1
|
Ainslie K. Modifying Post-Surgical Immunity: Controlled Release of TLR7/8 Agonist for Immune Mediated Clearance of Glioblastoma. RESEARCH SQUARE 2024:rs.3.rs-5024510. [PMID: 39399681 PMCID: PMC11469459 DOI: 10.21203/rs.3.rs-5024510/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Glioblastoma is an aggressive brain cancer with a dismal prognosis despite current therapeutic interventions. Tumor resection is standard-of-care for glioblastoma and has profound immunostimulatory effects. Resulting in a nadir in tumor burden, resection offers a unique opportunity to break local immune tolerance and mount an effective anti-tumor immune response. Here, we explore the effect of local and controlled release of TLR7/8 agonist from a polymer scaffold implanted at the time of tumor resection. We find that sustained release of TLR7/8 agonist leads to clearance of residual post-resection tumor, improved survival, and subsequent protection from tumor challenge in mice bearing orthotopic GL261 or CT2A gliomas. We show that scaffold therapy boosts resection-mediated disruption to the tumor microenvironment, leading to an early inflammatory innate immune response both in the brain and cervical lymph node. This is followed by an influx of activated NK cells in the brain and effector T cells in the lymph node and brain. In sum, sustained local TLR7/8 agonism within the context of tumor resection is a promising approach for glioblastoma.
Collapse
|
2
|
Hogan A, Mut M. Neurosteroids in Glioma: A Novel Therapeutic Concept. Life (Basel) 2024; 14:975. [PMID: 39202716 PMCID: PMC11355226 DOI: 10.3390/life14080975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/03/2024] Open
Abstract
Glioma, a diverse group of brain and spinal cord tumors arising from glial cells, is characterized by varying degrees of malignancy, with some types exhibiting highly aggressive behavior, rapid proliferation, and invasive growth patterns, posing significant therapeutic challenges. This review delves into the complex interactions between glioma cells, neurotransmitters, and neurosteroids, emphasizing their potential as therapeutic targets. Key neurotransmitters, like glutamate and gamma-aminobutyric acid (GABA), play crucial roles in glioma growth, invasion, and treatment response. This review examines the involvement of neurosteroids in glioma biology and explores innovative therapeutic strategies targeting these systems. It encompasses the biosynthesis and mechanisms of neurosteroids, interactions between gliomas and neurotransmitters, the spatial distribution of neurosteroid synthesis in gliomas, the role of ion channels, hormonal influences, enzyme modulation, and the neuroimmune system in glioma progression. Additionally, it highlights the potential of neurosteroids to modulate these pathways for therapeutic benefit.
Collapse
Affiliation(s)
- Ava Hogan
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903, USA;
| | - Melike Mut
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
3
|
Li F, Song B, Zhou WF, Chu LJ. Toll-Like Receptors 7/8: A Paradigm for the Manipulation of Immunologic Reactions for Immunotherapy. Viral Immunol 2023; 36:564-578. [PMID: 37751284 DOI: 10.1089/vim.2023.0077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
The innate immune system recognizes conserved features of viral and microbial pathogens through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are one type of PRR used by the innate immune system to mediate the secretion of proinflammatory cytokines and promote innate and adaptive immune responses. TLR family members TLR7 and TLR8 (referred to as TLR7/8 from herein) are endosomal transmembrane receptors that recognize purine-rich single-stranded RNA (ssRNA) and bacterial DNA, eliciting an immunologic reaction to pathogens. TLR7/8 were discovered to mediate the secretion of proinflammatory cytokines by activating immune cells. In addition, accumulating evidence has indicated that TLR7/8 may be closely related to numerous immune-mediated disorders, specifically several types of cancer, autoimmune disease, and viral disease. TLR7/8 agonists and antagonists, which are used as drugs or adjuvants, have been identified in preclinical studies and clinical trials as promising immune stimulators for the immunotherapy of these immune-mediated disorders. These results provided reasoning to further explore immunotherapy for the treatment of immune-mediated disorders. Nevertheless, numerous needs remain unmet, and the therapeutic effects of TLR7/8 agonists and antagonists are poor and exert strong immune-related toxicities. The present review aimed to provide an overview of the TLR family members, particularly TLR7/8, and address the underlying molecular mechanisms and clinical implications of TLR7/8 in immune-mediated disorders. The aim of the work is to discuss the underlying molecular mechanisms and clinical implications of TLR7/8 in immune-mediated disorders.
