1
|
Figg J, Chen D, Falceto Font L, Flores C, Jin D. In vivo mouse models for adult brain tumors: Exploring tumorigenesis and advancing immunotherapy development. Neuro Oncol 2024; 26:1964-1980. [PMID: 38990913 PMCID: PMC11534310 DOI: 10.1093/neuonc/noae131] [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: 03/28/2024] [Indexed: 07/13/2024] Open
Abstract
Brain tumors, particularly glioblastoma (GBM), are devastating and challenging to treat, with a low 5-year survival rate of only 6.6%. Mouse models are established to understand tumorigenesis and develop new therapeutic strategies. Large-scale genomic studies have facilitated the identification of genetic alterations driving human brain tumor development and progression. Genetically engineered mouse models (GEMMs) with clinically relevant genetic alterations are widely used to investigate tumor origin. Additionally, syngeneic implantation models, utilizing cell lines derived from GEMMs or other sources, are popular for their consistent and relatively short latency period, addressing various brain cancer research questions. In recent years, the success of immunotherapy in specific cancer types has led to a surge in cancer immunology-related research which specifically necessitates the utilization of immunocompetent mouse models. In this review, we provide a comprehensive summary of GEMMs and syngeneic mouse models for adult brain tumors, emphasizing key features such as model origin, genetic alteration background, oncogenic mechanisms, and immune-related characteristics. Our review serves as a valuable resource for the brain tumor research community, aiding in the selection of appropriate models to study cancer immunology.
Collapse
Affiliation(s)
- John Figg
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Dongjiang Chen
- Division of Neuro-Oncology, Department of Neurological Surgery and Neurology, USC Keck Brain Tumor Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Laura Falceto Font
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Catherine Flores
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Dan Jin
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
2
|
Liu SJ, Zou C, Pak J, Morse A, Pang D, Casey-Clyde T, Borah AA, Wu D, Seo K, O'Loughlin T, Lim DA, Ozawa T, Berger MS, Kamber RA, Weiss WA, Raleigh DR, Gilbert LA. In vivo perturb-seq of cancer and microenvironment cells dissects oncologic drivers and radiotherapy responses in glioblastoma. Genome Biol 2024; 25:256. [PMID: 39375777 PMCID: PMC11457336 DOI: 10.1186/s13059-024-03404-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Genetic perturbation screens with single-cell readouts have enabled rich phenotyping of gene function and regulatory networks. These approaches have been challenging in vivo, especially in adult disease models such as cancer, which include mixtures of malignant and microenvironment cells. Glioblastoma (GBM) is a fatal cancer, and methods of systematically interrogating gene function and therapeutic targets in vivo, especially in combination with standard of care treatment such as radiotherapy, are lacking. RESULTS Here, we iteratively develop a multiplex in vivo perturb-seq CRISPRi platform for single-cell genetic screens in cancer and tumor microenvironment cells that leverages intracranial convection enhanced delivery of sgRNA libraries into mouse models of GBM. Our platform enables potent silencing of drivers of in vivo growth and tumor maintenance as well as genes that sensitize GBM to radiotherapy. We find radiotherapy rewires transcriptional responses to genetic perturbations in an in vivo-dependent manner, revealing heterogenous patterns of treatment sensitization or resistance in GBM. Furthermore, we demonstrate targeting of genes that function in the tumor microenvironment, enabling alterations of ligand-receptor interactions between immune and stromal cells following in vivo CRISPRi perturbations that can affect tumor cell phagocytosis. CONCLUSION In sum, we demonstrate the utility of multiplexed perturb-seq for in vivo single-cell dissection of adult cancer and normal tissue biology across multiple cell types in the context of therapeutic intervention, a platform with potential for broad application.
Collapse
Affiliation(s)
- S John Liu
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Arc Institute, Palo Alto, CA, 94304, USA
| | - Christopher Zou
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Joanna Pak
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Alexandra Morse
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Dillon Pang
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Timothy Casey-Clyde
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ashir A Borah
- Arc Institute, Palo Alto, CA, 94304, USA
- Biological and Medical Informatics Graduate Program, University of California San Francisco, San Francisco, CA, 94143, USA
| | - David Wu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kyounghee Seo
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Thomas O'Loughlin
- Department of Neuroscience, Icahn School of Medicine, Mount Sinai, New York, NY, 10029, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Tomoko Ozawa
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Roarke A Kamber
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - William A Weiss
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Departments of Pediatrics, Neurology, and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Luke A Gilbert
- Arc Institute, Palo Alto, CA, 94304, USA.
- Department of Urology, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
3
|
Yao L, Hatami M, Ma W, Skutella T. Vaccine-based immunotherapy and related preclinical models for glioma. Trends Mol Med 2024; 30:965-981. [PMID: 39013724 DOI: 10.1016/j.molmed.2024.06.009] [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: 02/19/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/18/2024]
Abstract
Glioma, the most common primary malignant tumor in the central nervous system (CNS), lacks effective treatments, and >60% of cases are glioblastoma (GBM), the most aggressive form. Despite advances in immunotherapy, GBM remains highly resistant. Approaches that target tumor antigens expedite the development of immunotherapies, including personalized tumor-specific vaccines, patient-specific target selection, dendritic cell (DC) vaccines, and chimeric antigen receptor (CAR) and T cell receptor (TCR) T cells. Recent studies show promising results in treating GBM and lower-grade glioma (LGG), fostering hope for future immunotherapy. This review discusses tumor vaccines against glioma, preclinical models in immunological research, and the role of CD4+ T cells in vaccine-induced antitumor immunity. We also summarize clinical approaches, challenges, and future research for creating more effective vaccines.
Collapse
Affiliation(s)
- Longping Yao
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Maryam Hatami
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
4
|
Guo Y, Lee H, Kim C, Park C, Yamamichi A, Chuntova P, Gallus M, Bernabeu MO, Okada H, Jo H, Arvanitis C. Ultrasound frequency-controlled microbubble dynamics in brain vessels regulate the enrichment of inflammatory pathways in the blood-brain barrier. Nat Commun 2024; 15:8021. [PMID: 39271721 PMCID: PMC11399249 DOI: 10.1038/s41467-024-52329-y] [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: 11/20/2023] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Microbubble-enhanced ultrasound provides a noninvasive physical method to locally overcome major obstacles to the accumulation of blood-borne therapeutics in the brain, posed by the blood-brain barrier (BBB). However, due to the highly nonlinear and coupled behavior of microbubble dynamics in brain vessels, the impact of microbubble resonant effects on BBB signaling and function remains undefined. Here, combined theoretical and prospective experimental investigations reveal that microbubble resonant effects in brain capillaries can control the enrichment of inflammatory pathways that are sensitive to wall shear stress and promote differential expression of a range of transcripts in the BBB, supporting the notion that microbubble dynamics exerted mechanical stress can be used to establish molecular, in addition to spatial, therapeutic windows to target brain diseases. Consistent with these findings, a robust increase in cytotoxic T-cell accumulation in brain tumors was observed, demonstrating the functional relevance and potential clinical significance of the observed immuno-mechano-biological responses.
Collapse
Affiliation(s)
- Yutong Guo
- Georgia Institute of Technology, Woodruff School of Mechanical Engineering, Atlanta, USA
- Stanford University, Department of Radiology, Stanford, USA
| | - Hohyun Lee
- Georgia Institute of Technology, Woodruff School of Mechanical Engineering, Atlanta, USA
| | - Chulyong Kim
- Georgia Institute of Technology, Woodruff School of Mechanical Engineering, Atlanta, USA
| | - Christian Park
- Georgia Institute of Technology and Emory University, Coulter Department of Biomedical Engineering, Atlanta, USA
| | - Akane Yamamichi
- University of California San Francisco, Department of Neurological Surgery, San Francisco, USA
| | - Pavlina Chuntova
- University of California San Francisco, Department of Neurological Surgery, San Francisco, USA
| | - Marco Gallus
- University of California San Francisco, Department of Neurological Surgery, San Francisco, USA
| | - Miguel O Bernabeu
- The University of Edinburgh, Centre for Medical Informatics, Usher Institute, Edinburgh, United Kingdom
- The University of Edinburgh, The Bayes Centre, Edinburgh, United Kingdom
| | - Hideho Okada
- University of California San Francisco, Department of Neurological Surgery, San Francisco, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, USA
| | - Hanjoong Jo
- Georgia Institute of Technology and Emory University, Coulter Department of Biomedical Engineering, Atlanta, USA
- Emory University, Department of Medicine, Atlanta, USA
| | - Costas Arvanitis
- Georgia Institute of Technology, Woodruff School of Mechanical Engineering, Atlanta, USA.
- Georgia Institute of Technology and Emory University, Coulter Department of Biomedical Engineering, Atlanta, USA.
| |
Collapse
|
5
|
Vandecandelaere G, Ramapriyan R, Gaffey M, Richardson LG, Steuart SJ, Tazhibi M, Kalaw A, Grewal EP, Sun J, Curry WT, Choi BD. Pre-Clinical Models for CAR T-Cell Therapy for Glioma. Cells 2024; 13:1480. [PMID: 39273050 PMCID: PMC11394304 DOI: 10.3390/cells13171480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Immunotherapy represents a transformative shift in cancer treatment. Among myriad immune-based approaches, chimeric antigen receptor (CAR) T-cell therapy has shown promising results in treating hematological malignancies. Despite aggressive treatment options, the prognosis for patients with malignant brain tumors remains poor. Research leveraging CAR T-cell therapy for brain tumors has surged in recent years. Pre-clinical models are crucial in evaluating the safety and efficacy of these therapies before they advance to clinical trials. However, current models recapitulate the human tumor environment to varying degrees. Novel in vitro and in vivo techniques offer the opportunity to validate CAR T-cell therapies but also have limitations. By evaluating the strengths and weaknesses of various pre-clinical glioma models, this review aims to provide a roadmap for the development and pre-clinical testing of CAR T-cell therapies for brain tumors.
Collapse
Affiliation(s)
- Gust Vandecandelaere
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
- Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Rishab Ramapriyan
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Matthew Gaffey
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Leland Geoffrey Richardson
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Samuel Jeffrey Steuart
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Masih Tazhibi
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Adrian Kalaw
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Eric P. Grewal
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Jing Sun
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - William T. Curry
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Bryan D. Choi
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| |
Collapse
|
6
|
Jones AB, Tuy K, Hawkins CC, Quinn CH, Saad J, Gary SE, Beierle EA, Ding L, Rochlin KM, Lamb LS, Hjelmeland AB. Temozolomide and the PARP Inhibitor Niraparib Enhance Expression of Natural Killer Group 2D Ligand ULBP1 and Gamma-Delta T Cell Cytotoxicity in Glioblastoma. Cancers (Basel) 2024; 16:2852. [PMID: 39199623 PMCID: PMC11352900 DOI: 10.3390/cancers16162852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/18/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Glioblastoma (GBM) is an immunologically cold tumor, but several immunotherapy-based strategies show promise, including the administration of ex vivo expanded and activated cytotoxic gamma delta T cells. Cytotoxicity is partially mediated through interactions with natural killer group 2D ligands (NKG2DL) on tumor cells. We sought to determine whether the addition of the blood-brain barrier penetrant PARP inhibitor niraparib to the standard of care DNA alkylator temozolomide (TMZ) could upregulate NKG2DL, thereby improving immune cell recognition. Changes in viability were consistent with prior publications as there was a growth inhibitory effect of the combination of TMZ and niraparib. However, decreases in viability did not always correlate with changes in NKG2DL mRNA. ULBP1/Mult-1 mRNA was increased with the combination therapy in comparison to either drug alone in two of the three cell types tested, even though viability was consistently decreased. mRNA expression correlated with protein levels and ULBP1/MULT-1 cell surface protein was significantly increased with TMZ and niraparib treatment in four of the five cell types tested. Gamma delta T cell-mediated cytotoxicity at a 10:1 effector-to-target ratio was significantly increased upon pretreatment of cells derived from a GBM PDX with TMZ and niraparib in comparison to the control or either drug alone. Together, these data demonstrate that the combination of PARP inhibition, DNA alkylation, and gamma delta T cell therapy has the potential for the treatment of GBM.
Collapse
Affiliation(s)
- Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Kaysaw Tuy
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Cyntanna C. Hawkins
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Colin H. Quinn
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Joelle Saad
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Sam E. Gary
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Elizabeth A. Beierle
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Lei Ding
- In8Bio, Inc., New York, NY 10118, USA; (L.D.); (K.M.R.); (L.S.L.)
| | - Kate M. Rochlin
- In8Bio, Inc., New York, NY 10118, USA; (L.D.); (K.M.R.); (L.S.L.)
| | - Lawrence S. Lamb
- In8Bio, Inc., New York, NY 10118, USA; (L.D.); (K.M.R.); (L.S.L.)
