement. This review briefly summarizes the knowledge of the role of cytopathology in the analysis of PD-L1 by immunocytochemistry (ICC) and future directions of cytopathology in the immunotherapy setting.
Collapse
Affiliation(s)
- Eva Tejerina
- Department of Pathology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Laura Garca Tobar
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| | - Jos I Echeveste
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| | | | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mara D Lozano
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| |
Collapse
|
35
|
Wadee R, Wadee AA. The Pathology of Lymphocytes, Histiocytes, and Immune Mechanisms in Mycobacterium tuberculosis Granulomas. Am J Trop Med Hyg 2021; 104:1796-1802. [PMID: 33720848 PMCID: PMC8103466 DOI: 10.4269/ajtmh.20-1372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/24/2021] [Indexed: 12/20/2022] Open
Abstract
Granuloma formation is the pathologic hallmark of tuberculosis (TB). Few studies have detailed the exact production of cytokines in human granulomatous inflammation and little is known about accessory molecule expressions in tuberculous granulomas. We aimed to identify some of the components of the immune response in granulomas in HIV-positive and -negative lymph nodes. We investigated the immunohistochemical profiles of CD4+, CD8+, CD68+, Th-17, Forkhead box P3 (FOXP3) cells, accessory molecule expression (human leukocyte antigen [HLA] classes I and II), and selected cytokines (interleukins 2, 4, and 6 and interferon-γ) of various cells, in granulomas within lymph nodes from 10 HIV-negative (-) and 10 HIV-positive (+) cases. CD4+ lymphocyte numbers were retained in HIV- granulomas, whereas CD4+:CD8 + cell were reversed in HIV+ TB granulomas. CD68 stained all histiocytes. Granulomas from the HIV+ group demonstrated a significant increase in FOXP3 cells. Interleukin-2 cytoplasmic expression was similar in both groups. Interferon-gamma (IFN-γ) expression was moderately increased, IL-6 was statistically increased and IL-4 expression was marginally lower in cells from HIV- than HIV+ TB granulomas. Greater numbers of cells expressed IFN-γ and IL-6 than IL-2 and IL-4 in HIV- TB granulomas. This study highlights the varied cytokine production in HIV-positive and -negative TB granulomas and indicates the need to identify localized tissue factors that play a role in mounting an adequate immune response required to halt infection. Although TB mono-infection causes variation in cell marker expression and cytokines in granulomas, alterations in TB and HIV coinfection are greater, pointing toward evolution of microorganism synergism.
Collapse
Affiliation(s)
- Reubina Wadee
- Department of Anatomical Pathology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, and National Health Laboratory Service (NHLS), Johannesburg, South Africa;,Address correspondence to Reubina Wadee, University of the Witwatersrand/National Health Laboratory Service (NHLS), Rm. 3L30, University of the Witwatersrand, School of Pathology, 7 York Rd., Parktown, Johannesburg 2193, Republic of South Africa. E-mail:
| | | |
Collapse
|
36
|
Tobar LG, Villalba-Esparza M, Abengozar-Muela M, Alvarez Gigli L, Echeveste JI, de Andrea CE, Lozano MD. Utilisation of cytological samples for multiplex immunofluorescence assay. Cytopathology 2021; 32:611-616. [PMID: 33870575 DOI: 10.1111/cyt.12979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Understanding the immune environment of non-small cell lung cancer (NSCLC) is important for designing effective anticancer immunotherapies. We describe the use of multiplex immunofluorescence (mIF) assays to enable characterisation of the tumour-infiltrating immune cells and their interactions, both across and within immune subtypes. METHODS Six cytological samples of NSCLC taken by transoesophageal ultrasound-guided fine needle aspiration were tested with an mIF assay designed to detect the expression of key immune cell markers such as CD3, CD8, CD20, CD11b, and CD68. Pan-cytokeratin was used to detect the NSCLC cells. Fluorescence images were acquired on a Vectra-Polaris Automated Quantitative Pathology Imaging System (Akoya Biosciences). RESULTS MIF assay was able to reliably detect and quantify the myeloid cell markers CD11b, CD68, CD3+ and CD8+ T cells, and CD20+ B lymphocytes on cytological samples of NSCLC. Whole-tissue analysis and its correlation with the corresponding H&E stains allowed a better understanding of the tissue morphology and the relationship between tumour and stroma compartments. Additionally, a uniform, specific, and correct staining pattern was seen for every immune marker. CONCLUSION The implementation of mIF assay on cytological samples taken with minimally invasive methods seems feasible and can be used to explore the immune environment of NSCLC.
