1
|
Haines NA, Fowler MG, Zeh BG, Kriete CB, Bai Q, Wakefield MR, Fang Y. Unlocking the 'ova'-coming power: immunotherapy's role in shaping the future of ovarian cancer treatment. Med Oncol 2024; 41:67. [PMID: 38286890 DOI: 10.1007/s12032-023-02281-6] [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: 11/04/2023] [Accepted: 12/06/2023] [Indexed: 01/31/2024]
Abstract
Ovarian cancer is a prominent cancer worldwide with a relatively low survival rate for women diagnosed. Many individuals are diagnosed in the late stage of the disease and are prescribed a wide variety of treatment options. Current treatment options are primarily a combination of surgery and chemotherapy as well as a new but promising treatment involving immunotherapy. Nevertheless, contemporary therapeutic modalities exhibit a discernible lag in advancement when compared with the strides achieved in recent years in the context of other malignancies. Moreover, many surgery and chemotherapy options have a high risk for recurrence due to the late-stage diagnosis. Therefore, there is a necessity to further treatment options. There have been many new advancements in the field of immunotherapy. Immunotherapy has been approved for 16 various types of cancers and has shown significant treatment potential in many other cancers as well. Researchers have also found many promising outlooks for immunotherapy as a treatment for ovarian cancer. This review summarizes many of the new advancements in immunotherapy treatment options and could potentially offer valuable insights to gynecologists aimed at enhancing the efficacy of their treatment approaches for patients diagnosed with ovarian cancer.
Collapse
Affiliation(s)
- Nathan A Haines
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, 8025, Grand Ave, West Des Moines, IA, 50266, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mia G Fowler
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Benjamin G Zeh
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, 8025, Grand Ave, West Des Moines, IA, 50266, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Carter B Kriete
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, 8025, Grand Ave, West Des Moines, IA, 50266, USA.
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
| |
Collapse
|
2
|
Sato Y, Vatsan R, Joshi BH, Husain SR, Puri RK. A Novel Recombinant Modified Vaccinia Ankara Virus expressing Interleukin-13 Receptor α2 Antigen for Potential Cancer Immunotherapy. Curr Mol Med 2024; 24:758-770. [PMID: 36999709 DOI: 10.2174/1566524023666230331085007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND Genetically altered recombinant poxviruses hold great therapeutic promise in animal models of cancer. Poxviruses can induce effective cellmediated immune responses against tumor-associated antigens. Preventive and therapeutic vaccination with a DNA vaccine expressing IL-13Rα2 can mediate partial regression of established tumors in vivo, indicating that host immune responses against IL-13Rα2 need further augmentation. OBJECTIVE The aim of the study is developing a recombinant modified vaccinia Ankara (MVA) expressing IL-13Rα2 (rMVA-IL13Rα2) virus and study in vitro infectivity and efficacy against IL-13Rα2 positive cell lines. METHODS We constructed a recombinant MVA expressing IL-13Rα2 and a green fluorescent protein (GFP) reporter gene. Purified virus titration by infection of target cells and immunostaining using anti-vaccinia and anti-IL-13Rα2 antibodies was used to confirm the identity and purity of the rMVA-IL13Rα2. RESULTS Western Blot analysis confirmed the presence of IL-13Rα2 protein (~52 kDa). Flow cytometric analysis of IL-13Rα2 negative T98G glioma cells when infected with rMVA-IL13Rα2 virus demonstrated cell-surface expression of IL-13Rα2, indicating the infectivity of the recombinant virus. Incubation of T98G-IL13Rα2 cells with varying concentrations (0.1-100 ng/ml) of interleukin-13 fused to truncated Pseudomonas exotoxin (IL13-PE) resulted in depletion of GFP+ fluorescence in T98G-IL13Rα2 cells. IL13-PE (10-1000 ng/ml) at higher concentrations also inhibited the protein synthesis in T98G-IL13Rα2 cells compared to cells infected with the control pLW44-MVA virus. IL13- PE treatment of rMVA-IL13Rα2 infected chicken embryonic fibroblast and DF-1 cell line reduced virus titer compared to untreated cells. CONCLUSION rMVA-IL13Rα2 virus can successfully infect mammalian cells to express IL-13Rα2 in a biologically active form on the surface of infected cells. To evaluate the efficacy of rMVA-IL13Rα2, immunization studies are planned in murine tumor models.
Collapse
Affiliation(s)
- Yuki Sato
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
- Department of Research Promotion, Division of Cancer Research, Japan Agency for Medical Research and Development, 1-7-1, Otemachi, Chiyoda, Tokyo 100- 0004, Japan
| | - Ramjay Vatsan
- Gene Therapy Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Bharat H Joshi
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Syed R Husain
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
- Iovance Biotherapeutics, 825 Industrial Road, Suite 400, San Carlos, CA, California, 94070, USA
| | - Raj K Puri
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
- Iovance Biotherapeutics, 825 Industrial Road, Suite 400, San Carlos, CA, California, 94070, USA
| |
Collapse
|
3
|
Goutnik M, Iakovidis A, Still MEH, Moor RSF, Melnick K, Yan S, Abbas M, Huang J, Ghiaseddin AP. Advancements in chimeric antigen receptor-expressing T-cell therapy for glioblastoma multiforme: Literature review and future directions. Neurooncol Adv 2024; 6:vdae025. [PMID: 38486856 PMCID: PMC10939440 DOI: 10.1093/noajnl/vdae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive cancer that has been difficult to treat and often requires multimodal therapy consisting of surgery, radiotherapy, and chemotherapy. Chimeric antigen receptor-expressing (CAR-T) cells have been efficacious in treating hematological malignancies, resulting in several FDA-approved therapies. CAR-T cells have been more recently studied for the treatment of GBM, with some promising preclinical and clinical results. The purpose of this literature review is to highlight the commonly targeted antigens, results of clinical trials, novel modifications, and potential solutions for challenges that exist for CAR-T cells to become more widely implemented and effective in eradicating GBM.
