1
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [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: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
2
|
Strassheimer F, Elleringmann P, Ludmirski G, Roller B, Macas J, Alekseeva T, Cakmak P, Aliraj B, Krenzlin H, Demes MC, Mildenberger IC, Tonn T, Weber KJ, Reiss Y, Plate KH, Weigert A, Wels WS, Steinbach JP, Burger MC. CAR-NK cell therapy combined with checkpoint inhibition induces an NKT cell response in glioblastoma. Br J Cancer 2025; 132:849-860. [PMID: 40102596 PMCID: PMC12041480 DOI: 10.1038/s41416-025-02977-8] [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: 06/05/2024] [Revised: 02/02/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Glioblastoma is the most aggressive primary brain tumor with limited efficacy of established therapies, and a pronounced immunosuppressive tumor microenvironment. Targeting HER2 with local immunotherapy allows for high tumor specificity in the brain with physiologically very low expression. Monotherapy with CAR-NK cells targeted against HER2 has previously shown efficacy in medium-sized GL261/HER2 tumors. METHODS Advanced GL261/HER2 tumors were treated by local CAR-NK cell injection combined with systemic anti-PD-1 checkpoint blockade. Tumor growth and survival were monitored. In-depth characterization of the microenvironment was performed by multiplex immune fluorescence, spectral flow cytometry and RNAseq. RESULTS Untreated GL261/HER2 tumors were characterized by local immunosuppression and high PD-L1 expression. Combined treatment with NK-92/5.28.z and systemic anti-PD-1 induced robust anti-tumor response and long-term survival. Multiplex immunofluorescence and spectral flow cytometry showed increased CD4+ T cell infiltration in mice treated with CAR-NK cell and anti-PD-1 combination therapy. A cluster of T cells specifically emerging in the combination therapy group expressed markers of NKT cells, which was further verified by immunofluorescence staining. CONCLUSION The combination therapy reverted the immunosuppressive tumor microenvironment with increased T and NKT cell infiltration. This resulted in successful treatment of advanced orthotopic tumors refractory to CAR-NK cell monotherapy.
Collapse
Affiliation(s)
- F Strassheimer
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P Elleringmann
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - G Ludmirski
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
| | - B Roller
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - J Macas
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, Institute of Neurology (Edinger Institute), Goethe University Hospital, Frankfurt, Germany
| | - T Alekseeva
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - P Cakmak
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University, Institute of Neurology (Edinger Institute), Goethe University Hospital, Frankfurt, Germany
| | - B Aliraj
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, Institute of Biochemistry I, Faculty of Medicine, Frankfurt, Germany
| | - H Krenzlin
- University Medical Center Mainz, Department of Neurosurgery, Mainz, Germany
| | - M C Demes
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, Dr. Senckenberg Institute of Pathology, Goethe University Hospital, Frankfurt, Germany
| | - I C Mildenberger
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - T Tonn
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Donation Service Baden-Württemberg-Hessen and Goethe University Hospital, Frankfurt, Germany
| | - K J Weber
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University, Institute of Neurology (Edinger Institute), Goethe University Hospital, Frankfurt, Germany
| | - Y Reiss
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University, Institute of Neurology (Edinger Institute), Goethe University Hospital, Frankfurt, Germany
| | - K H Plate
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University, Institute of Neurology (Edinger Institute), Goethe University Hospital, Frankfurt, Germany
| | - A Weigert
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University, Institute of Biochemistry I, Faculty of Medicine, Frankfurt, Germany
| | - W S Wels
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - J P Steinbach
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M C Burger
- Goethe University, Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
3
|
Mulliqi E, Khelwatty S, Bagwan I, Kamaludin A, Morgan A, Long N, Ashkan K, Modjtahedi H. The Co-Expression and Cellular Location of HER Family Members, EGFRvIII, Putative Cancer Stem Cell Biomarkers CD44 and CD109 in Patients with Glioblastoma, and Their Impacts on Prognosis. Cancers (Basel) 2025; 17:1221. [PMID: 40227788 PMCID: PMC11987930 DOI: 10.3390/cancers17071221] [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: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES The aberrant expression and activation of HER family members is a known major oncogenic pathway for the proliferation, progression, and metastasis of a wide range of human malignancies. In this study, our aim was to examine the relative expression and prognostic significance of all members of the HER family, the type III EGFR mutant (EGFRvIII), and the putative stem cell markers CD44 and CD109 in patients with glioblastoma. METHODS The expression levels of wild-type EGFR (wtEGFR), HER2, HER3, HER4, EGFRvIII, CD44, and CD109 were determined in tumour specimens from 80 patients by immunohistochemistry. The staining was scored based on the percentage of positive tumour cells, the intensity, and the cellular location of immunostaining. The association between the expression level of the biomarkers and patient overall survival was evaluated using Chi-squared, Kaplan-Meier survival curves, and log-rank tests. RESULTS At a cut-off value of ≥5% with positive staining, 46% (wtEGFR), 75% (HER2), 19% (HER3), 71% (HER4), 85% (EGFRvIII), 95% (CD44), and 16% (CD109) of the cases were positive for these biomarkers. Interestingly, at the same cut-off value, the expression of wtEGFR in these patients was accompanied by co-expression with HER2 (35%), HER3 (0%), HER4 (30%), EGFRvIII (36%), CD44 (44%), HER2/EGFRvIII (28%), HER2/CD44 (31%), and EGFRvIII/CD44 (36%). In addition, the expression of EGFRvIII was accompanied by co-expression with HER2 (65%), HER3 (15%), HER4 (63%), CD44 (83%), CD109 (16%), wtEGFR/HER2 (28%), and 55% of the cases had co-expression of EGFRvIII/HER2/HER4/CD44. With the exception of HER2 expression, at cut-off values of ≥5% of tumour cells with positive staining, which was associated with better overall survival [HR = 0.57 (p = 0.038), HR = 0.56 (p = 0.034)], there was no significant association between the expression of other members of the HER family, EGFRvIII, CD44, and CD109 on the overall survival in both univariate and multivariate analysis. Conclusions Our results suggest that the co-expression of different members of the HER family, with EGFRvIII, CD44, and CD109, occurs in patients with glioblastoma. As the results of therapy with EGFR inhibitors have not been encouraging in patients with a brain tumour, further investigation should determine whether the co-expression of such biomarkers can be of predictive value for the response to the therapy with various types of HER inhibitors and their potential as therapeutic targets for co-targeted therapy.
Collapse
Affiliation(s)
- Ermira Mulliqi
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Said Khelwatty
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Izhar Bagwan
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
- Berkshire Surrey Pathology Services, Royal Surrey Hospital, Guildford GU2 7XX, UK
| | - Ahmad Kamaludin
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Anna Morgan
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Natalie Long
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Helmout Modjtahedi
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| |
Collapse
|
4
|
Sabahi M, Fathi Jouzdani A, Sadeghian Z, Dabbagh Ohadi MA, Sultan H, Salehipour A, Maniakhina L, Rezaei N, Adada B, Mansouri A, Borghei-Razavi H. CAR-engineered NK cells versus CAR T cells in treatment of glioblastoma; strength and flaws. J Neurooncol 2025; 171:495-530. [PMID: 39538038 DOI: 10.1007/s11060-024-04876-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary brain tumor that carries a grim prognosis. Because of the dearth of treatment options available for treatment of GBM, Chimeric Antigen Receptor (CAR)-engineered T cell and Natural Killer (NK) therapy could provide alternative strategies to address the challenges in GBM treatment. In these approaches, CAR T and NK cells are engineered for cancer-specific immunotherapy by recognizing surface antigens independently of major histocompatibility complex (MHC) molecules. However, the efficacy of CAR T cells is hindered by GBM's downregulation of its targeted antigens. CAR NK cells face similar challenges, but, in contrast, they offer advantages as off-the-shelf allogeneic products, devoid of graft-versus-host disease (GVHD) risk as well as anti-cancer activity beyond CAR specificity, potentially reducing the risk of relapse or resistance. Despite CAR T cell therapies being extensively studied in clinical settings, the use of CAR-modified NK cells in GBM treatment remains largely in the preclinical stage. This review aims to discuss recent advancements in NK cell and CAR T cell therapies for GBM, including methods for introducing CARs into both NK cells and T cells, addressing manufacturing challenges, and providing evidence supporting the efficacy of these approaches from preclinical and early-phase clinical studies. The comprehensive evaluation of CAR-engineered NK cells and CAR T cells seeks to identify the optimal therapeutic approach for GBM, contributing to the development of effective immunotherapies for this devastating disease.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Ali Fathi Jouzdani
- Neurosurgery Research Group (NRG), Hamadan University of Medical Sciences, Hamadan, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohre Sadeghian
- Department of Pathology & Laboratory Medicine, Cleveland Clinic Florida, Weston, FL, USA
| | | | - Hadi Sultan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Arash Salehipour
- Neurosurgery Research Group (NRG), Hamadan University of Medical Sciences, Hamadan, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Lana Maniakhina
- Department of Neurosurgery, Geisinger and Geisinger Commonwealth School of Medicine, Wilkes-Barre, PA, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Badih Adada
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| |
Collapse
|
5
|
Cooksey LC, Friesen DC, Mangan ED, Mathew PA. Prospective Molecular Targets for Natural Killer Cell Immunotherapy against Glioblastoma Multiforme. Cells 2024; 13:1567. [PMID: 39329751 PMCID: PMC11429815 DOI: 10.3390/cells13181567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of primary malignant brain tumor and has a dismal overall survival rate. To date, no GBM therapy has yielded successful results in survival for patients beyond baseline surgical resection, radiation, and chemotherapy. Immunotherapy has taken the oncology world by storm in recent years and there has been movement from researchers to implement the immunotherapy revolution into GBM treatment. Natural killer (NK) cell-based immunotherapies are a rising candidate to treat GBM from multiple therapeutic vantage points: monoclonal antibody therapy targeting tumor-associated antigens (TAAs), immune checkpoint inhibitors, CAR-NK cell therapy, Bi-specific killer cell engagers (BiKEs), and more. NK therapies often focus on tumor antigens for targeting. Here, we reviewed some common targets analyzed in the fight for GBM immunotherapy relevant to NK cells: EGFR, HER2, CD155, and IL-13Rα2. We further propose investigating the Lectin-like Transcript 1 (LLT1) and cell surface proliferating cell nuclear antigen (csPCNA) as targets for NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Luke C. Cooksey
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (L.C.C.); (D.C.F.); (E.D.M.)
- Department of Microbiology, Immunology and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Derek C. Friesen
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (L.C.C.); (D.C.F.); (E.D.M.)
| | - Enrique D. Mangan
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (L.C.C.); (D.C.F.); (E.D.M.)
| | - Porunelloor A. Mathew
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (L.C.C.); (D.C.F.); (E.D.M.)