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| |
Collapse
|
7
|
Roncali L, Marionneau-Lambot S, Roy C, Eychenne R, Gouard S, Avril S, Chouin N, Riou J, Allard M, Rousseau A, Guérard F, Hindré F, Chérel M, Garcion E. Brain intratumoural astatine-211 radiotherapy targeting syndecan-1 leads to durable glioblastoma remission and immune memory in female mice. EBioMedicine 2024; 105:105202. [PMID: 38905749 PMCID: PMC11246004 DOI: 10.1016/j.ebiom.2024.105202] [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: 02/14/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Glioblastoma (GB), the most aggressive brain cancer, remains a critical clinical challenge due to its resistance to conventional treatments. Here, we introduce a locoregional targeted-α-therapy (TAT) with the rat monoclonal antibody 9E7.4 targeting murine syndecan-1 (SDC1) coupled to the α-emitter radionuclide astatine-211 (211At-9E7.4). METHODS We orthotopically transplanted 50,000 GL261 cells of murine GB into the right striatum of syngeneic female C57BL/6JRj mice using stereotaxis. After MRI validation of tumour presence at day 11, TAT was injected at the same coordinates. Biodistribution, efficacy, toxicity, local and systemic responses were assessed following application of this protocol. The 9E7.4 monoclonal antibody was labelled with iodine-125 (125I) for biodistribution and with astatine-211 (211At) for the other experiments. FINDINGS The 211At-9E7.4 TAT demonstrated robust efficacy in reducing orthotopic tumours and achieved improved survival rates in the C57BL/6JRj model, reaching up to 70% with a minimal activity of 100 kBq. Targeting SDC1 ensured the cerebral retention of 211At over an optimal time window, enabling low-activity administration with a minimal toxicity profile. Moreover, TAT substantially reduced the occurrence of secondary tumours and provided resistance to new tumour development after contralateral rechallenge, mediated through the activation of central and effector memory T cells. INTERPRETATION The locoregional 211At-9E7.4 TAT stands as one of the most efficient TAT across all preclinical GB models. This study validates SDC1 as a pertinent therapeutic target for GB and underscores 211At-9E7.4 TAT as a promising advancement to improve the treatment and quality of life for patients with GB. FUNDING This work was funded by the French National Agency for Research (ANR) "France 2030 Investment Plan" Labex Iron [ANR-11-LABX-18-01], The SIRIC ILIAD [INCa-DGOS-INSERM-18011], the French program "Infrastructure d'Avenir en Biologie-Santé" (France Life Imaging) [ANR-11-INBS-0006], the PIA3 of the ANR, integrated to the "France 2030 Investment Plan" [ANR-21-RHUS-0012], and support from Inviscan SAS (Strasbourg, France). It was also related to: the ANR under the frame of EuroNanoMed III (project GLIOSILK) [ANR-19-ENM3-0003-01]; the "Région Pays-de-la-Loire" under the frame of the Target'In project; the "Ligue Nationale contre le Cancer" and the "Comité Départemental de Maine-et-Loire de la Ligue contre le Cancer" (CD49) under the frame of the FusTarG project and the "Tumour targeting, imaging and radio-therapies network" of the "Cancéropôle Grand-Ouest" (France). This work was also funded by the Institut National de la Santé et de la Recherche Médicale (INSERM), the University of Nantes, and the University of Angers.
Collapse
Affiliation(s)
- Loris Roncali
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Séverine Marionneau-Lambot
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CHU Nantes, Nantes Université, Service de médecine nucléaire, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Charlotte Roy
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France
| | - Romain Eychenne
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; GIP ARRONAX, F-44160, Saint-Herblain, France
| | - Sébastien Gouard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Sylvie Avril
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France
| | - Nicolas Chouin
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; ONIRIS, F-44000, Nantes, France
| | - Jérémie Riou
- CHU Angers, Université d'Angers, F-49000, Angers, France
| | - Mathilde Allard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Audrey Rousseau
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; CHU Angers, Université d'Angers, F-49000, Angers, France
| | - François Guérard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - François Hindré
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France
| | - Michel Chérel
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; Institut de Cancérologie de l'Ouest, Service de médecine nucléaire, F-44160, Saint-Herblain, France.
| | - Emmanuel Garcion
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France; PACEM (Plateforme d'Analyse Cellulaire et Moléculaire), Université d'Angers, SFR 4208, F-49000, Angers, France.
| |
Collapse
|
8
|
Chellen T, Bausart M, Maus P, Vanvarenberg K, Limaye N, Préat V, Malfanti A. In situ administration of STING-activating hyaluronic acid conjugate primes anti-glioblastoma immune response. Mater Today Bio 2024; 26:101057. [PMID: 38660475 PMCID: PMC11040137 DOI: 10.1016/j.mtbio.2024.101057] [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: 02/07/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor, with a highly immunosuppressive tumor immune microenvironment (TIME). In this work, we investigated the use of the STimulator of INterferon Genes (STING) pathway as an effective means to remodel the GBM TIME through the recruitment of both innate and adaptive immune cell populations. Using hyaluronic acid (HA), we developed a novel polymer-drug conjugate of a non-nucleotide STING agonist (MSA2), called HA-MSA2 for the in situ treatment of GBM. In JAWSII cells, HA-MSA2 exerted a greater increase of STING signaling and upregulation of STING-related downstream cyto-/chemokines in immune cells than the free drug. HA-MSA2 also elicited cancer cell-intrinsic immunostimulatory gene expression and promoted immunogenic cell death of GBM cells. In the SB28 GBM model, local delivery of HA-MSA2 induced a delay in tumor growth and a significant extension of survival. The analysis of the TIME showed a profound shift in the GBM immune landscape after HA-MSA2 treatment, with higher infiltration by innate and adaptive immune cells including dendritic, natural killer (NK) and CD8 T cell populations. The therapeutic potential of this novel polymer conjugate warrants further investigation, particularly with other chemo-immunotherapeutics or cancer vaccines as a promising combinatorial therapeutic approach.
Collapse
Affiliation(s)
- Teenesha Chellen
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Pierre Maus
- UCLouvain, de Duve Institute, Genetics of Autoimmune Diseases and Cancer, Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Nisha Limaye
- UCLouvain, de Duve Institute, Genetics of Autoimmune Diseases and Cancer, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
9
|
Chakraborty A, Yang C, Kresak JL, Silver AJ, Feier D, Tian G, Andrews M, Sobanjo OO, Hodge ED, Engelbart MK, Huang J, Harrison JK, Sarkisian MR, Mitchell DA, Deleyrolle LP. KR158 Spheres Harboring Slow-Cycling Cells Recapitulate High-Grade Glioma Features in an Immunocompetent System. Cells 2024; 13:938. [PMID: 38891070 PMCID: PMC11171638 DOI: 10.3390/cells13110938] [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: 03/04/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) poses a significant challenge in clinical oncology due to its aggressive nature, heterogeneity, and resistance to therapies. Cancer stem cells (CSCs) play a critical role in GBM, particularly in treatment resistance and tumor relapse, emphasizing the need to comprehend the mechanisms regulating these cells. Also, their multifaceted contributions to the tumor microenvironment (TME) underline their significance, driven by their unique properties. This study aimed to characterize glioblastoma stem cells (GSCs), specifically slow-cycling cells (SCCs), in an immunocompetent murine GBM model to explore their similarities with their human counterparts. Using the KR158 mouse model, we confirmed that SCCs isolated from this model exhibited key traits and functional properties akin to human SCCs. KR158 murine SCCs, expanded in the gliomasphere assay, demonstrated sphere forming ability, self-renewing capacity, positive tumorigenicity, enhanced stemness and resistance to chemotherapy. Together, our findings validate the KR158 murine model as a framework to investigate GSCs and SCCs in GBM pathology, and explore specifically the SCC-immune system communications, understand their role in disease progression, and evaluate the effect of therapeutic strategies targeting these specific connections.
Collapse
Affiliation(s)
- Avirup Chakraborty
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Changlin Yang
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Jesse L. Kresak
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Aryeh J. Silver
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Diana Feier
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Guimei Tian
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA
| | - Michael Andrews
- College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Olusegun O. Sobanjo
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Ethan D. Hodge
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Mia K. Engelbart
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Jianping Huang
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Jeffrey K. Harrison
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32603, USA
| | - Matthew R. Sarkisian
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Duane A. Mitchell
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Loic P. Deleyrolle
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
10
|
De Leo A, Ugolini A, Yu X, Scirocchi F, Scocozza D, Peixoto B, Pace A, D'Angelo L, Liu JKC, Etame AB, Rughetti A, Nuti M, Santoro A, Vogelbaum MA, Conejo-Garcia JR, Rodriguez PC, Veglia F. Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma. Immunity 2024; 57:1105-1123.e8. [PMID: 38703775 PMCID: PMC11114377 DOI: 10.1016/j.immuni.2024.04.006] [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: 03/21/2023] [Revised: 12/08/2023] [Accepted: 04/09/2024] [Indexed: 05/06/2024]
Abstract
Immunosuppressive macrophages restrict anti-cancer immunity in glioblastoma (GBM). Here, we studied the contribution of microglia (MGs) and monocyte-derived macrophages (MDMs) to immunosuppression and mechanisms underlying their regulatory function. MDMs outnumbered MGs at late tumor stages and suppressed T cell activity. Molecular and functional analysis identified a population of glycolytic MDM expressing GLUT1 with potent immunosuppressive activity. GBM-derived factors promoted high glycolysis, lactate, and interleukin-10 (IL-10) production in MDMs. Inhibition of glycolysis or lactate production in MDMs impaired IL-10 expression and T cell suppression. Mechanistically, intracellular lactate-driven histone lactylation promoted IL-10 expression, which was required to suppress T cell activity. GLUT1 expression on MDMs was induced downstream of tumor-derived factors that activated the PERK-ATF4 axis. PERK deletion in MDM abrogated histone lactylation, led to the accumulation of intratumoral T cells and tumor growth delay, and, in combination with immunotherapy, blocked GBM progression. Thus, PERK-driven glucose metabolism promotes MDM immunosuppressive activity via histone lactylation.
Collapse
Affiliation(s)
- Alessandra De Leo
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Alessio Ugolini
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA; Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatic, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Fabio Scirocchi
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Delia Scocozza
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Barbara Peixoto
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Angelica Pace
- Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Luca D'Angelo
- Department of Human Neurosciences, Neurosurgery Division, "Sapienza" University, AOU Policlinico Umberto I, Rome, Italy
| | - James K C Liu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Arnold B Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Aurelia Rughetti
- Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Antonio Santoro
- Department of Human Neurosciences, Neurosurgery Division, "Sapienza" University, AOU Policlinico Umberto I, Rome, Italy
| | | | - Jose R Conejo-Garcia
- Department of Integrative immunobiology, Duke School of Medicine, Durham, NC, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Filippo Veglia
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Department of Neuro-Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Peshoff MM, Gupta P, Oberai S, Trivedi R, Katayama H, Chakrapani P, Dang M, Migliozzi S, Gumin J, Kadri DB, Lin JK, Milam NK, Maynard ME, Vaillant BD, Parker-Kerrigan B, Lang FF, Huse JT, Iavarone A, Wang L, Clise-Dwyer K, Bhat KP. Triggering receptor expressed on myeloid cells 2 (TREM2) regulates phagocytosis in glioblastoma. Neuro Oncol 2024; 26:826-839. [PMID: 38237157 PMCID: PMC11066944 DOI: 10.1093/neuonc/noad257] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Glioblastomas (GBMs) are central nervous system tumors that resist standard-of-care interventions and even immune checkpoint blockade. Myeloid cells in the tumor microenvironment can contribute to GBM progression; therefore, emerging immunotherapeutic approaches include reprogramming these cells to achieve desirable antitumor activity. Triggering receptor expressed on myeloid cells 2 (TREM2) is a myeloid signaling regulator that has been implicated in a variety of cancers and neurological diseases with contrasting functions, but its role in GBM immunopathology and progression is still under investigation. METHODS Our reverse translational investigations leveraged single-cell RNA sequencing and cytometry of human gliomas to characterize TREM2 expression across myeloid subpopulations. Using 2 distinct murine glioma models, we examined the role of Trem2 on tumor progression and immune modulation of myeloid cells. Furthermore, we designed a method of tracking phagocytosis of glioma cells in vivo and employed in vitro assays to mechanistically understand the influence of TREM2 signaling on tumor uptake. RESULTS We discovered that TREM2 expression does not correlate with immunosuppressive pathways, but rather showed strong a positive association with the canonical phagocytosis markers lysozyme (LYZ) and macrophage scavenger receptor (CD163) in gliomas. While Trem2 deficiency was found to be dispensable for gliomagenesis, Trem2+ myeloid cells display enhanced tumor uptake compared to Trem2- cells. Mechanistically, we demonstrate that TREM2 mediates phagocytosis via Syk signaling. CONCLUSIONS These results indicate that TREM2 is not associated with immunosuppression in gliomas. Instead, TREM2 is an important regulator of phagocytosis that may be exploited as a potential therapeutic strategy for brain tumors.
