1
|
Nasir A, Rehman MU, Khan T, Husn M, Khan M, Khan A, Nuh AM, Jiang W, Farooqi HMU, Bai Q. Advances in nanotechnology-assisted photodynamic therapy for neurological disorders: a comprehensive review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:84-103. [PMID: 38235991 DOI: 10.1080/21691401.2024.2304814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
Neurological disorders such as neurodegenerative diseases and nervous system tumours affect more than one billion people throughout the globe. The physiological sensitivity of the nervous tissue limits the application of invasive therapies and leads to poor treatment and prognosis. One promising solution that has generated attention is Photodynamic therapy (PDT), which can potentially revolutionise the treatment landscape for neurological disorders. PDT attracted substantial recognition for anticancer efficacy and drug conjugation for targeted drug delivery. This review thoroughly explained the basic principles of PDT, scientific interventions and advances in PDT, and their complicated mechanism in treating brain-related pathologies. Furthermore, the merits and demerits of PDT in the context of neurological disorders offer a well-rounded perspective on its feasibility and challenges. In conclusion, this review encapsulates the significant potential of PDT in transforming the treatment landscape for neurological disorders, emphasising its role as a non-invasive, targeted therapeutic approach with multifaceted applications.
Collapse
Affiliation(s)
- Abdul Nasir
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mujeeb Ur Rehman
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Tamreez Khan
- Department of Zoology, Abdul Wali Khan University, Mardan, Pakistan
| | - Mansoor Husn
- Department of Biochemistry, Abdul Wali Khan University, Mardan, Pakistan
| | - Manzar Khan
- Department of Zoology, Hazara University Mansehra, Mansehra, Pakistan
| | - Ahmad Khan
- Department of Psychology, University of Karachi, Karachi, Pakistan
| | - Abdifatah Mohamed Nuh
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Qain Bai
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Ramar V, Guo S, Hudson B, Khedri A, Guo AA, Li J, Liu M. Interaction of NF-κB and FOSL1 drives glioma stemness. Cell Mol Life Sci 2024; 81:255. [PMID: 38856747 PMCID: PMC11335291 DOI: 10.1007/s00018-024-05293-1] [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: 01/20/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 06/11/2024]
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant primary brain tumor; GBM's inevitable recurrence suggests that glioblastoma stem cells (GSC) allow these tumors to persist. Our previous work showed that FOSL1, transactivated by the STAT3 gene, functions as a tumorigenic gene in glioma pathogenesis and acts as a diagnostic marker and potential drug target in glioma patients. Accumulating evidence shows that STAT3 and NF-κB cooperate to promote the development and progression of various cancers. The link between STAT3 and NF-κB suggests that NF-κB can also transcriptionally regulate FOSL1 and contribute to gliomagenesis. To investigate downstream molecules of FOSL1, we analyzed the transcriptome after overexpressing FOSL1 in a PDX-L14 line characterized by deficient FOSL1 expression. We then conducted immunohistochemical staining for FOSL1 and NF-κB p65 using rabbit polyclonal anti-FOSL1 and NF-κB p65 in glioma tissue microarrays (TMA) derived from 141 glioma patients and 15 healthy individuals. Next, mutants of the human FOSL1 promoter, featuring mutations in essential binding sites for NF-κB were generated using a Q5 site-directed mutagenesis kit. Subsequently, we examined luciferase activity in glioma cells and compared it to the wild-type FOSL1 promoter. Then, we explored the mutual regulation between NF-κB signaling and FOSL1 by modulating the expression of NF-κB or FOSL1. Subsequently, we assessed the activity of FOSL1 and NF-κB. To understand the role of FOSL1 in cell growth and stemness, we conducted a CCK-8 assay and cell cycle analysis, assessing apoptosis and GSC markers, ALDH1, and CD133 under varying FOSL1 expression conditions. Transcriptome analyses of downstream molecules of FOSL1 show that NF-κB signaling pathway is regulated by FOSL1. NF-κB p65 protein expression correlates to the expression of FOSL1 in glioma patients, and both are associated with glioma grades. NF-κB is a crucial transcription factor activating the FOSL1 promoter in glioma cells. Mutual regulation between NF-κB and FOSL1 contributes to glioma tumorigenesis and stemness through promoting G1/S transition and inhibiting apoptosis. Therefore, the FOSL1 molecular pathway is functionally connected to NF-κB activation, enhances stemness, and is indicative that FOSL1 may potentially be a novel GBM drug target.
Collapse
Affiliation(s)
- Vanajothi Ramar
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Shanchun Guo
- Department of Chemistry, Xavier University, 1 Drexel Dr, New Orleans, LA, USA
| | - Breanna Hudson
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Azam Khedri
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Alyssa A Guo
- Wake Forest University School of Medicine, 475 Vine Street, Winston-Salem, NC, USA
| | - Jason Li
- Wake Forest University School of Medicine, 475 Vine Street, Winston-Salem, NC, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, USA.
| |
Collapse
|
3
|
Leland P, Degheidy H, Lea A, Bauer SR, Puri RK, Joshi BH. Identification and characterisation of novel CAR-T cells to target IL13Rα2 positive human glioma in vitro and in vivo. Clin Transl Med 2024; 14:e1664. [PMID: 38685487 PMCID: PMC11058282 DOI: 10.1002/ctm2.1664] [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: 12/16/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Previously, we discovered that human solid tumours, but not normal human tissues, preferentially overexpress interleukin-13Receptor alpha2, a high binding receptor for IL-13. To develop novel anti-cancer approaches, we constructed a chimeric antigen receptor construct using a high binding and codon optimised scFv-IL-13Rα2 fragment fused with CD3ζ and co-stimulatory cytoplasmic domains of CD28 and 4-1BB. METHODS We developed a scFv clone, designated 14-1, by biopanning the bound scFv phages using huIL-13Rα2Fc chimeric protein and compared its binding with our previously published clone 4-1. We performed bioinformatic analyses for complementary determining regions (CDR) framework and residue analyses of the light and heavy chains. This construct was packaged with helper plasmids to produce CAR-lentivirus and transduced human Jurkat T or activated T cells from peripheral blood mononuclear cells (PBMCs) to produce CAR-T cells and tested for their quality attributes in vitro and in vivo. Serum enzymes including body weight from non-tumour bearing mice were tested for assessing general toxicity of CAR-T cells. RESULTS The binding of 14-1 clone is to IL-13Rα2Fc-chimeric protein is ∼5 times higher than our previous clone 4-1. The 14-1-CAR-T cells grew exponentially in the presence of cytokines and maintained phenotype and biological attributes such as cell viability, potency, migration and T cell activation. Clone 14-1 migrated to IL-13Rα2Fc and cell free supernatants only from IL-13Rα2+ve confluent glioma tumour cells in a chemotaxis assay. scFv-IL-13Rα2-CAR-T cells specifically killed IL-13Rα2+ve but not IL-13Rα2-ve tumour cells in vitro and selectively caused significant release of IFN-γ only from IL-13Rα2+ve co-cultures. These CAR-T cells regressed IL-13Rα2+ve glioma xenografts in vivo without any general toxicity. In contrast, the IL-13Rα2 gene knocked-down U251 and U87 xenografts failed to respond to the CAR-T therapy. CONCLUSION Taken together, we conclude that the novel scFv-IL-13Rα2 CAR-T cell therapy may offer an effective therapeutic option after designing a careful pre-clinical and clinical study.
Collapse
Affiliation(s)
- Pamela Leland
- Tumor Vaccine and Biotechnology BranchDivision of Cell Therapy IISilver SpringMarylandUSA
| | - Heba Degheidy
- Cellular and Tissue Therapy Branch, Office of Cellular Therapy & Human Tissues, Office of Therapeutic ProductsCenter for Biologics Evaluation and ResearchU.S. Food and Drug Administration, White OakSilver SpringMarylandUSA
| | - Ashley Lea
- Tumor Vaccine and Biotechnology BranchDivision of Cell Therapy IISilver SpringMarylandUSA
| | - Steven R. Bauer
- Cellular and Tissue Therapy Branch, Office of Cellular Therapy & Human Tissues, Office of Therapeutic ProductsCenter for Biologics Evaluation and ResearchU.S. Food and Drug Administration, White OakSilver SpringMarylandUSA
- Wake Forest Institute for Regenerative MedicineWinston‐SalemNorth CarolinaUSA
| | - Raj K. Puri
- Tumor Vaccine and Biotechnology BranchDivision of Cell Therapy IISilver SpringMarylandUSA
- Iovance Biotherapeutics, Inc.FrederickMarylandUSA
| | - Bharat H. Joshi
- Tumor Vaccine and Biotechnology BranchDivision of Cell Therapy IISilver SpringMarylandUSA
| |
Collapse
|
4
|
Yu C, Hsieh K, Cherry DR, Nehlsen AD, Resende Salgado L, Lazarev S, Sindhu KK. Immune Escape in Glioblastoma: Mechanisms of Action and Implications for Immune Checkpoint Inhibitors and CAR T-Cell Therapy. BIOLOGY 2023; 12:1528. [PMID: 38132354 PMCID: PMC10741174 DOI: 10.3390/biology12121528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Glioblastoma, the most common primary brain cancer in adults, is characterized by a poor prognosis and resistance to standard treatments. The advent of immunotherapy has revolutionized the treatment of several cancers in recent years but has failed to demonstrate benefit in patients with glioblastoma. Understanding the mechanisms by which glioblastoma exerts tumor-mediated immune suppression in both the tumor microenvironment and the systemic immune landscape is a critical step towards developing effective immunotherapeutic strategies. In this review, we discuss the current understanding of immune escape mechanisms in glioblastoma that compromise the efficacy of immunotherapies, with an emphasis on immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy. In parallel, we review data from preclinical studies that have identified additional therapeutic targets that may enhance overall treatment efficacy in glioblastoma when administered alongside existing immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kunal K. Sindhu
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.Y.); (D.R.C.); (A.D.N.); (L.R.S.); (S.L.)
| |
Collapse
|
5
|
Guo S, Ramar V, Guo AA, Saafir T, Akpobiyeri H, Hudson B, Li J, Liu M. TRPM7 transactivates the FOSL1 gene through STAT3 and enhances glioma stemness. Cell Mol Life Sci 2023; 80:270. [PMID: 37642779 PMCID: PMC10465393 DOI: 10.1007/s00018-023-04921-6] [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: 02/15/2023] [Revised: 07/20/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION We previously reported that TRPM7 regulates glioma cells' stemness through STAT3. In addition, we demonstrated that FOSL1 is a response gene for TRPM7, and the FOSL1 gene serves as an oncogene to promote glioma proliferation and invasion. METHODS In the present study, we determined the effects of FOSL1 on glioma stem cell (GSC) markers CD133 and ALDH1 by flow cytometry, and the maintenance of stem cell activity by extreme limiting dilution assays (ELDA). To further gain insight into the mechanism by which TRPM7 activates transcription of the FOSL1 gene to contribute to glioma stemness, we constructed a FOSL1 promoter and its GAS mutants followed by luciferase reporter assays and ChIP-qPCR in a glioma cell line and glioma patient-derived xenoline. We further examined GSC markers ALDH1 and TRPM7 as well as FOSL1 by immunohistochemistry staining (IHC) in brain tissue microarray (TMA) of glioma patients. RESULTS We revealed that FOSL1 knockdown reduces the expression of GSC markers CD133 and ALDH1, and FOSL1 is required to maintain stem cell activity in glioma cells. The experiments also showed that mutations of - 328 to - 336 and - 378 to - 386 GAS elements markedly reduced FOSL1 promoter activity. Constitutively active STAT3 increased while dominant-negative STAT3 decreased FOSL1 promoter activity. Furthermore, overexpression of TRPM7 enhanced while silencing of TRPM7 reduced FOSL1 promoter activity. ChIP-qPCR assays revealed that STAT3, present in nuclear lysates of glioma cells stimulated by constitutively activated STAT3, can bind to two GAS elements, respectively. We demonstrated that deacetylation of FOSL1 at the Lys-116 residue located within its DNA binding domain led to an increase in FOSL1 transcriptional activity. We found that the expression of TRPM7, ALDH1, and FOSL1 protein is associated with grades of malignant glioma, and TRPM7 protein expression correlates to the expression of ALDH1 and FOSL1 in glioma patients. CONCLUSIONS These combined results demonstrated that TRPM7 induced FOSL1 transcriptional activation, which is mediated by the action of STAT3, a mechanism shown to be important in glioma stemness. These results indicated that FOSL1, similar to GSC markers ALDH1 and TRPM7, is a diagnostic marker and potential drug target for glioma patients.