- Department of Microbiology, Immunology and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
6
|
Jones CM, Rohwedder A, Suen KM, Mohajerani SZ, Calabrese AN, Knipp S, Bedford MT, Ladbury JE. Affinity purification mass spectrometry characterisation of the interactome of receptor tyrosine kinase proline-rich motifs in cancer. Heliyon 2024; 10:e35480. [PMID: 39165974 PMCID: PMC11334840 DOI: 10.1016/j.heliyon.2024.e35480] [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: 02/26/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Receptor tyrosine kinase (RTK) overexpression is linked to the development and progression of multiple cancers. RTKs are classically considered to initiate cytoplasmic signalling pathways via ligand-induced tyrosine phosphorylation, however recent evidence points to a second tier of signalling contingent on interactions mediated by the proline-rich motif (PRM) regions of non-activated RTKs. The presence of PRMs on the C-termini of >40 % of all RTKs and the abundance of PRM-binding proteins encoded by the human genome suggests that there is likely to be a large number of previously unexplored interactions which add to the RTK intracellular interactome. Here, we explore the RTK PRM interactome and its potential significance using affinity purification mass spectrometry and in silico enrichment analyses. Peptides comprising PRM-containing C-terminal tail regions of EGFR, FGFR2 and HER2 were used as bait to affinity purify bound proteins from different cancer cell line lysates. 490 unique interactors were identified, amongst which proteins with metabolic, homeostatic and migratory functions were overrepresented. This suggests that PRMs from RTKs may sustain a diverse interactome in cancer cells. Since RTK overexpression is common in cancer, RTK PRM-derived signalling may be an important, but as yet underexplored, contributor to negative cancer outcomes including resistance to kinase inhibitors.
Collapse
Affiliation(s)
- Christopher M. Jones
- Department of Oncology, University of Cambridge, Cambridge, CB2 0XZ, UK
- Faculty of Biological Sciences, University of Leeds, Leeds, LJ2 9JT, UK
| | - Arndt Rohwedder
- Faculty of Biological Sciences, University of Leeds, Leeds, LJ2 9JT, UK
- Centre for Medical Research (ZMF), Johannes Kepler University, 4020 Linz, Austria
| | - Kin Man Suen
- Faculty of Biological Sciences, University of Leeds, Leeds, LJ2 9JT, UK
| | | | | | - Sabine Knipp
- Faculty of Biological Sciences, University of Leeds, Leeds, LJ2 9JT, UK
| | - Mark T. Bedford
- Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Centre, Houston, TX. TX 77230, USA
| | - John E. Ladbury
- Faculty of Biological Sciences, University of Leeds, Leeds, LJ2 9JT, UK
| |
Collapse
|
7
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
8
|
Huang B, Zhang J, Zong W, Chen S, Zong Z, Zeng X, Zhang H. Myeloidcells in the immunosuppressive microenvironment in glioblastoma: The characteristics and therapeutic strategies. Front Immunol 2023; 14:994698. [PMID: 36923402 PMCID: PMC10008967 DOI: 10.3389/fimmu.2023.994698] [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: 07/15/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
Glioblastoma (GBM) is the most common and lethal malignant tumor of the central nervous system in adults. Conventional therapies, including surgery, radiotherapy, and chemotherapy, have limited success in ameliorating patient survival. The immunosuppressive tumor microenvironment, which is infiltrated by a variety of myeloid cells, has been considered a crucial obstacle to current treatment. Recently, immunotherapy, which has achieved great success in hematological malignancies and some solid cancers, has garnered extensive attention for the treatment of GBM. In this review, we will present evidence on the features and functions of different populations of myeloid cells, and on current clinical advances in immunotherapies for glioblastoma.
Collapse
Affiliation(s)
- Boyuan Huang
- Department of Neurosurgery, Capital Medical University Electric Power Teaching Hospital/State Grid Beijing Electric Power Hospital, Beijing, China
| | - Jin Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Sisi Chen
- Department of neurosurgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, China
| | - Zhitao Zong
- Department of neurosurgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, China
| | - Xiaojun Zeng
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Hongbo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Strecker M, Wlotzka K, Strassheimer F, Roller B, Ludmirski G, König S, Röder J, Opitz C, Alekseeva T, Reul J, Sevenich L, Tonn T, Wels W, Steinbach J, Buchholz C, Burger M. AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma. Oncoimmunology 2022; 11:2127508. [PMID: 36249274 PMCID: PMC9559045 DOI: 10.1080/2162402x.2022.2127508] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated tumor-cell specific delivery of an anti-PD-1 immunoadhesin (aPD-1) via a targeted adeno-associated viral vector (AAV) as well as HER2-specific NK-92/5.28.z (anti-HER2.CAR/NK-92) cells as components for a combination immunotherapy. In co-culture experiments, target-activated anti-HER2.CAR/NK-92 cells modified surrounding tumor cells and bystander immune cells by triggering the release of inflammatory cytokines and upregulation of PD-L1. Tumor cell-specific delivery of aPD-1 was achieved by displaying a HER2-specific designed ankyrin repeat protein (DARPin) on the AAV surface. HER2-AAV mediated gene transfer into GB cells correlated with HER2 expression levels, without inducing anti-viral responses in transduced cells. Furthermore, AAV-transduction did not interfere with anti-HER2.CAR/NK-92 cell-mediated tumor cell lysis. After selective transduction of HER2+ cells, aPD-1 expression was detected at the mRNA and protein level. The aPD-1 immunoadhesin was secreted in a time-dependent manner, bound its target on PD-1-expressing cells and was able to re-activate T cells by efficiently disrupting the PD-1/PD-L1 axis. Moreover, high intratumoral and low systemic aPD-1 concentrations were achieved following local injection of HER2-AAV into orthotopic tumor grafts in vivo. aPD-1 was selectively produced in tumor tissue and could be detected up to 10 days after a single HER2-AAV injection. In subcutaneous GL261-HER2 and Tu2449-HER2 immunocompetent mouse models, administration of the combination therapy significantly prolonged survival, including complete tumor control in several animals in the GL261-HER2 model. In summary, local therapy with aPD-1 encoding HER2-AAVs in combination with anti-HER2.CAR/NK-92 cells may be a promising novel strategy for GB immunotherapy with the potential to enhance efficacy and reduce systemic side effects of immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- M.I. Strecker
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - K. Wlotzka
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - F. Strassheimer
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - B. Roller
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - G. Ludmirski
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - S. König
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - J. Röder
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - C. Opitz
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - T. Alekseeva
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - J. Reul
- Paul-Ehrlich-Institut, Molecular Biotechnology and Gene Therapy, Langen, Germany
| | - L. Sevenich
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - T. Tonn
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
| | - W.S. Wels
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - J.P. Steinbach
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - C.J. Buchholz
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- Paul-Ehrlich-Institut, Molecular Biotechnology and Gene Therapy, Langen, Germany
- German Cancer Consortium (DKTK), partner site Heidelberg, Heidelberg, Germany
| | - M.C. Burger
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| |
Collapse
|
10
|
Li H, Song W, Li Z, Zhang M. Preclinical and clinical studies of CAR-NK-cell therapies for malignancies. Front Immunol 2022; 13:992232. [PMID: 36353643 PMCID: PMC9637940 DOI: 10.3389/fimmu.2022.992232] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 12/01/2022] Open
Abstract
The development of chimeric antigen receptor T (CAR-T) cell therapy, a specific type of immunotherapy, in recent decades was a fantastic breakthrough for the treatment of hematological malignancies. However, difficulties in collecting normal T cells from patients and the time cost of manufacturing CAR-T cells have limited the application of CAR-T-cell therapy. In addition, the termination of related clinical trials on universal CAR-T cell therapy has made further research more difficult. Natural killer (NK) cells have drawn great attention in recent years. Chimeric antigen receptor-NK (CAR-NK) cell therapy is a promising strategy in the treatment of malignant tumors because of its lack of potential for causing graft-versus-host disease (GVHD). In this review, we will address the advances in and achievements of CAR-NK cell therapy.
Collapse
Affiliation(s)
- Hongwen Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Mingzhi Zhang,
| |
Collapse
|
11
|
Barish ME, Weng L, Awabdeh D, Zhai Y, Starr R, D'Apuzzo M, Rockne RC, Li H, Badie B, Forman SJ, Brown CE. Spatial organization of heterogeneous immunotherapy target antigen expression in high-grade glioma. Neoplasia 2022; 30:100801. [PMID: 35550513 PMCID: PMC9108993 DOI: 10.1016/j.neo.2022.100801] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
High-grade (WHO grades III-IV) glioma remains one of the most lethal human cancers. Adoptive transfer of tumor-targeting chimeric antigen receptor (CAR)-redirected T cells for high-grade glioma has revealed promising indications of anti-tumor activity, but objective clinical responses remain elusive for most patients. A significant challenge to effective immunotherapy is the highly heterogeneous structure of these tumors, including large variations in the magnitudes and distributions of target antigen expression, observed both within individual tumors and between patients. To obtain a more detailed understanding of immunotherapy target antigens within patient tumors, we immunochemically mapped at single cell resolution three clinically-relevant targets, IL13Rα2, HER2 and EGFR, on tumor samples drawn from a 43-patient cohort. We observed that within individual tumor samples, expression of these antigens was neither random nor uniform, but rather that they mapped into local neighborhoods - phenotypically similar cells within regions of cellular tumor - reflecting not well understood properties of tumor cells and their milieu. Notably, tumor cell neighborhoods of high antigen expression were not arranged independently within regions. For example, in cellular tumor regions, neighborhoods of high IL13Rα2 and HER2 expression appeared to be reciprocal to those of EGFR, while in areas of pseudopalisading necrosis, expression of IL13Rα2 and HER2, but not EGFR, appeared to reflect the radial organization of tumor cells around hypoxic cores. Other structural features affecting expression of immunotherapy target antigens remain to be elucidated. This structured but heterogeneous organization of antigen expression in high grade glioma is highly permissive for antigen escape, and combinatorial antigen targeting is a commonly suggested potential mitigating strategy. Deeper understanding of antigen expression within and between patient tumors will enhance optimization of combination immunotherapies, the most immediate clinical application of the observations presented here being the importance of including (wild-type) EGFR as a target antigen.