Collapse
Affiliation(s)
- Mekenzie M Peshoff
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Pravesh Gupta
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shivangi Oberai
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rakesh Trivedi
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hiroshi Katayama
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Prashanth Chakrapani
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Minghao Dang
- Department of Genomic Medicine, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Simona Migliozzi
- Department of Neurological Surgery, Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Joy Gumin
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Divya B Kadri
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jessica K Lin
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nancy K Milam
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark E Maynard
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| | - Brian D Vaillant
- Departments of Translational Molecular Pathology, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Brittany Parker-Kerrigan
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason T Huse
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Antonio Iavarone
- Department of Neurological Surgery, Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Linghua Wang
- Department of Genomic Medicine, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Karen Clise-Dwyer
- Department of Hematopoietic Biology & Malignancy, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Krishna P Bhat
- Department of Translational Molecular Pathology, Neurosurgery at the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
12
|
Ghosh S, Rothlin CV. TREM2 function in glioblastoma immune microenvironment: Can we distinguish reality from illusion? Neuro Oncol 2024; 26:840-842. [PMID: 38290471 PMCID: PMC11066908 DOI: 10.1093/neuonc/noae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Indexed: 02/01/2024] Open
Affiliation(s)
- Sourav Ghosh
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carla V Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Wang H, Medina R, Ye J, Zhang Y, Chakraborty S, Valenzuela A, Uher O, Hadrava Vanova K, Sun M, Sang X, Park DM, Zenka J, Gilbert MR, Pacak K, Zhuang Z. rWTC-MBTA Vaccine Induces Potent Adaptive Immune Responses Against Glioblastomas via Dynamic Activation of Dendritic Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308280. [PMID: 38298111 PMCID: PMC11005728 DOI: 10.1002/advs.202308280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Despite strides in immunotherapy, glioblastoma multiforme (GBM) remains challenging due to low inherent immunogenicity and suppressive tumor microenvironment. Converting "cold" GBMs to "hot" is crucial for immune activation and improved outcomes. This study comprehensively characterized a therapeutic vaccination strategy for preclinical GBM models. The vaccine consists of Mannan-BAM-anchored irradiated whole tumor cells, Toll-like receptor ligands [lipoteichoic acid (LTA), polyinosinic-polycytidylic acid (Poly (I:C)), and resiquimod (R-848)], and anti-CD40 agonistic antibody (rWTC-MBTA). Intracranial GBM models (GL261, SB28 cells) are used to evaluate the vaccine efficacy. A substantial number of vaccinated mice exhibited complete regression of GBM tumors in a T-cell-dependent manner, with no significant toxicity. Long-term tumor-specific immune memory is confirmed upon tumor rechallenge. In the vaccine-draining lymph nodes of the SB28 model, rWTC-MBTA vaccination triggered a major rise in conventional dendritic cell type 1 (cDC1) 12 h post-treatment, followed by an increase in conventional dendritic cell type 2 (cDC2), monocyte-derived dendritic cell (moDC), and plasmacytoid dendritic cell (pDC) on Day 5 and Day 13. Enhanced cytotoxicity of CD4+ and CD8+ T cells in vaccinated mice is verified in co-culture with tumor cells. Analyses of immunosuppressive signals (T-cell exhaustion, myeloid-derived suppressor cells (MDSC), M2 macrophages) in the GBM microenvironment suggest potential combinations with other immunotherapies for enhanced efficacy. In conclusion, the authors findings demonstrate that rWTC-MBTA induces potent and long-term adaptive immune responses against GBM.
Collapse
Affiliation(s)
- Herui Wang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Staff Scientist Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100437 Convent Dr.BethesdaMD20892USA
| | - Rogelio Medina
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Juan Ye
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Yaping Zhang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | | | - Alex Valenzuela
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Ondrej Uher
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Katerina Hadrava Vanova
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Mitchell Sun
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Xueyu Sang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Deric M. Park
- John Theurer Cancer CenterHUMCHackensack Meridian School of Medicine92 2nd StHackensackNJ07601USA
| | - Jan Zenka
- Department of Medical BiologyFaculty of ScienceUniversity of South BohemiaČeské Budějovice37005Czech Republic
| | - Mark R. Gilbert
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Zhengping Zhuang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Senior Investigator Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100037 Convent DrBethesdaMD20892USA
| |
Collapse
|
14
|
Ghasemi A, Martinez-Usatorre A, Li L, Hicham M, Guichard A, Marcone R, Fournier N, Torchia B, Martinez Bedoya D, Davanture S, Fernández-Vaquero M, Fan C, Janzen J, Mohammadzadeh Y, Genolet R, Mansouri N, Wenes M, Migliorini D, Heikenwalder M, De Palma M. Cytokine-armed dendritic cell progenitors for antigen-agnostic cancer immunotherapy. NATURE CANCER 2024; 5:240-261. [PMID: 37996514 PMCID: PMC10899110 DOI: 10.1038/s43018-023-00668-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 10/11/2023] [Indexed: 11/25/2023]
Abstract
Dendritic cells (DCs) are antigen-presenting myeloid cells that regulate T cell activation, trafficking and function. Monocyte-derived DCs pulsed with tumor antigens have been tested extensively for therapeutic vaccination in cancer, with mixed clinical results. Here, we present a cell-therapy platform based on mouse or human DC progenitors (DCPs) engineered to produce two immunostimulatory cytokines, IL-12 and FLT3L. Cytokine-armed DCPs differentiated into conventional type-I DCs (cDC1) and suppressed tumor growth, including melanoma and autochthonous liver models, without the need for antigen loading or myeloablative host conditioning. Tumor response involved synergy between IL-12 and FLT3L and was associated with natural killer and T cell infiltration and activation, M1-like macrophage programming and ischemic tumor necrosis. Antitumor immunity was dependent on endogenous cDC1 expansion and interferon-γ signaling but did not require CD8+ T cell cytotoxicity. Cytokine-armed DCPs synergized effectively with anti-GD2 chimeric-antigen receptor (CAR) T cells in eradicating intracranial gliomas in mice, illustrating their potential in combination therapies.
Collapse
Affiliation(s)
- Ali Ghasemi
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Amaia Martinez-Usatorre
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Luqing Li
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Mehdi Hicham
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Alan Guichard
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Rachel Marcone
- Agora Cancer Research Center, Lausanne, Switzerland
- Translational Data Science (TDS) Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Center, Lausanne, Switzerland
- Translational Data Science (TDS) Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Bruno Torchia
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Darel Martinez Bedoya
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Suzel Davanture
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mirian Fernández-Vaquero
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chaofan Fan
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Janzen
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yahya Mohammadzadeh
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Raphael Genolet
- Ludwig Institute for Cancer Research, Lausanne, Switzerland
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Nahal Mansouri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Mathias Wenes
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Denis Migliorini
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Oncology, Geneva University Hospital (HUG), Geneva, Switzerland
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- The M3 Research Center, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180), Eberhard Karls University, Tübingen, Germany
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
| |
Collapse
|
15
|
Chakraborty A, Yang C, Kresak JL, Silver A, Feier D, Tian G, Andrews M, Sobanjo OO, Hodge ED, Engelbart MK, Huang J, Harrison JK, Sarkisian MR, Mitchell DA, Deleyrolle LP. KR158 spheres harboring slow-cycling cells recapitulate GBM features in an immunocompetent system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577279. [PMID: 38501121 PMCID: PMC10945590 DOI: 10.1101/2024.01.26.577279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Glioblastoma (GBM) poses a significant challenge in clinical oncology due to its aggressive nature, heterogeneity, and resistance to therapies. Cancer stem cells (CSCs) play a critical role in GBM, particularly in treatment-resistance and tumor relapse, emphasizing the need to comprehend the mechanisms regulating these cells. Also, their multifaceted contributions to the tumor-microenvironment (TME) underline their significance, driven by their unique properties. This study aimed to characterize glioblastoma stem cells (GSCs), specifically slow-cycling cells (SCCs), in an immunocompetent murine GBM model to explore their similarities with their human counterparts. Using the KR158 mouse model, we confirmed that SCCs isolated from this model exhibited key traits and functional properties akin to human SCCs. KR158 murine SCCs, expanded in the gliomasphere assay, demonstrated sphere forming ability, self-renewing capacity, positive tumorigenicity, enhanced stemness and resistance to chemotherapy. Together, our findings validate the KR158 murine model as a framework to investigate GSCs and SCCs in GBM-pathology, and explore specifically the SCC-immune system communications, understand their role in disease progression, and evaluate the effect of therapeutic strategies targeting these specific connections.
Collapse
|
16
|
Wang Q, Wang Z. Serpin family H member 1 and its related collagen gene network are the potential prognostic biomarkers and anticancer targets for glioma. J Biochem Mol Toxicol 2024; 38:e23541. [PMID: 37712121 DOI: 10.1002/jbt.23541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/02/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023]
Abstract
Serpin family H member 1 (SERPINH1) is responsible for encoding the protein known as heat shock protein 47, which functions as a molecular chaperone specific to collagen (COL). This protein has been identified as a potential therapeutic target for COL-related disorders. In this study, we aimed to investigate the role of SERPINH1 in the tumorigenicity of gliomas. To achieve this, we utilized various bioinformatics tools to analyze gene expression, overall survival, protein-protein interactions, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and Gene Set Enrichment Analysis (GSEA). Based on The Cancer Genome Atlas database revealed that SERPINH1 and four COL family members (COL1A1, COL3A1, COL4A1, and COL4A2) expression are significantly upregulated in glioma tissues compared with normal nontumor tissues. GO, KEGG, and GSEA analyses exhibited that SERPINH1 is implicated in the establishment and degradation of COL-containing extracellular matrix (ECM), focal adhesion, and ECM-receptor interaction in glioma. SERPINH1 is an independent prognostic factor, exhibiting a positive association with the augmentation of neutrophils and macrophages, as well as the manifestation of immune checkpoint molecules within glioma. Experimental assessments conducted both in vitro and in vivo demonstrated that the suppression of SERPINH1 impeded the migratory, invasive, and proliferative capacities of glioma cells, while concurrently fostering cellular apoptosis. Consequently, SERPINH1 emerges as an oncogenic gene and an independent prognostic marker for glioma, potentially facilitating the advancement of immunotherapeutic interventions for the treatment of glioma.
Collapse
Affiliation(s)
- Qi Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhe Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
17
|
Yadav N, Purow BW. Understanding current experimental models of glioblastoma-brain microenvironment interactions. J Neurooncol 2024; 166:213-229. [PMID: 38180686 PMCID: PMC11056965 DOI: 10.1007/s11060-023-04536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
Glioblastoma (GBM) is a common and devastating primary brain tumor, with median survival of 16-18 months after diagnosis in the setting of substantial resistance to standard-of-care and inevitable tumor recurrence. Recent work has implicated the brain microenvironment as being critical for GBM proliferation, invasion, and resistance to treatment. GBM does not operate in isolation, with neurons, astrocytes, and multiple immune populations being implicated in GBM tumor progression and invasiveness. The goal of this review article is to provide an overview of the available in vitro, ex vivo, and in vivo experimental models for assessing GBM-brain interactions, as well as discuss each model's relative strengths and limitations. Current in vitro models discussed will include 2D and 3D co-culture platforms with various cells of the brain microenvironment, as well as spheroids, whole organoids, and models of fluid dynamics, such as interstitial flow. An overview of in vitro and ex vivo organotypic GBM brain slices is also provided. Finally, we conclude with a discussion of the various in vivo rodent models of GBM, including xenografts, syngeneic grafts, and genetically-engineered models of GBM.
Collapse
Affiliation(s)
- Niket Yadav
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Benjamin W Purow
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA.
| |
Collapse
|
18
|
Slika H, Karimov Z, Alimonti P, Abou-Mrad T, De Fazio E, Alomari S, Tyler B. Preclinical Models and Technologies in Glioblastoma Research: Evolution, Current State, and Future Avenues. Int J Mol Sci 2023; 24:16316. [PMID: 38003507 PMCID: PMC10671665 DOI: 10.3390/ijms242216316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma is the most common malignant primary central nervous system tumor and one of the most debilitating cancers. The prognosis of patients with glioblastoma remains poor, and the management of this tumor, both in its primary and recurrent forms, remains suboptimal. Despite the tremendous efforts that are being put forward by the research community to discover novel efficacious therapeutic agents and modalities, no major paradigm shifts have been established in the field in the last decade. However, this does not mirror the abundance of relevant findings and discoveries made in preclinical glioblastoma research. Hence, developing and utilizing appropriate preclinical models that faithfully recapitulate the characteristics and behavior of human glioblastoma is of utmost importance. Herein, we offer a holistic picture of the evolution of preclinical models of glioblastoma. We further elaborate on the commonly used in vitro and vivo models, delving into their development, favorable characteristics, shortcomings, and areas of potential improvement, which aids researchers in designing future experiments and utilizing the most suitable models. Additionally, this review explores progress in the fields of humanized and immunotolerant mouse models, genetically engineered animal models, 3D in vitro models, and microfluidics and highlights promising avenues for the future of preclinical glioblastoma research.