Collapse
Affiliation(s)
- Fang Li
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| | - Biao Song
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei-Feng Zhou
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| | - Li-Jin Chu
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| |
Collapse
|
4
|
Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neurooncol 2023; 162:93-108. [PMID: 36854924 DOI: 10.1007/s11060-023-04257-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Exosomes are nano-vesicular carriers capable of delivering cargoes for intercellular communication, which holds potential as biocompatible and high efficiency systems for drug delivery. In this study, we evaluated the potential effect of T7 peptide-decorated exosome-loaded Galectin-9 siRNA (T7-Exo/siGalectin-9) in the M1 polarization of macrophages and immunosuppression of glioblastoma (GBM). METHODS Differentially expressed genes in GBM were in silico predicted and then experimentally verified. Galectin-9 was knocked down by siRNA to assess its role in tumor-bearing mice. T7 peptide-decorated exosomes (derived from human embryonic kidney [HEK]-293T cells) targeting GBM were prepared, and loaded with Galectin-9 siRNA by electroporation to prepare nanoformulations (T7-Exo/siGalectin-9). The role of T7-Exo/siGalectin-9 in CD8+ T cell cytotoxicity to target GBM cells and polarization of macrophages was evaluated after artificial modulation of Galectin-9 expression. Anti-tumor effects of T7-Exo/siGalectin-9 were elucidated in vitro and in vivo. RESULTS Galectin-9 was highly expressed in GBM tissues and cell lines. The siRNA-mediated knockdown of Galectin-9 repressed the growth of xenografts of GBM cells in C57BL/6 mice and activated immune response in the tumor microenvironment. T7-Exo/siGalectin-9 effectively delivered siGalectin-9 to GBM cells. T7-Exo/siGalectin-9 contributed to activation of the TLR7-IRF5 pathway, which polarized macrophages to M1 phenotype. By this mechanism, phagocytosis of GBM cells by macrophages was increased, the anti-tumor effect of CD8+ T cells was enhanced and the inflammatory responses were suppressed. CONCLUSION Overall, T7-Exo/siGalectin-9 promotes macrophage repolarization and restricts the immunosuppression of GBM, thus providing novel insights into and drug delivery system of immunotherapy for GBM.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Ning Guan
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121000, Liaoning, People's Republic of China.
| |
Collapse
|
5
|
Meng W, Li Z, Zhang Y, Yang A, Wang Y, Zhou Y, Wu W, Qiu Y, Li L. ZhenQi FuZheng formula inhibits the growth of colorectal tumors by modulating intestinal microflora-mediated immune function. Aging (Albany NY) 2022; 14:4769-4785. [PMID: 35680568 PMCID: PMC9217701 DOI: 10.18632/aging.204111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/13/2022] [Indexed: 12/24/2022]
Abstract
Zhenqi Fuzheng formula (ZQFZ), of which the main ingredients are Astragalus membranaceus and Ligustrum lucidum, has immune system regulatory functions and potential anti-tumor bioactivity. The inhibition of colorectal tumor growth by ZQFZ was analyzed in inflammatory cells and B6/JGpt-Apcem1Cin(MinC)/Gpt (ApcMin/+) mice. ZQFZ exhibited anti-inflammatory activity by decreasing the phosphorylation of nuclear factor-kappa B (NF-κB) pathway-related proteins in lipopolysaccharide-induced RAW264.7 cells. After 56 days of treatment, ZQFZ alleviated the progression of colorectal cancer (CRC) and increased the body weight and thymic index values of the ApcMin/+ mice. An analysis of the intestinal microflora showed that ZQFZ affected the abundance of certain immune-related bacteria, which may explain its immunomodulatory effects. Moreover, the percentages of T cells and NK cells in peripheral blood were significantly increased and 15 immune-related cytokines were regulated in serum or the colon or both. ZQFZ upregulated the levels of CD4 and CD8 in the spleen and colorectal tumors and decreased the expression levels of cytotoxic T-lymphocyte-associated protein 4 and programmed death-ligand 1 in colorectal tumors. ZQFZ promoted an anti-tumor immune response and inhibited the occurrence and development of CRC by regulating the immune system. This study provides the experimental basis for the application of ZQFZ as a therapeutic agent for CRC.