Collapse
Affiliation(s)
- Michael E Barish
- Department of Stem Cell Biology & Regenerative Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States.
| | - Lihong Weng
- Department of Hematology & Hematopoietic Cell Transplantation, National Medical Center, City of Hope, Duarte, CA 91010, United States
| | - Dina Awabdeh
- Department of Stem Cell Biology & Regenerative Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States
| | - Yubo Zhai
- Department of Hematology & Hematopoietic Cell Transplantation, National Medical Center, City of Hope, Duarte, CA 91010, United States
| | - Renate Starr
- Department of Hematology & Hematopoietic Cell Transplantation, National Medical Center, City of Hope, Duarte, CA 91010, United States
| | - Massimo D'Apuzzo
- Department of Pathology, National Medical Center, City of Hope, Duarte, CA 91010, United States
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States
| | - Haiqing Li
- Integrative Genomics Core, Division of Translational Bioinformatics, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States
| | - Behnam Badie
- Department of Surgery, Division of Neurosurgery, National Medical Center, City of Hope, Duarte, CA 91010, United States
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, National Medical Center, City of Hope, Duarte, CA 91010, United States
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, National Medical Center, City of Hope, Duarte, CA 91010, United States; Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, United States.
| |
Collapse
|
12
|
Baghery Saghchy Khorasani A, Yousefi AM, Bashash D. CAR NK cell therapy in hematologic malignancies and solid tumors; obstacles and strategies to overcome the challenges. Int Immunopharmacol 2022; 110:109041. [PMID: 35839565 DOI: 10.1016/j.intimp.2022.109041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 02/08/2023]
Abstract
Adoptive cell treatment (ACT) utilizing chimeric antigen receptors (CAR) diverts the specificity of safe cells against a target-specific antigen and portrays exceptional potential for cancer treatment. While CAR T cell treatment has risen as a breakthrough with unprecedented results within the therapeutic procedures of human malignancies, different deficiencies including challenging and costly generation processes, strict patient qualification criteria, and undesirable toxicity have ruined its application. Unlike T cells, the application of natural killer (NK) cells has attracted consideration as a reasonable alternative owing to the major histocompatibility complex (MHC)-independency, shorter life expectancy, the potential to create an off-the-shelf immune product, and potent antitumor properties. In this article, we provide an updated review of the differences between CAR T and CAR NK cells, current enhancements in CAR NK design, the available sources for collecting NK cells, and strategies for the transduction step of the CARs to NK cells. Furthermore, we focus on the published and ongoing preclinical and clinical studies of CAR NK treatment strategies both in hematologic malignancies and solid tumors. We also discuss limitations and plausible solutions to improve the perseverance, function, safety, and efficacy of CAR NK cells with a special focus on solid tumors.
Collapse
Affiliation(s)
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Feldman L, Brown C, Badie B. Chimeric Antigen Receptor (CAR) T Cell Therapy for Glioblastoma. Neuromolecular Med 2022; 24:35-40. [PMID: 34665390 PMCID: PMC11220928 DOI: 10.1007/s12017-021-08689-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022]
Abstract
Glioblastoma (GBM) are the most common and aggressive primary brain tumors in adults. Current mainstay treatments include surgery, chemotherapy, and radiation; however, these are ineffective. As a result, immunotherapy treatment strategies are being developed to harness the body's natural defense mechanisms against gliomas. Adoptive cell therapy with chimeric antigen receptor (CAR) T cells uses patients' own T cells that are genetically modified to target tumor-associated antigens. These cells are harvested from patients, engineered to target specific proteins expressed by the tumor and re-injected into the patient with the goal of destroying tumor cells. In this mini review, we outline the history of CAR T cell therapy, describe current antigen targets, and review challenges this treatment faces specifically in targeting GBM.
Collapse
Affiliation(s)
- Lisa Feldman
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, CA, 91010, USA.
- Division of Neurosurgery, City of Hope National Medical Center, MOB 2001, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| | - Christine Brown
- Departments of Cancer Immunotherapy & Tumor Immunology and Hematology & Hematopoietic Call Transplantation, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Behnam Badie
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, CA, 91010, USA
| |
Collapse
|
14
|
da Silva LHR, Catharino LCC, da Silva VJ, Evangelista GCM, Barbuto JAM. The War Is on: The Immune System against Glioblastoma—How Can NK Cells Drive This Battle? Biomedicines 2022; 10:biomedicines10020400. [PMID: 35203609 PMCID: PMC8962431 DOI: 10.3390/biomedicines10020400] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that play an important role in immunosurveillance, acting alongside other immune cells in the response against various types of malignant tumors and the prevention of metastasis. Since their discovery in the 1970s, they have been thoroughly studied for their capacity to kill neoplastic cells without the need for previous sensitization, executing rapid and robust cytotoxic activity, but also helper functions. In agreement with this, NK cells are being exploited in many ways to treat cancer. The broad arsenal of NK-based therapies includes adoptive transfer of in vitro expanded and activated cells, genetically engineered cells to contain chimeric antigen receptors (CAR-NKs), in vivo stimulation of NK cells (by cytokine therapy, checkpoint blockade therapies, etc.), and tumor-specific antibody-guided NK cells, among others. In this article, we review pivotal aspects of NK cells’ biology and their contribution to immune responses against tumors, as well as providing a wide perspective on the many antineoplastic strategies using NK cells. Finally, we also discuss those approaches that have the potential to control glioblastoma—a disease that, currently, causes inevitable death, usually in a short time after diagnosis.
Collapse
Affiliation(s)
- Lucas Henrique Rodrigues da Silva
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508000, Brazil; (L.H.R.d.S.); (L.C.C.C.); (V.J.d.S.); (G.C.M.E.)
| | - Luana Correia Croda Catharino
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508000, Brazil; (L.H.R.d.S.); (L.C.C.C.); (V.J.d.S.); (G.C.M.E.)
| | - Viviane Jennifer da Silva
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508000, Brazil; (L.H.R.d.S.); (L.C.C.C.); (V.J.d.S.); (G.C.M.E.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Departamento de Hematologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 0124690, Brazil
| | - Gabriela Coeli Menezes Evangelista
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508000, Brazil; (L.H.R.d.S.); (L.C.C.C.); (V.J.d.S.); (G.C.M.E.)
| | - José Alexandre Marzagão Barbuto
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508000, Brazil; (L.H.R.d.S.); (L.C.C.C.); (V.J.d.S.); (G.C.M.E.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Departamento de Hematologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 0124690, Brazil
- Correspondence: ; Tel.: +55-11-3091-7375
| |
Collapse
|
15
|
Gatto L, Franceschi E, Di Nunno V, Maggio I, Lodi R, Brandes AA. Engineered CAR-T and novel CAR-based therapies to fight the immune evasion of glioblastoma: gutta cavat lapidem. Expert Rev Anticancer Ther 2021; 21:1333-1353. [PMID: 34734551 DOI: 10.1080/14737140.2021.1997599] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The field of cancer immunotherapy has achieved great advancements through the application of genetically engineered T cells with chimeric antigen receptors (CAR), that have shown exciting success in eradicating hematologic malignancies and have proved to be safe with promising early signs of antitumoral activity in the treatment of glioblastoma (GBM). AREAS COVERED We discuss the use of CAR T cells in GBM, focusing on limitations and obstacles to advancement, mostly related to toxicities, hostile tumor microenvironment, limited CAR T cells infiltration and persistence, target antigen loss/heterogeneity and inadequate trafficking. Furthermore, we introduce the refined strategies aimed at strengthening CAR T activity and offer insights in to novel immunotherapeutic approaches, such as the potential use of CAR NK or CAR M to optimize anti-tumor effects for GBM management. EXPERT OPINION With the progressive wide use of CAR T cell therapy, significant challenges in treating solid tumors, including central nervous system (CNS) tumors, are emerging, highlighting early disease relapse and cancer cell resistance issues, owing to hostile immunosuppressive microenvironment and tumor antigen heterogeneity. In addition to CAR T cells, there is great interest in utilizing other types of CAR-based therapies, such as CAR natural killer (CAR NK) or CAR macrophages (CAR M) cells for CNS tumors.
Collapse
Affiliation(s)
- Lidia Gatto
- Medical Oncology Department, Azienda USL, Bologna, Italy
| | - Enrico Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, Bologna, Italy
| | | | - Ilaria Maggio
- Medical Oncology Department, Azienda USL, Bologna, Italy
| | - Raffaele Lodi
- IrcssIstituto di Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alba Ariela Brandes
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, Bologna, Italy
| |
Collapse
|
16
|
Marei HE, Althani A, Afifi N, Hasan A, Caceci T, Pozzoli G, Cenciarelli C. Current progress in chimeric antigen receptor T cell therapy for glioblastoma multiforme. Cancer Med 2021; 10:5019-5030. [PMID: 34145792 PMCID: PMC8335808 DOI: 10.1002/cam4.4064] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the deadliest brain tumors with an unfavorable prognosis and overall survival of approximately 20 months following diagnosis. The current treatment for GBM includes surgical resections and chemo- and radiotherapeutic modalities, which are not effective. CAR-T immunotherapy has been proven effective for CD19-positive blood malignancies, and the application of CAR-T cell therapy for solid tumors including GBM offers great hope for this aggressive tumor which has a limited response to current treatments. CAR-T technology depends on the use of patient-specific T cells genetically engineered to express specific tumor-associated antigens (TAAs). Interaction of CAR-T cells with tumor cells triggers the destruction/elimination of these cells by the induction of cytotoxicity and the release of different cytokines. Despite the great promise of CAR-T cell-based therapy several challenges exist. These include the heterogeneity of GBM cancer cells, aberrant various signaling pathways involved in tumor progression, antigen escape, the hostile inhibitory GBM microenvironment, T cell dysfunction, blood-brain barrier, and defective antigen presentation. All need to be addressed before full application at the clinical level can begin. Herein we provide a focused review of the rationale for the use of different types of CAR-T cells (including FcγRs), the different GBM-associated antigens, the challenges still facing CAR-T-based therapy, and means to overcome such challenges. Finally, we enumerate currently completed and ongoing clinical trials, highlighting the different ways such trials are designed to overcome specific problems. Exploitation of the full potential of CAR-T cell therapy for GBM depends on their solution.