Collapse
Affiliation(s)
- Shanchun Guo
- Department of Chemistry, Xavier University, 1 Drexel Dr, New Orleans, LA, USA
| | - Vanajothi Ramar
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Alyssa A Guo
- University of South Carolina SOM Greenville, Greenville, SC, USA
| | - Talib Saafir
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Hannah Akpobiyeri
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Breanna Hudson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, USA
| | - Jason Li
- Wake Forest University School of Medicine, 475 Vine Street, Winston-Salem, NC, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, USA.
| |
Collapse
|
6
|
Witusik-Perkowska M, Głowacka P, Pieczonka AM, Świderska E, Pudlarz A, Rachwalski M, Szymańska J, Zakrzewska M, Jaskólski DJ, Szemraj J. Autophagy Inhibition with Chloroquine Increased Pro-Apoptotic Potential of New Aziridine-Hydrazide Hydrazone Derivatives against Glioblastoma Cells. Cells 2023; 12:1906. [PMID: 37508570 PMCID: PMC10378024 DOI: 10.3390/cells12141906] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Tumor therapy escape due to undesired side effects induced by treatment, such as prosurvival autophagy or cellular senescence, is one of the key mechanisms of resistance that eventually leads to tumor dormancy and recurrence. Glioblastoma is the most frequent and practically incurable neoplasm of the central nervous system; thus, new treatment modalities have been investigated to find a solution more effective than the currently applied standards based on temozolomide. The present study examined the newly synthesized compounds of aziridine-hydrazide hydrazone derivatives to determine their antineoplastic potential against glioblastoma cells in vitro. Although the output of our investigation clearly demonstrates their proapoptotic activity, the cytotoxic effect appeared to be blocked by treatment-induced autophagy, the phenomenon also detected in the case of temozolomide action. The addition of an autophagy inhibitor, chloroquine, resulted in a significant increase in apoptosis triggered by the tested compounds, as well as temozolomide. The new aziridine-hydrazide hydrazone derivatives, which present cytotoxic potential against glioblastoma cells comparable to or even higher than that of temozolomide, show promising results and, thus, should be further investigated as antineoplastic agents. Moreover, our findings suggest that the combination of an apoptosis inducer with an autophagy inhibitor could optimize chemotherapeutic efficiency, and the addition of an autophagy inhibitor should be considered as an optional adjunctive therapy minimizing the risk of tumor escape from treatment.
Collapse
Affiliation(s)
- Monika Witusik-Perkowska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Pola Głowacka
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Adam M Pieczonka
- Department of Organic and Applied Chemistry, Faculty of Chemistry, University of Lodz, Tamka 12, 91-403 Lodz, Poland
| | - Ewa Świderska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Agnieszka Pudlarz
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Michał Rachwalski
- Department of Organic and Applied Chemistry, Faculty of Chemistry, University of Lodz, Tamka 12, 91-403 Lodz, Poland
| | - Julia Szymańska
- Department of Organic and Applied Chemistry, Faculty of Chemistry, University of Lodz, Tamka 12, 91-403 Lodz, Poland
| | - Magdalena Zakrzewska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Pomorska 251, 92-216 Lodz, Poland
| | - Dariusz J Jaskólski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| |
Collapse
|
7
|
Santana-Viera L, Dassie JP, Rosàs-Lapeña M, Garcia-Monclús S, Chicón-Bosch M, Pérez-Capó M, Pozo LD, Sanchez-Serra S, Almacellas-Rabaiget O, Maqueda-Marcos S, López-Alemany R, Thiel WH, Giangrande PH, Tirado OM. Combination of protein and cell internalization SELEX identifies a potential RNA therapeutic and delivery platform to treat EphA2-expressing tumors. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:758-772. [PMID: 37251690 PMCID: PMC10213179 DOI: 10.1016/j.omtn.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023]
Abstract
The EphA2 receptor tyrosine kinase is overexpressed in most solid tumors and acts as the major driver of tumorigenesis. In this study, we developed a novel approach for targeting the EphA2 receptor using a 2'-fluoro-modified pyrimidine RNA aptamer termed ATOP. We identified the ATOP EphA2 aptamer using a novel bioinformatics strategy that compared aptamers enriched during a protein SELEX using recombinant human EphA2 and a cell-internalization SELEX using EphA2-expressing MDA231 tumor cells. When applied to EphA2-expressing tumor cell lines, the ATOP EphA2 aptamer attenuated tumor cell migration and clonogenicity. In a mouse model of spontaneous metastasis, the ATOP EphA2 aptamer slowed primary tumor growth and significantly reduced the number of lung metastases. The EphA2 ATOP aptamer represents a promising candidate for the development of next-generation targeted therapies that provide safer and more effective treatment of EphA2-overexpressing tumors.
Collapse
Affiliation(s)
- Laura Santana-Viera
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Justin P. Dassie
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, IA 52242, USA
| | - Marta Rosàs-Lapeña
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Silvia Garcia-Monclús
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Mariona Chicón-Bosch
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Marina Pérez-Capó
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Lidia del Pozo
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Sara Sanchez-Serra
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Olga Almacellas-Rabaiget
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Susana Maqueda-Marcos
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Roser López-Alemany
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - William H. Thiel
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, IA 52242, USA
| | - Paloma H. Giangrande
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, IA 52242, USA
| | - Oscar M. Tirado
- Sarcoma Research Group, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, Oncobell, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- CIBERONC, Carlos III Institute of Health (ISCIII), Madrid, Spain
- Institut Català d’Oncologia (ICO), L’Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
8
|
Lau A, Le N, Nguyen C, Kandpal RP. Signals transduced by Eph receptors and ephrin ligands converge on MAP kinase and AKT pathways in human cancers. Cell Signal 2023; 104:110579. [PMID: 36572189 DOI: 10.1016/j.cellsig.2022.110579] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Eph receptors, the largest known family of receptor tyrosine kinases, and ephrin ligands have been implicated in a variety of human cancers. The novel bidirectional signaling events initiated by binding of Eph receptors to their cognate ephrin ligands modulate many cellular processes such as proliferation, metastasis, angiogenesis, invasion, and apoptosis. The relationships between the abundance of a unique subset of Eph receptors and ephrin ligands with associated cellular processes indicate a key role of these molecules in tumorigenesis. The combinatorial expression of these molecules converges on MAP kinase and/or AKT/mTOR signaling pathways. The intracellular target proteins of the initial signal may, however, vary in some cancers. Furthermore, we have also described the commonality of up- and down-regulation of individual receptors and ligands in various cancers. The current state of research in Eph receptors illustrates MAP kinase and mTOR pathways as plausible targets for therapeutic interventions in various cancers.
Collapse
Affiliation(s)
- Andreas Lau
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Nghia Le
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Claudia Nguyen
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Raj P Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
9
|
Khristov V, Lin A, Freedman Z, Staub J, Shenoy G, Mrowczynski O, Rizk E, Zacharia B, Connor J. Tumor-Derived Biomarkers in Liquid Biopsy of Glioblastoma. World Neurosurg 2023; 170:182-194. [PMID: 36347463 DOI: 10.1016/j.wneu.2022.11.012] [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: 09/13/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/07/2022]
Abstract
There is a pressing clinical need for minimally invasive liquid biopsies to supplement imaging in the treatment of glioblastoma. Diagnostic imaging is often difficult to interpret and the medical community is divided on distinguishing among complete response, partial response, stable disease, and progressive disease. A minimally invasive liquid biopsy would supplement imaging and clinical findings and has the capacity to be helpful in several ways: 1) diagnosis, 2) selection of patients for specific treatments, 3) tracking of treatment response, and 4) prognostic value. The liquid biome is the combination of biological fluids including blood, urine, and cerebrospinal fluid that contain small amounts of tumor cells, DNA/RNA coding material, peptides, and metabolites. Within the liquid biome, 2 broad categories of biomarkers can exist: tumor-derived, which can be directly traced to the tumor, and tumor-associated, which can be traced back to the response of the body to disease. Although tumor-associated biomarkers are promising liquid biopsy candidates, recent advances in biomarker enrichment and detection have allowed concentration on a new class of biomarker: tumor-derived biomarkers. This review focuses on making the distinction between the 2 biomarker categories and highlights promising new direction.
Collapse
Affiliation(s)
- Vladimir Khristov
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA.
| | - Andrea Lin
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - Zachary Freedman
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - Jacob Staub
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - Ganesh Shenoy
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - Oliver Mrowczynski
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - Elias Rizk
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - Brad Zacharia
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| | - James Connor
- Department of Neurosurgery, Penn State Hershey College of Medicne, Hershey, Pennsylvania, USA
| |
Collapse
|
10
|
Knudson KM, Hwang S, McCann MS, Joshi BH, Husain SR, Puri RK. Recent Advances in IL-13Rα2-Directed Cancer Immunotherapy. Front Immunol 2022; 13:878365. [PMID: 35464460 PMCID: PMC9023787 DOI: 10.3389/fimmu.2022.878365] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/17/2022] [Indexed: 01/14/2023] Open
Abstract
Interleukin-13 receptor subunit alpha-2 (IL-13Rα2, CD213A), a high-affinity membrane receptor of the anti-inflammatory Th2 cytokine IL-13, is overexpressed in a variety of solid tumors and is correlated with poor prognosis in glioblastoma, colorectal cancer, adrenocortical carcinoma, pancreatic cancer, and breast cancer. While initially hypothesized as a decoy receptor for IL-13-mediated signaling, recent evidence demonstrates IL-13 can signal through IL-13Rα2 in human cells. In addition, expression of IL-13Rα2 and IL-13Rα2-mediated signaling has been shown to promote tumor proliferation, cell survival, tumor progression, invasion, and metastasis. Given its differential expression in tumor versus normal tissue, IL-13Rα2 is an attractive immunotherapy target, as both a targetable receptor and an immunogenic antigen. Multiple promising strategies, including immunotoxins, cancer vaccines, and chimeric antigen receptor (CAR) T cells, have been developed to target IL-13Rα2. In this mini-review, we discuss recent developments surrounding IL-13Rα2-targeted therapies in pre-clinical and clinical study, including potential strategies to improve IL-13Rα2-directed cancer treatment efficacy.
Collapse
|
11
|
Sobolev VV, Khashukoeva AZ, Evina OE, Geppe NA, Chebysheva SN, Korsunskaya IM, Tchepourina E, Mezentsev A. Role of the Transcription Factor FOSL1 in Organ Development and Tumorigenesis. Int J Mol Sci 2022; 23:1521. [PMID: 35163444 PMCID: PMC8835756 DOI: 10.3390/ijms23031521] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/25/2022] Open
Abstract
The transcription factor FOSL1 plays an important role in cell differentiation and tumorigenesis. Primarily, FOSL1 is crucial for the differentiation of several cell lineages, namely adipocytes, chondrocytes, and osteoblasts. In solid tumors, FOSL1 controls the progression of tumor cells through the epithelial-mesenchymal transformation. In this review, we summarize the available data on FOSL1 expression, stabilization, and degradation in the cell. We discuss how FOSL1 is integrated into the intracellular signaling mechanisms and provide a comprehensive analysis of FOSL1 influence on gene expression. We also analyze the pathological changes caused by altered Fosl1 expression in genetically modified mice. In addition, we dedicated a separate section of the review to the role of FOSL1 in human cancer. Primarily, we focus on the FOSL1 expression pattern in solid tumors, FOSL1 importance as a prognostic factor, and FOSL1 perspectives as a molecular target for anticancer therapy.