Collapse
Affiliation(s)
- Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Ziya Karimov
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
- Faculty of Medicine, Ege University, 35100 Izmir, Turkey
| | - Paolo Alimonti
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Tatiana Abou-Mrad
- Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon;
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emerson De Fazio
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| |
Collapse
|
19
|
Bartos LM, Kirchleitner SV, Kolabas ZI, Quach S, Beck A, Lorenz J, Blobner J, Mueller SA, Ulukaya S, Hoeher L, Horvath I, Wind-Mark K, Holzgreve A, Ruf VC, Gold L, Kunze LH, Kunte ST, Beumers P, Park HE, Antons M, Zatcepin A, Briel N, Hoermann L, Schaefer R, Messerer D, Bartenstein P, Riemenschneider MJ, Lindner S, Ziegler S, Herms J, Lichtenthaler SF, Ertürk A, Tonn JC, von Baumgarten L, Albert NL, Brendel M. Deciphering sources of PET signals in the tumor microenvironment of glioblastoma at cellular resolution. SCIENCE ADVANCES 2023; 9:eadi8986. [PMID: 37889970 PMCID: PMC10610915 DOI: 10.1126/sciadv.adi8986] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
Various cellular sources hamper interpretation of positron emission tomography (PET) biomarkers in the tumor microenvironment (TME). We developed an approach of immunomagnetic cell sorting after in vivo radiotracer injection (scRadiotracing) with three-dimensional (3D) histology to dissect the cellular allocation of PET signals in the TME. In mice with implanted glioblastoma, translocator protein (TSPO) radiotracer uptake per tumor cell was higher compared to tumor-associated microglia/macrophages (TAMs), validated by protein levels. Translation of in vitro scRadiotracing to patients with glioma immediately after tumor resection confirmed higher single-cell TSPO tracer uptake of tumor cells compared to immune cells. Across species, cellular radiotracer uptake explained the heterogeneity of individual TSPO-PET signals. In consideration of cellular tracer uptake and cell type abundance, tumor cells were the main contributor to TSPO enrichment in glioblastoma; however, proteomics identified potential PET targets highly specific for TAMs. Combining cellular tracer uptake measures with 3D histology facilitates precise allocation of PET signals and serves to validate emerging novel TAM-specific radioligands.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Alexander Beck
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Julia Lorenz
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Jens Blobner
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stephan A. Mueller
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- School of Computation, Information and Technology (CIT), TUM, Boltzmannstr. 3, 85748 Garching, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lukas Gold
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Philipp Beumers
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Melissa Antons
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Nils Briel
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Rebecca Schaefer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Denise Messerer
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F. Lichtenthaler
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
20
|
Zhang W, Zhang K, Ma Y, Song Y, Qi T, Xiong G, Zhang Y, Kan C, Zhang J, Han F, Sun X. Secreted frizzled-related proteins: A promising therapeutic target for cancer therapy through Wnt signaling inhibition. Biomed Pharmacother 2023; 166:115344. [PMID: 37634472 DOI: 10.1016/j.biopha.2023.115344] [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: 06/19/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023] Open
Abstract
The Wnt signaling system is a critical pathway that regulates embryonic development and adult homeostasis. Secreted frizzled-related proteins (SFRPs) are extracellular inhibitors of Wnt signaling that act by binding directly to Wnt ligands or Frizzled receptors. SFRPs can act as anti-Wnt agents and suppress cancer growth by blocking the action of Wnt ligands. However, SFRPs are often silenced by promoter methylation in cancer cells, resulting in hyperactivation of the Wnt pathway. Epigenetic modifiers can reverse this silencing and restore SFRPs expression. Despite the potential of SFRPs as a therapeutic target, the effects of SFRPs on tumor development remain unclear. Therefore, a review of the expression of various members of the SFRPs family in different cancers and their potential as therapeutic targets is warranted. This review aims to summarize the current knowledge of SFRPs in cancer, focusing on their expression patterns and their potential as novel therapeutic targets.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Yanhui Ma
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Guoji Xiong
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Yuanzhu Zhang
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China.
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, China.
| |
Collapse
|
21
|
Fan P, Xia J, Zhou M, Zhuo C, He H. Development and validation of a personalized classifier to predict the prognosis and response to immunotherapy in glioma based on glycolysis and the tumor microenvironment. PeerJ 2023; 11:e16066. [PMID: 37744243 PMCID: PMC10516100 DOI: 10.7717/peerj.16066] [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: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Background Glycolysis is closely associated with cancer progression and treatment outcomes. However, the role of glycolysis in the immune microenvironment, prognosis, and immunotherapy of glioma remains unclear. Methods This study investigated the role of glycolysis on prognosis and its relationship with the tumor microenvironment (TME). Subsequently, we developed and validated the glycolysis-related gene signature (GRS)-TME classifier using multiple independent cohorts. Furthermore, we also examined the prognostic value, somatic alterations, molecular characteristics, and potential benefits of immunotherapy based on GRS-TME classifier. Lastly, the effect of kinesin family member 20A (KIF20A) on the proliferation and migration of glioma cells was evaluated in vitro. Results Glycolysis was identified as a significant prognostic risk factor in glioma, and closely associated with an immunosuppressive microenvironment characterized by altered distribution of immune cells. Furthermore, a personalized GRS-TME classifier was developed and validated by combining the glycolysis (18 genes) and TME (seven immune cells) scores. Patients in the GRSlow/TMEhigh subgroup exhibited a more favorable prognosis compared to other subgroups. Distinct genomic alterations and signaling pathways were observed among different subgroups, which are closely associated with cell cycle, epithelial-mesenchymal transition, p53 signaling pathway, and interferon-alpha response. Additionally, we found that patients in the GRSlow/TMEhigh subgroup exhibit a higher response rate to immunotherapy, and the GRS-TME classifier can serve as a novel biomarker for predicting immunotherapy outcomes. Finally, high expression of KIF20A is associated with an unfavorable prognosis in glioma, and its knockdown can inhibit the proliferation and migration of glioma cells. Conclusions Our study developed a GRS-TME classifier for predicting the prognosis and potential benefits of immunotherapy in glioma patients. Additionally, we identified KIF20A as a prognostic and therapeutic biomarker for glioma.
Collapse
Affiliation(s)
- Pengfei Fan
- Department of Neurology, Changxing People’s Hospital, Huzhou, Zhejiang, China
| | - Jinjin Xia
- Department of Neurology, Changxing People’s Hospital, Huzhou, Zhejiang, China
| | - Meifeng Zhou
- Department of Neurology, Changxing People’s Hospital, Huzhou, Zhejiang, China
| | - Chao Zhuo
- Department of Pediatrics, Changxing People’s Hospital, Huzhou, Zhejiang, China
| | - Hui He
- Department of Pediatrics, Changxing People’s Hospital, Huzhou, Zhejiang, China
| |
Collapse
|
22
|
Chen D, Varanasi SK, Hara T, Traina K, Sun M, McDonald B, Farsakoglu Y, Clanton J, Xu S, Garcia-Rivera L, Mann TH, Du V, Chung HK, Xu Z, Tripple V, Casillas E, Ma S, O'Connor C, Yang Q, Zheng Y, Hunter T, Lemke G, Kaech SM. CTLA-4 blockade induces a microglia-Th1 cell partnership that stimulates microglia phagocytosis and anti-tumor function in glioblastoma. Immunity 2023; 56:2086-2104.e8. [PMID: 37572655 DOI: 10.1016/j.immuni.2023.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/14/2023] [Accepted: 07/20/2023] [Indexed: 08/14/2023]
Abstract
The limited efficacy of immunotherapies against glioblastoma underscores the urgency of better understanding immunity in the central nervous system. We found that treatment with αCTLA-4, but not αPD-1, prolonged survival in a mouse model of mesenchymal-like glioblastoma. This effect was lost upon the depletion of CD4+ T cells but not CD8+ T cells. αCTLA-4 treatment increased frequencies of intratumoral IFNγ-producing CD4+ T cells, and IFNγ blockade negated the therapeutic impact of αCTLA-4. The anti-tumor activity of CD4+ T cells did not require tumor-intrinsic MHC-II expression but rather required conventional dendritic cells as well as MHC-II expression on microglia. CD4+ T cells interacted directly with microglia, promoting IFNγ-dependent microglia activation and phagocytosis via the AXL/MER tyrosine kinase receptors, which were necessary for tumor suppression. Thus, αCTLA-4 blockade in mesenchymal-like glioblastoma promotes a CD4+ T cell-microglia circuit wherein IFNγ triggers microglia activation and phagocytosis and microglia in turn act as antigen-presenting cells fueling the CD4+ T cell response.
Collapse
Affiliation(s)
- Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Toshiro Hara
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kacie Traina
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ming Sun
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Biomedicine, University of Basel, Basel 4058, Switzerland
| | - Josh Clanton
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Lizmarie Garcia-Rivera
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Thomas H Mann
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Victor Du
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - H Kay Chung
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ziyan Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; School of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Eduardo Casillas
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Shixin Ma
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Carolyn O'Connor
- Flow Cytometry Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Qiyuan Yang
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ye Zheng
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Greg Lemke
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
Davy M, Genest L, Legrand C, Pelouin O, Froget G, Castagné V, Rupp T. Evaluation of Temozolomide and Fingolimod Treatments in Glioblastoma Preclinical Models. Cancers (Basel) 2023; 15:4478. [PMID: 37760448 PMCID: PMC10527257 DOI: 10.3390/cancers15184478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastomas are malignant brain tumors which remain lethal due to their aggressive and invasive nature. The standard treatment combines surgical resection, radiotherapy, and chemotherapy using Temozolomide, albeit with a minor impact on patient prognosis (15 months median survival). New therapies evaluated in preclinical translational models are therefore still required to improve patient survival and quality of life. In this preclinical study, we evaluated the effect of Temozolomide in different models of glioblastoma. We also aimed to investigate the efficacy of Fingolimod, an immunomodulatory drug for multiple sclerosis also described as an inhibitor of the sphingosine-1-phosphate (S1P)/S1P receptor axis. The effects of Fingolimod and Temozolomide were analyzed with in vitro 2D and 3D cellular assay and in vivo models using mouse and human glioblastoma cells implanted in immunocompetent or immunodeficient mice, respectively. We demonstrated both in in vitro and in vivo models that Temozolomide has a varied effect depending on the tumor type (i.e., U87MG, U118MG, U138MG, and GL261), demonstrating sensitivity, acquired resistance, and purely resistant tumor phenotypes, as observed in patients. Conversely, Fingolimod only reduced in vitro 2D tumor cell growth and increased cytotoxicity. Indeed, Fingolimod had little or no effect on 3D spheroid cytotoxicity and was devoid of effect on in vivo tumor progression in Temozolomide-sensitive models. These results suggest that the efficacy of Fingolimod is dependent on the glioblastoma tumor microenvironment. Globally, our data suggest that the response to Temozolomide varies depending on the cancer model, consistent with its clinical activity, whereas the potential activity of Fingolimod may merit further evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tristan Rupp
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France
| |
Collapse
|
24
|
Lee J, Nicosia M, Hong ES, Silver DJ, Li C, Bayik D, Watson DC, Lauko A, Kay KE, Wang SZ, Johnson S, McGraw M, Grabowski MM, Kish DD, Desai AB, Goodman WA, Cameron SJ, Okada H, Valujskikh A, Fairchild RL, Ahluwalia MS, Lathia JD. Sex-Biased T-cell Exhaustion Drives Differential Immune Responses in Glioblastoma. Cancer Discov 2023; 13:2090-2105. [PMID: 37378557 PMCID: PMC10481130 DOI: 10.1158/2159-8290.cd-22-0869] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 05/14/2023] [Accepted: 06/23/2023] [Indexed: 06/29/2023]
Abstract
Sex differences in glioblastoma (GBM) incidence and outcome are well recognized, and emerging evidence suggests that these extend to genetic/epigenetic and cellular differences, including immune responses. However, the mechanisms driving immunologic sex differences are not fully understood. Here, we demonstrate that T cells play a critical role in driving GBM sex differences. Male mice exhibited accelerated tumor growth, with decreased frequency and increased exhaustion of CD8+ T cells in the tumor. Furthermore, a higher frequency of progenitor exhausted T cells was found in males, with improved responsiveness to anti-PD-1 treatment. Moreover, increased T-cell exhaustion was observed in male GBM patients. Bone marrow chimera and adoptive transfer models indicated that T cell-mediated tumor control was predominantly regulated in a cell-intrinsic manner, partially mediated by the X chromosome inactivation escape gene Kdm6a. These findings demonstrate that sex-biased predetermined behavior of T cells is critical for inducing sex differences in GBM progression and immunotherapy response. SIGNIFICANCE Immunotherapies in patients with GBM have been unsuccessful due to a variety of factors, including the highly immunosuppressive tumor microenvironment in GBM. This study demonstrates that sex-biased T-cell behaviors are predominantly intrinsically regulated, further suggesting sex-specific approaches can be leveraged to potentially improve the therapeutic efficacy of immunotherapy in GBM. See related commentary by Alspach, p. 1966. This article is featured in Selected Articles from This Issue, p. 1949.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ellen S. Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Medical Scientist Training Program, Department of Medicine, Case Western Reserve University, Cleveland Ohio
| | - Daniel J. Silver
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Cathy Li
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Defne Bayik
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Dionysios C. Watson
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Hematology/Oncology Division, Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Adam Lauko
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Medical Scientist Training Program, Department of Medicine, Case Western Reserve University, Cleveland Ohio
| | - Kristen E. Kay
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Sabrina Z. Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Medical Scientist Training Program, Department of Medicine, Case Western Reserve University, Cleveland Ohio
| | - Sadie Johnson
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mary McGraw
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, Ohio
| | | | - Danielle D. Kish
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amar B. Desai
- Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Wendy A. Goodman
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Scott J. Cameron
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Hideho Okada
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
25
|
Zheng M, Wang Y, Fu F, Zhang K, Wang Y, Zhao S, Liu Q, Mu H, Zhang X, Miao L. Radioimmunotherapy Targeting B7-H3 in situ glioma models enhanced antitumor efficacy by Reconstructing the tumor microenvironment. Int J Biol Sci 2023; 19:4278-4290. [PMID: 37705739 PMCID: PMC10496502 DOI: 10.7150/ijbs.87763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/01/2023] [Indexed: 09/15/2023] Open
Abstract
Radionuclide drug conjugates (RDCs) with antibodies serve as a novel approach for the treatment of malignant tumors including glioblastoma. However, RDCs require optimal antibodies to work efficiently. Hu4G4, a novel B7-H3-targeting humanized monoclonal IgG1 antibody, is highly specific for the human B7-H3 protein (a marker of tumor cells, including glioblastoma cells). Herein, we established 131I-labeled hu4G4 (131I-hu4G4) and showed that it specifically bound to B7-H3 with high affinity (Kd = 0.99 ± 0.07 nM) and inhibited the growth of U87 cells in vitro. 131I-hu4G4 displayed potent in situ antitumor activity in a mouse model of glioma based on GL261 Red-Fluc-B7-H3 cells. More importantly, 131I-hu4G4 remodeled the tumor microenvironment and promoted the transformation of glioma from "cold" to "hot" tumors by promoting CD4+ and CD8+ T cell infiltration and the polarization of M2 to M1. Therefore, the antitumor activity observed with 131I-hu4G4, together with its ability to enhance antitumor immune responses, makes it a novel candidate for radioimmunotherapy of glioblastoma.