Collapse
Affiliation(s)
- Weiqi Meng
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China
| | - Zhiping Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, P.R. China.,School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China
| | - Yiting Zhang
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China
| | - Anhui Yang
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China
| | - Yanzhen Wang
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai, P.R. China
| | - Yulin Zhou
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China
| | - Wanyue Wu
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China
| | - Ye Qiu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Lanzhou Li
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R. China.,Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, Jilin, P.R. China
| |
Collapse
|
6
|
Alvarez-Arellano L, Eguía-Aguilar P, Piña-Sánchez P, González-García N, Palma-Guzman A, Perezpeña-Diazconti M, Maldonado-Bernal C. High expression of Toll-like receptor 7 is a survival factor in pediatric medulloblastoma. Childs Nerv Syst 2021; 37:3743-3752. [PMID: 34480601 DOI: 10.1007/s00381-021-05347-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Medulloblastoma is an embryonal brain tumor that predominantly occurs in childhood with a wide histological and molecular variability. Our aim was to investigate the expression of Toll-like receptors (TLRs), their association with the infiltration of immune cells and with the histological subgroups, and, also, with the overall survival of patients. METHODS Fifty-six paraffin-preserved biopsies from children with medulloblastoma of the classic, desmoplastic, and anaplastic subtypes were included. Microarrays of tissues were performed, and the infiltration of T and NK cells was quantified, as well as the expression of TLR7, TLR8, and TLR9. For all statistical analyses, significance was p < 0.05. RESULTS CD4 + and CD8 + T lymphocytes and NK cells were found infiltrating the tumor. The infiltration of NK and CD4 + cells was greater in the classic and desmoplastic subtypes than in anaplastic. We found an important expression of TLRs in all medulloblastomas, but TLR7 and TLR8 were considerably higher in classic and desmoplastic subtypes than in anaplastic. Importantly, we observed that TLR7 was a prognostic factor for survival. CONCLUSIONS Medulloblastomas present cellular infiltration and a differential expression of TLRs depending on the histological subtype. TLR7 is a prognostic factor of survival that is dependent on treatment and age.
Collapse
Affiliation(s)
| | - Pilar Eguía-Aguilar
- Laboratorio de Biología Molecular, Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Patricia Piña-Sánchez
- Laboratorio de Oncología Molecular, Unidad de Investigación Médica en Enfermedades Oncológicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Nadia González-García
- Laboratorio de Neurociencias, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Alam Palma-Guzman
- Laboratorio Nacional de Microscopia Avanzada, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Mario Perezpeña-Diazconti
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Carmen Maldonado-Bernal
- Unidad de Investigación en Inmunología y Proteómica , Hospital Infantil de México Federico Gómez, Mexico City, Mexico.
| |
Collapse
|
7
|
Xun Y, Yang H, Kaminska B, You H. Toll-like receptors and toll-like receptor-targeted immunotherapy against glioma. J Hematol Oncol 2021; 14:176. [PMID: 34715891 PMCID: PMC8555307 DOI: 10.1186/s13045-021-01191-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023] Open
Abstract
Glioma represents a fast proliferating and highly invasive brain tumor which is resistant to current therapies and invariably recurs. Despite some advancements in anti-glioma therapies, patients’ prognosis remains poor. Toll-like receptors (TLRs) act as the first line of defense in the immune system being the detectors of those associated with bacteria, viruses, and danger signals. In the glioma microenvironment, TLRs are expressed on both immune and tumor cells, playing dual roles eliciting antitumoral (innate and adaptive immunity) and protumoral (cell proliferation, migration, invasion, and glioma stem cell maintenance) responses. Up to date, several TLR-targeting therapies have been developed aiming at glioma bulk and stem cells, infiltrating immune cells, the immune checkpoint axis, among others. While some TLR agonists exhibited survival benefit in clinical trials, it attracts more attention when they are involved in combinatorial treatment with radiation, chemotherapy, immune vaccination, and immune checkpoint inhibition in glioma treatment. TLR agonists can be used as immune modulators to enhance the efficacy of other treatment, to avoid dose accumulation, and what brings more interests is that they can potentiate immune checkpoint delayed resistance to PD-1/PD-L1 blockade by upregulating PD-1/PD-L1 overexpression, thus unleash powerful antitumor responses when combined with immune checkpoint inhibitors. Herein, we focus on recent developments and clinical trials exploring TLR-based treatment to provide a picture of the relationship between TLR and glioma and their implications for immunotherapy.