Collapse
Affiliation(s)
- Vladimir V. Sobolev
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| | - Asiat Z. Khashukoeva
- Federal State Autonomous Educational Institution of Higher Education, N.I. Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia;
| | - Olga E. Evina
- “JSC DK Medsi”, Medical and Diagnostics Center, 125284 Moscow, Russia;
| | - Natalia A. Geppe
- NF Filatov Clinical Institute of Children’s Health, I.M. Sechenov First MSMU, 119435 Moscow, Russia; (N.A.G.); (S.N.C.)
| | - Svetlana N. Chebysheva
- NF Filatov Clinical Institute of Children’s Health, I.M. Sechenov First MSMU, 119435 Moscow, Russia; (N.A.G.); (S.N.C.)
| | - Irina M. Korsunskaya
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| | - Ekaterina Tchepourina
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| | - Alexandre Mezentsev
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| |
Collapse
|
12
|
Márquez-Ortiz RA, Contreras-Zárate MJ, Tesic V, Alvarez-Eraso KLF, Kwak G, Littrell Z, Costello JC, Sreekanth V, Ormond DR, Karam SD, Kabos P, Cittelly DM. IL13Rα2 Promotes Proliferation and Outgrowth of Breast Cancer Brain Metastases. Clin Cancer Res 2021; 27:6209-6221. [PMID: 34544797 PMCID: PMC8595859 DOI: 10.1158/1078-0432.ccr-21-0361] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE The survival of women with brain metastases (BM) from breast cancer remains very poor, with over 80% dying within a year of their diagnosis. Here, we define the function of IL13Rα2 in outgrowth of breast cancer brain metastases (BCBM) in vitro and in vivo, and postulate IL13Rα2 as a suitable therapeutic target for BM. EXPERIMENTAL DESIGN We performed IHC staining of IL13Rα2 in BCBM to define its prognostic value. Using inducible shRNAs in TNBC and HER2+ breast-brain metastatic models, we assessed IL13Rα2 function in vitro and in vivo. We performed RNAseq and functional studies to define the molecular mechanisms underlying IL13Rα2 function in BCBM. RESULTS High IL13Rα2 expression in BCBM predicted worse survival after BM diagnoses. IL13Rα2 was essential for cancer-cell survival, promoting proliferation while repressing invasion. IL13Rα2 KD resulted in FAK downregulation, repression of cell cycle and proliferation mediators, and upregulation of Ephrin B1 signaling. Ephrin-B1 (i) promoted invasion of BC cells in vitro, (ii) marked micrometastasis and invasive fronts in BCBM, and (iii) predicted shorter disease-free survival and BM-free survival (BMFS) in breast primary tumors known to metastasize to the brain. In experimental metastases models, which bypass early tumor invasion, downregulation of IL13Rα2 before or after tumor seeding and brain intravasation decreased BMs, suggesting that IL13Rα2 and the promotion of a proliferative phenotype is critical to BM progression. CONCLUSIONS Non-genomic phenotypic adaptations at metastatic sites are critical to BM progression and patients' prognosis. This study opens the road to use IL13Rα2 targeting as a therapeutic strategy for BM.
Collapse
Affiliation(s)
| | | | - Vesna Tesic
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | | | - Gina Kwak
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Zachary Littrell
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - James C Costello
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Varsha Sreekanth
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - D Ryan Ormond
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Peter Kabos
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Diana M Cittelly
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
13
|
Sanders S, Herpai DM, Rodriguez A, Huang Y, Chou J, Hsu FC, Seals D, Mott R, Miller LD, Debinski W. The Presence and Potential Role of ALDH1A2 in the Glioblastoma Microenvironment. Cells 2021; 10:2485. [PMID: 34572134 PMCID: PMC8468822 DOI: 10.3390/cells10092485] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive malignant glioma. Therapeutic targeting of GBM is made more difficult due to its heterogeneity, resistance to treatment, and diffuse infiltration into the brain parenchyma. Better understanding of the tumor microenvironment should aid in finding more effective management of GBM. GBM-associated macrophages (GAM) comprise up to 30% of the GBM microenvironment. Therefore, exploration of GAM activity/function and their specific markers are important for developing new therapeutic agents. In this study, we identified and evaluated the expression of ALDH1A2 in the GBM microenvironment, and especially in M2 GAM, though it is also expressed in reactive astrocytes and multinucleated tumor cells. We demonstrated that M2 GAM highly express ALDH1A2 when compared to other ALDH1 family proteins. Additionally, GBM samples showed higher expression of ALDH1A2 when compared to low-grade gliomas (LGG), and this expression was increased upon tumor recurrence both at the gene and protein levels. We demonstrated that the enzymatic product of ALDH1A2, retinoic acid (RA), modulated the expression and activity of MMP-2 and MMP-9 in macrophages, but not in GBM tumor cells. Thus, the expression of ALDH1A2 may promote the progressive phenotype of GBM.
Collapse
Affiliation(s)
- Stephanie Sanders
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA; (S.S.); (D.M.H.); (Y.H.); (J.C.); (L.D.M.)
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston Salem, NC 27157, USA
| | - Denise M. Herpai
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA; (S.S.); (D.M.H.); (Y.H.); (J.C.); (L.D.M.)
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston Salem, NC 27157, USA
| | - Analiz Rodriguez
- Department of Neurosurgery, Jackson T. Stephens Spine and Neuroscience Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yue Huang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA; (S.S.); (D.M.H.); (Y.H.); (J.C.); (L.D.M.)
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston Salem, NC 27157, USA
| | - Jeff Chou
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA; (S.S.); (D.M.H.); (Y.H.); (J.C.); (L.D.M.)
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, NC 27157, USA;
| | - Darren Seals
- Biology Department, Appalachian State University, Boone, NC 28608, USA;
| | - Ryan Mott
- Department of Pathology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA;
| | - Lance D. Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA; (S.S.); (D.M.H.); (Y.H.); (J.C.); (L.D.M.)
| | - Waldemar Debinski
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA; (S.S.); (D.M.H.); (Y.H.); (J.C.); (L.D.M.)
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston Salem, NC 27157, USA
| |
Collapse
|
14
|
Ulasov IV, Borovjagin A, Laevskaya A, Kamynina M, Timashev P, Cerchia L, Rozhkova EA. The IL13α 2R paves the way for anti-glioma nanotherapy. Genes Dis 2021; 10:89-100. [PMID: 37013057 PMCID: PMC10066331 DOI: 10.1016/j.gendis.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive (grade IV) gliomas characterized by a high rate of recurrence, resistance to therapy and a grim survival prognosis. The long-awaited improvement in GBM patients' survival rates essentially depends on advances in the development of new therapeutic approaches. Recent preclinical studies show that nanoscale materials could greatly contribute to the improvement of diagnosis and management of brain cancers. In the current review, we will discuss how specific features of glioma pathobiology can be employed for designing efficient targeting approaches. Moreover, we will summarize the main evidence for the potential of the IL-13R alpha 2 receptor (IL13α2R) targeting in GBM early diagnosis and experimental therapy.
Collapse
Affiliation(s)
- Ilya V. Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Corresponding author.
| | - Anton Borovjagin
- Department of BioMedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, 4 Kosygin St, Moscow 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow 119991, Russia
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples 80131, Italy
| | - Elena A. Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, Argonne, IL 60439, USA
| |
Collapse
|
15
|
Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, Downey KM, Yu W, Carrera DA, Celli A, Cho J, Briones JD, Duecker JM, Goretsky YE, Dannenfelser R, Cardarelli L, Troyanskaya O, Sidhu SS, Roybal KT, Okada H, Lim WA. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med 2021; 13:13/591/eabe7378. [PMID: 33910979 DOI: 10.1126/scitranslmed.abe7378] [Citation(s) in RCA: 246] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/26/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
Treatment of solid cancers with chimeric antigen receptor (CAR) T cells is plagued by the lack of ideal target antigens that are both absolutely tumor specific and homogeneously expressed. We show that multi-antigen prime-and-kill recognition circuits provide flexibility and precision to overcome these challenges in the context of glioblastoma. A synNotch receptor that recognizes a specific priming antigen, such as the heterogeneous but tumor-specific glioblastoma neoantigen epidermal growth factor receptor splice variant III (EGFRvIII) or the central nervous system (CNS) tissue-specific antigen myelin oligodendrocyte glycoprotein (MOG), can be used to locally induce expression of a CAR. This enables thorough but controlled tumor cell killing by targeting antigens that are homogeneous but not absolutely tumor specific. Moreover, synNotch-regulated CAR expression averts tonic signaling and exhaustion, maintaining a higher fraction of the T cells in a naïve/stem cell memory state. In immunodeficient mice bearing intracerebral patient-derived xenografts (PDXs) with heterogeneous expression of EGFRvIII, a single intravenous infusion of EGFRvIII synNotch-CAR T cells demonstrated higher antitumor efficacy and T cell durability than conventional constitutively expressed CAR T cells, without off-tumor killing. T cells transduced with a synNotch-CAR circuit primed by the CNS-specific antigen MOG also exhibited precise and potent control of intracerebral PDX without evidence of priming outside of the brain. In summary, by using circuits that integrate recognition of multiple imperfect but complementary antigens, we improve the specificity, completeness, and persistence of T cells directed against glioblastoma, providing a general recognition strategy applicable to other solid tumors.
Collapse
Affiliation(s)
- Joseph H Choe
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Payal B Watchmaker
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Milos S Simic
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ryan D Gilbert
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Aileen W Li
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nira A Krasnow
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kira M Downey
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Wei Yu
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Diego A Carrera
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Anna Celli
- Department of Veterans' Affairs Medical Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Juhyun Cho
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jessica D Briones
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jason M Duecker
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yitzhar E Goretsky
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ruth Dannenfelser
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA.,Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| | - Lia Cardarelli
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Olga Troyanskaya
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA.,Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| | - Sachdev S Sidhu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Kole T Roybal
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA. .,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.,Helen Diller Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA. .,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94158, USA.,Helen Diller Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Wendell A Lim
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA. .,Helen Diller Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.,Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| |
Collapse
|
16
|
Rossmeisl JH, Herpai D, Quigley M, Cecere TE, Robertson JL, D'Agostino RB, Hinckley J, Tatter SB, Dickinson PJ, Debinski W. Phase I trial of convection-enhanced delivery of IL13RA2 and EPHA2 receptor targeted cytotoxins in dogs with spontaneous intracranial gliomas. Neuro Oncol 2021; 23:422-434. [PMID: 32812637 PMCID: PMC7992889 DOI: 10.1093/neuonc/noaa196] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background The interleukin-13 receptor alpha 2 (IL13RA2) and ephrin type A receptor 2 (EPHA2) are attractive therapeutic targets, being expressed in ~90% of canine and human gliomas, and absent in normal brain. Clinical trials using an earlier generation IL-13 based cytotoxin showed encouraging clinical effects in human glioma, but met with technical barriers associated with the convection-enhanced delivery (CED) method. In this study, IL-13 mutant and ephrin A1 (EFNA1)–based bacterial cytotoxins targeted to IL13RA2 and EPHA2 receptors, respectively, were administered locoregionally by CED to dogs with intracranial gliomas to evaluate their safety and preliminary efficacy. Methods In this phase I, 3 + 3 dose escalation trial, cytotoxins were infused by CED in 17 dogs with gliomas expressing IL13RA2 or EPHA2 receptors. CED was performed using a shape-fitting therapeutic planning algorithm, reflux-preventing catheters, and real-time intraoperative MRI monitoring. The primary endpoint was to determine the maximum tolerated dose of the cytotoxic cocktail in dogs with gliomas. Results Consistent intratumoral delivery of the cytotoxic cocktail was achieved, with a median target coverage of 70% (range, 40–94%). Cytotoxins were well tolerated over a dose range of 0.012–1.278 μg/mL delivered to the target volume (median, 0.099 μg/mL), with no dose limiting toxicities observed. Objective tumor responses, up to 94% tumor volume reduction, were observed in 50% (8/16) of dogs, including at least one dog in each dosing cohort >0.05 μg/mL. Conclusions This study provides preclinical data fundamental to the translation of this multireceptor targeted therapeutic approach to the human clinic.
Collapse
Affiliation(s)
- John H Rossmeisl
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Veterinary and Comparative Neurooncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.,Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.,Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia
| | - Denise Herpai
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina
| | - Mindy Quigley
- Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Thomas E Cecere
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - John L Robertson
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Veterinary and Comparative Neurooncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.,Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia
| | - Ralph B D'Agostino
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Jonathan Hinckley
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina
| | - Stephen B Tatter
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Department of Neurosurgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Peter J Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California (P.J.D.)
| | - Waldemar Debinski
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia.,Department of Cancer Biology of Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
17
|
Zhang X. The Expression Profile and Prognostic Values of EPHA Family Members in Breast Cancer. Front Oncol 2021; 11:619949. [PMID: 34221956 PMCID: PMC8250424 DOI: 10.3389/fonc.2021.619949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/30/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND EphAs are a class of ephrin receptors that belong to the membrane-bound receptor tyrosine kinases group. Accumulating experimental evidence has shown that the EphA family is involved in tumor progression, namely in cell proliferation, invasiveness, and metastasis. EphAs are a promising target for anticancer therapy. However, their role in breast cancer (BC) is still not well understood. MATERIALS AND METHODS We used a series of bioinformatic approaches to analyze the expression of the EphA family members and investigate their prognostic value in BC. RESULTS Lower expression levels of EphA2, EphA3, EphA4, EphA5, and EphA7 and higher expression levels of EphA10 were found in BC tissues compared to those in normal tissues. The expression levels of the EphA family genes were correlated with molecular subtyping but not with tumor stage. High expression levels of most EphAs indicated a better prognosis in BC. CONCLUSIONS This study suggested that EphA2, EphA3, EphA4, and EphA5 can act as tumor-inhibiting factors as well as biomarkers for the prognosis of BC.