Collapse
MESH Headings
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/therapeutic use
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Blood-Brain Barrier
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- Cell Movement/immunology
- Cell Movement/physiology
- Clinical Trials as Topic
- Disease Progression
- ErbB Receptors/immunology
- Forecasting
- Glioblastoma/immunology
- Glioblastoma/therapy
- Humans
- Immune Checkpoint Inhibitors/metabolism
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Interleukin-13 Receptor alpha2 Subunit/immunology
- Lymphocyte Activation
- Lymphocyte Depletion
- Receptor, ErbB-2/immunology
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/therapeutic use
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- T-Lymphocytes/physiology
- Tumor Escape
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Hany E. Marei
- Department of Cytology and HistologyFaculty of Veterinary MedicineMansoura UniversityMansouraEgypt
| | | | | | - Anwarul Hasan
- Department of Mechanical and Industrial EngineeringCollege of EngineeringQatar UniversityDohaQatar
| | - Thomas Caceci
- Biomedical SciencesVirginia Maryland College of Veterinary MedicineBlacksburgVirginiaUSA
| | - Giacomo Pozzoli
- Pharmacology UnitFondazione Policlinico A. GemelliIRCCSRomeItaly
| | | |
Collapse
|
17
|
Feldman L, Brown C, Badie B. Chimeric Antigen Receptor T-Cell Therapy: Updates in Glioblastoma Treatment. Neurosurgery 2021; 88:1056-1064. [PMID: 33575786 DOI: 10.1093/neuros/nyaa584] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) are the most common and among the deadliest brain tumors in adults. Current mainstay treatments are insufficient to treat this tumor, and therefore, more effective therapies are desperately needed. Immunotherapy, which takes advantage of the body's natural defense mechanism, is an exciting emerging field in neuro-oncology. Adoptive cell therapy with chimeric antigen receptor (CAR) T cells provides a treatment strategy based on using patients' own selected and genetically engineered cells that target tumor-associated antigens. These cells are harvested from patients, modified to target specific proteins expressed by the tumor, and re-introduced into the patient with the goal of destroying tumor cells. Here, we review the history of CAR T-cell therapy, and describe the characteristics of various generations of CAR T therapies, and the challenges inherent to treatment of GBM. Finally, we describe recent and current CAR T clinical trials designed to combat GBM.
Collapse
Affiliation(s)
- Lisa Feldman
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, California
| | - Christine Brown
- Department of Cancer Immunotherapy & Tumor Immunology, City of Hope National Medical Center, Duarte, California.,Department of Hematology & Hematopoietic Call Transplantation, City of Hope National Medical Center, Duarte, California
| | - Behnam Badie
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
18
|
Chokshi CR, Brakel BA, Tatari N, Savage N, Salim SK, Venugopal C, Singh SK. Advances in Immunotherapy for Adult Glioblastoma. Cancers (Basel) 2021; 13:cancers13143400. [PMID: 34298615 PMCID: PMC8305609 DOI: 10.3390/cancers13143400] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Therapy failure and disease recurrence are hallmarks of glioblastoma (GBM), the most common and lethal tumor in adults that originates in the brain. Despite aggressive standards of care, tumor recurrence is inevitable with no standardized second-line therapy. Recent clinical studies evaluating therapies that augment the anti-tumor immune response (i.e., immunotherapies) have yielded promising results in subsets of GBM patients. Here, we summarize clinical studies in the past decade that evaluate vaccines, immune checkpoint inhibitors and chimeric antigen receptor (CAR) T cells for treatment of GBM. Although immunotherapies have yet to return widespread efficacy for the majority of GBM patients, critical insights from completed and ongoing clinical trials are informing development of the next generation of therapies, with the goal to alleviate disease burden and extend patient survival. Abstract Despite aggressive multimodal therapy, glioblastoma (GBM) remains the most common malignant primary brain tumor in adults. With the advent of therapies that revitalize the anti-tumor immune response, several immunotherapeutic modalities have been developed for treatment of GBM. In this review, we summarize recent clinical and preclinical efforts to evaluate vaccination strategies, immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells. Although these modalities have shown long-term tumor regression in subsets of treated patients, the underlying biology that may predict efficacy and inform therapy development is being actively investigated. Common to all therapeutic modalities are fundamental mechanisms of therapy evasion by tumor cells, including immense intratumoral heterogeneity, suppression of the tumor immune microenvironment and low mutational burden. These insights have led efforts to design rational combinatorial therapies that can reignite the anti-tumor immune response, effectively and specifically target tumor cells and reliably decrease tumor burden for GBM patients.
Collapse
Affiliation(s)
- Chirayu R. Chokshi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Benjamin A. Brakel
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Nazanin Tatari
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Sabra K. Salim
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Chitra Venugopal
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Sheila K. Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
- Correspondence:
| |
Collapse
|
19
|
Zhu G, Zhang Q, Zhang J, Liu F. Targeting Tumor-Associated Antigen: A Promising CAR-T Therapeutic Strategy for Glioblastoma Treatment. Front Pharmacol 2021; 12:661606. [PMID: 34248623 PMCID: PMC8264285 DOI: 10.3389/fphar.2021.661606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/09/2021] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor T cells (CAR-T) therapy is a prospective therapeutic strategy for blood cancers tumor, especially leukemia, but it is not effective for solid tumors. Glioblastoma (GBM) is a highly immunosuppressive and deadly malignant tumor with poor responses to immunotherapies. Although CAR-T therapeutic strategies were used for glioma in preclinical trials, the current proliferation activity of CAR-T is not sufficient, and malignant glioma usually recruit immunosuppressive cells to form a tumor microenvironment that hinders CAR-T infiltration, depletes CAR-T, and impairs their efficacy. Moreover, specific environments such as hypoxia and nutritional deficiency can hinder the killing effect of CAR-T, limiting their therapeutic effect. The normal brain lack lymphocytes, but CAR-T usually can recognize specific antigens and regulate the tumor immune microenvironment to increase and decrease pro- and anti-inflammatory factors, respectively. This increases the number of T cells and ultimately enhances anti-tumor effects. CAR-T therapy has become an indispensable modality for glioma due to the specific tumor-associated antigens (TAAs). This review describes the characteristics of CAR-T specific antigen recognition and changing tumor immune microenvironment, as well as ongoing research into CAR-T therapy targeting TAAs in GBM and their potential clinical application.
Collapse
Affiliation(s)
- Guidong Zhu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China.,Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan, China
| | - Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China
| |
Collapse
|
20
|
Gatto L, Nunno VD, Franceschi E, Brandes AA. Chimeric antigen receptor macrophage for glioblastoma immunotherapy: the way forward. Immunotherapy 2021; 13:879-883. [PMID: 34078139 DOI: 10.2217/imt-2021-0054] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Lidia Gatto
- Department of Medical Oncology, Azienda USL of Bologna, Bologna, Italy
| | - Vincenzo Di Nunno
- Department of Medical Oncology, Azienda USL of Bologna, Bologna, Italy
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda USL of Bologna, Bologna, Italy
| | | |
Collapse
|
21
|
Daher M, Melo Garcia L, Li Y, Rezvani K. CAR-NK cells: the next wave of cellular therapy for cancer. Clin Transl Immunology 2021; 10:e1274. [PMID: 33959279 PMCID: PMC8080297 DOI: 10.1002/cti2.1274] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
T cells engineered to express chimeric antigen receptors (CARs) have revolutionised the field of cellular therapy for cancer. Despite its success, this strategy has some recognised limitations and toxicities. Hence, there is growing interest in developing novel cellular therapies based on non-αβ T-cell immune effector cells, including NK cells that offer clear advantages in cancer immunotherapy. As a result, NK cells are being explored as an alternative platform for CAR engineering and are becoming recognised as important players in the next generation of cellular therapies targeting cancer. In this review, we highlight preclinical and clinical studies of CAR-NK cells derived from different sources and discuss strategies under investigation to enhance the antitumor activity of these engineered innate immune cells.
Collapse
Affiliation(s)
- May Daher
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Luciana Melo Garcia
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy The University of Texas MD Anderson Cancer Center Houston TX USA
| |
Collapse
|
22
|
Huang B, Li X, Li Y, Zhang J, Zong Z, Zhang H. Current Immunotherapies for Glioblastoma Multiforme. Front Immunol 2021; 11:603911. [PMID: 33767690 PMCID: PMC7986847 DOI: 10.3389/fimmu.2020.603911] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/29/2020] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant tumor found in the central nervous system. Currently, standard treatments in the clinic include maximal safe surgical resection, radiation, and chemotherapy and are mostly limited by low therapeutic efficiency correlated with poor prognosis. Immunotherapy, which predominantly focuses on peptide vaccines, dendritic cell vaccines, chimeric antigen receptor T cells, checkpoint inhibitor therapy, and oncolytic virotherapy, have achieved some promising results in both preclinical and clinical trials. The future of immune therapy for GBM requires an integrated effort with rational combinations of vaccine therapy, cell therapy, and radio- and chemotherapy as well as molecule therapy targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Boyuan Huang
- Department of Neurosurgery, Beijing Electric Power Hospital, Beijing, China
| | - Xuesong Li
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, China
| | - Yuntao Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Guangzhou, China
| | - Jin Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zhitao Zong
- Department of Neurosurgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, China
| | - Hongbo Zhang
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, China.,Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Guangzhou, China.,Department of Neurosurgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, China
| |
Collapse
|
23
|
Abstract
Chimeric antigen receptor T (CAR-T) cells, an immunotherapy that demonstrates marked success in treatment of hematologic malignancies, are an emergent therapeutic for patients with glioblastoma (GBM). GBM CAR-T trials have focused on targeting well-characterized antigens in the pathogenesis of GBM. Early stage trials demonstrate initial success in terms of safety and tolerability. There is preliminary evidence of antitumor activity and localization of the CAR-T product to tumoral sites. There are mixed results regarding patient outcomes. Ongoing GBM CAR-T trials will target novel antigens, explore CAR-T combination therapy, design multivalent CAR constructs, and assess the impact of lymphodepletion before CAR-T delivery.
Collapse
Affiliation(s)
- Thilan Tudor
- University of Pennsylvania, 3600 Hamilton Walk, Stemmler Hall, Room 176, Philadelphia, PA 19104
| | - Zev A Binder
- University of Pennsylvania, 3600 Hamilton Walk, Stemmler Hall, Room 176, Philadelphia, PA 19104.
| | - Donald M O'Rourke
- John Templeton, Jr. M.D. Professor in Neurosurgery, Hospital of the University of Pennsylvania, 3400 Spruce St. Philadelphia, PA 19104, USA. https://twitter.com/DrORourke2
| |
Collapse
|
24
|
Kim HJ, Kim DY. Present and Future of Anti-Glioblastoma Therapies: A Deep Look into Molecular Dependencies/Features. Molecules 2020; 25:molecules25204641. [PMID: 33053763 PMCID: PMC7587213 DOI: 10.3390/molecules25204641] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is aggressive malignant tumor residing within the central nervous system. Although the standard treatment options, consisting of surgical resection followed by combined radiochemotherapy, have long been established for patients with GBM, the prognosis is still poor. Despite recent advances in diagnosis, surgical techniques, and therapeutic approaches, the increased patient survival after such interventions is still sub-optimal. The unique characteristics of GBM, including highly infiltrative nature, hard-to-access location (mainly due to the existence of the blood brain barrier), frequent and rapid recurrence, and multiple drug resistance mechanisms, pose challenges to the development of an effective treatment. To overcome current limitations on GBM therapy and devise ideal therapeutic strategies, efforts should focus on an improved molecular understanding of GBM pathogenesis. In this review, we summarize the molecular basis for the development and progression of GBM as well as some emerging therapeutic approaches.