Collapse
Affiliation(s)
- Yang Xun
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong Province, China
| | - Hua Yang
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong Province, China
| | - Bozena Kaminska
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou, 510095, China.,Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Hua You
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou, 510095, China.
| |
Collapse
|
8
|
Macrophage polarization synergizes with oxaliplatin in lung cancer immunotherapy via enhanced tumor cell phagocytosis. Transl Oncol 2021; 14:101202. [PMID: 34390893 PMCID: PMC8365514 DOI: 10.1016/j.tranon.2021.101202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
LLC cells pretreated with OXA were more prone to be phagocytized by M1 than M2 macrophages. M2 repolarized to M1 by R848 in vitro showed enhanced phagocytic ability to OXA-treated LLC cells. Macrophage polarization from pro-tumor M2 to anti-tumor M1 synergizes with OXA.
Calreticulin (CALR) exposure is required for most immunogenic cell death (ICD) in the anti-tumor immunity induced by chemotherapeutic agents. The present study aimed to explore the anti-tumor efficacy of the combined administration of oxaliplatin (OXA) and R848 (an agent for macrophage polarization) in lung cancer cells. Flow cytometry and immunostaining assays were performed to evaluate CALR exposure induced by OXA in the murine Lewis lung carcinoma (LLC) cells. The phagocytosis of macrophages was determined using flow cytometry and western blotting assays. The anti-tumor efficacy of the OXA and R848 combination was evaluated using flow cytometry and western blotting in vitro and in vivo. OXA induced CALR exposure on the surface of LLC cells after low dose and short duration of treatment (20 μM OXA for 24 h). LLC cells pretreated with OXA were more prone to be phagocytized by M1 than M2 macrophages. M2 macrophages repolarized to M1 by R848 in vitro showed enhanced phagocytic ability to OXA-treated LLC cells. Finally, combined administration of OXA and R848 exhibited a synergistic anti-tumor effect than single agent applied in vitro and in vivo. Macrophage polarization from pro-tumor M2 to anti-tumor M1 synergizes with OXA in lung cancer immunotherapy via enhanced tumor cell phagocytosis.
Collapse
|
9
|
Shah D, Comba A, Faisal SM, Kadiyala P, Baker GJ, Alghamri MS, Doherty R, Zamler D, Nuñez G, Castro MG, Lowenstein PR. A novel miR1983-TLR7-IFNβ circuit licenses NK cells to kill glioma cells, and is under the control of galectin-1. Oncoimmunology 2021; 10:1939601. [PMID: 34249474 PMCID: PMC8244780 DOI: 10.1080/2162402x.2021.1939601] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/31/2021] [Indexed: 02/08/2023] Open
Abstract
Although pharmacological stimulation of TLRs has anti-tumor effects, it has not been determined whether endogenous stimulation of TLRs can lead to tumor rejection. Herein, we demonstrate the existence of an innate anti-glioma NK-mediated circuit initiated by glioma-released miR-1983 within exosomes, and which is under the regulation of galectin-1 (Gal-1). We demonstrate that miR-1983 is an endogenous TLR7 ligand that activates TLR7 in pDCs and cDCs through a 5'-UGUUU-3' motif at its 3' end. TLR7 activation and downstream signaling through MyD88-IRF5/IRF7 stimulates secretion of IFN-β. IFN-β then stimulates NK cells resulting in the eradication of gliomas. We propose that successful immunotherapy for glioma could exploit this endogenous innate immune circuit to activate TLR7 signaling and stimulate powerful anti-glioma NK activity, at least 10-14 days before the activation of anti-tumor adaptive immunity.