Collapse
Affiliation(s)
- Meng Zheng
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| | - Yan Wang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, SZ, China
| | - Fengqing Fu
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, SZ, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
| | - Kaijie Zhang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| | - Yanan Wang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| | - Shandong Zhao
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| | - Qingfeng Liu
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| | - Huiwen Mu
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| | - Xueguang Zhang
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, SZ, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- SuZhou Bright Scistar Antibody Biotech co., Ltd, 303-305, Bldg 15, NO.8, Jinfeng Road, Suzhou, SZ, China
| | - Liyan Miao
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, SZ, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, SZ, China
| |
Collapse
|
26
|
Nejo T, Krishna S, Jimenez C, Yamamichi A, Young JS, Lakshmanachetty S, Chen T, Phyu SSS, Ogino H, Watchmaker P, Diebold D, Choudhury A, Daniel AGS, Raleigh DR, Hervey-Jumper SL, Okada H. Glioma-neuronal circuit remodeling induces regional immunosuppression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.548295. [PMID: 37577659 PMCID: PMC10418167 DOI: 10.1101/2023.08.04.548295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Neuronal activity-driven mechanisms impact glioblastoma cell proliferation and invasion 1-7 , and glioblastoma remodels neuronal circuits 8,9 . Distinct intratumoral regions maintain functional connectivity via a subpopulation of malignant cells that mediate tumor-intrinsic neuronal connectivity and synaptogenesis through their transcriptional programs 8 . However, the effects of tumor-intrinsic neuronal activity on other cells, such as immune cells, remain unknown. Here we show that regions within glioblastomas with elevated connectivity are characterized by regional immunosuppression. This was accompanied by different cell compositions and inflammatory status of tumor-associated macrophages (TAMs) in the tumor microenvironment. In preclinical intracerebral syngeneic glioblastoma models, CRISPR/Cas9 gene knockout of Thrombospondin-1 (TSP-1/ Thbs1 ), a synaptogenic factor critical for glioma-induced neuronal circuit remodeling, in glioblastoma cells suppressed synaptogenesis and glutamatergic neuronal hyperexcitability, while simultaneously restoring antigen-presentation and pro-inflammatory responses. Moreover, TSP-1 knockout prolonged survival of immunocompetent mice harboring intracerebral syngeneic glioblastoma, but not of immunocompromised mice, and promoted infiltrations of pro-inflammatory TAMs and CD8+ T-cells in the tumor microenvironment. Notably, pharmacological inhibition of glutamatergic excitatory signals redirected tumor-associated macrophages toward a less immunosuppressive phenotype, resulting in prolonged survival. Altogether, our results demonstrate previously unrecognized immunosuppression mechanisms resulting from glioma-neuronal circuit remodeling and suggest future strategies targeting glioma-neuron-immune crosstalk may open up new avenues for immunotherapy.
Collapse
|
27
|
Chen CH, Chin RL, Hartley GP, Lea ST, Engel BJ, Hsieh CE, Prasad R, Roszik J, Shingu T, Lizee GA, Heimberger AB, Millward SW, Hu J, Hong DS, Curran MA. Novel murine glioblastoma models that reflect the immunotherapy resistance profile of a human disease. Neuro Oncol 2023; 25:1415-1427. [PMID: 36705543 PMCID: PMC10398813 DOI: 10.1093/neuonc/noad025] [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: 03/27/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The lack of murine glioblastoma models that mimic the immunobiology of human disease has impeded basic and translational immunology research. We, therefore, developed murine glioblastoma stem cell lines derived from Nestin-CreERT2QkL/L; Trp53L/L; PtenL/L (QPP) mice driven by clinically relevant genetic mutations common in human glioblastoma. This study aims to determine the immune sensitivities of these QPP lines in immunocompetent hosts and their underlying mechanisms. METHODS The differential responsiveness of QPP lines was assessed in the brain and flank in untreated, anti-PD-1, or anti-CTLA-4 treated mice. The impact of genomic landscape on the responsiveness of each tumor was measured through whole exome sequencing. The immune microenvironments of sensitive (QPP7) versus resistant (QPP8) lines were compared in the brain using flow cytometry. Drivers of flank sensitivity versus brain resistance were also measured for QPP8. RESULTS QPP lines are syngeneic to C57BL/6J mice and demonstrate varied sensitivities to T cell immune checkpoint blockade ranging from curative responses to complete resistance. Infiltrating tumor immune analysis of QPP8 reveals improved T cell fitness and augmented effector-to-suppressor ratios when implanted subcutaneously (sensitive), which are absent on implantation in the brain (resistant). Upregulation of PD-L1 across the myeloid stroma acts to establish this state of immune privilege in the brain. In contrast, QPP7 responds to checkpoint immunotherapy even in the brain likely resulting from its elevated neoantigen burden. CONCLUSIONS These syngeneic QPP models of glioblastoma demonstrate clinically relevant profiles of immunotherapeutic sensitivity and potential utility for both mechanistic discovery and evaluation of immune therapies.
Collapse
Affiliation(s)
- Chao-Hsien Chen
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Neurology, Houston Methodist Neurological Institute, Houston, Texas 77030, USA
| | - Renee L Chin
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Genevieve P Hartley
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Spencer T Lea
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Brian J Engel
- Departement of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Cheng-En Hsieh
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Rishika Prasad
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jason Roszik
- Departement of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| | - Takashi Shingu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Gregory A Lizee
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Departement of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Steven W Millward
- Departement of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Michael A Curran
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
28
|
Mondal I, Das O, Sun R, Gao J, Yu B, Diaz A, Behnan J, Dubey A, Meng Z, Eskandar E, Xu B, Lu RO, Ho WS. PP2Ac Deficiency Enhances Tumor Immunogenicity by Activating STING-Type I Interferon Signaling in Glioblastoma. Cancer Res 2023; 83:2527-2542. [PMID: 37219874 PMCID: PMC10525036 DOI: 10.1158/0008-5472.can-22-3382] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/31/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
Glioblastoma (GBM) is an immunologically "cold" tumor that does not respond to current immunotherapy. Here, we demonstrate a fundamental role for the α-isoform of the catalytic subunit of protein phosphatase-2A (PP2Ac) in regulating glioma immunogenicity. Genetic ablation of PP2Ac in glioma cells enhanced double-stranded DNA (dsDNA) production and cGAS-type I IFN signaling, MHC-I expression, and tumor mutational burden. In coculture experiments, PP2Ac deficiency in glioma cells promoted dendritic cell (DC) cross-presentation and clonal expansion of CD8+ T cells. In vivo, PP2Ac depletion sensitized tumors to immune-checkpoint blockade and radiotherapy treatment. Single-cell analysis demonstrated that PP2Ac deficiency increased CD8+ T-cell, natural killer cell, and DC accumulation and reduced immunosuppressive tumor-associated macrophages. Furthermore, loss of PP2Ac increased IFN signaling in myeloid and tumor cells and reduced expression of a tumor gene signature associated with worse patient survival in The Cancer Genome Atlas. Collectively, this study establishes a novel role for PP2Ac in inhibiting dsDNA-cGAS-STING signaling to suppress antitumor immunity in glioma. SIGNIFICANCE PP2Ac deficiency promotes cGAS-STING signaling in glioma to induce a tumor-suppressive immune microenvironment, highlighting PP2Ac as a potential therapeutic target to enhance tumor immunogenicity and improve response to immunotherapy.
Collapse
Affiliation(s)
- Isha Mondal
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Oishika Das
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Raymond Sun
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Jian Gao
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Bohyeon Yu
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Aaron Diaz
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Jinan Behnan
- The Leo M. Davidoff Department of Neurological Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abhishek Dubey
- The Leo M. Davidoff Department of Neurological Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zhipeng Meng
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Emad Eskandar
- The Leo M. Davidoff Department of Neurological Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rongze Olivia Lu
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Winson S. Ho
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
29
|
Sun F, Lv H, Feng B, Sun J, Zhang L, Dong B. Identification of natural killer cell-related characteristics to predict the clinical prognosis and immune microenvironment of patients with low-grade glioma. Aging (Albany NY) 2023; 15:6264-6291. [PMID: 37405952 PMCID: PMC10373982 DOI: 10.18632/aging.204850] [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: 03/08/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Individuals with low-grade glioma (LGG) have a dismal prognosis, and most patients will eventually progress to high-grade disease. Therefore, it is crucial to accurately determine their prognoses. METHODS Seventy-nine NK cell genes were downloaded from the LM22 database and univariate Cox regression analysis was utilized to detect NK cell-related genes affecting prognosis. Molecular types were established for LGG using the "ConsensusClusterPlus" R package. The results from a functional enrichment analysis and the immune microenvironment were intensively explored to determine molecular heterogeneity and immune characteristics across distinct subtypes. Furthermore, a RiskScore model was developed and verified using expression profiles of NK cells, and a nomogram consisting of the RiskScore model and clinical traits was constructed. Moreover, pan-cancer traits of NK cells were also investigated. RESULTS The C1 subtype included the greatest amount of immune infiltration and the poorest prognosis among well-established subtypes. The majority of enriched pathways were those involved in tumor progression, including epithelial-mesenchymal transition and cell cycle pathways. Differentially expressed genes among distinct subtypes were determined and used to develop a novel RiskScore model. This model was able to distinguish low-risk patients with LGG from those with high-risk disease. An accurate nomogram including the RiskScore, disease grade and patient's age was constructed to predict clinical outcomes of LGG patients. Finally, a pan-cancer analysis further highlighted the crucial roles of NK cell-related genes in the tumor microenvironment. CONCLUSIONS An NK cell-related RiskScore model can accurately predict the prognoses of patients with LGG and provide valuable insights into personalized medicine.
Collapse
Affiliation(s)
- Fei Sun
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Neurosurgery, Xinhua Hospital Affiliated to Dalian University, Dalian, Liaoning, China
| | - Hongtao Lv
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Baozhi Feng
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Linyun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bin Dong
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
30
|
Nisnboym M, Vincze SR, Xiong Z, Sneiderman CT, Raphael RA, Li B, Jaswal AP, Sever RE, Day KE, LaToche JD, Foley LM, Karimi H, Hitchens TK, Agnihotri S, Hu B, Rajasundaram D, Anderson CJ, Blumenthal DT, Pearce TM, Uttam S, Nedrow JR, Panigrahy A, Pollack IF, Lieberman FS, Drappatz J, Raphael I, Edwards WB, Kohanbash G. Immuno-PET Imaging of CD69 Visualizes T-Cell Activation and Predicts Survival Following Immunotherapy in Murine Glioblastoma. CANCER RESEARCH COMMUNICATIONS 2023; 3:1173-1188. [PMID: 37426447 PMCID: PMC10324623 DOI: 10.1158/2767-9764.crc-22-0434] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023]
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Immunotherapy may be promising for the treatment of some patients with GBM; however, there is a need for noninvasive neuroimaging techniques to predict immunotherapeutic responses. The effectiveness of most immunotherapeutic strategies requires T-cell activation. Therefore, we aimed to evaluate an early marker of T-cell activation, CD69, for its use as an imaging biomarker of response to immunotherapy for GBM. Herein, we performed CD69 immunostaining on human and mouse T cells following in vitro activation and post immune checkpoint inhibitors (ICI) in an orthotopic syngeneic mouse glioma model. CD69 expression on tumor-infiltrating leukocytes was assessed using single-cell RNA sequencing (scRNA-seq) data from patients with recurrent GBM receiving ICI. Radiolabeled CD69 Ab PET/CT imaging (CD69 immuno-PET) was performed on GBM-bearing mice longitudinally to quantify CD69 and its association with survival following immunotherapy. We show CD69 expression is upregulated upon T-cell activation and on tumor-infiltrating lymphocytes (TIL) in response to immunotherapy. Similarly, scRNA-seq data demonstrated elevated CD69 on TILs from patients with ICI-treated recurrent GBM as compared with TILs from control cohorts. CD69 immuno-PET studies showed a significantly higher tracer uptake in the tumors of ICI-treated mice compared with controls. Importantly, we observed a positive correlation between survival and CD69 immuno-PET signals in immunotherapy-treated animals and established a trajectory of T-cell activation by virtue of CD69-immuno-PET measurements. Our study supports the potential use of CD69 immuno-PET as an immunotherapy response assessment imaging tool for patients with GBM. Significance Immunotherapy may hold promise for the treatment of some patients with GBM. There is a need to assess therapy responsiveness to allow the continuation of effective treatment in responders and to avoid ineffective treatment with potential adverse effects in the nonresponders. We demonstrate that noninvasive PET/CT imaging of CD69 may allow early detection of immunotherapy responsiveness in patients with GBM.