Collapse
Affiliation(s)
- Michael Goutnik
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Alexandria Iakovidis
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Megan E H Still
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Rachel S F Moor
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Kaitlyn Melnick
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sandra Yan
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Muhammad Abbas
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jianping Huang
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ashley P Ghiaseddin
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
4
|
Rechberger JS, Porath KA, Zhang L, Nesvick CL, Schrecengost RS, Sarkaria JN, Daniels DJ. IL-13Rα2 Status Predicts GB-13 (IL13.E13K-PE4E) Efficacy in High-Grade Glioma. Pharmaceutics 2022; 14:922. [PMID: 35631512 PMCID: PMC9143740 DOI: 10.3390/pharmaceutics14050922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/14/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
High-grade gliomas (HGG) are devastating diseases in children and adults. In the pediatric population, diffuse midline gliomas (DMG) harboring H3K27 alterations are the most aggressive primary malignant brain tumors. With no effective therapies available, children typically succumb to disease within one year of diagnosis. In adults, glioblastoma (GBM) remains largely intractable, with a median survival of approximately 14 months despite standard clinical care of radiation and temozolomide. Therefore, effective therapies for these tumors remain one of the most urgent and unmet needs in modern medicine. Interleukin 13 receptor subunit alpha 2 (IL-13Rα2) is a cell-surface transmembrane protein upregulated in many HGGs, including DMG and adult GBM, posing a potentially promising therapeutic target for these tumors. In this study, we investigated the pharmacological effects of GB-13 (also known as IL13.E13K-PE4E), a novel peptide-toxin conjugate that contains a targeting moiety designed to bind IL-13Rα2 with high specificity and a point-mutant cytotoxic domain derived from Pseudomonas exotoxin A. Glioma cell lines demonstrated a spectrum of IL-13Rα2 expression at both the transcript and protein level. Anti-tumor effects of GB-13 strongly correlated with IL-13Rα2 expression and were reflected in apoptosis induction and decreased cell proliferation in vitro. Direct intratumoral administration of GB-13 via convection-enhanced delivery (CED) significantly decreased tumor burden and resulted in prolonged survival in IL-13Rα2-upregulated orthotopic xenograft models of HGG. In summary, administration of GB-13 demonstrated a promising pharmacological response in HGG models both in vitro and in vivo in a manner strongly associated with IL-13Rα2 expression, underscoring the potential of this IL-13Rα2-targeted therapy in a subset of HGG with increased IL-13Rα2 levels.
Collapse
Affiliation(s)
- Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Kendra A. Porath
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.P.); (J.N.S.)
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
| | - Cody L. Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
| | | | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.P.); (J.N.S.)
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Kang JH, Desjardins A. Convection-enhanced delivery for high-grade glioma. Neurooncol Pract 2021; 9:24-34. [DOI: 10.1093/nop/npab065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Glioblastoma (GBM) is the most common adult primary malignant brain tumor and is associated with a dire prognosis. Despite multi-modality therapies of surgery, radiation, and chemotherapy, its 5-year survival rate is 6.8%. The presence of the blood-brain barrier (BBB) is one factor that has made GBM difficult to treat. Convection-enhanced delivery (CED) is a modality that bypasses the BBB, which allows the intracranial delivery of therapies that would not otherwise cross the BBB and avoids systemic toxicities. This review will summarize prior and ongoing studies and highlights practical considerations related to clinical care to aid providers caring for a high-grade glioma patient being treated with CED. Although not the main scope of this paper, this review also touches upon relevant technical considerations of using CED, an area still under much development.
Collapse
Affiliation(s)
- Jennifer H Kang
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
6
|
Kang YJ, Holley CK, Abidian MR, Madhankumar AB, Connor J, Majd S. Tumor Targeted Delivery of an Anti-Cancer Therapeutic: An In Vitro and In Vivo Evaluation. Adv Healthc Mater 2021; 10:e2001261. [PMID: 33191612 DOI: 10.1002/adhm.202001261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/01/2020] [Indexed: 02/01/2023]
Abstract
The limited effectiveness of current therapeutics against malignant brain gliomas has led to an urgent need for development of new formulations against these tumors. Chelator Dp44mT (di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone) presents a promising candidate to defeat gliomas due to its exceptional anti-tumor activity and its unique ability to overcome multidrug resistance. The goal of this study is to develop a targeted nano-carrier for Dp44mT delivery to glioma tumors and to assess its therapeutic efficacy in vitro and in vivo. Dp44mT is loaded into poly(ethylene glycol) (PEG)ylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) decorated with glioma-targeting ligand Interlukin 13 (IL13). IL13-conjugation enhanced the NP uptake by glioma cells and also improved their transport across an in vitro blood-brain-barrier (BBB) model. This targeted formulation showed an outstanding toxicity towards glioma cell lines and patient-derived stem cells in vitro, with IC50 values less than 125 nM, and caused no significant death in healthy brain microvascular endothelial cells. In vivo, when tested on a xenograft mouse model, IL13-conjugated Dp44mT-NPs reduced the glioma tumor growth by ≈62% while their untargeted counterparts reduced the tumor growth by only ≈16%. Notably, this formulation does not cause any significant weight loss or kidney/liver toxicity in mice, demonstrating its great therapeutic potential.