Collapse
Affiliation(s)
- Diana Shah
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Syed M. Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gregory J. Baker
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert Doherty
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel Zamler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Nuñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MIUSA
- Cancer Biology Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Keane L, Cheray M, Blomgren K, Joseph B. Multifaceted microglia - key players in primary brain tumour heterogeneity. Nat Rev Neurol 2021; 17:243-259. [PMID: 33692572 DOI: 10.1038/s41582-021-00463-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 01/31/2023]
Abstract
Microglia are the resident innate immune cells of the immune-privileged CNS and, as such, represent the first line of defence against tissue injury and infection. Given their location, microglia are undoubtedly the first immune cells to encounter a developing primary brain tumour. Our knowledge of these cells is therefore important to consider in the context of such neoplasms. As the heterogeneous nature of the most aggressive primary brain tumours is thought to underlie their poor prognosis, this Review places a special emphasis on the heterogeneity of the tumour-associated microglia and macrophage populations present in primary brain tumours. Where available, specific information on microglial heterogeneity in various types and subtypes of brain tumour is included. Emerging evidence that highlights the importance of considering the heterogeneity of both the tumour and of microglial populations in providing improved treatment outcomes for patients is also discussed.
Collapse
Affiliation(s)
- Lily Keane
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
11
|
Bahmani B, Gong H, Luk BT, Haushalter KJ, DeTeresa E, Previti M, Zhou J, Gao W, Bui JD, Zhang L, Fang RH, Zhang J. Intratumoral immunotherapy using platelet-cloaked nanoparticles enhances antitumor immunity in solid tumors. Nat Commun 2021; 12:1999. [PMID: 33790276 PMCID: PMC8012593 DOI: 10.1038/s41467-021-22311-z] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 03/05/2021] [Indexed: 12/22/2022] Open
Abstract
Intratumoral immunotherapy is an emerging modality for the treatment of solid tumors. Toll-like receptor (TLR) agonists have shown promise for eliciting immune responses, but systemic administration often results in the development of adverse side effects. Herein, we investigate whether localized delivery of the TLR agonist, resiquimod (R848), via platelet membrane-coated nanoparticles (PNP-R848) elicits antitumor responses. The membrane coating provides a means of enhancing interactions with the tumor microenvironment, thereby maximizing the activity of R848. Intratumoral administration of PNP-R848 strongly enhances local immune activation and leads to complete tumor regression in a colorectal tumor model, while providing protection against repeated tumor re-challenges. Moreover, treatment of an aggressive breast cancer model with intratumoral PNP-R848 delays tumor growth and inhibits lung metastasis. Our findings highlight the promise of locally delivering immunostimulatory payloads using biomimetic nanocarriers, which possess advantages such as enhanced biocompatibility and natural targeting affinities.
Collapse
Affiliation(s)
| | - Hua Gong
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Brian T Luk
- Cello Therapeutics, Inc., San Diego, CA, 92121, USA
| | | | | | - Mark Previti
- Cello Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Jiarong Zhou
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jack D Bui
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Jie Zhang
- Cello Therapeutics, Inc., San Diego, CA, 92121, USA.
| |
Collapse
|
12
|
Kos S, Lopes A, Preat V, Cemazar M, Lampreht Tratar U, Ucakar B, Vanvarenberg K, Sersa G, Vandermeulen G. Intradermal DNA vaccination combined with dual CTLA-4 and PD-1 blockade provides robust tumor immunity in murine melanoma. PLoS One 2019; 14:e0217762. [PMID: 31150505 PMCID: PMC6544376 DOI: 10.1371/journal.pone.0217762] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/19/2019] [Indexed: 01/01/2023] Open
Abstract
We aimed to explore whether the combination of intradermal DNA vaccination, to boost immune response against melanoma antigens, and immune checkpoint blockade, to alleviate immunosuppression, improves antitumor effectiveness in a murine B16F10 melanoma tumor model. Compared to single treatments, a combination of intradermal DNA vaccination (ovalbumin or gp100 plasmid adjuvanted with IL12 plasmid) and immune checkpoint CTLA-4/PD-1 blockade resulted in a significant delay in tumor growth and prolonged survival of treated mice. Strong activation of the immune response induced by combined treatment resulted in a significant antigen-specific immune response, with elevated production of antigen-specific IgG antibodies and increased intratumoral CD8+ infiltration. These results indicate a potential application of the combined DNA vaccination and immune checkpoint blockade, specifically, to enhance the efficacy of DNA vaccines and to overcome the resistance to immune checkpoint inhibitors in certain cancer types.