Collapse
|
18
|
Hao Y, Li G. Role of EFNA1 in tumorigenesis and prospects for cancer therapy. Biomed Pharmacother 2020; 130:110567. [PMID: 32745910 DOI: 10.1016/j.biopha.2020.110567] [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] [Received: 05/06/2020] [Revised: 07/14/2020] [Accepted: 07/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the major threats to human health. It is of vital importance to reveal the mechanisms of tumorigenesis, identify effective biomarkers and develop novel treatments to improve patient outcome. EFNA1 (ephrinA1) is a member of the EFN family, and it has been studied extensively since its discovery in 1990. Increasing evidence indicates that EFNA1 plays a pivotal role in the pathogenesis of tumors. We provide a detailed overview of the expression and prognostic value of EFNA1 in different types of human malignancies. We briefly discuss the mechanisms of EFNA1 induction in hypoxic environments and its pro-angiogenic function in different cancer cells. We describe the effects of EFNA1 on tumor growth, invasiveness and metastasis. We summarize recent advances in EFNA1-associated cancer therapeutics with emphasis on the prospect of novel anti-tumor methods based on EFNA1.
Collapse
Affiliation(s)
- Yongping Hao
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China.
| | - Guang Li
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China.
| |
Collapse
|
19
|
Wang Q, Han B, Huang W, Qi C, Liu F. Identification of KIF15 as a potential therapeutic target and prognostic factor for glioma. Oncol Rep 2020; 43:1035-1044. [PMID: 32323839 PMCID: PMC7057805 DOI: 10.3892/or.2020.7510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/18/2019] [Indexed: 02/02/2023] Open
Abstract
Glioma is the most commonly diagnosed primary intracranial malignant tumor with rapid growth, easy recurrence and thus poor prognosis. In the present study, the role of kinesin‑12 (KIF15) in glioma was revealed. Immunohistochemical staining and western blot analysis were used to detect the protein expression. An MTT assay was performed to evaluate cell proliferation. Flow cytometric analysis was utilized to assess cell apoptosis and the cell cycle. A mouse xenograft model was constructed for in vivo study. The results indicated that KIF15 was significantly upregulated in glioma tumor tissues and positively correlated with pathological staging, recurrence risk and poor prognosis. Silencing of KIF15 could inhibit cell proliferation and stemness of glioma cells, arrest cells in the G2 phase and induce cell apoptosis. The in vivo study verified the inhibitory effect of KIF15 knockdown on tumor growth. The mechanism study demonstrated the regulation of apoptosis‑ and cycle‑related proteins in the KIF15 KD‑induced inhibition of glioma. KIF15 was revealed to function as a tumor promoter in the development and progression of glioma. KIF15 also served as a prognostic indicator for glioma and may be a therapeutic target for glioma therapy.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Neurosurgery, Nanjing Medical University Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Bin Han
- Department of Neurosurgery, Nanjing Medical University Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Wu Huang
- Department of Neurosurgery, Nanjing Medical University Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Chunjian Qi
- Department of Central Lab, Nanjing Medical University Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Fang Liu
- Department of Neurosurgery, Nanjing Medical University Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
20
|
Shen SH, Woroniecka K, Barbour AB, Fecci PE, Sanchez-Perez L, Sampson JH. CAR T cells and checkpoint inhibition for the treatment of glioblastoma. Expert Opin Biol Ther 2020; 20:579-591. [PMID: 32027536 DOI: 10.1080/14712598.2020.1727436] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Glioblastoma (GBM) is a highly aggressive brain tumor and is one of the most lethal human cancers. Chimeric antigen receptor (CAR) T cell therapy has markedly improved survival in previously incurable disease; however, this vanguard treatment still faces challenges in GBM. Likewise, checkpoint blockade therapies have not enjoyed the same victories against GBM. As it becomes increasingly evident that a mono-therapeutic approach is unlikely to provide anti-tumor efficacy, there evolves a critical need for combined treatment strategies.Areas covered: This review highlights the clinical successes observed with CAR T cell therapy as well the current efforts to overcome its perceived limitations. The review also explores employed combinations of CAR T cell approaches with immune checkpoint blockade strategies, which aim to potentiate immunotherapeutic benefits while restricting the impact of tumor heterogeneity and T cell exhaustion.Expert opinion: Barriers such as tumor heterogeneity and T cell exhaustion have exposed the weaknesses of various mono-immunotherapeutic approaches to GBM, including CAR T cell and checkpoint blockade strategies. Combining these potentially complementary strategies, however, may proffer a rational means of mitigating these barriers and advancing therapeutic successes against GBM and other solid tumors.
Collapse
Affiliation(s)
- Steven H Shen
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Karolina Woroniecka
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Andrew B Barbour
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Pathology, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
21
|
Sharma P, Sonawane P, Herpai D, D’Agostino R, Rossmeisl J, Tatter S, Debinski W. Multireceptor targeting of glioblastoma. Neurooncol Adv 2020; 2:vdaa107. [PMID: 33150335 PMCID: PMC7596893 DOI: 10.1093/noajnl/vdaa107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Treatment for glioblastoma (GBM) remains an unmet need in medicine. Novel therapies that address GBM complexity and heterogeneity in particular are warranted. To this end, we target 4 tumor-associated receptors at a time that span virtually all of the GBM microenvironment including bulk tumor cells, infiltrating tumor cells, neovasculature, and tumor-infiltrating cells with one pharmaceutical agent delivering a cytotoxic load. METHODS We engineered multivalent ligand-based vector proteins termed QUAD with an ability to bind to 4 of the following GBM-associated receptors: IL-13RA2, EphA2, EphA3, and EphB2. We conjugated QUAD with a modified bacterial toxin PE38QQR and tested it in vitro and in vivo. RESULTS The QUAD variants preserved functional characteristics of the respective ligands for the 4 receptors. The QUAD 3.0 variant conjugate was highly cytotoxic to GBM cells, but it was nontoxic in mice, and the conjugate exhibited strong antitumor effect in a dog with spontaneous GBM. CONCLUSION The QUAD addresses, to a large extent, the issues of intra- and intertumoral heterogeneity and, at the same time, it targets several pathophysiologically important tumor compartments in GBM through multiple receptors overexpressed in tumors allowing for what we call "molecular resection." QUAD-based targeted agents warrant further pre- and clinical development.
Collapse
Affiliation(s)
- Puja Sharma
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| | - Poonam Sonawane
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
- Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Denise Herpai
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| | - Ralph D’Agostino
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - John Rossmeisl
- Neurology and Neurosurgery, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Stephen Tatter
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
- Department of Neurosurgery, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Waldemar Debinski
- Brain Tumor Center of Excellence, Wake Forest Baptist Medical Center Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| |
Collapse
|
22
|
Expression and function of FRA1 protein in tumors. Mol Biol Rep 2019; 47:737-752. [PMID: 31612408 DOI: 10.1007/s11033-019-05123-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022]
Abstract
AP-1 is a dimeric complex that is composed of JUN, FOS, ATF and MAF protein families. FOS-related antigen 1 (FRA1) which encoded by FOSL1 gene, belongs to the FOS protein family, and mainly forms an AP-1 complex with the protein of the JUN family to exert an effect. Regulation of FRA1 occurs at levels of transcription and post-translational modification, and phosphorylation is the major post-translational modification. FRA1 is mainly regulated by the mitogen-activated protein kinases signaling pathway and is degraded by ubiquitin-independent proteasomes. FRA1 can affect biological functions, such as tumor proliferation, differentiation, invasion and apoptosis. Studies have demonstrated that FRA1 is abnormally expressed in many tumors and plays a relevant role, but the specific condition varies from the target organs. FRA1 is overexpressed in breast cancer, lung cancer, colorectal cancer, prostate cancer, nasopharyngeal cancer, thyroid cancer and other tumors. However, the expression of FRA1 is decreased in cervical cancer, and the expression of FRA1 in ovarian cancer and oral squamous cell carcinoma is still controversial. In this review, we present a detailed description of the regulatory factors and functions of FRA1, also, the expression of FRA1 in various tumors and its function in relative tumor.
Collapse
|
23
|
Targeted Delivery of Nanoparticulate Cytochrome C into Glioma Cells Through the Proton-Coupled Folate Transporter. Biomolecules 2019; 9:biom9040154. [PMID: 31003476 PMCID: PMC6523331 DOI: 10.3390/biom9040154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 01/14/2023] Open
Abstract
In this study, we identified the proton-coupled folate transporter (PCFT) as a route for targeted delivery of drugs to some gliomas. Using the techniques of confocal imaging, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and small interfering (siRNA) knockdown against the PCFT, we demonstrated that Gl261 and A172 glioma cells, but not U87 and primary cultured astrocytes, express the PCFT, which provides selective internalization of folic acid (FA)-conjugated cytochrome c-containing nanoparticles (FA-Cyt c NPs), followed by cell death. The FA-Cyt c NPs (100 µg/mL), had no cytotoxic effects in astrocytes but caused death in glioma cells, according to their level of expression of PCFT. Whole-cell patch clamp recording revealed FA-induced membrane currents in FA-Cyt c NPs-sensitive gliomas, that were reduced by siRNA PCFT knockdown in a similar manner as by application of FA-Cyt c NPs, indicating that the PCFT is a route for internalization of FA-conjugated NPs in these glioma cells. Analysis of human glioblastoma specimens revealed that at least 25% of glioblastomas express elevated level of either PCFT or folate receptor (FOLR1). We conclude that the PCFT provides a mechanism for targeted delivery of drugs to some gliomas as a starting point for the development of efficient methods for treating gliomas with high expression of PCFT and/or FOLR1.
Collapse
|
24
|
Irradiation to Improve the Response to Immunotherapeutic Agents in Glioblastomas. Adv Radiat Oncol 2018; 4:268-282. [PMID: 31011672 PMCID: PMC6460102 DOI: 10.1016/j.adro.2018.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022] Open
Abstract
Purpose Glioblastoma (GBM) remains an incurable disease despite extensive treatment with surgical resection, irradiation, and temozolomide. In line with many other forms of aggressive cancers, GBM is currently under consideration as a target for immunotherapy. However, GBM tends to be nonimmunogenic and exhibits a microenvironment with few or no effector T cells, a relatively low nonsynonymous somatic mutational load, and a low predicted neoantigen burden. GBM also exploits a multitude of immunosuppressive strategies. Methods and Materials A number of immunotherapeutic approaches have been tested with disappointing results. A rationale exists to combine immunotherapy and radiation therapy, which can induce an immunogenic form of cell death with T-cell activation and tumor infiltration. Results Various immunotherapy agents, including immune checkpoint modulators, transforming growth factor beta receptor inhibitors, and indoleamine-2,3-dioxygenase inhibitors, have been evaluated with irradiation in preclinical GBM models, with promising results, and are being further tested in clinical trials. Conclusions This review aims to present the basic rationale behind this emerging complementary therapeutic approach in GBM, appraise the current preclinical and clinical data, and discuss the future challenges in improving the antitumor immune response.