Collapse
Affiliation(s)
- You Jung Kang
- Department of Biomedical Engineering Pennsylvania State University University Park PA 16802 USA
| | - Claire K. Holley
- Department of Biomedical Engineering University of Houston Houston TX 77204 USA
| | | | | | - James Connor
- Department of Neurosurgery Penn State University College of Medicine Hershey PA 17033 USA
| | - Sheereen Majd
- Department of Biomedical Engineering University of Houston Houston TX 77204 USA
| |
Collapse
|
7
|
Prolonged survival in secondary glioblastoma following local injection of targeted alpha therapy with 213Bi-substance P analogue. Eur J Nucl Med Mol Imaging 2018; 45:1636-1644. [PMID: 29713762 PMCID: PMC6061489 DOI: 10.1007/s00259-018-4015-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/09/2018] [Indexed: 12/14/2022]
Abstract
Background Glioblastoma multiforme (GBM), the most common malignant brain tumor, mainly manifests as a primary de novo and less frequently as a secondary glial neoplasm. GBM has been demonstrated to overexpress the NK-1 receptor and substance P can be used as a ligand for targeted therapy. Alpha emitters, e.g. 213Bi, that deposit their high energy within a short range allow the selective irradiation of tumor cells while sparing adjacent neuronal structures. Material and methods Among 50 glioma patients of different subtypes that have to date been treated with targeted alpha therapy at the Medical University Warsaw, we report here the data on nine patients with secondary GBM. Following surgery, chemo- and radiotherapy, recurrent GBM was treated by intracavitary injection of 1–6 doses of 0.9–2.3 GBq 213Bi- DOTA-[Thi8,Met(O2)11]-substance P (213Bi-DOTA-SP) in 2-month intervals. 68Ga-DOTA-[Thi8,Met(O2)11]-substance P (68Ga-DOTA-SP) was co-injected with the therapeutic doses to assess biodistribution using PET/CT. Therapeutic response was monitored with MRI. Results Treatment with activities ranging from 1.4 to 9.7 (median 5.8) GBq 213Bi- DOTA-SP was well tolerated with only mild transient adverse reactions, mainly headaches due to a transient perfocal edema reaction. The median progression free survival and overall survival time following the initiation of alpha therapy was 5.8 and 16.4 months, respectively. The median overall survival time from the first diagnosis was 52.3 months. Two out of nine patients are still alive 39 and 51 months, respectively, after the initiation of the therapy. Conclusions Targeted alpha therapy of secondary GBM with 213Bi-DOTA-SP is safe and well tolerated and may evolve as a promising novel therapeutic option for secondary GBM.
Collapse
|
8
|
Abstract
Background: The prognosis for most patients with primary brain tumors remains poor. Recent advances in molecular and cell biology have led to a greater understanding of molecular alterations in brain tumors. These advances are being translated into new therapies that will hopefully improve the prognosis for patients with brain tumors. Methods: We reviewed the literature on small molecule targeted agents and monoclonal antibodies used in brain tumor research and brain tumor clinical trials for the past 20 years. Results: Brain tumors commonly express molecular abnormalities. These alterations can lead to the activation of cell pathways involved in cell proliferation. This knowledge has led to interest in novel anti-brain-tumor therapies targeting key components of these pathways. Many drugs and monoclonal antibodies have been developed that modulate these pathways and are in various stages of testing. Conclusions: The use of targeted therapies against brain tumors promises to improve the prognosis for patients with brain tumors. However, as the molecular pathogenesis of brain tumors has not been linked to a single genetic defect or target, molecular agents may need to be used in combinations or in tandem with cytotoxic agents. Further study of these agents in well-designed cooperative clinical trials is needed.
Collapse
Affiliation(s)
- Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA 94143-0350, USA
| | | |
Collapse
|
9
|
Muro K, Das S, Raizer JJ. Convection-Enhanced and Local Delivery of Targeted Cytotoxins in the Treatment of Malignant Gliomas. Technol Cancer Res Treat 2016; 5:201-13. [PMID: 16700617 DOI: 10.1177/153303460600500304] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Despite advances in our knowledge about the genesis, molecular biology, and natural history of malignant gliomas and the use of a multi-disciplinary approach to their treatment, patients harboring this diagnosis continue to face a grim prognosis. At the time of diagnosis, patients typically undergo surgery for the establishment of a histologic diagnosis, the reduction of tumor burden, and the relief of mass effect, with the maintenance of the patient's neurological function in mind. This is followed by the administration of adjuvant therapeutics, including radiation therapy and chemotherapy. Many investigational agents with laboratory evidence of efficacy against malignant gliomas have not met their promise in the clinical setting, largely due to the barriers that they must overcome to reach the tumor at a therapeutically meaningful concentration for a durable period of time. The relevant aspects of the blood-brain barrier, blood-tumor barrier, and blood-cerebrospinal fluid barrier, as they pertain to the delivery of agents to the tumor, will be discussed along with the strategies devised to circumvent them. This discussion will be followed by a description of agents currently in preclinical and clinical development, many of which are the result of intense ongoing research into the molecular biology of gliomas.
Collapse
Affiliation(s)
- Kenji Muro
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Abbott Hall, Suite 1123, 710 N Lake Shore Drive, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
10
|
Ung TH, Malone H, Canoll P, Bruce JN. Convection-enhanced delivery for glioblastoma: targeted delivery of antitumor therapeutics. CNS Oncol 2015; 4:225-34. [PMID: 26103989 DOI: 10.2217/cns.15.12] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is the most common primary brain tumor in adults and carries a dismal prognosis despite advancements in treatment. Diffuse tumor infiltration precludes curative surgical resection and necessitates advancements in drug delivery mechanisms. Convection-enhanced delivery (CED) enables continuous local drug delivery for a diverse population of antitumor agents. Importantly, CED circumvents therapeutic challenges posed by the blood-brain barrier by facilitating concentrated local therapeutic drug delivery with limited systemic effects. Here, we present a concise review of properties essential for safe and efficient convection-enhanced drug delivery, as well as a focused review of clinical studies evaluating CED in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Timothy H Ung
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Hani Malone
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter Canoll
- Department of Pathology & Cellular Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
11
|
Pahle J, Aumann J, Kobelt D, Walther W. Oncoleaking: Use of the Pore-Forming Clostridium perfringens Enterotoxin (CPE) for Suicide Gene Therapy. Methods Mol Biol 2015; 1317:69-85. [PMID: 26072402 DOI: 10.1007/978-1-4939-2727-2_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Suicide gene therapy has been shown to be very efficient in tumor eradication. Numerous suicide genes were tested in vitro and in vivo demonstrating their therapeutic potential in clinical trials. Apart from this, still growing efforts are made to generate more targeted and more effective suicide gene systems for cancer gene therapy. In this regard bacterial toxins are an alternative, which add to the broad spectrum of different suicide strategies. In this context, the claudin-targeted bacterial Clostridium perfringens enterotoxin (CPE) is an attractive new type of suicide oncoleaking gene, which as pore-forming protein exerts specific and rapid toxicity towards claudin-3- and -4-overexpressing cancers. In this chapter we describe the generation and use of CPE-expressing vectors for the effective tumor cell killing as novel suicide gene approach particularly for treatment of therapy refractory tumors.