Collapse
Affiliation(s)
- Spela Kos
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Alessandra Lopes
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Veronique Preat
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
- * E-mail: (GS); (VP)
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Ursa Lampreht Tratar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
- * E-mail: (GS); (VP)
| | - Gaelle Vandermeulen
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
13
|
Toll-Like Receptors as Therapeutic Targets in Central Nervous System Tumors. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5286358. [PMID: 31240216 PMCID: PMC6556293 DOI: 10.1155/2019/5286358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/08/2019] [Indexed: 12/24/2022]
Abstract
In recent years, progress has been made in understanding the pathological, genetic, and molecular heterogeneity of central nervous system (CNS) tumors. However, improvements in risk classification, prognosis, and treatment have not been sufficient. Currently, great importance has been placed to the tumor microenvironment and the immune system, which are very important components that influence the establishment and development of tumors. Toll-like receptors (TLRs) are innate immunite system sensors of a wide variety of molecules, such as those associated with microorganisms and danger signals. TLRs are expressed on many cells, including immune cells and nonimmune cells such as neurons and cancer cells. In the tumor microenvironment, activation of TLRs plays dual antitumoral (dendritic cells, cytotoxic T cells, and natural killer cells activation) and protumoral effects (tumor cell proliferation, survival, and resistance to chemotherapy) and constitutes an area of opportunities and challenges in the development of new therapeutic strategies. Several clinical trials have been carried out, and others are currently in process; however, the results obtained to date have been contradictory and have not led to a definitive position about the use of TLR agonists in adjuvant therapy during the treatment of central nervous system (CNS) tumors. In this review, we focus on recent advances in TLR agonists as immunotherapies for treatment of CNS tumors.
Collapse
|
14
|
Wu J, Waxman DJ. Immunogenic chemotherapy: Dose and schedule dependence and combination with immunotherapy. Cancer Lett 2019; 419:210-221. [PMID: 29414305 DOI: 10.1016/j.canlet.2018.01.050] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Conventional cytotoxic cancer chemotherapy is often immunosuppressive and associated with drug resistance and tumor regrowth after a short period of tumor shrinkage or growth stasis. However, certain cytotoxic cancer chemotherapeutic drugs, including doxorubicin, mitoxantrone, and cyclophosphamide, can kill tumor cells by an immunogenic cell death pathway, which activates robust innate and adaptive anti-tumor immune responses and has the potential to greatly increase the efficacy of chemotherapy. Here, we review studies on chemotherapeutic drug-induced immunogenic cell death, focusing on how the choice of a conventional cytotoxic agent and its dose and schedule impact anti-tumor immune responses. We propose a strategy for effective immunogenic chemotherapy that employs a modified metronomic schedule for drug delivery, which we term medium-dose intermittent chemotherapy (MEDIC). Striking responses have been seen in preclinical cancer models using MEDIC, where an immunogenic cancer chemotherapeutic agent is administered intermittently and at an intermediate dose, designed to impart strong and repeated cytotoxic damage to tumors, and on a schedule compatible with activation of a sustained anti-tumor immune response, thereby maximizing anti-cancer activity. We also discuss strategies for combination chemo-immunotherapy, and we outline approaches to identify new immunogenic chemotherapeutic agents for drug development.
Collapse
Affiliation(s)
- Junjie Wu
- Department of Biology, Division of Cell and Molecular Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Department of Biology, Division of Cell and Molecular Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
15
|
Bota DA, Alexandru-Abrams D, Pretto C, Hofman FM, Chen TC, Fu B, Carrillo JA, Schijns VE, Stathopoulos A. Use of ERC-1671 Vaccine in a Patient with Recurrent Glioblastoma Multiforme after Progression during Bevacizumab Therapy: First Published Report. Perm J 2015; 19:41-6. [PMID: 25785641 DOI: 10.7812/tpp/14-042] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Glioblastoma multiforme is a highy aggressive tumor that recurs despite resection, focal beam radiation, and temozolamide chemotherapy. ERC-1671 is an experimental treatment strategy that uses the patient's own immune system to attack the tumor cells. The authors report preliminary data on the first human administration of ERC-1671 vaccination under a single-patient, compassionate-use protocol. The patient survived for ten months after the vaccine administration without any other adjuvant therapy and died of complications related to his previous chemotherapies.