Collapse
|
25
|
|
26
|
Karim ME, Tha KK, Othman I, Borhan Uddin M, Chowdhury EH. Therapeutic Potency of Nanoformulations of siRNAs and shRNAs in Animal Models of Cancers. Pharmaceutics 2018; 10:E65. [PMID: 29861465 PMCID: PMC6026921 DOI: 10.3390/pharmaceutics10020065] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/19/2018] [Accepted: 05/22/2018] [Indexed: 02/07/2023] Open
Abstract
RNA Interference (RNAi) has brought revolutionary transformations in cancer management in the past two decades. RNAi-based therapeutics including siRNA and shRNA have immense scope to silence the expression of mutant cancer genes specifically in a therapeutic context. Although tremendous progress has been made to establish catalytic RNA as a new class of biologics for cancer management, a lot of extracellular and intracellular barriers still pose a long-lasting challenge on the way to clinical approval. A series of chemically suitable, safe and effective viral and non-viral carriers have emerged to overcome physiological barriers and ensure targeted delivery of RNAi. The newly invented carriers, delivery techniques and gene editing technology made current treatment protocols stronger to fight cancer. This review has provided a platform about the chronicle of siRNA development and challenges of RNAi therapeutics for laboratory to bedside translation focusing on recent advancement in siRNA delivery vehicles with their limitations. Furthermore, an overview of several animal model studies of siRNA- or shRNA-based cancer gene therapy over the past 15 years has been presented, highlighting the roles of genes in multiple cancers, pharmacokinetic parameters and critical evaluation. The review concludes with a future direction for the development of catalytic RNA vehicles and design strategies to make RNAi-based cancer gene therapy more promising to surmount cancer gene delivery challenges.
Collapse
Affiliation(s)
- Md Emranul Karim
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia.
| | - Kyi Kyi Tha
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia.
| | - Mohammad Borhan Uddin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia.
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
27
|
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: 2.0] [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
|
28
|
Bielamowicz K, Fousek K, Byrd TT, Samaha H, Mukherjee M, Aware N, Wu MF, Orange JS, Sumazin P, Man TK, Joseph SK, Hegde M, Ahmed N. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro Oncol 2018; 20:506-518. [PMID: 29016929 PMCID: PMC5909636 DOI: 10.1093/neuonc/nox182] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common primary malignant brain cancer, and is currently incurable. Chimeric antigen receptor (CAR) T cells have shown promise in GBM treatment. While we have shown that combinatorial targeting of 2 glioma antigens offsets antigen escape and enhances T-cell effector functions, the interpatient variability in surface antigen expression between patients hinders the clinical impact of targeting 2 antigen pairs. This study addresses targeting 3 antigens using a single CAR T-cell product for broader application. Methods We analyzed the surface expression of 3 targetable glioma antigens (human epidermal growth factor receptor 2 [HER2], interleukin-13 receptor subunit alpha-2 [IL13Rα2], and ephrin-A2 [EphA2]) in 15 primary GBM samples. Accordingly, we created a trivalent T-cell product armed with 3 CAR molecules specific for these validated targets encoded by a single universal (U) tricistronic transgene (UCAR T cells). Results Our data showed that co-targeting HER2, IL13Rα2, and EphA2 could overcome interpatient variability by a tendency to capture nearly 100% of tumor cells in most tumors tested in this cohort. UCAR T cells made from GBM patients' blood uniformly expressed all 3 CAR molecules with distinct antigen specificity. UCAR T cells mediated robust immune synapses with tumor targets forming more polarized microtubule organizing centers and exhibited improved cytotoxicity and cytokine release over best monospecific and bispecific CAR T cells per patient tumor profile. Lastly, low doses of UCAR T cells controlled established autologous GBM patient derived xenografts (PDXs) and improved survival of treated animals. Conclusion UCAR T cells can overcome antigenic heterogeneity in GBM and lead to improved treatment outcomes.
Collapse
Affiliation(s)
- Kevin Bielamowicz
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Kristen Fousek
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Interdepartmental Graduate Program, Baylor College of Medicine, Houston, Texas, USA
| | - Tiara T Byrd
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Hebatalla Samaha
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Children’s Cancer Hospital of Egypt, Cairo, Egypt
| | - Malini Mukherjee
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital Center for Human Immunobiology, Houston, Texas, USA
| | - Nikita Aware
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Meng-Fen Wu
- The Biostatistics and Informatics Shared Resource, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Jordan S Orange
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital Center for Human Immunobiology, Houston, Texas, USA
| | - Pavel Sumazin
- Texas Children’s Cancer and Hematology Centers, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Tsz-Kwong Man
- Texas Children’s Cancer and Hematology Centers, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Program of Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Sujith K Joseph
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers, Houston, Texas, USA
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Children’s Cancer Hospital of Egypt, Cairo, Egypt
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
29
|
Newman JP, Wang GY, Arima K, Guan SP, Waters MR, Cavenee WK, Pan E, Aliwarga E, Chong ST, Kok CYL, Endaya BB, Habib AA, Horibe T, Ng WH, Ho IAW, Hui KM, Kordula T, Lam PYP. Interleukin-13 receptor alpha 2 cooperates with EGFRvIII signaling to promote glioblastoma multiforme. Nat Commun 2017; 8:1913. [PMID: 29203859 PMCID: PMC5715073 DOI: 10.1038/s41467-017-01392-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/14/2017] [Indexed: 01/09/2023] Open
Abstract
The interleukin-13 receptor alpha2 (IL-13Rα2) is a cancer-associated receptor overexpressed in human glioblastoma multiforme (GBM). This receptor is undetectable in normal brain which makes it a highly suitable target for diagnostic and therapeutic purposes. However, the pathological role of this receptor in GBM remains to be established. Here we report that IL-13Rα2 alone induces invasiveness of human GBM cells without affecting their proliferation. In contrast, in the presence of the mutant EGFR (EGFRvIII), IL-13Rα2 promotes GBM cell proliferation in vitro and in vivo. Mechanistically, the cytoplasmic domain of IL-13Rα2 specifically binds to EGFRvIII, and this binding upregulates the tyrosine kinase activity of EGFRvIII and activates the RAS/RAF/MEK/ERK and STAT3 pathways. Our findings support the "To Go or To Grow" hypothesis whereby IL-13Rα2 serves as a molecular switch from invasion to proliferation, and suggest that targeting both receptors with STAT3 signaling inhibitor might be a therapeutic approach for the treatment of GBM.
Collapse
Affiliation(s)
- Jennifer P Newman
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore
| | - Grace Y Wang
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore.,Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, 94609, USA
| | - Kazuhiko Arima
- Department of Biomolecular Sciences, Saga Medical School, Saga, 840-8502, Japan
| | - Shou P Guan
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore
| | - Michael R Waters
- School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | | | - Edward Pan
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Edita Aliwarga
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore
| | - Siao T Chong
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore
| | - Catherine Y L Kok
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore
| | - Berwini B Endaya
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222, Queensland, Australia
| | - Amyn A Habib
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center and the North Texas VA Medical Center, Dallas, 75390, USA
| | - Tomohisa Horibe
- Department of Pharmacoepidemiology, Kyoto University School of Public Health, Kyoto, 606-8501, Japan
| | - Wai H Ng
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Ivy A W Ho
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore.,National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Kam M Hui
- Bek Chai Heah Laboratory of Cancer Genomics, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore, 169610, Singapore.,Cancer and Stem Cells Biology Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore, 169857, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Dr, Singapore, 117596, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Proteos, 61 Biopolis Dr, Singapore, 138673, Singapore
| | - Tomasz Kordula
- School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Paula Y P Lam
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore, 169610, Singapore. .,Cancer and Stem Cells Biology Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore, 169857, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, Singapore, 117593, Singapore.
| |
Collapse
|
30
|
Monteiro AR, Hill R, Pilkington GJ, Madureira PA. The Role of Hypoxia in Glioblastoma Invasion. Cells 2017; 6:E45. [PMID: 29165393 PMCID: PMC5755503 DOI: 10.3390/cells6040045] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), a grade IV astrocytoma, is the most common and deadly type of primary malignant brain tumor, with a patient's median survival rate ranging from 15 to 17 months. The current treatment for GBM involves tumor resection surgery based on MRI image analysis, followed by radiotherapy and treatment with temozolomide. However, the gradual development of tumor resistance to temozolomide is frequent in GBM patients leading to subsequent tumor regrowth/relapse. For this reason, the development of more effective therapeutic approaches for GBM is of critical importance. Low tumor oxygenation, also known as hypoxia, constitutes a major concern for GBM patients, since it promotes cancer cell spreading (invasion) into the healthy brain tissue in order to evade this adverse microenvironment. Tumor invasion not only constitutes a major obstacle to surgery, radiotherapy, and chemotherapy, but it is also the main cause of death in GBM patients. Understanding how hypoxia triggers the GBM cells to become invasive is paramount to developing novel and more effective therapies against this devastating disease. In this review, we will present a comprehensive examination of the available literature focused on investigating how GBM hypoxia triggers an invasive cancer cell phenotype and the role of these invasive proteins in GBM progression.
Collapse
Affiliation(s)
- Ana Rita Monteiro
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, Room 3.4, 8005-139 Faro, Portugal.
| | - Richard Hill
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| | - Geoffrey J Pilkington
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| | - Patrícia A Madureira
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, Room 3.4, 8005-139 Faro, Portugal.
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| |
Collapse
|
31
|
Brown CE, Aguilar B, Starr R, Yang X, Chang WC, Weng L, Chang B, Sarkissian A, Brito A, Sanchez JF, Ostberg JR, D'Apuzzo M, Badie B, Barish ME, Forman SJ. Optimization of IL13Rα2-Targeted Chimeric Antigen Receptor T Cells for Improved Anti-tumor Efficacy against Glioblastoma. Mol Ther 2017; 26:31-44. [PMID: 29103912 DOI: 10.1016/j.ymthe.2017.10.002] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/28/2017] [Accepted: 10/01/2017] [Indexed: 12/27/2022] Open
Abstract
T cell immunotherapy is emerging as a powerful strategy to treat cancer and may improve outcomes for patients with glioblastoma (GBM). We have developed a chimeric antigen receptor (CAR) T cell immunotherapy targeting IL-13 receptor α2 (IL13Rα2) for the treatment of GBM. Here, we describe the optimization of IL13Rα2-targeted CAR T cells, including the design of a 4-1BB (CD137) co-stimulatory CAR (IL13BBζ) and a manufacturing platform using enriched central memory T cells. Utilizing orthotopic human GBM models with patient-derived tumor sphere lines in NSG mice, we found that IL13BBζ-CAR T cells improved anti-tumor activity and T cell persistence as compared to first-generation IL13ζ-CAR CD8+ T cells that had shown evidence for bioactivity in patients. Investigating the impact of corticosteroids, given their frequent use in the clinical management of GBM, we demonstrate that low-dose dexamethasone does not diminish CAR T cell anti-tumor activity in vivo. Furthermore, we found that local intracranial delivery of CAR T cells elicits superior anti-tumor efficacy as compared to intravenous administration, with intraventricular infusions exhibiting possible benefit over intracranial tumor infusions in a multifocal disease model. Overall, these findings help define parameters for the clinical translation of CAR T cell therapy for the treatment of brain tumors.
Collapse
Affiliation(s)
- Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA.
| | - Brenda Aguilar
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Renate Starr
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Xin Yang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Wen-Chung Chang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Lihong Weng
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Brenda Chang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Aniee Sarkissian
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Alfonso Brito
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - James F Sanchez
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Julie R Ostberg
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Massimo D'Apuzzo
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Behnam Badie
- Department of Neurosurgery, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Michael E Barish
- Department of Developmental and Stem Cell Biology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
32
|
Kuramitsu S, Yamamichi A, Ohka F, Motomura K, Hara M, Natsume A. Adoptive immunotherapy for the treatment of glioblastoma: progress and possibilities. Immunotherapy 2017; 8:1393-1404. [PMID: 28000534 DOI: 10.2217/imt-2016-0076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Patients with glioblastoma have a very poor prognosis. Adoptive cellular therapy (ACT) is defined as the collection of circulating or tumor-infiltrating lymphocytes, their selection, modification, expansion and activation, and their re-administration to patients in order to induce antitumor activity. Although various ACTs have been attempted, most failed to improve the outcome. Immune checkpoint blockade antibodies and T cell engineering with tumor-specific chimeric antigen receptors suggest the emergence of a new era of immunotherapy. Here, we summarize approaches with ACTs using genetically modified T cells, which have been improved by enhancing their antitumor activity, and discuss strategies to develop these therapies. The mechanisms by which gliomas modulate and evade the immune system are also discussed.