Collapse
Affiliation(s)
- Jessica Pahle
- Experimental and Clinical Research Center (ECRC), Charité University Medicine Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
12
|
Fagan-Solis KD, Reaves DK, Rangel MC, Popoff MR, Stiles BG, Fleming JM. Challenging the roles of CD44 and lipolysis stimulated lipoprotein receptor in conveying Clostridium perfringens iota toxin cytotoxicity in breast cancer. Mol Cancer 2014; 13:163. [PMID: 24990559 PMCID: PMC4086999 DOI: 10.1186/1476-4598-13-163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/24/2014] [Indexed: 01/01/2023] Open
Abstract
Background Translational exploration of bacterial toxins has come to the forefront of research given their potential as a chemotherapeutic tool. Studies in select tissues have demonstrated that Clostridium perfringens iota toxin binds to CD44 and lipolysis stimulated lipoprotein receptor (LSR) cell-surface proteins. We recently demonstrated that LSR expression correlates with estrogen receptor positive breast cancers and that LSR signaling directs aggressive, tumor-initiating cell behaviors. Herein, we identify the mechanisms of iota toxin cytotoxicity in a tissue-specific, breast cancer model with the ultimate goal of laying the foundation for using iota toxin as a targeted breast cancer therapy. Methods In vitro model systems were used to determine the cytotoxic effect of iota toxin on breast cancer intrinsic subtypes. The use of overexpression and knockdown technologies confirmed the roles of LSR and CD44 in regulating iota toxin endocytosis and induction of cell death. Lastly, cytotoxicity assays were used to demonstrate the effect of iota toxin on a validated set of tamoxifen resistant breast cancer cell lines. Results Treatment of 14 breast cancer cell lines revealed that LSR+/CD44- lines were highly sensitive, LSR+/CD44+ lines were slightly sensitive, and LSR-/CD44+ lines were resistant to iota cytotoxicity. Reduction in LSR expression resulted in a significant decrease in toxin sensitivity; however, overexpression of CD44 conveyed toxin resistance. CD44 overexpression was correlated with decreased toxin-stimulated lysosome formation and decreased cytosolic levels of iota toxin. These findings indicated that expression of CD44 drives iota toxin resistance through inhibition of endocytosis in breast cancer cells, a role not previously defined for CD44. Moreover, tamoxifen-resistant breast cancer cells exhibited robust expression of LSR and were highly sensitive to iota-induced cytotoxicity. Conclusions Collectively, these data are the first to show that iota toxin has the potential to be an effective, targeted therapy for breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Jodie M Fleming
- Department of Biology, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
13
|
IL-13Rα2 mediates PNR-induced migration and metastasis in ERα-negative breast cancer. Oncogene 2014; 34:1596-607. [PMID: 24747967 DOI: 10.1038/onc.2014.53] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/30/2014] [Accepted: 02/11/2014] [Indexed: 12/21/2022]
Abstract
Emerging evidence has linked photoreceptor cell-specific nuclear receptor (PNR/NR2E3), an orphan nuclear hormone receptor, to human breast cancer. PNR was shown to be a transcriptional activator of estrogen receptor-α (ERα) in ERα-positive breast cancer cell lines and high-level expression of PNR correlates with favorable response of ERα-positive breast cancer patients to tamoxifen. Interestingly, gene expression microarray study shows that PNR regulates distinct genes from those regulated by ERα, suggesting that PNR could have ERα-independent functions. Herein, we investigated the function of PNR in ERα-negative breast cancer cells. Our results showed that PNR-induced cell migration and metastasis of ERα-negative breast cancer cells both in vitro and in vivo, and the effect was attributed to the upregulation of interleukin (IL)-13Rα2, a high-affinity receptor for IL-13 that regulates tumor growth, invasion and metastasis of various human cancers. Mechanistically, PNR activated transcription of IL-13Rα2 through direct recruitment to IL-13Rα2 promoter. Upon stimulation with IL-13, IL-13Rα2 increased the extracellular signal-regulated kinases 1 and 2 phosphorylation, which led to breast cancer migration and metastasis. The IL-13 triggered signal cascade was specific to IL-13Rα2, as the closely related IL-13Rα1 was not regulated by PNR. IL-13Rα2 is a novel tumor antigen that is overexpressed in a variety of solid tumor types. This study presents the first evidence that PNR could promote ERα-negative breast cancer metastasis through activation of IL-13Rα2-mediated signaling pathway.
Collapse
|
14
|
Wen PY, Kesari S, Drappatz J. Malignant gliomas: strategies to increase the effectiveness of targeted molecular treatment. Expert Rev Anticancer Ther 2014; 6:733-54. [PMID: 16759164 DOI: 10.1586/14737140.6.5.733] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, there has been increasing interest in the use of targeted molecular agents for the treatment of malignant gliomas. These agents are generally well tolerated but have demonstrated only modest activity. In this article, the current status of targeted molecular agents for malignant gliomas will be reviewed and strategies to improve their effectiveness will be discussed.