Collapse
Affiliation(s)
- Daniela A Bota
- Associate Professor of Neurology and Neurosurgery at the University of California, Irvine in the City of Orange.
| | - Daniela Alexandru-Abrams
- Neurosurgery Resident at the University of California, Irvine Medical Center in the City of Orange.
| | - Chrystel Pretto
- Laboratory Manager for Epitopoietic Research Corporation at the Scientific Park Crealys in Gemblous, Les Ines, Belgium.
| | - Florence M Hofman
- Professor of Pathology at the Keck School of Medicine, University of Southern California, Los Angeles.
| | - Thomas C Chen
- Chief Medical Officer of Epitopoietic Research Corporation and a Professor of Neurosurgery at the Keck School of Medicine, University of Southern California, Los Angeles.
| | - Beverly Fu
- Neurological Oncology Nurse Practitioner in the Neurology Department of the University of California, Irvine Medical Center in the City of Orange.
| | - Jose A Carrillo
- Assistant Professor of Neurology at the University of California, Irvine Medical Center in the City of Orange.
| | - Virgil Ejc Schijns
- Chief Security Officer of Epitopoietic Research Corporation and a Professor of Immune Intervention in the Department of Cell Biology and Immunology at the Wageningen University in Netherlands.
| | - Apostolos Stathopoulos
- Chief Executive Officer for Epitopoietic Research Corporation at the Scientific Park Crealys in Gemblous, Les Ines, Belgium and Chief Neurosurgeon, Department of Neurosurgery, Arlon Hospital, Belgium.
| |
Collapse
|
16
|
Wu J, Waxman DJ. Metronomic cyclophosphamide eradicates large implanted GL261 gliomas by activating antitumor Cd8 + T-cell responses and immune memory. Oncoimmunology 2015; 4:e1005521. [PMID: 26137402 PMCID: PMC4485826 DOI: 10.1080/2162402x.2015.1005521] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/02/2015] [Accepted: 01/06/2015] [Indexed: 11/13/2022] Open
Abstract
Cancer chemotherapy using cytotoxic drugs can induce immunogenic tumor cell death; however, dosing regimens and schedules that enable single-agent chemotherapy to induce adaptive immune-dependent ablation of large, established tumors with activation of long-term immune memory have not been identified. Here, we investigate this issue in a syngeneic, implanted GL261 glioma model in immune-competent mice given cyclophosphamide on a 6-day repeating metronomic schedule. Two cycles of metronomic cyclophosphamide treatment induced sustained upregulation of tumor-associated CD8+ cytotoxic T lymphocyte (CTL) cells, natural killer (NK) cells, macrophages, and other immune cells. Expression of CTL- and NK–cell-shared effectors peaked on Day 6, and then declined by Day 9 after the second cyclophosphamide injection and correlated inversely with the expression of the regulatory T cell (Treg) marker Foxp3. Sustained tumor regression leading to tumor ablation was achieved after several cyclophosphamide treatment cycles. Tumor ablation required CD8+ T cells, as shown by immunodepletion studies, and was associated with immunity to re-challenge with GL261 glioma cells, but not B16-F10 melanoma or Lewis lung carcinoma cells. Rejection of GL261 tumor re-challenge was associated with elevated CTLs in blood and increased CTL infiltration in tumors, consistent with the induction of long-term, specific CD8+ T-cell anti-GL261 tumor memory. Co-depletion of CD8+ T cells and NK cells did not inhibit tumor regression beyond CD8+ T-cell depletion alone, suggesting that the metronomic cyclophosphamide-activated NK cells function via CD8a+ T cells. Taken together, these findings provide proof-of-concept that single-agent chemotherapy delivered on an optimized metronomic schedule can eradicate large, established tumors and induce long-term immune memory.