Collapse
Affiliation(s)
- Michal Nisnboym
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Sarah R. Vincze
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zujian Xiong
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Chaim T. Sneiderman
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rebecca A. Raphael
- Department of Computational and Systems Biology, UPMC Hillman Cancer Center, Cancer Biology Program, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bo Li
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ambika P. Jaswal
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - ReidAnn E. Sever
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kathryn E. Day
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Joseph D. LaToche
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Lesley M. Foley
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Hanieh Karimi
- Department of Biochemistry, University of Missouri, Columbia, Missouri
| | - T. Kevin Hitchens
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sameer Agnihotri
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Baoli Hu
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dhivyaa Rajasundaram
- Division of Health Informatics, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Deborah T. Blumenthal
- Neuro-oncology Division, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Thomas M. Pearce
- Division of Neuropathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shikhar Uttam
- Department of Computational and Systems Biology, UPMC Hillman Cancer Center, Cancer Biology Program, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessie R. Nedrow
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Ashok Panigrahy
- Department of Radiology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ian F. Pollack
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Frank S. Lieberman
- Neuro-oncology Program, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Jan Drappatz
- Neuro-oncology Program, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Itay Raphael
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wilson B. Edwards
- Department of Biochemistry, University of Missouri, Columbia, Missouri
| | - Gary Kohanbash
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Ma L, Gonzalez-Junca A, Chou W, Barcellos-Hoff MH. Monitoring TGFβ signaling in irradiated tumors. Methods Cell Biol 2023; 180:49-67. [PMID: 37890932 DOI: 10.1016/bs.mcb.2023.02.009] [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] [Indexed: 10/29/2023]
Abstract
Transforming growth factor β (TGFβ) is exquisitely regulated under physiological conditions but its activity is highly dysregulated in cancer. All cells make TGFβ and have receptors for the ligand, which is sequestered in the extracellular matrix in a latent form. Ionizing radiation elicits rapid release of TGFβ from these stores, so-called activation, over a wide range of doses and exposures, including low dose (<1Gy) whole-body irradiation, creating an extraordinarily potent signal in the irradiated tissue or tumor. Hence, accurate evaluation of TGFβ activity is complicated because of its ubiquitous distribution as a latent complex. Here we describe conditions for assays that reveal TGFβ activity in situ using either tissue preparations or functional imaging.
Collapse
Affiliation(s)
- Lin Ma
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Alba Gonzalez-Junca
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - William Chou
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
32
|
Geels SN, Moshensky A, Sousa RS, Walker BL, Singh R, Gutierrez G, Hwang M, Mempel TR, Nie Q, Othy S, Marangoni F. Interruption of the Intratumor CD8:Treg Crosstalk Improves the Efficacy of PD-1 Immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540889. [PMID: 37292782 PMCID: PMC10245792 DOI: 10.1101/2023.05.15.540889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PD-1 blockade unleashes the potent antitumor activity of CD8 cells but can also promote immunosuppressive T regulatory (Treg) cells, which may worsen response to immunotherapy. Tumor Treg inhibition is a promising strategy to overcome therapeutic resistance; however, the mechanisms supporting tumor Tregs during PD-1 immunotherapy are largely unexplored. Here, we report that PD-1 blockade increases tumor Tregs in mouse models of immunogenic tumors, including melanoma, and metastatic melanoma patients. Unexpectedly, Treg accumulation was not caused by Treg-intrinsic inhibition of PD-1 signaling but instead depended on an indirect effect of activated CD8 cells. CD8 cells colocalized with Tregs within tumors and produced IL-2, especially after PD-1 immunotherapy. IL-2 upregulated the anti-apoptotic protein ICOS on tumor Tregs, causing their accumulation. ICOS signaling inhibition before PD-1 immunotherapy resulted in increased control of immunogenic melanoma. Thus, interrupting the intratumor CD8:Treg crosstalk is a novel strategy that may enhance the efficacy of immunotherapy in patients.
Collapse
|
33
|
Eng J, Bucher E, Hu Z, Sanders M, Chakravarthy B, Gonzalez P, Pietenpol JA, Gibbs SL, Sears RC, Chin K. Robust biomarker discovery through multiplatform multiplex image analysis of breast cancer clinical cohorts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.525753. [PMID: 36778343 PMCID: PMC9915596 DOI: 10.1101/2023.01.31.525753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Spatial profiling of tissues promises to elucidate tumor-microenvironment interactions and enable development of spatial biomarkers to predict patient response to immunotherapy and other therapeutics. However, spatial biomarker discovery is often carried out on a single patient cohort or imaging technology, limiting statistical power and increasing the likelihood of technical artifacts. In order to analyze multiple patient cohorts profiled on different platforms, we developed methods for comparative data analysis from three disparate multiplex imaging technologies: 1) cyclic immunofluorescence data we generated from 102 breast cancer patients with clinical follow-up, in addition to publicly available 2) imaging mass cytometry and 3) multiplex ion-beam imaging data. We demonstrate similar single-cell phenotyping results across breast cancer patient cohorts imaged with these three technologies and identify cellular abundance and proximity-based biomarkers with prognostic value across platforms. In multiple platforms, we identified lymphocyte infiltration as independently associated with longer survival in triple negative and high-proliferation breast tumors. Then, a comparison of nine spatial analysis methods revealed robust spatial biomarkers. In estrogen receptor-positive disease, quiescent stromal cells close to tumor were more abundant in good prognosis tumors while tumor neighborhoods of mixed fibroblast phenotypes were enriched in poor prognosis tumors. In triple-negative breast cancer (TNBC), macrophage proximity to tumor and B cell proximity to T cells were greater in good prognosis tumors, while tumor neighborhoods of vimentin-positive fibroblasts were enriched in poor prognosis tumors. We also tested previously published spatial biomarkers in our ensemble cohort, reproducing the positive prognostic value of isolated lymphocytes and lymphocyte occupancy and failing to reproduce the prognostic value of tumor-immune mixing score in TNBC. In conclusion, we demonstrate assembly of larger clinical cohorts from diverse platforms to aid in prognostic spatial biomarker identification and validation.
Collapse
Affiliation(s)
- Jennifer Eng
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Zhi Hu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Melinda Sanders
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Bapsi Chakravarthy
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
| | - Paula Gonzalez
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
| | - Jennifer A. Pietenpol
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Summer L. Gibbs
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Koei Chin
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, 97239, USA
| |
Collapse
|
34
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [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: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
McCornack C, Woodiwiss T, Hardi A, Yano H, Kim AH. The function of histone methylation and acetylation regulators in GBM pathophysiology. Front Oncol 2023; 13:1144184. [PMID: 37205197 PMCID: PMC10185819 DOI: 10.3389/fonc.2023.1144184] [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: 01/13/2023] [Accepted: 03/29/2023] [Indexed: 05/21/2023] Open
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain malignancy and is characterized by a high degree of intra and intertumor cellular heterogeneity, a starkly immunosuppressive tumor microenvironment, and nearly universal recurrence. The application of various genomic approaches has allowed us to understand the core molecular signatures, transcriptional states, and DNA methylation patterns that define GBM. Histone posttranslational modifications (PTMs) have been shown to influence oncogenesis in a variety of malignancies, including other forms of glioma, yet comparatively less effort has been placed on understanding the transcriptional impact and regulation of histone PTMs in the context of GBM. In this review we discuss work that investigates the role of histone acetylating and methylating enzymes in GBM pathogenesis, as well as the effects of targeted inhibition of these enzymes. We then synthesize broader genomic and epigenomic approaches to understand the influence of histone PTMs on chromatin architecture and transcription within GBM and finally, explore the limitations of current research in this field before proposing future directions for this area of research.
Collapse
Affiliation(s)
- Colin McCornack
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, United States
| | - Timothy Woodiwiss
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa, IA, United States
| | - Angela Hardi
- Bernard Becker Medical Library, Washington University School of Medicine, St. Louis, MO, United States
| | - Hiroko Yano
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Albert H. Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
36
|
van Hooren L, Handgraaf SM, Kloosterman DJ, Karimi E, van Mil LWHG, Gassama AA, Solsona BG, de Groot MHP, Brandsma D, Quail DF, Walsh LA, Borst GR, Akkari L. CD103 + regulatory T cells underlie resistance to radio-immunotherapy and impair CD8 + T cell activation in glioblastoma. NATURE CANCER 2023; 4:665-681. [PMID: 37081259 DOI: 10.1038/s43018-023-00547-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 03/20/2023] [Indexed: 04/22/2023]
Abstract
Glioblastomas are aggressive primary brain tumors with an inherent resistance to T cell-centric immunotherapy due to their low mutational burden and immunosuppressive tumor microenvironment. Here we report that fractionated radiotherapy of preclinical glioblastoma models induce a tenfold increase in T cell content. Orthogonally, spatial imaging mass cytometry shows T cell enrichment in human recurrent tumors compared with matched primary glioblastoma. In glioblastoma-bearing mice, α-PD-1 treatment applied at the peak of T cell infiltration post-radiotherapy results in a modest survival benefit compared with concurrent α-PD-1 administration. Following α-PD-1 therapy, CD103+ regulatory T cells (Tregs) with upregulated lipid metabolism accumulate in the tumor microenvironment, and restrain immune checkpoint blockade response by repressing CD8+ T cell activation. Treg targeting elicits tertiary lymphoid structure formation, enhances CD4+ and CD8+ T cell frequency and function and unleashes radio-immunotherapeutic efficacy. These results support the rational design of therapeutic regimens limiting the induction of immunosuppressive feedback pathways in the context of T cell immunotherapy in glioblastoma.
Collapse
Affiliation(s)
- Luuk van Hooren
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Shanna M Handgraaf
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daan J Kloosterman
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Lotte W H G van Mil
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Awa A Gassama
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Beatriz Gomez Solsona
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marnix H P de Groot
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Gerben R Borst
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- Department of Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK.
| | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
37
|
Ho WS, Mondal I, Xu B, Das O, Sun R, Chiou P, Cai X, Tahmasebinia F, McFadden E, Wu CYJ, Wu Z, Matsui W, Lim M, Meng Z, Lu RO. PP2Ac/STRN4 negatively regulates STING-type I IFN signaling in tumor-associated macrophages. J Clin Invest 2023; 133:e162139. [PMID: 36757811 PMCID: PMC10014107 DOI: 10.1172/jci162139] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Stimulator of IFN genes type I (STING-Type I) IFN signaling in myeloid cells plays a critical role in effective antitumor immune responses, but STING agonists as monotherapy have shown limited efficacy in clinical trials. The mechanisms that downregulate STING signaling are not fully understood. Here, we report that protein phosphatase 2A (PP2A), with its specific B regulatory subunit Striatin 4 (STRN4), negatively regulated STING-Type I IFN in macrophages. Mice with macrophage PP2A deficiency exhibited reduced tumor progression. The tumor microenvironment showed decreased immunosuppressive and increased IFN-activated macrophages and CD8+ T cells. Mechanistically, we demonstrated that Hippo kinase MST1/2 was required for STING activation. STING agonists induced dissociation of PP2A from MST1/2 in normal macrophages, but not in tumor conditioned macrophages. Furthermore, our data showed that STRN4 mediated PP2A binding to and dephosphorylation of Hippo kinase MST1/2, resulting in stabilization of YAP/TAZ to antagonize STING activation. In human patients with glioblastoma (GBM), YAP/TAZ was highly expressed in tumor-associated macrophages but not in nontumor macrophages. We also demonstrated that PP2A/STRN4 deficiency in macrophages reduced YAP/TAZ expression and sensitized tumor-conditioned macrophages to STING stimulation. In summary, we demonstrated that PP2A/STRN4-YAP/TAZ has, in our opinion, been an unappreciated mechanism that mediates immunosuppression in tumor-associated macrophages, and targeting the PP2A/STRN4-YAP/TAZ axis can sensitize tumors to immunotherapy.
Collapse
Affiliation(s)
- Winson S. Ho
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Isha Mondal
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Oishika Das
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Raymond Sun
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Pochin Chiou
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Xiaomin Cai
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Foozhan Tahmasebinia
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| | - Elizabeth McFadden
- Department of Molecular Sciences, University of Texas at Austin, Austin, Texas, USA
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Stanford University, Stanford, California, USA
| | - Zhihao Wu
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| | - William Matsui
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Stanford, California, USA
| | - Zhipeng Meng
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rongze Olivia Lu
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
- Helen Diller Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| |
Collapse
|
38
|
Frederico SC, Zhang X, Hu B, Kohanbash G. Pre-clinical models for evaluating glioma targeted immunotherapies. Front Immunol 2023; 13:1092399. [PMID: 36700223 PMCID: PMC9870312 DOI: 10.3389/fimmu.2022.1092399] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Gliomas have an extremely poor prognosis in both adult and pediatric patient populations as these tumors are known to grow aggressively and respond poorly to standard of care treatment. Currently, treatment for gliomas involves surgical resection followed by chemoradiation therapy. However, some gliomas, such as diffuse midline glioma, have more limited treatment options such as radiotherapy alone. Even with these interventions, the prognosis for those diagnosed with a glioma remains poor. Immunotherapy is highly effective for some cancers and there is great interest in the development of effective immunotherapies for the treatment of gliomas. Clinical trials evaluating the efficacy of immunotherapies targeted to gliomas have largely failed to date, and we believe this is partially due to the poor choice in pre-clinical mouse models that are used to evaluate these immunotherapies. A key consideration in evaluating new immunotherapies is the selection of pre-clinical models that mimic the glioma-immune response in humans. Multiple pre-clinical options are currently available, each one with their own benefits and limitations. Informed selection of pre-clinical models for testing can facilitate translation of more promising immunotherapies in the clinical setting. In this review we plan to present glioma cell lines and mouse models, as well as alternatives to mouse models, that are available for pre-clinical glioma immunotherapy studies. We plan to discuss considerations of model selection that should be made for future studies as we hope this review can serve as a guide for investigators as they choose which model is best suited for their study.