Collapse
Affiliation(s)
- Hyeon Ji Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea;
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41940, Korea
- Correspondence: ; Tel.: +82-53-660-6880
| |
Collapse
|
25
|
An immunohistochemical study of HER2 expression in primary brain tumors. Biomedicine (Taipei) 2020; 10:21-27. [PMID: 33854909 PMCID: PMC7608844 DOI: 10.37796/2211-8039.1001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/18/2019] [Indexed: 12/24/2022] Open
Abstract
Background Primary brain tumors (PBTs) include any tumor in the brain whose prognosis is weak because of their histological characteristics. Aim Herein, this study aimed to assess the HER2 tumor marker frequency in PBTs. Materials and methods This study was done on the samples of primary brain tumor diagnosis from 2008 to 2015. Results Out of 107 patients of brain tumor that had a mean age of 40.4 years (61.7% men), the most common location of the tumor was in the supratentorial region (63.85% cases). The prevalence of high-grade astrocytoma (HGA) and low-grade astrocytoma (LGA) at diagnosis was 43.9% and 37.4%, respectively. With regard to HER2 score, HER2-positive (scores 2 & 3) was in 42.1% of patients. On the other hand, HER2-negative (−) was in 57.9%, 2+ in 33.6%, and 3+ in 8.4% of patients. The patients of LGA had significantly younger ages, lower HER2 positivity, and lower HER2 percent compared with the HGA patients. Conclusions The type of brain tumors can impact on HER2 expression that high HER2 expression in HGA may be helpful for therapeutic aims. Further studies are required to support these results with a higher volume of patients in the world.
Collapse
|
26
|
Salinas RD, Durgin JS, O'Rourke DM. Potential of Glioblastoma-Targeted Chimeric Antigen Receptor (CAR) T-Cell Therapy. CNS Drugs 2020; 34:127-145. [PMID: 31916100 DOI: 10.1007/s40263-019-00687-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the established efficacy of chimeric antigen receptor (CAR) T-cell therapy in hematologic malignancies, translating CAR T therapy to solid tumors has remained investigational. Glioblastoma, the most aggressive and lethal form of primary brain tumor, has recently been among the malignancies being trialed clinically with CAR T cells. Glioblastoma in particular holds several unique features that have hindered clinical translation, including its vast intertumoral and intratumoral heterogeneity, associated immunosuppressive environment, and lack of clear experimental models to predict response and analyze resistant phenotypes. Here, we review the history of CAR T therapy development, its current progress in treating glioblastoma, as well as the current challenges and future directions in establishing CAR T therapy as a viable alternative to the current standard of care. Tremendous efforts are currently ongoing to identify novel CAR targets and target combinations for glioblastoma, to modify T cells to enhance their efficacy and to enable them to resist tumor-mediated immunosuppression, and to utilize adjunct therapies such as lymphodepletion, checkpoint inhibition, and bi-specific engagers to improve CAR T persistence. Furthermore, new preclinical models of CAR T therapy are being developed that better reflect the clinical features seen in human trials. Current clinical trials that rapidly incorporate key preclinical findings to patient translation are emerging.
Collapse
Affiliation(s)
- Ryan D Salinas
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Joseph S Durgin
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Glioblastoma Translational Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
27
|
Shahsavari S, Shaghaghi Z, Abedi SM, Hosseinimehr SJ. Evaluation of 99mTc-HYNIC-(ser)3-LTVPWY peptide for glioblastoma imaging. Int J Radiat Biol 2019; 96:502-509. [DOI: 10.1080/09553002.2020.1704906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Shima Shahsavari
- Faculty of Pharmacy, Department of Radiopharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Faculty of Pharmacy, Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Shaghaghi
- Faculty of Pharmacy, Department of Radiopharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Nuclear Medicine and Molecular Imaging, Clinical Development Research Unit of Farshchian Heart Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Mohammad Abedi
- Faculty of Medicine, Department of Radiology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Faculty of Pharmacy, Department of Radiopharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
28
|
Burger MC, Zhang C, Harter PN, Romanski A, Strassheimer F, Senft C, Tonn T, Steinbach JP, Wels WS. CAR-Engineered NK Cells for the Treatment of Glioblastoma: Turning Innate Effectors Into Precision Tools for Cancer Immunotherapy. Front Immunol 2019; 10:2683. [PMID: 31798595 PMCID: PMC6868035 DOI: 10.3389/fimmu.2019.02683] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/31/2019] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GB) is the most common and aggressive primary brain tumor in adults and currently incurable. Despite multimodal treatment regimens, median survival in unselected patient cohorts is <1 year, and recurrence remains almost inevitable. Escape from immune surveillance is thought to contribute to the development and progression of GB. While GB tumors are frequently infiltrated by natural killer (NK) cells, these are actively suppressed by the GB cells and the GB tumor microenvironment. Nevertheless, ex vivo activation with cytokines can restore cytolytic activity of NK cells against GB, indicating that NK cells have potential for adoptive immunotherapy of GB if potent cytotoxicity can be maintained in vivo. NK cells contribute to cancer immune surveillance not only by their direct natural cytotoxicity which is triggered rapidly upon stimulation through germline-encoded cell surface receptors, but also by modulating T-cell mediated antitumor immune responses through maintaining the quality of dendritic cells and enhancing the presentation of tumor antigens. Furthermore, similar to T cells, specific recognition and elimination of cancer cells by NK cells can be markedly enhanced through expression of chimeric antigen receptors (CARs), which provides an opportunity to generate NK-cell therapeutics of defined specificity for cancer immunotherapy. Here, we discuss effects of the GB tumor microenvironment on NK-cell functionality, summarize early treatment attempts with ex vivo activated NK cells, and describe relevant CAR target antigens validated with CAR-T cells. We then outline preclinical approaches that employ CAR-NK cells for GB immunotherapy, and give an overview on the ongoing clinical development of ErbB2 (HER2)-specific CAR-NK cells currently applied in a phase I clinical trial in glioblastoma patients.
Collapse
Affiliation(s)
- Michael C Burger
- Institute for Neurooncology, Goethe University, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Congcong Zhang
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Patrick N Harter
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Annette Romanski
- German Red Cross Blood Donation Service Baden-Württemberg-Hessen, Frankfurt am Main, Germany
| | - Florian Strassheimer
- Institute for Neurooncology, Goethe University, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Christian Senft
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Torsten Tonn
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Red Cross Blood Donation Service North-East, Dresden, Germany.,Transfusion Medicine, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Joachim P Steinbach
- Institute for Neurooncology, Goethe University, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Winfried S Wels
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Vengoji R, Macha MA, Nimmakayala RK, Rachagani S, Siddiqui JA, Mallya K, Gorantla S, Jain M, Ponnusamy MP, Batra SK, Shonka N. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:266. [PMID: 31215502 PMCID: PMC6582495 DOI: 10.1186/s13046-019-1264-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022]
Abstract
Background Glioblastoma (GBM) is an aggressive brain tumor with universal recurrence and poor prognosis. The recurrence is largely driven by chemoradiation resistant cancer stem cells (CSCs). Epidermal growth factor receptor (EGFR) and its mutant EGFRvIII are amplified in ~ 60% and ~ 30% of GBM patients, respectively; however, therapies targeting EGFR have failed to improve disease outcome. EGFRvIII-mediated cross-activation of tyrosine kinase receptor, cMET, regulates GBM CSC maintenance and promote tumor recurrence. Here, we evaluated the efficacy of pan-EGFR inhibitor afatinib and Temozolomide (TMZ) combination on GBM in vitro and in vivo. Methods We analyzed the effect of afatinib and temozolomide (TMZ) combination on GBM cells U87MG and U251 engineered to express wild type (WT) EGFR, EGFRvIII or EGFRvIII dead kinase, CSCs isolated from U87 and U87EGFRvIII in vitro. The therapeutic utility of the drug combination was investigated on tumor growth and progression using intracranially injected U87EGFRvIII GBM xenografts. Results Afatinib and TMZ combination synergistically inhibited the proliferation, clonogenic survival, motility, invasion and induced senescence of GBM cells compared to monotherapy. Mechanistically, afatinib decreased U87EGFRvIII GBM cell proliferation and motility/invasion by inhibiting EGFRvIII/AKT, EGFRvIII/JAK2/STAT3, and focal adhesion kinase (FAK) signaling pathways respectively. Interestingly, afatinib specifically inhibited EGFRvIII-cMET crosstalk in CSCs, resulting in decreased expression of Nanog and Oct3/4, and in combination with TMZ significantly decreased their self-renewal property in vitro. More interestingly, afatinib and TMZ combination significantly decreased the xenograft growth and progression compared to single drug alone. Conclusion Our study demonstrated significant inhibition of GBM tumorigenicity, CSC maintenance in vitro, and delayed tumor growth and progression in vivo by combination of afatinib and TMZ. Our results warrant evaluation of this drug combination in EGFR and EGFRvIII amplified GBM patients. Electronic supplementary material The online version of this article (10.1186/s13046-019-1264-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
30
|
Filley AC, Henriquez M, Dey M. CART Immunotherapy: Development, Success, and Translation to Malignant Gliomas and Other Solid Tumors. Front Oncol 2018; 8:453. [PMID: 30386740 PMCID: PMC6199385 DOI: 10.3389/fonc.2018.00453] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/26/2018] [Indexed: 12/26/2022] Open
Abstract
T cell chimeric antigen receptor (CAR) technology has allowed for the introduction of a high degree of tumor selectivity into adoptive cell transfer therapies. Evolution of this technology has produced a robust antitumor immunotherapeutic strategy that has resulted in dramatic outcomes in liquid cancers. CAR-expressing T-cells (CARTs) targeting CD19 and CD20 have been successfully used in the treatment of hematologic malignancies, producing sustained tumor regressions in a majority of treated patients. These encouraging results have led to a historic and unprecedented FDA approval of CTL019, Novartis' CAR T-cell therapy for the treatment of children and young adults with relapsed or refractory B-cell acute lymphoblastic leukemia (ALL). However, the translation of this technology to solid tumors, like malignant gliomas (MG), has thus far been unsuccessful. This review provides a timely analysis of the factors leading to the success of CART immunotherapy in the setting of hematologic malignancies, barriers limiting its success in the treatment of solid tumors, and approaches to overcome these challenges and allow the application of CART immunotherapy as a treatment modality for refractory tumors, like malignant gliomas, that are in desperate need of effective therapies.