Collapse
Key Words
- B6, C57BL/6 mouse strain
- CD8+ T cells
- CPA, cyclophosphamide
- CPA-90 and CPA-140, metronomic CPA scheduling at 90 and 140 mg CPA/kg body weight, respectively, repeated every 6 days
- CTL, cytotoxic T lymphocyte
- FACS, fluorescence-activated cell sorting
- GL261 glioma
- LLC, Lewis lung carcinoma
- MDSC, myeloid-derived suppressor cells
- MTD, maximum-tolerated dose
- NK cell, natural killer cell
- NK cells
- PEB, phosphate-EDTA buffer
- chemoimmunotherapy
- chemotherapy
- cyclophosphamide
- drug scheduling
- glioblastoma
- immune memory
- metronomic chemotherapy
- qPCR, quantitative real-time polymerase chain reaction
Collapse
Affiliation(s)
- Junjie Wu
- Division of Cell and Molecular Biology; Department of Biology and Graduate Program in Molecular and Translational Medicine; Boston University ; Boston, MA USA
| | - David J Waxman
- Division of Cell and Molecular Biology; Department of Biology and Graduate Program in Molecular and Translational Medicine; Boston University ; Boston, MA USA
| |
Collapse
|
17
|
Recent advances in the role of toll-like receptors and TLR agonists in immunotherapy for human glioma. Protein Cell 2014; 5:899-911. [PMID: 25411122 PMCID: PMC4259890 DOI: 10.1007/s13238-014-0112-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
Gliomas are extremely aggressive brain tumors with a very poor prognosis. One of the more promising strategies for the treatment of human gliomas is targeted immunotherapy where antigens that are unique to the tumors are exploited to generate vaccines. The approach, however, is complicated by the fact that human gliomas escape immune surveillance by creating an immune suppressed microenvironment. In order to oppose the glioma imposed immune suppression, molecules and pathways involved in immune cell maturation, expansion, and migration are under intensive clinical investigation as adjuvant therapy. Toll-like receptors (TLRs) mediate many of these functions in immune cell types, and TLR agonists, thus, are currently primary candidate molecules to be used as important adjuvants in a variety of cancers. In animal models for glioma, TLR agonists have exhibited antitumor properties by facilitating antigen presentation and stimulating innate and adaptive immunity. In clinical trials, several TLR agonists have achieved survival benefit, and many more trials are recruiting or ongoing. However, a second complicating factor is that TLRs are also expressed on cancer cells where they can participate instead in a variety of tumor promoting activities including cell growth, proliferation, invasion, migration, and even stem cell maintenance. TLR agonists can, therefore, possibly play dual roles in tumor biology. Here, how TLRs and TLR agonists function in glioma biology and in anti-glioma therapies is summarized in an effort to provide a current picture of the sophisticated relationship of glioma with the immune system and the implications for immunotherapy.
Collapse
|
18
|
Vacchelli E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists for cancer therapy. Oncoimmunology 2013; 2:e25238. [PMID: 24083080 PMCID: PMC3782517 DOI: 10.4161/onci.25238] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 05/31/2013] [Indexed: 12/19/2022] Open
Abstract
Toll-like receptors (TLRs) have long been known for their ability to initiate innate immune responses upon exposure to conserved microbial components such as lipopolysaccharide (LPS) and double-stranded RNA. More recently, this family of pattern recognition receptors has been attributed a critical role in the elicitation of anticancer immune responses, raising interest in the development of immunochemotherapeutic regimens based on natural or synthetic TLR agonists. In spite of such an intense wave of preclinical and clinical investigation, only three TLR agonists are currently licensed by FDA for use in cancer patients: bacillus Calmette-Guérin (BCG), an attenuated strain of Mycobacterium bovis that operates as a mixed TLR2/TLR4 agonist; monophosphoryl lipid A (MPL), a derivative of Salmonella minnesota that functions as a potent agonist of TLR4; and imiquimod, a synthetic imidazoquinoline that activates TLR7. One year ago, in the August and September issues of OncoImmunology, we described the main biological features of TLRs and discussed the progress of clinical studies evaluating the safety and therapeutic potential of TLR agonists in cancer patients. Here, we summarize the latest developments in this exciting area of research, focusing on preclinical studies that have been published during the last 13 mo and clinical trials launched in the same period to investigate the antineoplastic activity of TLR agonists.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre; Paris, France
- INSERM, U848; Villejuif, France
| | | | - Catherine Sautès-Fridman
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris; Paris, France
| | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 15, Centre de Recherche des Cordeliers; Paris, France
- INSERM, U872; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Laurence Zitvogel
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre; Paris, France
- INSERM, U1015; Villejuif, France
| | - Guido Kroemer
- INSERM, U848; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Metabolomics and Cell Biology Platform; Institut Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Institut Gustave Roussy; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|