Collapse
Affiliation(s)
- Laura Garcia Tobar
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - María Villalba-Esparza
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Marta Abengozar-Muela
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Laura Alvarez Gigli
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - José I Echeveste
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| | - María D Lozano
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Anatomy, Physiology and Pathology, University of Navarra, Pamplona, Spain
| |
Collapse
|
37
|
Garcia-Moure M, Gonzalez-Huarriz M, Labiano S, Guruceaga E, Bandres E, Zalacain M, Marrodan L, de Andrea C, Villalba M, Martinez-Velez N, Laspidea V, Puigdelloses M, Gallego Perez-Larraya J, Iñigo-Marco I, Stripecke R, Chan JA, Raabe EH, Kool M, Gomez-Manzano C, Fueyo J, Patiño-García A, Alonso MM. Delta-24-RGD, an Oncolytic Adenovirus, Increases Survival and Promotes Proinflammatory Immune Landscape Remodeling in Models of AT/RT and CNS-PNET. Clin Cancer Res 2021; 27:1807-1820. [PMID: 33376098 PMCID: PMC7617079 DOI: 10.1158/1078-0432.ccr-20-3313] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/14/2020] [Accepted: 12/22/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Atypical teratoid/rhabdoid tumors (AT/RT) and central nervous system primitive neuroectodermal tumors (CNS-PNET) are pediatric brain tumors with poor survival and life-long negative side effects. Here, the aim was to characterize the efficacy and safety of the oncolytic adenovirus, Delta-24-RGD, which selectively replicates in and kills tumor cells. EXPERIMENTAL DESIGN Delta-24-RGD determinants for infection and replication were evaluated in patient expression datasets. Viral replication and cytotoxicity were assessed in vitro in a battery of CNS-PNET and AT/RT cell lines. In vivo, efficacy was determined in different orthotopic mouse models, including early and established tumor models, a disseminated AT/RT lesion model, and immunocompetent humanized mouse models (hCD34+-NSG-SGM3). RESULTS Delta-24-RGD infected and replicated efficiently in all the cell lines tested. In addition, the virus induced dose-dependent cytotoxicity [IC50 value below 1 plaque-forming unit (PFU)/cell] and the release of immunogenic markers. In vivo, a single intratumoral Delta-24-RGD injection (107 or 108 PFU) significantly increased survival and led to long-term survival in AT/RT and PNET models. Delta-24-RGD hindered the dissemination of AT/RTs and increased survival, leading to 70% of long-term survivors. Of relevance, viral administration to established tumor masses (30 days after engraftment) showed therapeutic benefit. In humanized immunocompetent models, Delta-24-RGD significantly extended the survival of mice bearing AT/RTs or PNETs (ranging from 11 to 27 days) and did not display any toxicity associated with inflammation. Immunophenotyping of Delta-24-RGD-treated tumors revealed increased CD8+ T-cell infiltration. CONCLUSIONS Delta-24-RGD is a feasible therapeutic option for AT/RTs and CNS-PNETs. This work constitutes the basis for potential translation to the clinical setting.
Collapse
Affiliation(s)
- Marc Garcia-Moure
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain.
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Elizabeth Guruceaga
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Bioinformatics Platform, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Navarra, Spain
| | - Eva Bandres
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Immunology Unit, Department of Hematology, Complejo Hospitalario de Navarra, Pamplona, Navarra, Spain
| | - Marta Zalacain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lucia Marrodan
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Carlos de Andrea
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Naiara Martinez-Velez
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Virginia Laspidea
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Montse Puigdelloses
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Jaime Gallego Perez-Larraya
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Iñigo-Marco
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Renata Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Laboratory of Regenerative Immune Therapies Applied of the Research Network REBIRTH, German Centre for Infection Research (DZIF), partner site Hannover, Hannover, Germany
| | - Jennifer A Chan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eric H Raabe
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Division of Pediatric Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center (KITZ), Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Candelaria Gomez-Manzano
- Department of NeuroOncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juan Fueyo
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana Patiño-García
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Marta M Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain.
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| |
Collapse
|
38
|
Salas-Benito D, Conde E, Tamayo-Uria I, Mancheño U, Elizalde E, Garcia-Ros D, Aramendia JM, Muruzabal JC, Alcaide J, Guillen-Grima F, Minguez JA, Amores-Tirado J, Gonzalez-Martin A, Sarobe P, Lasarte JJ, Ponz-Sarvise M, De Andrea CE, Hervas-Stubbs S. The mutational load and a T-cell inflamed tumour phenotype identify ovarian cancer patients rendering tumour-reactive T cells from PD-1 + tumour-infiltrating lymphocytes. Br J Cancer 2021; 124:1138-1149. [PMID: 33402737 PMCID: PMC7961070 DOI: 10.1038/s41416-020-01218-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Adoptive immunotherapy with tumour-infiltrating lymphocytes (TIL) may benefit from the use of selective markers, such as PD-1, for tumour-specific T-cell enrichment, and the identification of predictive factors that help identify those patients capable of rendering tumour-reactive TILs. We have investigated this in ovarian cancer (OC) patients as candidates for TIL therapy implementation. METHODS PD-1- and PD-1+ CD8 TILs were isolated from ovarian tumours and expanded cells were tested against autologous tumour cells. Baseline tumour samples were examined using flow cytometry, multiplexed immunofluorescence and Nanostring technology, for gene expression analyses, as well as a next-generation sequencing gene panel, for tumour mutational burden (TMB) calculation. RESULTS Tumour-reactive TILs were detected in half of patients and were exclusively present in cells derived from the PD-1+ fraction. Importantly, a high TIL density in the fresh tumour, the presence of CD137+ cells within the PD-1+CD8+ TIL subset and their location in the tumour epithelium, together with a baseline T-cell-inflamed genetic signature and/or a high TMB, are features that identify patients rendering tumour-reactive TIL products. CONCLUSION We have demonstrated that PD-1 identifies ovarian tumour-specific CD8 TILs and has uncovered predictive factors that identify OC patients who are likely to render tumour-specific cells from PD-1+ TILs.