Collapse
Affiliation(s)
- Shunichiro Kuramitsu
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Akane Yamamichi
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Fumiharu Ohka
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Kazuya Motomura
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Masahito Hara
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
33
|
Witusik-Perkowska M, Zakrzewska M, Sikorska B, Papierz W, Jaskolski DJ, Szemraj J, Liberski PP. Glioblastoma-derived cells in vitro unveil the spectrum of drug resistance capability - comparative study of tumour chemosensitivity in different culture systems. Biosci Rep 2017; 37:BSR20170058. [PMID: 28522553 PMCID: PMC5964726 DOI: 10.1042/bsr20170058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/05/2017] [Accepted: 05/18/2017] [Indexed: 12/28/2022] Open
Abstract
Resistance to cancer drugs is a complex phenomenon which could be influenced by in vitro conditions. However, tumour-derived cell cultures are routinely used for studies related to mechanisms of drug responsiveness or the search for new therapeutic approaches. The purpose of our work was to identify the potential differences in drug resistance and response to treatment of glioblastoma with the use of three in vitro models: traditional adherent culture, serum-free spheroid culture and novel adherent serum-free culture.The experimental models were evaluated according to 'stemness state' and epithelial-to-mesenchymal transition (EMT) status, invasion capability and their expression pattern of genes related to the phenomenon of tumour drug resistance. Additionally, the response to drug treatments of three different culture models was compared with regard to the type of cell death.Multi-gene expression profiling revealed differences between examined culture types with regard to the expression pattern of the selected genes. Functionally, the examined genes were related to drug resistance and metabolism, DNA damage and repair and cell cycle control, and included potential therapeutic targets.Cytotoxicity analyses confirmed that environmental factors can influence not only the molecular background of glioblastoma drug-resistance and efficiency of treatment, but also the mechanisms/pathways of cell death, which was reflected by a distinct intensification of apoptosis and autophagy observed in particular culture models. Our results suggest that parallel exploitation of different in vitro experimental models can be used to reveal the spectrum of cancer cell resistance capability, especially regarding intra-heterogeneous glioblastomas.
Collapse
Affiliation(s)
- Monika Witusik-Perkowska
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Magdalena Zakrzewska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Beata Sikorska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Wielislaw Papierz
- Department of Pathomorphology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Dariusz J Jaskolski
- Department of Neurosurgery and Oncology of Central Nervous System, Medical University of Lodz, Barlicki University Hospital, Kopcińskiego 22, Lodz 90-153, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| |
Collapse
|
34
|
Tanaka T, Yamada H, Kuroki M, Kodama S, Tamura K, Takamatsu Y. A Modified Adenovirus Vector-Mediated Antibody Screening Method Identifies EphA2 as a Cancer Target. Transl Oncol 2017; 10:476-484. [PMID: 28505517 PMCID: PMC5430157 DOI: 10.1016/j.tranon.2017.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/03/2017] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND: We constructed a genetically modified adenovirus vector incorporating an IgG Fc-binding motif from staphylococcal protein A, Z33 (Adv-FZ33). Adv-FZ33 allows an antibody to redirect the vector to a target molecule on the cell surface. We attempted to search for target antigen candidates and antibodies that allowed highly selective gene transduction into malignant tumors. METHODS: Hybridoma libraries producing monoclonal antibodies (mAbs) were screened that increased transduction efficiency in cancer cell lines after cross-linking with Adv-FZ33. Target antigens of the mAbs were identified by immunoprecipitation and mass spectrometry. Of these mAbs, we noted a clone, F2-27, that recognized the receptor tyrosine kinase EphA2. Next, we generated an adenovirus vector, Ax3CMTK-FZ33, that expressed a herpes simplex virus thymidine kinase (HSV-TK). The therapeutic efficacy of F2-27–mediated HSV-TK gene transduction, followed by ganciclovir (GCV) administration, was studied in vitro. The inhibitory effect of F2-27 on cancer cell invasion was investigated by a three-dimensional spheroid formation assay. RESULTS: In vitro reporter gene expression after Adv-FZ33 infection via F2-27 was 146 times higher than with control mAb in EphA2-expressing cancer cell lines. F2-27–mediated Ax3CMTK-FZ33 infection induced the HSV-TK gene in an F2-27–dependent manner and had a highly effective cytotoxic effect in a GCV-dependent manner. Additionally, F2-27 independently inhibited migration of EphA2-positive breast cancer cell lines in three-dimensional culture. CONCLUSION: Our modified adenovirus and hybridoma screening system is useful for the development of targeted cancer therapy, and F2-27 has the potential to be an antibody-based therapy for various EphA2-positive cancers.
Collapse
Affiliation(s)
- Toshihiro Tanaka
- Division of Oncology, Hematology and Infectious Diseases, Department of Internal Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Hiromi Yamada
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Masahide Kuroki
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kazuo Tamura
- Division of Oncology, Hematology and Infectious Diseases, Department of Internal Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yasushi Takamatsu
- Division of Oncology, Hematology and Infectious Diseases, Department of Internal Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
35
|
Sonawane P, Choi YA, Pandya H, Herpai DM, Fokt I, Priebe W, Debinski W. Novel Molecular Multilevel Targeted Antitumor Agents. CANCER TRANSLATIONAL MEDICINE 2017; 3:69-79. [PMID: 28825042 PMCID: PMC5558462 DOI: 10.4103/ctm.ctm_12_17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A multifunctional fusion protein, IL-13.E13K-D2-NLS, effectively recognizes glioblastoma (GBM) cells and delivers its portion to the cell nucleus. IL-13.E13K-D2-NLS is composed of a cancer cell targeting ligand (IL-13.E13K), specialized cytosol translocation bacterial toxin domain 2 of Pseudomonas exotoxin A (D2) and SV40 T antigen nuclear localization signal (NLS). We have now tested whether we can produce proteins that would serve as a delivery vehicle to lysosomes and mitochondria as well. Moreover, we examined whether IL-13.E13K-D2-NLS can deliver anti-cancer drugs like doxorubicin to their nuclear site of action in cancer cells. We have thus constructed two novel proteins: IL-13.E13K-D2-LLS which incorporates lysosomal localization signal (LLS) of a human lysosomal associated membrane protein (LAMP-1) for targeting to lysosomes and IL-13-D2-KK2, which incorporates a pro-apoptotic peptide (KLAKLAK)2 (KK2) exerting its action in mitochondria. Furthermore, we have produced IL-13.E13K-D2-NLS and IL-13.E13K-D2-LLS versions containing a cysteine for site-specific conjugation with a modified doxorubicin, WP936. We found that single-chain recombinant proteins IL-13.E13K-D2-LLS and IL-13-D2-KK2 are internalized and localized mostly to the lysosomal and mitochondrial compartments, respectively, without major trafficking to cells' nuclei. We also determined that IL-13.E13K-D2-NLS-cys[WP936], IL-13.E13K-D2-LAMP-cys[WP936] and IL-13-D2-KK2 were cytotoxic to GBM cells overexpressing IL-13RA2, while much less cytotoxic to GBM cell lines expressing low levels of the receptor. IL-13.E13K-D2-NLS-cys[WP936] was the most potent of the tested anti-tumor agents including free WP936. We believe that our receptor-directed intracellular organelle-targeted proteins can be employed for numerous specific and safer treatment applications when drugs have specific intracellular sites of their action.
Collapse
Affiliation(s)
- Poonam Sonawane
- Department of Cancer Biology, Brain Tumor Center of Excellence,
Comprehensive Cancer Center of Wake Baptist Medical Center, Medical Center Boulevard,
Winston-Salem, NC 27157, USA
| | - Young A. Choi
- Department of Cancer Biology, Brain Tumor Center of Excellence,
Comprehensive Cancer Center of Wake Baptist Medical Center, Medical Center Boulevard,
Winston-Salem, NC 27157, USA
| | - Hetal Pandya
- National Institutes of Health, Bethesda, MD, USA
| | - Denise M. Herpai
- Department of Cancer Biology, Brain Tumor Center of Excellence,
Comprehensive Cancer Center of Wake Baptist Medical Center, Medical Center Boulevard,
Winston-Salem, NC 27157, USA
| | | | | | - Waldemar Debinski
- Department of Cancer Biology, Brain Tumor Center of Excellence,
Comprehensive Cancer Center of Wake Baptist Medical Center, Medical Center Boulevard,
Winston-Salem, NC 27157, USA
| |
Collapse
|
36
|
Liu R, Li X, Xiao W, Lam KS. Tumor-targeting peptides from combinatorial libraries. Adv Drug Deliv Rev 2017; 110-111:13-37. [PMID: 27210583 DOI: 10.1016/j.addr.2016.05.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major and leading causes of death worldwide. Two of the greatest challenges in fighting cancer are early detection and effective treatments with no or minimum side effects. Widespread use of targeted therapies and molecular imaging in clinics requires high affinity, tumor-specific agents as effective targeting vehicles to deliver therapeutics and imaging probes to the primary or metastatic tumor sites. Combinatorial libraries such as phage-display and one-bead one-compound (OBOC) peptide libraries are powerful approaches in discovering tumor-targeting peptides. This review gives an overview of different combinatorial library technologies that have been used for the discovery of tumor-targeting peptides. Examples of tumor-targeting peptides identified from each combinatorial library method will be discussed. Published tumor-targeting peptide ligands and their applications will also be summarized by the combinatorial library methods and their corresponding binding receptors.
Collapse
Affiliation(s)
- Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; Division of Hematology & Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
37
|
Kamran N, Calinescu A, Candolfi M, Chandran M, Mineharu Y, Asad AS, Koschmann C, Nunez FJ, Lowenstein PR, Castro MG. Recent advances and future of immunotherapy for glioblastoma. Expert Opin Biol Ther 2016; 16:1245-64. [PMID: 27411023 PMCID: PMC5014608 DOI: 10.1080/14712598.2016.1212012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/08/2016] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Outcome for glioma (GBM) remains dismal despite advances in therapeutic interventions including chemotherapy, radiotherapy and surgical resection. The overall survival benefit observed with immunotherapies in cancers such as melanoma and prostate cancer has fuelled research into evaluating immunotherapies for GBM. AREAS COVERED Preclinical studies have brought a wealth of information for improving the prognosis of GBM and multiple clinical studies are evaluating a wide array of immunotherapies for GBM patients. This review highlights advances in the development of immunotherapeutic approaches. We discuss the strategies and outcomes of active and passive immunotherapies for GBM including vaccination strategies, gene therapy, check point blockade and adoptive T cell therapies. We also focus on immunoediting and tumor neoantigens that can impact the efficacy of immunotherapies. EXPERT OPINION Encouraging results have been observed with immunotherapeutic strategies; some clinical trials are reaching phase III. Significant progress has been made in unraveling the molecular and genetic heterogeneity of GBM and its implications to disease prognosis. There is now consensus related to the critical need to incorporate tumor heterogeneity into the design of therapeutic approaches. Recent data also indicates that an efficacious treatment strategy will need to be combinatorial and personalized to the tumor genetic signature.
Collapse
Affiliation(s)
- Neha Kamran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Alexandra Calinescu
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Marianela Candolfi
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Mayuri Chandran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Yohei Mineharu
- d Department of Neurosurgery , Kyoto University Graduate School of Medicine , Kyoto , Japan
| | - Antonela S Asad
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Carl Koschmann
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Felipe J Nunez
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Pedro R Lowenstein
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Maria G Castro
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| |
Collapse
|
38
|
Ferluga S, Tomé CML, Herpai DM, D'Agostino R, Debinski W. Simultaneous targeting of Eph receptors in glioblastoma. Oncotarget 2016; 7:59860-59876. [PMID: 27494882 PMCID: PMC5312354 DOI: 10.18632/oncotarget.10978] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/16/2016] [Indexed: 12/20/2022] Open
Abstract
Eph tyrosine kinase receptors are frequently overexpressed and functional in many cancers, and they are attractive candidates for targeted therapy. Here, we analyzed the expression of Eph receptor A3, one of the most up-regulated factors in glioblastoma cells cultured under tumorsphere-forming conditions, together with EphA2 and EphB2 receptors. EphA3 was overexpressed in up to 60% of glioblastoma tumors tested, but not in normal brain. EphA3 was localized in scattered areas of the tumor, the invasive ring, and niches near tumor vessels. EphA3 co-localized with macrophage/leukocyte markers, suggesting EphA3 expression on tumor-infiltrating cells of bone marrow origin. We took advantage of the fact that ephrinA5 (eA5) is a ligand that binds EphA3, EphA2 and EphB2 receptors, and used it to construct a novel targeted anti-glioblastoma cytotoxin. The eA5-based cytotoxin potently and specifically killed glioblastoma cells with an IC50 of at least 10-11 M. This and similar cytotoxins will simultaneously target different compartments of glioblastoma tumors while mitigating tumor heterogeneity.