Collapse
Affiliation(s)
- Patrick Y Wen
- Harvard Medical School, Dana-Farber/Brigham and Women's Cancer Center, SW430D, Boston, MA 02115, USA.
| | | | | |
Collapse
|
15
|
Lefranc F, Sadeghi N, Camby I, Metens T, Dewitte O, Kiss R. Present and potential future issues in glioblastoma treatment. Expert Rev Anticancer Ther 2014; 6:719-32. [PMID: 16759163 DOI: 10.1586/14737140.6.5.719] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The treatment of glioblastomas requires a multidisciplinary approach that takes the presently incurable nature of the disease into consideration. Treatments are multimodal and include surgery, radiotherapy and chemotherapy. Current recommendations are that patients with glioblastomas should undergo maximum surgical resection, followed by concurrent radiation and chemotherapy with the novel alkylating drug temozolomide. This is then to be followed by additional adjuvant temozolomide for a period of up to 6 months. Major advances in surgical and imaging technologies used to treat glioblastoma patients are described. These technologies include magnetic resonance imaging and metabolic data that are helpful in the diagnosis and guiding of surgical resection. However, glioblastomas almost invariably recur near their initial sites. Disease progression usually occurs within 6 months and leads rapidly to death. A number of signaling pathways can be activated constitutively in migrating glioma cells, thus rendering these cells resistant to proapoptotic insults, such as conventional chemotherapies. Therefore, the molecular and cellular therapies and local drug delivery that could be used to complement conventional treatments are described, and some of the currently ongoing clinical trials are reviewed, with respect to these new approaches.
Collapse
Affiliation(s)
- Florence Lefranc
- Departments of Neurosurgery, Erasme University Hospital, Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
16
|
Sato Y, Vatsan R, Joshi BH, Husain SR, Puri RK. Generation of interleukin-13 receptor alpha2 antigen expressing modified vaccinia ankara recombinant virus for potential cancer immunotherapy. J Immunother Cancer 2014. [PMCID: PMC4288700 DOI: 10.1186/2051-1426-2-s3-p58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
17
|
Hall B, Nakashima H, Sun ZJ, Sato Y, Bian Y, Husain SR, Puri RK, Kulkarni AB. Targeting of interleukin-13 receptor α2 for treatment of head and neck squamous cell carcinoma induced by conditional deletion of TGF-β and PTEN signaling. J Transl Med 2013; 11:45. [PMID: 23421960 PMCID: PMC3598213 DOI: 10.1186/1479-5876-11-45] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/11/2013] [Indexed: 11/19/2022] Open
Abstract
Background The sixth leading class of cancer worldwide is head and neck cancer, which typically arise within the squamous epithelium of the oral mucosa. Human head and neck squamous cell carcinoma (HNSCC) is known to be difficult to treat and has only a 50% five-year survival rate. With HNSCC, novel therapeutics are needed along with a means of rapidly screening anti-cancer agents in vivo, such as mouse models. Methods In order to develop new animal models of cancer to test safety and efficacy of novel therapeutic agents for human HNSCC, tumors resembling clinical cases of human HNSCC were induced in the head and neck epithelium of a genetically engineered mouse model. This mouse model was generated by conditional deletion of two tumor suppressors, Transforming Growth Factor-β Receptor 1 (TGFβRI) and Phosphatase and Tensin homolog (PTEN), in the oral epithelium. We discovered that the tumors derived from these Tgfbr1/Pten double conditional knockout (2cKO) mice over-expressed IL-13Rα2, a high affinity receptor for IL-13 that can function as a tumor antigen. To demonstrate a proof-of-concept that targeted therapy against IL-13Rα2 expression would have any antitumor efficacy in this spontaneous tumor model, these mice were treated systemically with IL-13-PE, a recombinant immunotoxin consisting of IL-13 fused to the Pseudomonas exotoxin A. Results Tgfbr1/Pten 2cKO mice when treated with IL-13-PE displayed significantly increased survival when compared to the untreated control mice. The untreated mice exhibited weight loss, particularly with the rapid onset of tongue tumors, but the treated mice gained weight while on IL-13-PE therapy and showed no clinical signs of toxicity due to the immunotoxin. Expression of IL-13Rα2 in tumors was significantly decreased with IL-13-PE treatment as compared to the controls and the number of myeloid-derived suppressor cells (MDSC) was also significantly reduced in the spleens of the IL-13-PE treated mice. Conclusions Our study demonstrates that the Tgfbr1/Pten 2cKO mouse model of human HNSCC is a useful model for assessing antitumor activity of new cancer therapeutic agents, and that IL-13-PE has therapeutic potential to treat human head and neck cancer.