Collapse
Affiliation(s)
- Diego Salas-Benito
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Conde
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Ibon Tamayo-Uria
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Uxua Mancheño
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Edurne Elizalde
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - David Garcia-Ros
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Department Pathology, Anatomy and Physiology, Universidad de Navarra, Pamplona, Spain
| | - Jose M Aramendia
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Juan C Muruzabal
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Gynecologic Oncology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Julia Alcaide
- Department of Oncology, Hospital Costa del Sol, Marbella, Spain
| | - Francisco Guillen-Grima
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Preventive Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Jose A Minguez
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Obstetrics and Gynecology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Antonio Gonzalez-Martin
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- GEICO Study Group, Madrid, Spain
| | - Pablo Sarobe
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan J Lasarte
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Mariano Ponz-Sarvise
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
- Program of Solid Tumors, CIMA, University of Navarra, Pamplona, Spain.
| | - Carlos E De Andrea
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Department Pathology, Anatomy and Physiology, Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
| | - Sandra Hervas-Stubbs
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
39
|
Proteomic Analysis of Low-Grade, Early-Stage Endometrial Carcinoma Reveals New Dysregulated Pathways Associated with Cell Death and Cell Signaling. Cancers (Basel) 2021; 13:cancers13040794. [PMID: 33672863 PMCID: PMC7917913 DOI: 10.3390/cancers13040794] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Low-grade, early-stage endometrial cancer (EC) is the most frequent malignant tumor of the uterine corpus. Our study aimed to assess dysregulated pathways in this specific subset of EC through proteomic analysis. We describe and validate the dysregulation of the SLIT/ROBO signaling pathway, as well as cellular death processes such as necroptosis and ferroptosis. We identify several immune-related pathways, with a dominance of innate immune response associated pathways. Our findings reveal the singular biology of low-grade, early-stage ECs and could guide future research in the field. Abstract Low-grade, early-stage endometrial carcinoma (EC) is the most frequent malignant tumor of the uterine corpus. However, the molecular alterations that underlie these tumors are far from being fully understood. The purpose of this study is to describe dysregulated molecular pathways from EC patients. Sixteen samples of tumor tissue and paired healthy controls were collected and both were subjected to mass spectrometry (MS)/MS proteomic analysis. Gene ontology and pathway analysis was performed to discover dysregulated pathways and/or proteins using different databases and bioinformatic tools. Dysregulated pathways were cross-validated in an independent external cohort. Cell signaling, immune response, and cell death-associated pathways were robustly identified. The SLIT/ROBO signaling pathway demonstrated dysregulation at the proteomic and transcriptomic level. Necroptosis and ferroptosis were cell death-associated processes aberrantly regulated, in addition to apoptosis. Immune response-associated pathways showed a dominance of innate immune responses. Tumor immune infiltrates measured by immunofluorescence demonstrated diverse lymphoid and myeloid populations. Our results suggest a role of SLIT/ROBO, necroptosis, and ferroptosis, as well as a prominent role of innate immune response in low-grade, early-stage EC. These results could guide future research in this group of tumors.
Collapse
|
40
|
Humphries M, Maxwell P, Salto-Tellez M. QuPath: The global impact of an open source digital pathology system. Comput Struct Biotechnol J 2021; 19:852-859. [PMID: 33598100 PMCID: PMC7851421 DOI: 10.1016/j.csbj.2021.01.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
QuPath, originally created at the Centre for Cancer Research & Cell Biology at Queen's University Belfast as part of a research programme in digital pathology (DP) funded by Invest Northern Ireland and Cancer Research UK, is arguably the most wildly used image analysis software program in the world. On the back of the explosion of DP and a need to comprehensively visualise and analyse whole slides images (WSI), QuPath was developed to address the many needs associated with tissue based image analysis; these were several fold and, predominantly, translational in nature: from the requirement to visualise images containing billions of pixels from files several GBs in size, to the demand for high-throughput reproducible analysis, which the paradigm of routine visual pathological assessment continues to struggle to deliver. Resultantly, large-scale biomarker quantification must increasingly be augmented with DP. Here we highlight the impact of the open source Quantitative Pathology & Bioimage Analysis DP system since its inception, by discussing the scope of scientific research in which QuPath has been cited, as the system of choice for researchers.
Collapse
Affiliation(s)
- M.P. Humphries
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
| | - P. Maxwell
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
| | - M. Salto-Tellez
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
- Integrated Pathology Programme, Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|