Collapse
Affiliation(s)
- Sara Ferluga
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Carla Maria Lema Tomé
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Denise Mazess Herpai
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Ralph D'Agostino
- Department of Biostatistical Sciences, Section on Biostatistics, Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA
| | - Waldemar Debinski
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
39
|
Hegde M, Mukherjee M, Grada Z, Pignata A, Landi D, Navai SA, Wakefield A, Fousek K, Bielamowicz K, Chow KK, Brawley VS, Byrd TT, Krebs S, Gottschalk S, Wels WS, Baker ML, Dotti G, Mamonkin M, Brenner MK, Orange JS, Ahmed N. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest 2016; 126:3036-52. [PMID: 27427982 PMCID: PMC4966331 DOI: 10.1172/jci83416] [Citation(s) in RCA: 498] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 05/13/2016] [Indexed: 12/24/2022] Open
Abstract
In preclinical models of glioblastoma, antigen escape variants can lead to tumor recurrence after treatment with CAR T cells that are redirected to single tumor antigens. Given the heterogeneous expression of antigens on glioblastomas, we hypothesized that a bispecific CAR molecule would mitigate antigen escape and improve the antitumor activity of T cells. Here, we created a CAR that joins a HER2-binding scFv and an IL13Rα2-binding IL-13 mutein to make a tandem CAR exodomain (TanCAR) and a CD28.ζ endodomain. We determined that patient TanCAR T cells showed distinct binding to HER2 or IL13Rα2 and had the capability to lyse autologous glioblastoma. TanCAR T cells exhibited activation dynamics that were comparable to those of single CAR T cells upon encounter of HER2 or IL13Rα2. We observed that TanCARs engaged HER2 and IL13Rα2 simultaneously by inducing HER2-IL13Rα2 heterodimers, which promoted superadditive T cell activation when both antigens were encountered concurrently. TanCAR T cell activity was more sustained but not more exhaustible than that of T cells that coexpressed a HER2 CAR and an IL13Rα2 CAR, T cells with a unispecific CAR, or a pooled product. In a murine glioblastoma model, TanCAR T cells mitigated antigen escape, displayed enhanced antitumor efficacy, and improved animal survival. Thus, TanCAR T cells show therapeutic potential to improve glioblastoma control by coengaging HER2 and IL13Rα2 in an augmented, bivalent immune synapse that enhances T cell functionality and reduces antigen escape.
Collapse
Affiliation(s)
- Meenakshi Hegde
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Malini Mukherjee
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Texas Children’s Hospital Center for Human Immunobiology, Houston, Texas, USA
| | - Zakaria Grada
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Antonella Pignata
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Daniel Landi
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Shoba A. Navai
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Amanda Wakefield
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Kristen Fousek
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Kevin Bielamowicz
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Kevin K.H. Chow
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Vita S. Brawley
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Tiara T. Byrd
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Simone Krebs
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Winfried S. Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Matthew L. Baker
- National Center for Macromolecular Imaging, Department of Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Gianpietro Dotti
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Malcolm K. Brenner
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Jordan S. Orange
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Texas Children’s Hospital Center for Human Immunobiology, Houston, Texas, USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
40
|
Burvenich IJG, Parakh S, Gan HK, Lee FT, Guo N, Rigopoulos A, Lee ST, Gong S, O'Keefe GJ, Tochon-Danguy H, Kotsuma M, Hasegawa J, Senaldi G, Scott AM. Molecular Imaging and Quantitation of EphA2 Expression in Xenograft Models with 89Zr-DS-8895a. J Nucl Med 2016; 57:974-80. [PMID: 26940768 DOI: 10.2967/jnumed.115.169839] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/29/2016] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Subtype A2 of the erythropoietin-producing hepatocellular tyrosine kinase (EphA2) cell surface receptor is expressed in a range of epithelial cancers. This study evaluated the molecular imaging of EphA2 expression in vivo in mouse tumor models using SPECT/MR and PET/MR and a humanized anti-EphA2 antibody, DS-8895a. METHODS DS-8895a was labeled with (111)In, (125)I, and (89)Zr and assessed for radiochemical purity, immunoreactivity (Lindmo analysis), antigen-binding affinity (Scatchard analysis), and serum stability in vitro. In vivo biodistribution, imaging, and pharmacokinetic studies were performed with SPECT/MR and PET/MR. A dose-escalation study was also performed to determine EphA2 receptor saturability through tissue and imaging quantitative analysis. RESULTS All conjugates demonstrated good serum stability and specific binding to EphA2-expressing cells in vitro. In vivo biodistribution studies showed high uptake of (111)In-CHX-A″-DTPA-DS-8895a and (89)Zr-Df-Bz-NCS-DS-8895a in EphA2-expressing xenograft models, with no specific uptake in normal tissues. In comparison, retention of (125)I-DS-8895a in tumors was lower because of internalization of the radioconjugate and dehalogenation. These results were confirmed by SPECT/MR and PET/MR. EphA2 receptor saturation was observed at the 30 mg/kg dose. CONCLUSION Molecular imaging of tumor uptake of DS-8895a allows noninvasive measurement of EphA2 expression in tumors in vivo and determination of receptor saturation. (89)Zr-Df-Bz-NCS-DS-8895a is suited for human bioimaging trials on the basis of superior imaging characteristics and will inform DS-8895a dose assessment and patient response evaluation in clinical trials.
Collapse
Affiliation(s)
- Ingrid J G Burvenich
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Sagun Parakh
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia School of Cancer Medicine, La Trobe University, Melbourne, Australia Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia
| | - Hui K Gan
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia School of Cancer Medicine, La Trobe University, Melbourne, Australia Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia
| | - Fook-Thean Lee
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Nancy Guo
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Sze-Ting Lee
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Sylvia Gong
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Graeme J O'Keefe
- Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Henri Tochon-Danguy
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Masakatsu Kotsuma
- Translational Medicine and Clinical Pharmacology Department, Daiichi-Sankyo Co., Ltd., Tokyo, Japan
| | - Jun Hasegawa
- Biologics Pharmacology Research Laboratories, Daiichi-Sankyo Co., Ltd., Tokyo, Japan
| | - Giorgio Senaldi
- Department of Translational Medicine and Clinical Pharmacology, Daiichi-Sankyo Pharma Development, Edison, New Jersey; and
| | - Andrew M Scott
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia Department of Medicine, University of Melbourne, Melbourne, Australia
| |
Collapse
|
41
|
Stuckey DW, Hingtgen SD, Karakas N, Rich BE, Shah K. Engineering toxin-resistant therapeutic stem cells to treat brain tumors. Stem Cells 2015; 33:589-600. [PMID: 25346520 DOI: 10.1002/stem.1874] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/22/2014] [Accepted: 09/18/2014] [Indexed: 01/14/2023]
Abstract
Pseudomonas exotoxin (PE) potently blocks protein synthesis by catalyzing the inactivation of elongation factor-2 (EF-2). Targeted PE-cytotoxins have been used as antitumor agents, although their effective clinical translation in solid tumors has been confounded by off-target delivery, systemic toxicity, and short chemotherapeutic half-life. To overcome these limitations, we have created toxin-resistant stem cells by modifying endogenous EF-2, and engineered them to secrete PE-cytotoxins that target specifically expressed (interleukin-13 receptor subunit alpha-2) or overexpressed (epidermal growth factor receptor) in glioblastomas (GBM). Molecular analysis correlated efficacy of PE-targeted cytotoxins with levels of cognate receptor expression, and optical imaging was applied to simultaneously track the kinetics of protein synthesis inhibition and GBM cell viability in vivo. The release of IL13-PE from biodegradable synthetic extracellular matrix (sECM) encapsulated stem cells in a clinically relevant GBM resection model led to increased long-term survival of mice compared to IL13-PE protein infusion. Moreover, multiple patient-derived GBM lines responded to treatment, underscoring its clinical relevance. In sum, integrating stem cell-based engineering, multimodal imaging, and delivery of PE-cytotoxins in a clinically relevant GBM model represents a novel strategy and a potential advancement in GBM therapy.
Collapse
Affiliation(s)
- Daniel W Stuckey
- Molecular Neurotherapy and Imaging Laboratory; Department of Radiology
| | | | | | | | | |
Collapse
|
42
|
Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, Naranjo A, Starr R, Wagner J, Wright C, Zhai Y, Bading JR, Ressler JA, Portnow J, D'Apuzzo M, Forman SJ, Jensen MC. Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin Cancer Res 2015; 21:4062-72. [PMID: 26059190 DOI: 10.1158/1078-0432.ccr-15-0428] [Citation(s) in RCA: 535] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/17/2015] [Indexed: 01/27/2023]
Abstract
PURPOSE A first-in-human pilot safety and feasibility trial evaluating chimeric antigen receptor (CAR)-engineered, autologous primary human CD8(+) cytotoxic T lymphocytes (CTL) targeting IL13Rα2 for the treatment of recurrent glioblastoma (GBM). EXPERIMENTAL DESIGN Three patients with recurrent GBM were treated with IL13(E13Y)-zetakine CD8(+) CTL targeting IL13Rα2. Patients received up to 12 local infusions at a maximum dose of 10(8) CAR-engineered T cells via a catheter/reservoir system. RESULTS We demonstrate the feasibility of manufacturing sufficient numbers of autologous CTL clones expressing an IL13(E13Y)-zetakine CAR for redirected HLA-independent IL13Rα2-specific effector function for a cohort of patients diagnosed with GBM. Intracranial delivery of the IL13-zetakine(+) CTL clones into the resection cavity of 3 patients with recurrent disease was well-tolerated, with manageable temporary brain inflammation. Following infusion of IL13-zetakine(+) CTLs, evidence for transient anti-glioma responses was observed in 2 of the patients. Analysis of tumor tissue from 1 patient before and after T-cell therapy suggested reduced overall IL13Rα2 expression within the tumor following treatment. MRI analysis of another patient indicated an increase in tumor necrotic volume at the site of IL13-zetakine(+) T-cell administration. CONCLUSIONS These findings provide promising first-in-human clinical experience for intracranial administration of IL13Rα2-specific CAR T cells for the treatment of GBM, establishing a foundation on which future refinements of adoptive CAR T-cell therapies can be applied.
Collapse
Affiliation(s)
- Christine E Brown
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California.
| | - Behnam Badie
- Department of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Michael E Barish
- Department of Neurosciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Lihong Weng
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Julie R Ostberg
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Wen-Chung Chang
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Araceli Naranjo
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Renate Starr
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Jamie Wagner
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Christine Wright
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Yubo Zhai
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - James R Bading
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Julie A Ressler
- Department of Diagnostic Radiology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Jana Portnow
- Department of Medical Oncology and Therapeutics Research, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Massimo D'Apuzzo
- Department of Pathology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Stephen J Forman
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Michael C Jensen
- Center for Childhood Cancer, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
43
|
Young JS, Kim JW, Ahmed AU, Lesniak MS. Therapeutic cell carriers: a potential road to cure glioma. Expert Rev Neurother 2015; 14:651-60. [PMID: 24852229 DOI: 10.1586/14737175.2014.917964] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many different experimental molecular therapeutic approaches have been evaluated in an attempt to treat brain cancer. However, despite the success of these experimental molecular therapies, research has shown that the specific and efficient delivery of therapeutic agents to tumor cells is a limitation. In this regard, cell carrier systems have garnered significant attraction due to their capacity to be loaded with therapeutic agents and carry them specifically to tumor sites. Furthermore, cell carriers can be genetically modified to express therapeutic agents that can directly eradicate cancerous cells or can modulate tumor microenvironments. This review describes the current state of cell carriers, their use as vehicles for the delivery of therapeutic agents to brain tumors, and future directions that will help overcome the present obstacles to cell carrier mediated therapy for brain cancer.
Collapse
Affiliation(s)
- Jacob S Young
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Eph receptor tyrosine kinases and the corresponding ephrin ligands play a pivotal role in the glioma development and progression. Aberrant protein expression levels of the Eph receptors and ephrins are often associated with higher tumor grade and poor prognosis. Their function in tumorigenesis is complex due to the intricate network of possible co-occurring interactions between neighboring tumor cells and tumor microenvironment. Both Ephs and ephrins localize on the surface of tumor cells, tumor vasculature, glioma stem cells, tumor cells infiltrating brain, and immune cells infiltrating tumors. They can both promote and inhibit tumorigenicity depending on the downstream forward and reverse signalling generated. All the above-mentioned features make the Ephs/ephrins system an intriguing candidate for the development of new therapeutic strategies in glioma treatment. This review will give a general overview on the structure and the function of Ephs and ephrins, with a particular emphasis on the state of the knowledge of their role in malignant gliomas.