Collapse
Affiliation(s)
- Stephen C. Frederico
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xiaoran Zhang
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Gary Kohanbash,
| |
Collapse
|
39
|
The Role of Cellular Immunity and Adaptive Immunity in Pathophysiology of Brain and Spinal Cord Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1394:51-72. [PMID: 36587381 DOI: 10.1007/978-3-031-14732-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Major advances have been made in our understanding of CNS tumors, especially glioma, however, the survival of patients with malignant glioma remains poor. While radiation and chemotherapy have increased overall survival, glioblastoma multiforme (GBM) still has one of the worst 5-year survival rates of all human cancers. Here, in this chapter, the authors review the abrogation of the immune system in the tumor setting, revealing many plausible targets for therapy and the current immunotherapy treatment strategies employed. Notably, glioma has also been characterized as a subset of primary spinal cord tumor and current treatment recommendations are outlined here.
Collapse
|
40
|
Bayik D, Bartels CF, Lovrenert K, Watson DC, Zhang D, Kay K, Lee J, Lauko A, Johnson S, Lo A, Silver DJ, McGraw M, Grabowski M, Mohammadi AM, Veglia F, Fan Y, Vogelbaum MA, Scacheri P, Lathia JD. Distinct Cell Adhesion Signature Defines Glioblastoma Myeloid-Derived Suppressor Cell Subsets. Cancer Res 2022; 82:4274-4287. [PMID: 36126163 PMCID: PMC9664137 DOI: 10.1158/0008-5472.can-21-3840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/27/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023]
Abstract
In multiple types of cancer, an increased frequency in myeloid-derived suppressor cells (MDSC) is associated with worse outcomes and poor therapeutic response. In the glioblastoma (GBM) microenvironment, monocytic (m) MDSCs represent the predominant subset. However, the molecular basis of mMDSC enrichment in the tumor microenvironment compared with granulocytic (g) MDSCs has yet to be determined. Here we performed the first broad epigenetic profiling of MDSC subsets to define underlying cell-intrinsic differences in behavior and found that enhanced gene accessibility of cell adhesion programs in mMDSCs is linked to their tumor-accelerating ability in GBM models upon adoptive transfer. Mouse and human mMDSCs expressed higher levels of integrin β1 and dipeptidyl peptidase-4 (DPP-4) compared with gMDSCs as part of an enhanced cell adhesion signature. Integrin β1 blockade abrogated the tumor-promoting phenotype of mMDSCs and altered the immune profile in the tumor microenvironment, whereas treatment with a DPP-4 inhibitor extended survival in preclinical GBM models. Targeting DPP-4 in mMDSCs reduced pERK signaling and their migration towards tumor cells. These findings uncover a fundamental difference in the molecular basis of MDSC subsets and suggest that integrin β1 and DPP-4 represent putative immunotherapy targets to attenuate myeloid cell-driven immune suppression in GBM. SIGNIFICANCE Epigenetic profiling uncovers cell adhesion programming as a regulator of the tumor-promoting functions of monocytic myeloid-derived suppressor cells in glioblastoma, identifying therapeutic targets that modulate the immune response and suppress tumor growth.
Collapse
Affiliation(s)
- Defne Bayik
- Lerner Research Institute, Cleveland Clinic, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Cynthia F. Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Katreya Lovrenert
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Dionysios C. Watson
- Lerner Research Institute, Cleveland Clinic, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kristen Kay
- Lerner Research Institute, Cleveland Clinic, Ohio
| | - Juyeun Lee
- Lerner Research Institute, Cleveland Clinic, Ohio
| | - Adam Lauko
- Lerner Research Institute, Cleveland Clinic, Ohio
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
- Case Western Reserve University, Medical Science Training Program, Cleveland, Ohio
| | | | - Alice Lo
- Lerner Research Institute, Cleveland Clinic, Ohio
| | - Daniel J. Silver
- Lerner Research Institute, Cleveland Clinic, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Mary McGraw
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Ohio
| | | | | | - Filippo Veglia
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Peter Scacheri
- Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Justin D. Lathia
- Lerner Research Institute, Cleveland Clinic, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Ohio
| |
Collapse
|
41
|
Fathi M, Razavi SM, Sojoodi M, Ahmadi A, Ebrahimi F, Namdar A, Hojjat-Farsangi M, Gholamin S, Jadidi-Niaragh F. Targeting the CTLA-4/B7 axes in glioblastoma: preclinical evidence and clinical interventions. Expert Opin Ther Targets 2022; 26:949-961. [PMID: 36527817 DOI: 10.1080/14728222.2022.2160703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Glioblastoma Multiforme (GBM) is one of the fatal cancers of the Central Nervous System (CNS). A variety of reasons exist for why previous immunotherapy strategies, especially Immune Checkpoint Blockers (ICBs), did not work in treating GBM patients. The cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a key immune checkpoint receptor. Its overexpression in cancer and immune cells causes tumor cell progression. CTLA-4 suppresses anti-tumor responses inside the GBM tumor-immune microenvironment. AREAS COVERED It has been attempted to explain the immunobiology of CTLA-4 as well as its interaction with different immune cells and cancer cells that lead to GBM progression. Additionally, CTLA-4 targeting studies have been reviewed and CTLA-4 combination therapy, as a promising therapeutic target and strategy for GBM immunotherapy, is recommended. EXPERT OPINION CTLA-4 could be a possible supplement for future cancer immunotherapies of GBM. However, many challenges remain such as the high toxicity of CTLA-4 blockers, and the unresponsiveness of most patients to immunotherapy. For the future clinical success of CTLA-4 blocker therapy, combination approaches with other targeted treatments would be a potentially effective strategy. Going forward, predictive biomarkers can be used to reduce trial timelines and increase the chance of success.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed-Mostafa Razavi
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mozhdeh Sojoodi
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Armin Ahmadi
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, AL, USA
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Afshin Namdar
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Sharareh Gholamin
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Bartos LM, Kirchleitner SV, Blobner J, Wind K, Kunze LH, Holzgreve A, Gold L, Zatcepin A, Kolabas ZI, Ulukaya S, Weidner L, Quach S, Messerer D, Bartenstein P, Tonn JC, Riemenschneider MJ, Ziegler S, von Baumgarten L, Albert NL, Brendel M. 18 kDa translocator protein positron emission tomography facilitates early and robust tumor detection in the immunocompetent SB28 glioblastoma mouse model. Front Med (Lausanne) 2022; 9:992993. [PMID: 36325388 PMCID: PMC9621314 DOI: 10.3389/fmed.2022.992993] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/02/2022] [Indexed: 10/29/2023] Open
Abstract
Introduction The 18 kDa translocator protein (TSPO) receives growing interest as a biomarker in glioblastoma. Mouse models can serve as an important tool for the investigation of biomarkers in glioblastoma, but several glioblastoma models indicated only low TSPO-PET signals in contrast to high TSPO-PET signals of human glioblastoma. Thus, we aimed to investigate TSPO-PET imaging in the syngeneic immunocompetent SB28 mouse model, which is thought to closely represent the tumor microenvironment (TME) of human glioblastoma. Methods Dynamic TSPO-PET/CT imaging was performed for 60 min after injection of 13.6 ± 4.2 MBq [18F]GE-180. Contrast enhanced CT (ceCT) was acquired prior to PET and served for assessment of tumor volumes and attenuation correction. SB28 and sham mice were imaged at an early (week-1; n = 6 SB28, n = 6 sham) and a late time-point (week-3; n = 8 SB28, n = 9 sham) after inoculation. Standard of truth ex vivo tumor volumes were obtained for SB28 mice at the late time-point. Tracer kinetics were analyzed for the lesion site and the carotid arteries to establish an image derived input function (IDIF). TSPO-PET and ceCT lesion volumes were compared with ex vivo volumes by calculation of root-mean-square-errors (RMSE). Volumes of distribution (VTmax/mean) in the lesion were calculated using carotid IDIF and standardized uptake values (SUVmax/mean) were obtained for a 40-60 min time frame. Results Higher uptake rate constants (K1) were observed for week-1 SB28 tumor lesions when compared to week-3 SB28 tumor lesions. Highest agreement between TSPO-PET lesion volumes and ex vivo tumor volumes was achieved with a 50% maximum threshold (RMSE-VT: 39.7%; RMSE-SUV: 34.4%), similar to the agreement of ceCT tumor volumes (RMSE: 30.1%). Lesions of SB28 mice had higher PET signal when compared to sham mice at week-1 (VTmax 6.6 ± 2.9 vs. 3.9 ± 0.8, p = 0.035; SUVmax 2.3 ± 0.5 vs. 1.2 ± 0.1, p < 0.001) and PET signals remained at a similar level at week-3 (VTmax 5.0 ± 1.6 vs. 2.7 ± 0.8, p = 0.029; SUVmax 1.9 ± 0.5 vs. 1.2 ± 0.2, p = 0.0012). VTmax correlated with SUVmax (R 2 = 0.532, p < 0.001). Conclusion TSPO-PET imaging of immunocompetent SB28 mice facilitates early detection of tumor signals over sham lesions. SB28 tumors mirror high TSPO-PET signals of human glioblastoma and could serve as a valuable translational model to study TSPO as an imaging biomarker.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Jens Blobner
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lukas Gold
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Zeynep Ilgin Kolabas
- Helmholtz Center, Institute for Tissue Engineering and Regenerative Medicine (iTERM), Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig- Maximilians University Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Selin Ulukaya
- Helmholtz Center, Institute for Tissue Engineering and Regenerative Medicine (iTERM), Munich, Germany
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Lorraine Weidner
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Denise Messerer
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
| |
Collapse
|
43
|
Karami Fath M, Babakhaniyan K, Anjomrooz M, Jalalifar M, Alizadeh SD, Pourghasem Z, Abbasi Oshagh P, Azargoonjahromi A, Almasi F, Manzoor HZ, Khalesi B, Pourzardosht N, Khalili S, Payandeh Z. Recent Advances in Glioma Cancer Treatment: Conventional and Epigenetic Realms. Vaccines (Basel) 2022; 10:1448. [PMID: 36146527 PMCID: PMC9501259 DOI: 10.3390/vaccines10091448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most typical and aggressive form of primary brain tumor in adults, with a poor prognosis. Successful glioma treatment is hampered by ineffective medication distribution across the blood-brain barrier (BBB) and the emergence of drug resistance. Although a few FDA-approved multimodal treatments are available for glioblastoma, most patients still have poor prognoses. Targeting epigenetic variables, immunotherapy, gene therapy, and different vaccine- and peptide-based treatments are some innovative approaches to improve anti-glioma treatment efficacy. Following the identification of lymphatics in the central nervous system, immunotherapy offers a potential method with the potency to permeate the blood-brain barrier. This review will discuss the rationale, tactics, benefits, and drawbacks of current glioma therapy options in clinical and preclinical investigations.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Kimiya Babakhaniyan
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran 1996713883, Iran
| | - Mehran Anjomrooz
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1411713135, Iran
| | | | | | - Zeinab Pourghasem
- Department of Microbiology, Islamic Azad University of Lahijan, Gilan 4416939515, Iran
| | - Parisa Abbasi Oshagh
- Department of Biology, Faculty of Basic Sciences, Malayer University, Malayer 6571995863, Iran
| | - Ali Azargoonjahromi
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz 7417773539, Iran
| | - Faezeh Almasi
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 1411734115, Iran
| | - Hafza Zahira Manzoor
- Experimental and Translational Medicine, University of Insubria, Via jean Henry Dunant 3, 21100 Varese, Italy
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
| | - Navid Pourzardosht
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht 4193713111, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran
| | - Zahra Payandeh
- Department of Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, SE-17177 Stockholm, Sweden
| |
Collapse
|
44
|
Marx S, Godicelj A, Wucherpfennig KW. A Conceptual Framework for Inducing T Cell-Mediated Immunity Against Glioblastoma. Semin Immunopathol 2022; 44:697-707. [PMID: 35505129 PMCID: PMC9942346 DOI: 10.1007/s00281-022-00945-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/20/2022] [Indexed: 12/12/2022]
Abstract
Glioblastoma is a highly aggressive brain tumor with limited treatment options. Several major challenges have limited the development of novel therapeutics, including the extensive heterogeneity of tumor cell states within each glioblastoma and the ability of glioma cells to diffusely infiltrate into neighboring healthy brain tissue, including the contralateral hemisphere. A T cell-mediated immune response could deal with these challenges based on the ability of polyclonal T cell populations to recognize diverse tumor antigens and perform surveillance throughout tissues. Here we will discuss the major pathways that inhibit T cell-mediated immunity against glioblastoma, with an emphasis on receptor-ligand systems by which glioma cells and recruited myeloid cells inhibit T cell function. A related challenge is that glioblastomas tend to be poorly infiltrated by T cells, which is not only caused by inhibitory molecular pathways but also currently utilized drugs, in particular high-dose corticosteroids that kill activated, proliferating T cells. We will discuss innovative approaches to induce glioblastoma-directed T cell responses, including neoantigen-based vaccines and sophisticated CAR T cell approaches that can target heterogeneous glioblastoma cell populations. Finally, we will propose a conceptual framework for the future development of T cell-based immunotherapies for glioblastoma.