Collapse
Affiliation(s)
- Anna C Filley
- Department of Neurosurgery, IU Simon Cancer Center, IU School of Medicine, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Mario Henriquez
- Department of Neurosurgery, IU Simon Cancer Center, IU School of Medicine, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Mahua Dey
- Department of Neurosurgery, IU Simon Cancer Center, IU School of Medicine, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| |
Collapse
|
31
|
Prinzing BL, Gottschalk SM, Krenciute G. CAR T-cell therapy for glioblastoma: ready for the next round of clinical testing? Expert Rev Anticancer Ther 2018; 18:451-461. [PMID: 29533108 PMCID: PMC6191291 DOI: 10.1080/14737140.2018.1451749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The outcome for patients with glioblastoma (GBM) remains poor, and there is an urgent need to develop novel therapeutic approaches. T cells genetically modified with chimeric antigen receptors (CARs) hold the promise to improve outcomes since they recognize and kill cells through different mechanisms than conventional therapeutics. Areas covered: This article reviews CAR design, tumor associated antigens expressed by GBMs that can be targeted with CAR T cells, preclinical and clinical studies conducted with CAR T cells, and genetic approaches to enhance their effector function. Expert commentary: While preclinical studies have highlighted the potent anti-GBM activity of CAR T cells, the initial foray of CAR T-cell therapies into the clinic resulted only in limited benefits for GBM patients. Additional genetic modification of CAR T cells has resulted in a significant increase in their anti-GBM activity in preclinical models. We are optimistic that clinical testing of these enhanced CAR T cells will be safe and result in improved anti-glioma activity in GBM patients.
Collapse
Affiliation(s)
- Brooke L. Prinzing
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, Texas 77030
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Stephen M. Gottschalk
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Giedre Krenciute
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
32
|
Schneider JR, Kwan K, Boockvar JA. Use of HER2-Specific Chimeric Antigen Receptor-Modified Virus-Specific T Cells as a Potential Therapeutic for Progressive HER2-Positive Glioblastoma. Neurosurgery 2017; 81:N42-N43. [PMID: 29088463 DOI: 10.1093/neuros/nyx449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Julia R Schneider
- Brain Tumor Center Department of Neurosurgery Lenox Hill Hospital & Hofstra Northwell School of Medicine New York, New York
| | - Kevin Kwan
- Brain Tumor Center Department of Neurosurgery Lenox Hill Hospital & Hofstra Northwell School of Medicine New York, New York
| | - John A Boockvar
- Brain Tumor Center Department of Neurosurgery Lenox Hill Hospital & Hofstra Northwell School of Medicine New York, New York.,New York Head and Neck Institute Hofstra Northwell School of Medicine New York, New York
| |
Collapse
|
33
|
Evolution of anti-HER2 therapies for cancer treatment. Cancer Treat Rev 2017; 59:1-21. [PMID: 28715775 DOI: 10.1016/j.ctrv.2017.06.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 01/13/2023]
Abstract
The development of HER2-directed monoclonal antibodies and tyrosine kinase inhibitors have provided benefits to cancer patients, as well as produced many insights into the biology of the ErbB receptor family. Current therapies based on ErbB family members have resulted in improved overall survival with associated improvements in quality of life for the cancer patients that respond to treatment. Compared to monotherapy using either two antibodies to block the HER2 receptor blockade or combinatorial approaches with HER2 antibodies and standard therapies has provided additional benefits. Despite the therapeutic success of existing HER2 therapies, personalising treatment and overcoming resistance to these therapies remains a significant challenge. The heterogeneous intra-tumoural HER2 expression and lack of fully predictive and prognostic biomarkers remain significant barriers to improving the use of HER2 antibodies. Imaging modalities using radiolabelled pertuzumab and trastuzumab allow quantitative assessment of intra-tumoural HER2 expression, HER2 antibody saturation and the success of different drug delivery systems to be assessed. Molecular imaging with HER2 antibodies has the potential to be a non-invasive, predictive and prognostic technique capable of influencing therapeutic decisions, predicting response and failure of treatments as well as providing insights into receptor recycling and signalling. Similarly, conjugating HER2 antibodies with novel toxic payloads or combining HER2 antibodies with cellular immunotherapy provide exciting new opportunities for the management of tumours overexpressing HER2. Future research will lead to higher therapeutic responses, lower toxicities and providing insight into the mechanisms of resistance to HER2-targeted treatments.
Collapse
|
34
|
Zhang L, Ren J, Zhang H, Cheng G, Xu Y, Yang S, Dong C, Fang D, Zhang J, Yang A. HER2-targeted recombinant protein immuno-caspase-6 effectively induces apoptosis in HER2-overexpressing GBM cells in vitro and in vivo. Oncol Rep 2016; 36:2689-2696. [DOI: 10.3892/or.2016.5088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 07/18/2016] [Indexed: 11/05/2022] Open
|
35
|
Abstract
Gliomas are the most common primary brain tumors of the central nervous system, and carry a grim prognosis. Novel approaches utilizing the immune system as adjuvant therapy are quickly emerging as viable and effective options. Immunotherapeutic strategies being investigated to treat glioblastoma include: vaccination therapy targeted against either specific tumor antigens or whole tumor lysate, adoptive cellular therapy with cytotoxic T lymphocytes, chimeric antigen receptors and bi-specific T-cell engaging antibodies allowing circumvention of major histocompatibility complex restriction, aptamer therapy with aims for more efficient target delivery, and checkpoint blockade in order to release the tumor-mediated inhibition of the immune system. Given the heterogeneity of glioblastoma and its ability to gain mutations throughout the disease course, multifaceted treatment strategies utilizing multiple forms of immunotherapy in combination with conventional therapy will be most likely to succeed moving forward.
Collapse
Affiliation(s)
- Brandon D Liebelt
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Houston Methodist Neurological Institute, Houston, TX, USA
| | - Gaetano Finocchiaro
- Department of Neuro-oncology, IRCCS Istituto Neurologico Besta, Milan, Italy
| | - Amy B Heimberger
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
36
|
Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, Hattingen E, Harter PN, Mittelbronn M, Tonn T, Steinbach JP, Wels WS. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J Natl Cancer Inst 2015; 108:djv375. [PMID: 26640245 DOI: 10.1093/jnci/djv375] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/02/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and malignant intracranial tumor in adults and currently incurable. To specifically target natural killer (NK) cell activity to GBM, we employed NK-92/5.28.z cells that are continuously expanding human NK cells expressing an ErbB2-specific chimeric antigen receptor (CAR). METHODS ErbB2 expression in 56 primary tumors, four primary cell cultures, and seven established cell lines was assessed by immunohistochemistry and flow cytometry. Cell killing activity of NK-92/5.28.z cells was analyzed in in vitro cytotoxicity assays. In vivo antitumor activity was evaluated in NOD-SCID IL2Rγ(null) (NSG) mice carrying orthotopic human GBM xenografts (6 to 11 mice per group) and C57BL/6 mice carrying subcutaneous and orthotopic ErbB2-expressing murine GBM tumors (5 to 8 mice per group). Statistical tests were two-sided. RESULTS We found elevated ErbB2 protein expression in 41% of primary GBM samples and in the majority of GBM cell lines investigated. In in vitro assays, NK-92/5.28.z in contrast to untargeted NK-92 cells lysed all ErbB2-positive established and primary GBM cells analyzed. Potent in vivo antitumor activity of NK-92/5.28.z was observed in orthotopic GBM xenograft models in NSG mice, leading to a marked extension of symptom-free survival upon repeated stereotactic injection of CAR NK cells into the tumor area (median survival of 200.5 days upon treatment with NK-92/5.28.z vs 73 days upon treatment with parental NK-92 cells, P < .001). In immunocompetent mice, local therapy with NK-92/5.28.z cells resulted in cures of transplanted syngeneic GBM in four of five mice carrying subcutaneous tumors and five of eight mice carrying intracranial tumors, induction of endogenous antitumor immunity, and long-term protection against tumor rechallenge at distant sites. CONCLUSIONS Our data demonstrate the potential of ErbB2-specific NK-92/5.28.z cells for adoptive immunotherapy of glioblastoma, justifying evaluation of this approach for the treatment of ErbB2-positive GBM in clinical studies.
Collapse
Affiliation(s)
- Congcong Zhang
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Michael C Burger
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Lukas Jennewein
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Sabrina Genßler
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Kurt Schönfeld
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Pia Zeiner
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Elke Hattingen
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Patrick N Harter
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Michel Mittelbronn
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Torsten Tonn
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Joachim P Steinbach
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Winfried S Wels
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| |
Collapse
|
37
|
Abstract
BRCA is a tumor suppressor gene implicated in the major mechanisms of cellular stability in every type of cell. Its mutations are described in numerous cancers, mainly breast and ovarian in women. It was also found an increase of lifetime risk of pancreas, colon, prostate cancer or lymphoma in men carriers. We report the cases of two female patients aged 40 and 58-years-old female patients and one 35-years-old male patient, with brain or medullar gliomas, carriers of a germline mutation of BRCA gene. Those gliomas were particularly aggressive and were not responding to the standard treatment, with chemo and radiotherapy. The very unusual characteristics in location and evolutive profile of these central nervous system tumors raise the question of a genetical underlying mechanism, maybe linked to the BRCA gene mutation that carry these patients. In addition, a non-fortuitous association between germline mutation of BRCA and occurrence of a glioma can be evoked according to the embryological, epidemiological and biomolecular findings noted in the literature. Other clinical and experimental studies are necessary to precise the physiopathological link existing between BRCA mutations and the occurrence of a glioma; this could have therapeutical and clinical implications in the future.
Collapse
|
38
|
Fan Y, Potdar AA, Gong Y, Eswarappa SM, Donnola S, Lathia JD, Hambardzumyan D, Rich JN, Fox PL. Profilin-1 phosphorylation directs angiocrine expression and glioblastoma progression through HIF-1α accumulation. Nat Cell Biol 2014; 16:445-56. [PMID: 24747440 PMCID: PMC4036069 DOI: 10.1038/ncb2954] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/20/2014] [Indexed: 12/18/2022]
Abstract
The tumour vascular microenvironment supports tumorigenesis not only by supplying oxygen and diffusible nutrients but also by secreting soluble factors that promote tumorigenesis. Here we identify a feedforward mechanism in which endothelial cells (ECs), in response to tumour-derived mediators, release angiocrines driving aberrant vascularization and glioblastoma multiforme (GBM) progression through a hypoxia-independent induction of hypoxia-inducible factor (HIF)-1α. Phosphorylation of profilin-1 (Pfn-1) at Tyr 129 in ECs induces binding to the tumour suppressor protein von Hippel-Lindau (VHL), and prevents VHL-mediated degradation of prolyl-hydroxylated HIF-1α, culminating in HIF-1α accumulation even in normoxia. Elevated HIF-1α induces expression of multiple angiogenic factors, leading to vascular abnormality and tumour progression. In a genetic model of GBM, mice with an EC-specific defect in Pfn-1 phosphorylation exhibit reduced tumour angiogenesis, normalized vasculature and improved survival. Moreover, EC-specific Pfn-1 phosphorylation is associated with tumour aggressiveness in human glioma. These findings suggest that targeting Pfn-1 phosphorylation may offer a selective strategy for therapeutic intervention of malignant solid tumours.