Collapse
Affiliation(s)
- Bradford Hall
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Building 30, Room 130, Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ma X, Lv P, Ye S, Zhang Y, Li S, Kan C, Fan L, Liu R, Luo D, Wang A, Yang W, Yang S, Bai X, Lu Y, Ma D, Xi L, Wang S. DT390-triTMTP1, a Novel Fusion Protein of Diphtheria Toxin with Tandem Repeat TMTP1 Peptide, Preferentially Targets Metastatic Tumors. Mol Pharm 2012. [PMID: 23198829 DOI: 10.1021/mp300125k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xiangyi Ma
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Peng Lv
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuangmei Ye
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Yiqun Zhang
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Shu Li
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Chunyi Kan
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Liangsheng Fan
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Ronghua Liu
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Danfeng Luo
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Aiping Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Wanhua Yang
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Shuhong Yang
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Xiangyang Bai
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Yunping Lu
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Ding Ma
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Ling Xi
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| | - Shixuan Wang
- Cancer Biology
Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue,
Wuhan, Hubei 430030, People's Republic of China
| |
Collapse
|
19
|
Kong S, Sengupta S, Tyler B, Bais AJ, Ma Q, Doucette S, Zhou J, Sahin A, Carter BS, Brem H, Junghans RP, Sampath P. Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells. Clin Cancer Res 2012; 18:5949-60. [PMID: 22966020 PMCID: PMC4337849 DOI: 10.1158/1078-0432.ccr-12-0319] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) remains highly incurable, with frequent recurrences after standard therapies of maximal surgical resection, radiation, and chemotherapy. To address the need for new treatments, we have undertaken a chimeric antigen receptor (CAR) "designer T cell" (dTc) immunotherapeutic strategy by exploiting interleukin (IL)13 receptor α-2 (IL13Rα2) as a GBM-selective target. EXPERIMENTAL DESIGN We tested a second-generation IL13 "zetakine" CAR composed of a mutated IL13 extracellular domain linked to intracellular signaling elements of the CD28 costimulatory molecule and CD3ζ. The aim of the mutation (IL13.E13K.R109K) was to enhance selectivity of the CAR for recognition and killing of IL13Rα2(+) GBMs while sparing normal cells bearing the composite IL13Rα1/IL4Rα receptor. RESULTS Our aim was partially realized with improved recognition of tumor and reduced but persisting activity against normal tissue IL13Rα1(+) cells by the IL13.E13K.R109K CAR. We show that these IL13 dTcs were efficient in killing IL13Rα2(+) glioma cell targets with abundant secretion of cytokines IL2 and IFNγ, and they displayed enhanced tumor-induced expansion versus control unmodified T cells in vitro. In an in vivo test with a human glioma xenograft model, single intracranial injections of IL13 dTc into tumor sites resulted in marked increases in animal survivals. CONCLUSIONS These data raise the possibility of immune targeting of diffusely invasive GBM cells either via dTc infusion into resection cavities to prevent GBM recurrence or via direct stereotactic injection of dTcs to suppress inoperable or recurrent tumors. Systemic administration of these IL13 dTc could be complicated by reaction against normal tissues expressing IL13Ra1.
Collapse
Affiliation(s)
- Seogkyoung Kong
- Brain Tumor Lab, Department of Neurosurgery, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Sadhak Sengupta
- Brain Tumor Lab, Department of Neurosurgery, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Betty Tyler
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anthony J. Bais
- Biotherapeutics Development Lab, Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Qiangzhong Ma
- Biotherapeutics Development Lab, Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Saryn Doucette
- Department of Pathology, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Jinyuan Zhou
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ayguen Sahin
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Henry Brem
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard P. Junghans
- Biotherapeutics Development Lab, Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Prakash Sampath
- Brain Tumor Lab, Department of Neurosurgery, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island
| |
Collapse
|
20
|
Vogelbaum MA, Iannotti CA. Convection-enhanced delivery of therapeutic agents into the brain. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:355-62. [PMID: 22230453 DOI: 10.1016/b978-0-444-52138-5.00023-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
CED of therapeutic agents remains a promising strategy for treating malignant gliomas and non-neoplastic neurological diseases. Although initial clinical trials have failed to show survival benefit for new agents delivered via this approach, multiple earlier stage trials have addressed only a fraction of the myriad of technical and technological issues that surround this novel approach. Development of CED has been limited by the fact that both new technologies and novel therapeutic agents are being developed simultaneously.New trials are being planned to investigate agents that can be coinfused with radiographic tracers, as well as novel catheters that avoid problems with backflow and potentially will provide more reliable drug distribution.
Collapse
|
21
|
Nakashima H, Husain SR, Puri RK. IL-13 receptor-directed cancer vaccines and immunotherapy. Immunotherapy 2012; 4:443-51. [DOI: 10.2217/imt.12.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Many immunotherapy approaches including therapeutic cancer vaccines targeting specific tumor-associated antigens are at various stages of development. Although the significance of overexpression of (IL-13Rα2) in cancer is being actively investigated, we have reported that IL-13Rα2 is a novel tumor-associated antigen. The IL-13Rα2-directed cancer vaccine is one of the most promising approaches to tumor immunotherapy, because of the selective expression of IL-13Rα2 in various solid tumor types but not in normal tissues. In this article, we will summarize its present status and potential strategies to improve IL-13Rα2-directed cancer vaccines for an optimal therapy of cancer.
Collapse
Affiliation(s)
- Hideyuki Nakashima
- Tumor Vaccines & Biotechnology Branch, Division of Cellular & Gene Therapies, Food & Drug Administration, Center for Biologics Evaluation & Research, NIH Building 29B, Room 2NN20, 29 Lincoln Drive, Bethesda, MD 20892, USA
| | - Syed R Husain
- Tumor Vaccines & Biotechnology Branch, Division of Cellular & Gene Therapies, Food & Drug Administration, Center for Biologics Evaluation & Research, NIH Building 29B, Room 2NN20, 29 Lincoln Drive, Bethesda, MD 20892, USA
| | - Raj K Puri
- Tumor Vaccines & Biotechnology Branch, Division of Cellular & Gene Therapies, Food & Drug Administration, Center for Biologics Evaluation & Research, NIH Building 29B, Room 2NN20, 29 Lincoln Drive, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
The legacy of nanotechnology: revolution and prospects in neurosurgery. Int J Surg 2011; 9:608-14. [PMID: 22027280 DOI: 10.1016/j.ijsu.2011.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/26/2011] [Accepted: 10/11/2011] [Indexed: 10/16/2022]
Abstract
Nanotechnology has been an ever-growing field since the discovery of carbon fullerenes, and is being assimilated progressively into a variety of other disciplines including medical science. The association with neurosurgery had initially been less well characterized compared to other organ systems, but has recently offered promising future potential for a wide range of utilities including new therapeutic options for Glioblastoma Multiforme, neurprotection against oxidative stress, nerve nanorepair, nanodiagnosis of Alzheimer's disease, nanoimaging with nanoparticles and quantum dots, nanomanipulation of CNS with surgical nanobots, and nanoneuromodulation with nanofibres & nanowires. This article examines such potentials as well as others, of the utility of nanotechnology in Neurosurgery.