Collapse
Affiliation(s)
- Sascha Marx
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Anze Godicelj
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Kai W. Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Immunology, Harvard Medical School, Boston, MA 02115, USA,Program in Immunology, Harvard Medical School, Boston, MA 02115, USA,Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02215, USA
| |
Collapse
|
45
|
Berger G, Knelson EH, Jimenez-Macias JL, Nowicki MO, Han S, Panagioti E, Lizotte PH, Adu-Berchie K, Stafford A, Dimitrakakis N, Zhou L, Chiocca EA, Mooney DJ, Barbie DA, Lawler SE. STING activation promotes robust immune response and NK cell-mediated tumor regression in glioblastoma models. Proc Natl Acad Sci U S A 2022; 119:e2111003119. [PMID: 35787058 PMCID: PMC9282249 DOI: 10.1073/pnas.2111003119] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 05/08/2022] [Indexed: 01/07/2023] Open
Abstract
Immunotherapy has had a tremendous impact on cancer treatment in the past decade, with hitherto unseen responses at advanced and metastatic stages of the disease. However, the aggressive brain tumor glioblastoma (GBM) is highly immunosuppressive and remains largely refractory to current immunotherapeutic approaches. The stimulator of interferon genes (STING) DNA sensing pathway has emerged as a next-generation immunotherapy target with potent local immune stimulatory properties. Here, we investigated the status of the STING pathway in GBM and the modulation of the brain tumor microenvironment (TME) with the STING agonist ADU-S100. Our data reveal the presence of STING in human GBM specimens, where it stains strongly in the tumor vasculature. We show that human GBM explants can respond to STING agonist treatment by secretion of inflammatory cytokines. In murine GBM models, we show a profound shift in the tumor immune landscape after STING agonist treatment, with massive infiltration of the tumor-bearing hemisphere with innate immune cells including inflammatory macrophages, neutrophils, and natural killer (NK) populations. Treatment of established murine intracranial GL261 and CT-2A tumors by biodegradable ADU-S100-loaded intracranial implants demonstrated a significant increase in survival in both models and long-term survival with immune memory in GL261. Responses to treatment were abolished by NK cell depletion. This study reveals therapeutic potential and deep remodeling of the TME by STING activation in GBM and warrants further examination of STING agonists alone or in combination with other immunotherapies such as cancer vaccines, chimeric antigen receptor T cells, NK therapies, and immune checkpoint blockade.
Collapse
Affiliation(s)
- Gilles Berger
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Microbiology, Bioorganic and Macromolecular Chemistry, Faculty of Pharmacy, Université Libre de Bruxelles, Brussels 1050, Belgium
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - Erik H. Knelson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Jorge L. Jimenez-Macias
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Michal O. Nowicki
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Saemi Han
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Eleni Panagioti
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Patrick H. Lizotte
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
- Human Tumor Profiling Group, Belfer Center for Applied Cancer Science, Boston, MA 02115
| | - Kwasi Adu-Berchie
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - Alexander Stafford
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - Lanlan Zhou
- Legorreta Cancer Center, Brown University, Providence, RI 02912
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912
| | - E. Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - David J. Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Sean E. Lawler
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
46
|
Zamler DB, Shingu T, Kahn LM, Huntoon K, Kassab C, Ott M, Tomczak K, Liu J, Li Y, Lai I, Zorilla-Veloz R, Yee C, Rai K, Kim BY, Watowich SS, Heimberger AB, Draetta GF, Hu J. Immune landscape of a genetically engineered murine model of glioma compared with human glioma. JCI Insight 2022; 7:e148990. [PMID: 35653194 PMCID: PMC9309065 DOI: 10.1172/jci.insight.148990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/04/2022] [Indexed: 11/25/2022] Open
Abstract
Novel therapeutic strategies targeting glioblastoma (GBM) often fail in the clinic, partly because preclinical models in which hypotheses are being tested do not recapitulate human disease. To address this challenge, we took advantage of our previously developed spontaneous Qk/Trp53/Pten (QPP) triple-knockout model of human GBM, comparing the immune microenvironment of QPP mice with that of patient-derived tumors to determine whether this model provides opportunity for gaining insights into tumor physiopathology and preclinical evaluation of therapeutic agents. Immune profiling analyses and single-cell sequencing of implanted and spontaneous tumors from QPP mice and from patients with glioma revealed intratumoral immune components that were predominantly myeloid cells (e.g., monocytes, macrophages, and microglia), with minor populations of T, B, and NK cells. When comparing spontaneous and implanted mouse samples, we found more neutrophils and T and NK cells in the implanted model. Neutrophils and T and NK cells were increased in abundance in samples derived from human high-grade glioma compared with those derived from low-grade glioma. Overall, our data demonstrate that our implanted and spontaneous QPP models recapitulate the immunosuppressive myeloid-dominant nature of the tumor microenvironment of human gliomas. Our model provides a suitable tool for investigating the complex immune compartment of gliomas.
Collapse
Affiliation(s)
- Daniel B. Zamler
- Department of Genomic Medicine
- Department of Cancer Biology, and
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Laura M. Kahn
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Immunology
| | | | | | | | | | - Jintan Liu
- Department of Genomic Medicine
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yating Li
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Melanoma Medical Oncology, and
| | - Ivy Lai
- Department of Melanoma Medical Oncology, and
| | - Rocio Zorilla-Veloz
- Department of Cancer Biology, and
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cassian Yee
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Melanoma Medical Oncology, and
| | - Kunal Rai
- Department of Genomic Medicine
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Stephanie S. Watowich
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Immunology
| | - Amy B. Heimberger
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Giulio F. Draetta
- Department of Genomic Medicine
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jian Hu
- Department of Cancer Biology, and
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
47
|
Combination of DNA Vaccine and Immune Checkpoint Blockades Improves the Immune Response in an Orthotopic Unresectable Glioblastoma Model. Pharmaceutics 2022; 14:pharmaceutics14051025. [PMID: 35631612 PMCID: PMC9145362 DOI: 10.3390/pharmaceutics14051025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
Combination immunotherapy has emerged as a promising strategy to increase the immune response in glioblastoma (GBM) and overcome the complex immunosuppression occurring in its microenvironment. In this study, we hypothesized that combining DNA vaccines—to stimulate a specific immune response—and dual immune checkpoint blockade (ICB)—to decrease the immunosuppression exerted on T cells—will improve the immune response and the survival in an orthotopic unresectable GL261 model. We first highlighted the influence of the insertion position of a GBM epitope sequence in a plasmid DNA vaccine encoding a vesicular stomatitis virus glycoprotein (VSV-G) (here referred to as pTOP) in the generation of a specific and significant IFN-γ response against the GBM antigen TRP2 by inserting a CD8 epitope sequence in specific permissive sites. Then, we combined the pTOP vaccine with anti-PD-1 and anti-CTLA-4 ICBs. Immune cell analysis revealed an increase in effector T cell to Treg ratios in the spleens and an increase in infiltrated IFN-γ-secreting CD8 T cell frequency in the brains following combination therapy. Even if the survival was not significantly different between dual ICB and combination therapy, we offer a new immunotherapeutic perspective by improving the immune landscape in an orthotopic unresectable GBM model.
Collapse
|
48
|
Sahu U, Barth RF, Otani Y, McCormack R, Kaur B. Rat and Mouse Brain Tumor Models for Experimental Neuro-Oncology Research. J Neuropathol Exp Neurol 2022; 81:312-329. [PMID: 35446393 PMCID: PMC9113334 DOI: 10.1093/jnen/nlac021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Rodent brain tumor models have been useful for developing effective therapies for glioblastomas (GBMs). In this review, we first discuss the 3 most commonly used rat brain tumor models, the C6, 9L, and F98 gliomas, which are all induced by repeated injections of nitrosourea to adult rats. The C6 glioma arose in an outbred Wistar rat and its potential to evoke an alloimmune response is a serious limitation. The 9L gliosarcoma arose in a Fischer rat and is strongly immunogenic, which must be taken into consideration when using it for therapy studies. The F98 glioma may be the best of the 3 but it does not fully recapitulate human GBMs because it is weakly immunogenic. Next, we discuss a number of mouse models. The first are human patient-derived xenograft gliomas in immunodeficient mice. These have failed to reproduce the tumor-host interactions and microenvironment of human GBMs. Genetically engineered mouse models recapitulate the molecular alterations of GBMs in an immunocompetent environment and “humanized” mouse models repopulate with human immune cells. While the latter are rarely isogenic, expensive to produce, and challenging to use, they represent an important advance. The advantages and limitations of each of these brain tumor models are discussed. This information will assist investigators in selecting the most appropriate model for the specific focus of their research.
Collapse
Affiliation(s)
- Upasana Sahu
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Yoshihiro Otani
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ryan McCormack
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Balveen Kaur
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
49
|
Žilionytė K, Bagdzevičiūtė U, Mlynska A, Urbštaitė E, Paberalė E, Dobrovolskienė N, Krasko JA, Pašukonienė V. Functional antigen processing and presentation mechanism as a prerequisite factor of response to treatment with dendritic cell vaccines and anti-PD-1 in preclinical murine LLC1 and GL261 tumor models. Cancer Immunol Immunother 2022; 71:2691-2700. [PMID: 35364740 DOI: 10.1007/s00262-022-03190-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/14/2022] [Indexed: 12/15/2022]
Abstract
Low efficacy of cancer immunotherapy encourages the search for possible resistance mechanisms and biomarkers that would predict the outcome of immunotherapy in oncology patients. Most cancer immunotherapies act on T lymphocytes, which can specifically recognize and kill tumor cells. However, for immunotherapy-activated T lymphocytes to be able to perform these functions, proper tumor Ag processing and surface presentation by MHC-I molecule is important. Knowing the significance of Ag processing and presentation mechanism (APM) in anti-tumor immune response, we sought to evaluate how the functionality of APM affects tumor immune microenvironment and response to dendritic cell vaccines (DCV) and anti-PD-1. By comparing murine Lewis lung carcinoma LLC1 and glioma GL261 models a decreased expression of APM-related genes, such as Psmb8, Psmb9, Psmb10, Tap1, Tap2, Erap1, B2m, and low expression of surface MHC-I molecule were found in LLC1 cells. Changes in APM-related gene expression affected the ability of T lymphocytes to recognize and kill LLC1 cells, resulting in the absence of cytotoxic immune response and resistance to DCV and anti-PD-1. An emerging cytotoxic immune reaction and sensitivity to DCV and anti-PD-1 were observed in GL261 tumors where APM remained functional. This study demonstrates that one of the possible mechanisms of tumor resistance to immunotherapy is a dysfunctional APM and reveals a predictive potential of APM-related gene set expression for the personalization of dendritic cell vaccine and anti-PD-1 therapies in murine pre-treated tumors.
Collapse
Affiliation(s)
- Karolina Žilionytė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania. .,Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Ugnė Bagdzevičiūtė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Agata Mlynska
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | | | - Emilija Paberalė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Jan Aleksander Krasko
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Vita Pašukonienė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| |
Collapse
|
50
|
Farshbaf M, Mojarad-Jabali S, Hemmati S, Khosroushahi AY, Motasadizadeh H, Zarebkohan A, Valizadeh H. Enhanced BBB and BBTB penetration and improved anti-glioma behavior of Bortezomib through dual-targeting nanostructured lipid carriers. J Control Release 2022; 345:371-384. [PMID: 35301054 DOI: 10.1016/j.jconrel.2022.03.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/12/2022] [Accepted: 03/10/2022] [Indexed: 12/19/2022]
Abstract
The effective treatment of glioma through conventional chemotherapy is proved to be a great challenge in clinics. The main reason is due to the existence of two physiological and pathological barriers respectively including the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) that prevent most of the chemotherapeutics from efficient delivery to the brain tumors. To address this challenge, an ideal drug delivery system would efficiently traverse the BBB and BBTB and deliver the therapeutics into the glioma cells with high selectivity. Herein, a targeted delivery system was developed based on nanostructured lipid carriers (NLCs) modified with two proteolytically stable D-peptides, D8 and RI-VAP (Dual NLCs). D8 possesses high affinity towards nicotine acetylcholine receptors (nAChRs), overexpressed on brain capillary endothelial cells (BCECs), and can penetrate through BBB with high efficiency. RI-VAP is a specific ligand of cell surface GRP78 (csGRP78), a specific angiogenesis and cancer cell-surface marker, capable of circumventing the BBTB with superior glioma-homing property. Dual NLCs could internalize into BCECs, tumor neovascular endothelial cells, and glioma cells with high specificity and could penetrate through in vitro BBB and BBTB models with excellent efficiency compared to non-targeted or mono-targeted NLCs. In vivo whole-animal imaging and ex vivo imaging further confirmed the superior targeting capability of Dual NLCs towards intracranial glioma. When loaded with Bortezomib (BTZ), Dual NLCs attained the highest therapeutic efficiency by means of in vitro cytotoxicity and apoptosis and prolonged survival rate and anti-glioma behavior in intracranial glioma bearing mice. Collectively, the designed targeting platform in this study could overcome multiple barriers and effectively deliver BTZ to glioma cells, which represent its potential for advanced brain cancer treatment with promising therapeutic outcomes.
Collapse
Affiliation(s)
- Masoud Farshbaf
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Mojarad-Jabali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamidreza Motasadizadeh
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|