Collapse
Affiliation(s)
- Yi Fan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania, USA 19104
| | - Alka A. Potdar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio, USA 44106
| | - Yanqing Gong
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania, USA 19104
| | - Sandeepa M. Eswarappa
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Shannon Donnola
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Justin D. Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Dolores Hambardzumyan
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Jeremy N. Rich
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| | - Paul L. Fox
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio, USA 44195
| |
Collapse
|
39
|
Yan M, Parker BA, Schwab R, Kurzrock R. HER2 aberrations in cancer: implications for therapy. Cancer Treat Rev 2014; 40:770-80. [PMID: 24656976 DOI: 10.1016/j.ctrv.2014.02.008] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 02/25/2014] [Accepted: 02/27/2014] [Indexed: 02/08/2023]
Abstract
Although anti-HER2 (human epidermal growth factor receptor 2) therapy is currently approved for breast, gastric, and gastroesophageal cancers overexpressing the HER2 protein or amplified for the HER2 gene, HER2 aberrations (gene amplification, gene mutations, and protein overexpression) are reported in other diverse malignancies. Indeed, about 1-37% of tumors of the following types harbor HER2 aberrations: bladder, cervix, colon, endometrium, germ cell, glioblastoma, head and neck, liver, lung, ovarian, pancreas, and salivary duct. Four HER2-targeted therapies have been approved for HER2-positive breast cancer: two antibodies (trastuzumab and pertuzumab), an antibody-drug conjugate (ado-trastuzumab emtansine), and a small molecule kinase inhibitor (lapatinib). In addition, afatinib, a small molecule kinase inhibitor that causes irreversible inhibition of EGFR (epidermal growth factor receptor) and HER2, was recently approved for EGFR-mutated non-small cell lung cancer. A large number of novel HER2-targeted agents are also in clinical trials. Herein we discuss the state of the art in understanding and targeting HER2 across anatomic tumor types.
Collapse
Affiliation(s)
- Min Yan
- Division of Hematology and Oncology, University of California, Moores Cancer Center, United States.
| | - Barbara A Parker
- Division of Hematology and Oncology, University of California, Moores Cancer Center, United States
| | - Richard Schwab
- Division of Hematology and Oncology, University of California, Moores Cancer Center, United States
| | - Razelle Kurzrock
- Division of Hematology and Oncology, University of California, Moores Cancer Center, United States
| |
Collapse
|
40
|
Pavlova GV, Baklaushev VP, Ivanova MA, Goriaĭnov SA, Rybalkina EI, Kopylov AM, Chekhonin VP, Potapov AA, Konovalov AN. Modern molecular approaches to diagnosis and treatment of high-grade brain gliomas. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2014; 78:85-100. [PMID: 25874291 DOI: 10.17116/neiro201478685-100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The review analyzes the current state of the problem of diagnosis and therapy of high-grade gliomas on the basis of the most promising present-day approaches. The diagnostic and treatment perspectives of the molecular genetic analysis of glioblastoma markers located on the tumor cell surface are considered. Gene therapy and the use of dendritic cells and oncolytic viruses are considered as the most interesting approaches to therapy of high-grade gliomas.
Collapse
Affiliation(s)
| | - V P Baklaushev
- Natsional'nyĭ issledovatel'skiĭ meditsinskiĭ universitet im. N.I. Pirogova Minzdrava Rossii; Gosudarstvennyĭ nauchnyĭ tsentr sotsial'noĭ i sudebnoĭ psikhiatrii im. V.P. Serbskogo Minzdrava Rossii
| | - M A Ivanova
- Khimicheskiĭ fakul'tet Moskovskogo gosudarstvennogo universiteta im. M.V. Lomonosova
| | - S A Goriaĭnov
- FGBNU "Nauchno-issledovatel'skiĭ institut neĭrokhirurgii im. akad. N.N. Burdenko"
| | - E Iu Rybalkina
- FGBNU "Rossiĭskiĭ onkologicheskiĭ nauchnyĭ tsentr im. N.N. Blokhina", Moskva
| | - A M Kopylov
- Khimicheskiĭ fakul'tet Moskovskogo gosudarstvennogo universiteta im. M.V. Lomonosova
| | - V P Chekhonin
- Natsional'nyĭ issledovatel'skiĭ meditsinskiĭ universitet im. N.I. Pirogova Minzdrava Rossii; Gosudarstvennyĭ nauchnyĭ tsentr sotsial'noĭ i sudebnoĭ psikhiatrii im. V.P. Serbskogo Minzdrava Rossii
| | - A A Potapov
- FGBNU "Nauchno-issledovatel'skiĭ institut neĭrokhirurgii im. akad. N.N. Burdenko"
| | - A N Konovalov
- FGBNU "Nauchno-issledovatel'skiĭ institut neĭrokhirurgii im. akad. N.N. Burdenko"
| |
Collapse
|
41
|
Waage IS, Vreim I, Torp SH. C-erbB2/HER2 in human gliomas, medulloblastomas, and meningiomas: a minireview. Int J Surg Pathol 2013; 21:573-82. [PMID: 23842006 DOI: 10.1177/1066896913492196] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
C-erbB2/HER2 serves as an important prognostic and predictive biomarker in various human tumors, especially in breast cancer, whereas its role in human intracranial tumors is more uncertain. We therefore performed a search in PubMed to get an update. This literature review comprises immunohistochemical studies on the clinical significance of c-erbB2/HER2 overexpression in gliomas, medulloblastomas, and meningiomas. In general, the findings were discrepant with regard to correlations between overexpression, tumor grade, and prognosis. Use of various antibodies may be a contributing factor to these discrepancies. Standardization of the immunohistochemical procedures is a relevant topic for discussion.
Collapse
|
42
|
Fermented mistletoe extract as a multimodal antitumoral agent in gliomas. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:501796. [PMID: 23133496 PMCID: PMC3485514 DOI: 10.1155/2012/501796] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 09/05/2012] [Indexed: 01/03/2023]
Abstract
In Europe, commercially available extracts from the white-berry mistletoe (Viscum album L.) are widely used as a complementary cancer therapy. Mistletoe lectins have been identified as main active components and exhibit cytotoxic effects as well as immunomodulatory activity. Since it is still not elucidated in detail how mistle toe extracts such as ISCADOR communicate their effects, we analyzed the mechanisms that might be responsible for their antitumoral function on a molecular and functional level. ISCADOR-treated glioblastoma (GBM) cells down-regulate central genes involved in glioblastoma progression and malignancy such as the cytokine TGF-β and matrix-metalloproteinases. Using in vitro glioblastoma/immune cell co-cultivation assays as well as measurement of cell migration and invasion, we could demonstrate that in glioblastoma cells, lectin-rich ISCADOR M and ISCADOR Q significantly enforce NK-cell-mediated GBM cell lysis. Beside its immune stimulatory effect, ISCADOR reduces the migratory and invasive potential of glioblastoma cells. In a syngeneic as well as in a xenograft glioblastoma mouse model, both pretreatment of tumor cells and intratumoral therapy of subcutaneously growing glioblastoma cells with ISCADOR Q showed delayed tumor growth. In conclusion, ISCADOR Q, showing multiple positive effects in the treatment of glioblastoma, may be a candidate for concomitant treatment of this cancer.
Collapse
|
43
|
Differential distribution of erbB receptors in human glioblastoma multiforme: expression of erbB3 in CD133-positive putative cancer stem cells. J Neuropathol Exp Neurol 2010; 69:606-22. [PMID: 20467331 DOI: 10.1097/nen.0b013e3181e00579] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Glioblastomas are the most common primary central nervous system tumors in adults, and they remain resistant to current treatments. erbB1 signaling is frequently altered in glioblastomas, suggesting thaterbB receptor family members may represent targets for molecular therapy. We performed a comprehensive analysis of erbB receptor and ligand expression profiles in a panel of 9 glioblastomas andcompared them to nonneoplastic cerebral tissue containing neocortex and adjacent white matter. Quantitative reverse transcription-polymerase chain reaction and Western blot analysis showed that erbB1signaling and erbB2 receptors exhibited highly variable deregulation profiles in the tumors, with patterns ranging from underexpression to overexpression; in contrast, erbB3 and erbB4 were downregulated. We next performed immunohistochemistry to determinethe distribution patterns of erbB receptors among the main neuralcell types in the tumors with special reference to the putative tumor stem cell population. Results revealed intertumoral and intratumoral heterogeneity in all 4 erbB expression profiles, but each receptor exhibited a distinct distribution pattern among glial fibrillary acidic protein-, Olig2-, NeuN-, and CD133-positive populations. Although erbB1 immunoreactivity was detected in only small subsets of CD133-positive putative tumor stem cells, erbB3 immunoreactivity was prominent in this population, suggesting that erbB3 may represent a new potential therapeutic target.
Collapse
|
44
|
Gulati S, Ytterhus B, Granli US, Gulati M, Lydersen S, Torp SH. Overexpression of c-erbB2 is a negative prognostic factor in anaplastic astrocytomas. Diagn Pathol 2010; 5:18. [PMID: 20331873 PMCID: PMC2859381 DOI: 10.1186/1746-1596-5-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 03/23/2010] [Indexed: 12/04/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) family, consisting of four tyrosine kinase receptors, c-erbB1-4, seems to be influential in gliomagenesis. The aim of this study was to investigate EGFR gene amplification and expression of c-erbB1-4 receptor proteins in human anaplastic astrocytomas. Formalin-fixed and paraffin-embedded sections from 31 cases were investigated by standard immunohistochemical procedures for expression of c-erbB1-4 receptor proteins using commercial antibodies. EGFR gene amplification was studied by fluorescence in situ hybridization using paraffin-embedded tissues. Two monoclonal antibodies, NCL-EGFR-384 and NCL-EGFR, were used for EGFR detection and they displayed positive immunoreactivity in 97% and 71%, respectively. For c-erbB2 detection three monoclonal antibodies, CB11, 3B5, and 5A2, were applied and they displayed positive immunoreactivity in 45%, 100%, and 52%, respectively. Positive immunostaining for c-erbB3 and c-erbB4 was encountered in 97% and 74%, respectively. The EGFR gene was amplified in 9 out of 31 tumors (29%). After adjusting for age, Karnofsky performance status, and extent of surgical resection, Cox multiple regression analysis with overall survival as the dependent variable revealed that c-erbB2 overexpression detected by the monoclonal antibody clone CB11 was a statistically significant poor prognostic factor (P = 0.004). This study shows the convenience and feasibility of immunohistochemistry when determining the expression of receptor proteins in tissue sections of human astrocytomas. The synchronous overexpression of c-erbB1-4 proteins in anaplastic astrocytomas supports their role in the pathogenesis of these tumors. Further, c-erbB2 overexpression seems to predict aggressive behaviour.