Collapse
Affiliation(s)
- Sara Ferluga
- Department of Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Waldemar Debinski
- Department of Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- To whom correspondence should be addressed: Waldemar Debinski, M.D., Ph.D., Director of Brain Tumor Center of Excellence, Thomas K. Hearn Jr. Brain Tumor Research Center, Professor of Neurosurgery, Radiation Oncology, and Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157, Phone: (336) 716-9712, Fax: (336) 713-7639,
| |
Collapse
|
45
|
Rajh T, Dimitrijevic NM, Bissonnette M, Koritarov T, Konda V. Titanium Dioxide in the Service of the Biomedical Revolution. Chem Rev 2014; 114:10177-216. [DOI: 10.1021/cr500029g] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tijana Rajh
- Center
for Nanoscale Materials, Argonne National Laboratory, 9700 South
Cass Avenue, Argonne, Illinois 60540, United States
| | - Nada M. Dimitrijevic
- Center
for Nanoscale Materials, Argonne National Laboratory, 9700 South
Cass Avenue, Argonne, Illinois 60540, United States
| | - Marc Bissonnette
- Department
of Medicine, The University of Chicago Medicine, 5841 South Maryland Avenue, MC 4076, Chicago, Illinois 60637, United States
| | - Tamara Koritarov
- Center
for Nanoscale Materials, Argonne National Laboratory, 9700 South
Cass Avenue, Argonne, Illinois 60540, United States
- School
of Medicine, Boston University, 72 East Concord Street, Boston, Massachusetts 02118, United States
| | - Vani Konda
- Department
of Medicine, The University of Chicago Medicine, 5841 South Maryland Avenue, MC 4076, Chicago, Illinois 60637, United States
| |
Collapse
|
46
|
Witusik-Perkowska M, Zakrzewska M, Szybka M, Papierz W, Jaskolski DJ, Liberski PP, Sikorska B. Astrocytoma-associated antigens - IL13Rα2, Fra-1, and EphA2 as potential markers to monitor the status of tumour-derived cell cultures in vitro. Cancer Cell Int 2014; 14:82. [PMID: 25788865 PMCID: PMC4364051 DOI: 10.1186/s12935-014-0082-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 11/18/2022] Open
Abstract
Background The molecular heterogeneity of high-grade astrocytomas underlies the difficulties in the development of representative and valuable in vitro experimental models for their studies. The purpose of our study was to estimate the value of astrocytoma-associated antigens (AAAs) - IL13Rα2, Fra-1, EphA2 - and the most common molecular aberrations typical for astrocytomas as potential markers to screen the status of tumour-derived cell cultures in vitro. Methods The tumour-derived cell cultures were established from high-grade astrocytomas. The expression analyses of the tested genes were performed via semi-quantitative real-time PCR and subsequently verified by immunohistochemical and immunocytochemical technique. The analyses of molecular aberrations at DNA level included gene dosage status evaluation based on real-time PCR, sequencing analysis, and loss of heterozygosity (LOH) assay. Results The expression analyses based on semi-quantitative real-time PCR showed that in the final stage of culture the expression level of all tested AAAs was significantly higher or at least comparable to that of primary tumours; however, two expression patterns were observed during cell culture establishment. Analysis at the single cell level via immunocytochemistry also demonstrated an increase of the level of tested proteins and/or selection of tumour cell populations strongly positive for AAAs vs. other cell types including admixed non-tumoural cells. Confrontation of AAA expression data with the results of molecular analyses at DNA level seems to support the latter, revealing that the expression pattern of astrocytoma-associated antigens in tumour-derived cells in subsequent stages of culture is convergent with changes in the molecular profile of examined cell populations. Conclusions The consistency of the obtained results seems to support the use of the selected AAAs, in particular IL13Rα2 and Fra-1, as tools facilitating the establishment of tumour-derived cultures. However, the intratumoural heterogeneity of high-grade astrocytomas may require further detailed characterisation of the molecular profile of a tumour in order to evaluate the value of the experimental model in relation to the individual context of particular studies.
Collapse
Affiliation(s)
- Monika Witusik-Perkowska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10 str, Lodz, 92-216, Poland
| | - Magdalena Zakrzewska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10 str, Lodz, 92-216, Poland
| | - Malgorzata Szybka
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10 str, Lodz, 92-216, Poland
| | - Wielislaw Papierz
- Department of Pathomorphology, Medical University of Lodz, Czechoslowacka 8/10 str, Lodz, 92-216, Poland
| | - Dariusz J Jaskolski
- Department of Neurosurgery, Medical University of Lodz, Kopcinskiego 22 str, Lodz, 90-153, Poland
| | - Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10 str, Lodz, 92-216, Poland
| | - Beata Sikorska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10 str, Lodz, 92-216, Poland
| |
Collapse
|
47
|
Eph receptors as therapeutic targets in glioblastoma. Br J Cancer 2014; 111:1255-61. [PMID: 25144626 PMCID: PMC4183860 DOI: 10.1038/bjc.2014.73] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 12/20/2022] Open
Abstract
The dismal outlook for patients with the most aggressive and common form of adult brain cancer, glioblastoma (GBM), motivates a search for new therapeutic strategies and targets for this aggressive disease. Here we review the findings to date on the role of Eph family receptor tyrosine kinases and their ephrin ligands in brain cancer. Expression of the Eph family of cell surface proteins is generally downregulated to very low levels in normal adult tissues making them particularly attractive for directed therapeutic targeting. Recent Eph targeting studies in pre-clinical models of GBM have been very encouraging and may provide an avenue to treat these highly refractory aggressive tumours.
Collapse
|
48
|
A new hope in immunotherapy for malignant gliomas: adoptive T cell transfer therapy. J Immunol Res 2014; 2014:326545. [PMID: 25009822 PMCID: PMC4070364 DOI: 10.1155/2014/326545] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/02/2014] [Accepted: 05/18/2014] [Indexed: 11/18/2022] Open
Abstract
Immunotherapy emerged as a promising therapeutic approach to highly incurable malignant gliomas due to tumor-specific cytotoxicity, minimal side effect, and a durable antitumor effect by memory T cells. But, antitumor activities of endogenously activated T cells induced by immunotherapy such as vaccination are not sufficient to control tumors because tumor-specific antigens may be self-antigens and tumors have immune evasion mechanisms to avoid immune surveillance system of host. Although recent clinical results from vaccine strategy for malignant gliomas are encouraging, these trials have some limitations, particularly their failure to expand tumor antigen-specific T cells reproducibly and effectively. An alternative strategy to overcome these limitations is adoptive T cell transfer therapy, in which tumor-specific T cells are expanded ex vivo rapidly and then transferred to patients. Moreover, enhanced biologic functions of T cells generated by genetic engineering and modified immunosuppressive microenvironment of host by homeostatic T cell expansion and/or elimination of immunosuppressive cells and molecules can induce more potent antitumor T cell responses and make this strategy hold promise in promoting a patient response for malignant glioma treatment. Here we will review the past and current progresses and discuss a new hope in adoptive T cell therapy for malignant gliomas.
Collapse
|
49
|
Brown CE, Warden CD, Starr R, Deng X, Badie B, Yuan YC, Forman SJ, Barish ME. Glioma IL13Rα2 is associated with mesenchymal signature gene expression and poor patient prognosis. PLoS One 2013; 8:e77769. [PMID: 24204956 PMCID: PMC3800130 DOI: 10.1371/journal.pone.0077769] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 09/04/2013] [Indexed: 11/19/2022] Open
Abstract
A major challenge for successful immunotherapy against glioma is the identification and characterization of validated targets. We have taken a bioinformatics approach towards understanding the biological context of IL-13 receptor α2 (IL13Rα2) expression in brain tumors, and its functional significance for patient survival. Querying multiple gene expression databases, we show that IL13Rα2 expression increases with glioma malignancy grade, and expression for high-grade tumors is bimodal, with approximately 58% of WHO grade IV gliomas over-expressing this receptor. By several measures, IL13Rα2 expression in patient samples and low-passage primary glioma lines most consistently correlates with the expression of signature genes defining mesenchymal subclass tumors and negatively correlates with proneural signature genes as defined by two studies. Positive associations were also noted with proliferative signature genes, whereas no consistent associations were found with either classical or neural signature genes. Probing the potential functional consequences of this mesenchymal association through IPA analysis suggests that IL13Rα2 expression is associated with activation of proinflammatory and immune pathways characteristic of mesenchymal subclass tumors. In addition, survival analyses indicate that IL13Rα2 over-expression is associated with poor patient prognosis, a single gene correlation ranking IL13Rα2 in the top ~1% of total gene expression probes with regard to survival association with WHO IV gliomas. This study better defines the functional consequences of IL13Rα2 expression by demonstrating association with mesenchymal signature gene expression and poor patient prognosis. It thus highlights the utility of IL13Rα2 as a therapeutic target, and helps define patient populations most likely to respond to immunotherapy in present and future clinical trials.
Collapse
Affiliation(s)
- Christine E. Brown
- Department of Cancer Immunotherapy & Tumor Immunology and Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail:
| | - Charles D. Warden
- Department of Molecular Medicine, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| | - Renate Starr
- Department of Cancer Immunotherapy & Tumor Immunology and Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| | - Xutao Deng
- Department of Molecular Medicine, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| | - Behnam Badie
- Department of Neurosurgery, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| | - Yate-Ching Yuan
- Department of Molecular Medicine, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| | - Stephen J. Forman
- Department of Cancer Immunotherapy & Tumor Immunology and Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| | - Michael E. Barish
- Department of Neurosciences, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, United States of America
| |
Collapse
|
50
|
Debinski W, Dickinson P, Rossmeisl JH, Robertson J, Gibo DM. New agents for targeting of IL-13RA2 expressed in primary human and canine brain tumors. PLoS One 2013; 8:e77719. [PMID: 24147065 PMCID: PMC3797726 DOI: 10.1371/journal.pone.0077719] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 09/11/2013] [Indexed: 11/20/2022] Open
Abstract
Interleukin 13 receptor alpha 2 (IL-13RA2) is over-expressed in a vast majority of human patients with high-grade astrocytomas like glioblastoma. Spontaneous astrocytomas in dogs resemble human disease and have been proposed as translational model system for investigation of novel therapeutic strategies for brain tumors. We have generated reagents for both detection and therapeutic targeting of IL-13RA2 in human and canine brain tumors. Peptides from three different regions of IL-13RA2 with 100% sequence identity between human and canine receptors were used as immunogens for generation of monoclonal antibodies. Recombinant canine mutant IL-13 (canIL-13.E13K) and canIL-13.E13K based cytotoxin were also produced. The antibodies were examined for their immunoreactivities in western blots, immunohistochemistry, immunofluorescence and cell binding assays using human and canine tumor specimen sections, tissue lysates and established cell lines; the cytotoxin was tested for specific cell killing. Several isolated MAbs were immunoreactive to IL-13RA2 in western blots of cell and tissue lysates from glioblastomas from both human and canine patients. Human and canine astrocytomas and oligodendrogliomas were also positive for IL-13RA2 to various degrees. Interestingly, both human and canine meningiomas also exhibited strong reactivity. Normal human and canine brain samples were virtually negative for IL-13RA2 using the newly generated MAbs. MAb 1E10B9 uniquely worked on tissue specimens and western blots, bound live cells and was internalized in GBM cells over-expressing IL-13RA2. The canIL-13.E13K cytotoxin was very potent and specific in killing canine GBM cell lines. Thus, we have obtained several monoclonal antibodies against IL-13RA2 cross-reacting with human and canine receptors. In addition to GBM, other brain tumors, such as high grade oligodendrogliomas, meningiomas and canine choroid plexus papillomas, appear to express the receptor at high levels and thus may be appropriate candidates for IL-13RA2-targeted imaging/therapies. Canine spontaneous primary brain tumors represent an excellent translational model for human counterparts.
Collapse
Affiliation(s)
- Waldemar Debinski
- The Brain Tumor Center of Excellence, Thomas K. Hearn Brain Tumor Research Center, Departments of Neurosurgery, Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Peter Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California at Davis, Davis, California, United States of America
| | - John H. Rossmeisl
- Department of Small Animal Clinical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - John Robertson
- Department of Small Animal Clinical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Denise M. Gibo
- The Brain Tumor Center of Excellence, Thomas K. Hearn Brain Tumor Research Center, Departments of Neurosurgery, Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| |
Collapse
|