Collapse
|
23
|
Walther W, Petkov S, Kuvardina ON, Aumann J, Kobelt D, Fichtner I, Lemm M, Piontek J, Blasig IE, Stein U, Schlag PM. Novel Clostridium perfringens enterotoxin suicide gene therapy for selective treatment of claudin-3- and -4-overexpressing tumors. Gene Ther 2011; 19:494-503. [PMID: 21975465 DOI: 10.1038/gt.2011.136] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bacterial toxins are known to be effective for cancer therapy. Clostridium perfringens enterotoxin (CPE) is produced by the bacterial Clostridium type A strain. The transmembrane proteins claudin-3 and -4, often overexpressed in numerous human epithelial tumors (for example, colon, breast, pancreas, prostate and ovarian), are the targeted receptors for CPE. CPE binding to them triggers formation of membrane pore complexes leading to rapid cell death. In this study, we aimed at selective tumor cell killing by CPE gene transfer. We generated expression vectors bearing the bacterial wild-type CPE cDNA (wtCPE) or translation-optimized CPE (optCPE) cDNA for in vitro and in vivo gene therapy of claudin-3- and -4-overexpressing tumors. The CPE expression analysis at messenger RNA and protein level revealed more efficient expression of optCPE compared with wtCPE. Expression of optCPE showed rapid cytotoxic activity, hightened by CPE release as bystander effect. Cytotoxicity of up to 100% was observed 72 h after gene transfer and is restricted to claudin-3-and -4-expressing tumor lines. MCF-7 and HCT116 cells with high claudin-4 expression showed dramatic sensitivity toward CPE toxicity. The claudin-negative melanoma line SKMel-5, however, was insensitive toward CPE gene transfer. The non-viral intratumoral in vivo gene transfer of optCPE led to reduced tumor growth in MCF-7 and HCT116 tumor-bearing mice compared with the vector-transfected control groups. This novel approach demonstrates that CPE gene transfer can be employed for a targeted suicide gene therapy of claudin-3- and -4-overexpressing tumors, leading to the rapid and efficient tumor cell killing in vitro and in vivo.
Collapse
Affiliation(s)
- W Walther
- Experimental and Clinical Research Center, Charité University Medicine Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Magnetically-enabled and MR-monitored selective brain tumor protein delivery in rats via magnetic nanocarriers. Biomaterials 2011; 32:6245-53. [PMID: 21632104 DOI: 10.1016/j.biomaterials.2011.05.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 05/01/2011] [Indexed: 11/20/2022]
Abstract
The delivery of bioactive proteins to tumors is associated with many difficulties that have impeded clinical translation of these promising therapeutics. Herein we present an approach, including (1) use of magnetically-responsive and MRI-visible nanoparticles as drug carriers, (2) topography-optimized intra-arterial magnetic targeting, (3) MRI-guided subject alignment within the magnetic field, and (4) surface modification of the protein drug with membrane-permeable polyethyleneimine (PEI), to prevail over the obstacles in protein delivery. Applying these methodologies, we demonstrated the delivery of a significant quantity of β-galactosidase selectively into brain tumors of glioma-bearing rats, while limiting the exposure of normal brain regions. Clinical viability of the technologies utilized, and the ability to deliver proteins at high nanomolar-range tumor concentrations, sufficient to completely eradicate a tumor lesion with existing picomolar-potency protein toxins, renders the prospect of enabling protein-based cancer therapy extremely promising.
Collapse
|
25
|
Yoon DJ, Liu CT, Quinlan DS, Nafisi PM, Kamei DT. Intracellular trafficking considerations in the development of natural ligand-drug molecular conjugates for cancer. Ann Biomed Eng 2011; 39:1235-51. [PMID: 21350890 PMCID: PMC3069328 DOI: 10.1007/s10439-011-0280-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 02/12/2011] [Indexed: 12/01/2022]
Abstract
Overexpressed receptors, characteristic of many cancers, have been targeted by various researchers to achieve a more specific treatment for cancer. A common approach is to use the natural ligand for the overexpressed receptor as a cancer-targeting agent which can deliver a chemically or genetically conjugated toxic molecule. However, it has been found that the therapeutic efficacy of such ligand-drug molecular conjugates can be limited, since they naturally follow the intracellular trafficking pathways of the endogenous ligands. Therefore, a thorough understanding of the intracellular trafficking properties of these ligands can lead to novel design criteria for engineering ligands to be more effective drug carriers. This review presents a few commonly used ligand/receptor systems where intracellular trafficking considerations can potentially improve the therapeutic efficacy of the ligand-drug molecular conjugates.
Collapse
Affiliation(s)
- Dennis J. Yoon
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Christina T. Liu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Devin S. Quinlan
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Parsa M. Nafisi
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Daniel T. Kamei
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
26
|
Arko L, Katsyv I, Park GE, Luan WP, Park JK. Experimental approaches for the treatment of malignant gliomas. Pharmacol Ther 2010; 128:1-36. [PMID: 20546782 PMCID: PMC2939300 DOI: 10.1016/j.pharmthera.2010.04.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 04/28/2010] [Indexed: 12/13/2022]
Abstract
Malignant gliomas, which include glioblastomas and anaplastic astrocytomas, are the most common primary tumors of the brain. Over the past 30 years, the standard treatment for these tumors has evolved to include maximal safe surgical resection, radiation therapy and temozolomide chemotherapy. While the median survival of patients with glioblastomas has improved from 6 months to 14.6 months, these tumors continue to be lethal for the vast majority of patients. There has, however, been recent substantial progress in our mechanistic understanding of tumor development and growth. The translation of these genetic, epigenetic and biochemical findings into therapies that have been tested in clinical trials is the subject of this review.