Collapse
Affiliation(s)
- Sasha Gulati
- Department of Neurosurgery, St Olavs University Hospital, Trondheim, Norway.
| | | | | | | | | | | |
Collapse
|
45
|
Ahmed N, Salsman VS, Kew Y, Shaffer D, Powell S, Zhang YJ, Grossman RG, Heslop HE, Gottschalk S. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res 2010; 16:474-85. [PMID: 20068073 DOI: 10.1158/1078-0432.ccr-09-1322] [Citation(s) in RCA: 305] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most aggressive human primary brain tumor and is currently incurable. Immunotherapies have the potential to target GBM stem cells, which are resistant to conventional therapies. Human epidermal growth factor receptor 2 (HER2) is a validated immunotherapy target, and we determined if HER2-specific T cells can be generated from GBM patients that will target autologous HER2-positive GBMs and their CD133-positive stem cell compartment. EXPERIMENTAL DESIGN HER2-specific T cells from 10 consecutive GBM patients were generated by transduction with a retroviral vector encoding a HER2-specific chimeric antigen receptor. The effector function of HER2-specific T cells against autologous GBM cells, including CD133-positive stem cells, was evaluated in vitro and in an orthotopic murine xenograft model. RESULTS Stimulation of HER2-specific T cells with HER2-positive autologous GBM cells resulted in T-cell proliferation and secretion of IFN-gamma and interleukin-2 in a HER2-dependent manner. Patients' HER2-specific T cells killed CD133-positive and CD133-negative cells derived from primary HER2-positive GBMs, whereas HER2-negative tumor cells were not killed. Injection of HER2-specific T cells induced sustained regression of autologous GBM xenografts established in the brain of severe combined immunodeficient mice. CONCLUSIONS Gene transfer allows the reliable generation of HER2-specific T cells from GBM patients, which have potent antitumor activity against autologous HER2-positive tumors including their putative stem cells. Hence, the adoptive transfer of HER2-redirected T cells may be a promising immunotherapeutic approach for GBM.
Collapse
Affiliation(s)
- Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gardner SL, Ahmed N, Okada H. Immunotherapy for pediatric central nervous system tumors. Biol Blood Marrow Transplant 2009; 16:S75-81. [PMID: 19896544 DOI: 10.1016/j.bbmt.2009.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sharon L Gardner
- Division of Pediatric Hematology/Oncology, Steven D. Hassenfeld Children's Center for Cancer and Blood Disorders, New York University, New York, New York, USA.
| | | | | |
Collapse
|
47
|
Balik V, Mirossay P, Bohus P, Sulla I, Mirossay L, Sarissky M. Flow cytometry analysis of neural differentiation markers expression in human glioblastomas may predict their response to chemotherapy. Cell Mol Neurobiol 2009; 29:845-58. [PMID: 19288188 PMCID: PMC11505807 DOI: 10.1007/s10571-009-9366-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 02/11/2009] [Indexed: 01/28/2023]
Abstract
Glioblastoma multiforme (GBM) represents an extremely chemoresistant tumour type. Here, authors analysed the immunophenotype of GBM tumours by flow cytometry and correlated the immunophenotypic characteristics with sensitivity to chemotherapy. The expression of selected neural and non-neural differentiation markers including A2B5, CD34, CD45, CD56, CD117, CD133, EGFR, GFAP, Her-2/neu, LIFR, nestin, NGFR, Pgp and vimentin was analysed by flow cytometry in eleven GBM (WHO gr.IV) patients. The sensitivity of tumour cells to a panel of chemotherapeutic agents was tested by the MTT assay. All tumours were positive for A2B5, CD56, nestin and vimentin. CD133, EGFR, LIFR, NGFR and Pgp were expressed only by minor tumour cell subpopulations. CD34, CD45, CD117, GFAP and Her-2/neu were constantly negative. Direct correlations were found between the immunophenotypic markers and chemosensitivity: A2B5 vs lomustine (r(2) = 0.642, P = 0.033), CD56 vs cisplatin (r(2) = 0.745, P = 0.013), %Pgp(+) vs vincristine (r(2) = 0.846, P = 0.008), and %NGFR(+) vs daunorubicine (r(2) = 0.672, P = 0.047) and topotecan (r(2) = 0.792, P = 0.011). In contrast, inverse correlations were observed between: EGFR vs paclitaxel (r(2) = -0.676, P = 0.046), CD133 vs dacarbazine (r(2) = -0.636, P = 0.048) and LIFR vs daunorubicine (r(2) = -0.878, P = 0.004). Finally, significant associations were also found among sensitivities to different chemotherapeutic agents and among different immunophenotypic markers. In conclusion, histopathologically identical GBM tumours displayed a marked immunophenotypic heterogeneity. The expression of A2B5, CD56, NGFR and Pgp appeared to be associated with chemoresistance whereas CD133, EGFR and LIFR expression was characteristic of chemosensitive tumours. We suggest that flow cytometric imunophenotypic analysis of GBM may predict chemoresponsiveness and help to identify patients who could potentially benefit from chemotherapy.
Collapse
Affiliation(s)
- Vladimir Balik
- Department of Neurosurgery, Faculty of Medicine and Louis Pasteur University Hospital, P.J. Safarik University, Kosice, Slovak Republic
| | - Peter Mirossay
- Department of Pharmacology, Faculty of Medicine, P.J. Safarik University, Trieda SNP 1, 04011 Kosice, Slovak Republic
| | - Peter Bohus
- Department of Pathology, Louis Pasteur University Hospital, Kosice, Slovak Republic
| | - Igor Sulla
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine, Kosice, Slovak Republic
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, P.J. Safarik University, Trieda SNP 1, 04011 Kosice, Slovak Republic
| | - Marek Sarissky
- Department of Pharmacology, Faculty of Medicine, P.J. Safarik University, Trieda SNP 1, 04011 Kosice, Slovak Republic
| |
Collapse
|
48
|
Rappl A, Piontek G, Schlegel J. EGFR-dependent migration of glial cells is mediated by reorganisation of N-cadherin. J Cell Sci 2008; 121:4089-97. [PMID: 19033391 DOI: 10.1242/jcs.027995] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Receptor tyrosine kinases of the EGFR family exert their various effects on cellular function through the formation of different dimeric receptor complexes. To investigate the functional impact of EGFR-HER2 heterodimers on migration of glial tumour cells, we stably transfected different HER2 constructs, including a constitutively active (HER2VE) and a dominant-negative (HER2VEKA) receptor, in the EGFR-overexpressing human glioma cell line LN18. Interference of EGFR activation through HER2VEKA inhibited cellular migration, whereas EGFR activation through HER2VE increased migration. These results were corroborated by inhibition of EGFR-HER2 signalling with tyrosine kinase inhibitors, because only the blocking of both receptors in HER2VE-cells with the bi-specific inhibitor AEE788 downregulated migration to levels comparable with those in HER2VEKA cells. The non-migratory phenotype was mediated through upregulation of N-cadherin and its recruitment to the cell membrane in HER2VEKA cells; downregulation of N-cadherin by RNAi restored migration in HER2VEKA cells and N-cadherin was also downregulated in migrating HER2VE-cells. Downregulation of N-cadherin levels in the plasma membrane was accompanied by a direct interaction of the EGFR-HER2 and N-cadherin-beta-catenin complexes, leading to tyrosine phosphorylation of beta-catenin. These results indicate that HER2 affects glial-cell migration by modulating EGFR-HER2 signal transduction, and that this effect is mediated by N-cadherin.
Collapse
Affiliation(s)
- Anne Rappl
- Division of Neuropathology, Institute of Pathology, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany
| | | | | |
Collapse
|
49
|
The role of fascin in the migration and invasiveness of malignant glioma cells. Neoplasia 2008; 10:149-59. [PMID: 18283337 DOI: 10.1593/neo.07909] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 11/28/2007] [Accepted: 11/28/2007] [Indexed: 11/18/2022] Open
Abstract
Malignant glioma is the most common primary brain tumor, and its ability to invade the surrounding brain parenchyma is a leading cause of tumor recurrence and treatment failure. Whereas the molecular mechanisms of glioma invasion are incompletely understood, there is growing evidence that cytoskeletal-matrix interactions contribute to this process. Fascin, an actin-bundling protein, induces parallel actin bundles in cell protrusions and increases cell motility in multiple human malignancies. The role of fascin in glioma invasion remains unclear. We demonstrate that fascin is expressed in a panel of human malignant glioma cell lines, and downregulation of fascin expression in glioma cell lines by small interfering RNA (siRNA) is associated with decreased cellular attachment to extracellular matrix (ECM) and reduced migration. Using immunofluorescence analysis, we show that fascin depletion results in a reduced number of filopodia as well as altered glioma cell shape. In vitro invasiveness of U251, U87, and SNB19 glioma cells was inhibited by fascin siRNA treatment by 52.2%, 40.3%, and 23.8% respectively. Finally, we show a decreased invasiveness of U251-GFP cells by fascin knockdown in an ex vivo rat brain slice model system. This is the first study to demonstrate a role for fascin in glioma cell morphology, motility, and invasiveness.
Collapse
|
50
|
Abstract
BACKGROUND The HER-2/neu oncogene encodes for a transmembrane glycoprotein with intracellular tyrosine kinase activity. The HER-2/neu receptor belongs to the family of epidermal growth factor receptors that are crucial in the activation of subcellular signal transduction pathways controlling epithelial cell growth and differentiation. Overexpression of HER-2/neu is observed in 20% to 40% of breast cancers and other solid tumors. Although information is limited, one study suggested that 15% of glioblastoma multiforme (GBM) express HER-2/neu by immunohistochemistry (IHC); gene amplification by fluorescence in situ hybridization (FISH) was not investigated. Studies in this area are potentially significant owing to the role of recombinant monoclonal anti-HER-2/neu antibody traztuzumab (Herceptin) in the treatment of tumors. DESIGN A retrospective clinicopathologic review of 49 patients with GBM with HER-2/neu IHC staining and HER-2/neu gene amplification by FISH was performed. RESULTS The study included 44 patients (17 women, 27 men; age range 20 to 79 y, mean 57.9 y). Initial surgery involved tumor debulking or subtotal resection in 34 patients. Thirty-six patients received adjuvant radiation therapy and 19 patients received adjuvant chemotherapy. At follow-up (range 1.0 to 49.5 mo, mean 10.5 mo), 40 patients died with tumor and 4 patients were lost to follow-up. All tumors were negative for HER-2/neu protein by IHC and for HER-2/neu gene amplification by FISH. CONCLUSIONS No GBM demonstrates HER-2/neu protein by IHC or amplification of the HER-2/neu gene by FISH. The HER-2/neu oncogene does not seem to play a role in the pathogenesis of GBM.
Collapse
|