Collapse
Affiliation(s)
- Leopold Arko
- Surgical and Molecular Neuro-oncology Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
27
|
Saka M, Amano T, Kajiwara K, Yoshikawa K, Ideguchi M, Nomura S, Fujisawa H, Kato S, Fujii M, Ueno K, Hinoda Y, Suzuki M. Vaccine therapy with dendritic cells transfected with Il13ra2 mRNA for glioma in mice. J Neurosurg 2010; 113:270-9. [PMID: 19895199 DOI: 10.3171/2009.9.jns09708] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The Il13ra2 gene is often overexpressed in brain tumors, making Il13ra2 one of the vaccine targets for immunotherapy of glioma. In this study, using a mouse glioma model, the authors tested the hypothesis that vaccination using dendritic cells transfected with Il13ra2 mRNA induces strong immunological antitumor effects. METHODS A plasmid was constructed for transduction of the mRNAs transcribed in vitro into dendritic cells. This was done to transport the intracellular protein efficiently into major histocompatibility complex class II compartments by adding a late endosomal/lysosomal sorting signal to the Il13ra2 gene. The dendritic cells transfected with this Il13ra2 mRNA were injected intraperitoneally into the mouse glioma model at 3 and 10 days after tumor cell implantation. The antitumor effects were estimated based on the survival rate, results of histological analysis, and immunohistochemical findings for immune cells. RESULTS The group treated by vaccination therapy with dendritic cells transfected with Il13ra2 mRNA survived significantly longer than did the control groups. Immunohistochemical analysis revealed that greater numbers of T lymphocytes containing CD4+ and CD8+ T cells were found in the group vaccinated with dendritic cells transfected with Il13ra2 mRNA. CONCLUSIONS These results demonstrate the therapeutic potential of vaccination with dendritic cells transfected with Il13ra2 mRNA for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Makoto Saka
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
He H, Xu J, Nelson PS, Marshall FF, Chung LWK, Zhau HE, He D, Wang R. Differential expression of the alpha2 chain of the interleukin-13 receptor in metastatic human prostate cancer ARCaPM cells. Prostate 2010; 70:993-1001. [PMID: 20166133 PMCID: PMC3180892 DOI: 10.1002/pros.21133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The alpha2 chain of the interleukin-13 receptor (IL13Ralpha2) is a high-affinity receptor and a candidate target for cytotoxic killing of cancer cells. Availability of a human prostate cancer cell line with high level of IL13Ralpha2 expression will facilitate the development of therapeutic modalities. METHODS ARCaP(E) and ARCaP(M) human prostate cancer cell lines were subjected to comparative analyses of gene expression. Expression of the IL13Ralpha2 protein was confirmed by Western blotting and immunostaining. IL13Ralpha2 proteins in xenograft tumors and clinical human prostate cancer specimens were detected by specific antibodies. LNCaP prostate cancer cells stably transfected with IL13Ralpha2 were examined for accelerated growth in athymic mice. RESULTS We found that IL13Ralpha2 proteins could be detected in both the ARCaP(E) and ARCaP(M) cells, but the expression level in ARCaP(M) was more than 17-fold higher than in ARCaP(E) cells. Importantly, the ARCaP lineage represented the only human prostate cancer cell line that expresses IL13Ralpha2 proteins at the level detectable by Western blotting. Expression of IL13Ralpha2 was accompanied by resistance to the anti-tumor activity of interleukin-13 (IL-13). ARCaP cells were found to be insensitive to growth inhibition upon IL-13 treatment, while overexpression of IL13Ralpha2 in LNCaP cells promoted intratibial tumor growth in athymic mice. CONCLUSIONS Differential IL13Ralpha2 expression may account for the high tumorigenic and metastatic potential of ARCaP(M) cells. The unique expression of IL13Ralpha2 makes ARCaP lineage an attractive model for evaluating the targeting efficacy of therapeutic agents based on IL13Ralpha2 protein expression.
Collapse
Affiliation(s)
- Hui He
- Department of Urology, the First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China 710061
- Molecular Urology and Therapeutics, Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322
| | - Jianchun Xu
- Molecular Urology and Therapeutics, Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Fray F. Marshall
- Molecular Urology and Therapeutics, Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322
| | - Leland W. K. Chung
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Haiyen E. Zhau
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Dalin He
- Department of Urology, the First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China 710061
| | - Ruoxiang Wang
- Molecular Urology and Therapeutics, Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322
- Corresponding to: Dr. Ruoxiang Wang, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, NE, Suite B5103, Atlanta, GA 30322, Tel. (404) 778-5116, FAX. (404) 778-3965,
| |
Collapse
|
29
|
Luther N, Cheung NK, Souliopoulos EP, Karampelas I, Karempelas I, Bassiri D, Edgar MA, Guo HF, Pastan I, Gutin PH, Souweidane MM. Interstitial infusion of glioma-targeted recombinant immunotoxin 8H9scFv-PE38. Mol Cancer Ther 2010; 9:1039-46. [PMID: 20371725 DOI: 10.1158/1535-7163.mct-09-0996] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies have the potential to target therapy for high-grade gliomas. Monoclonal antibody 8H9 is specific for membrane protein B7H3 and is reactive with most human high-grade gliomas. We tested the 8H9scFv-PE38 recombinant Pseudomonas immunotoxin in a preclinical model of high-grade glioma. The half maximal inhibitory concentration (IC(50)) of 8H9scFv-PE38 in vitro was determined using glioblastoma cell lines U87 and U251. Maximum tolerated infusion dose of 8H9scFv-PE38 following interstitial infusion to the striatum and pons was defined using athymic rats. Maximum tolerated infusion dose of 8H9scFv-PE38 or PBS control were interstitially delivered to athymic rats xenografted with U87 in the striatum or brain stem. Radiographic response and survivals were measured and compared between treatment groups. The in vitro IC(50) of 8H9scFv-PE38 for U87 was 1,265 ng/mL and, for U251, 91 ng/mL. The maximum tolerated infusion doses of interstitially infused 8H9scFv-PE38 to the striatum and brain stem were 0.75 and 1.8 mug, respectively. For rats harboring intracranial U87 xenografts, infusion of 8H9scFv-PE38 increased mean survival (striatum, 43.4 versus 24.6 days; brain stem, 80.6 versus 45.5 days; n = 28 total) and produced three long-term survivors past 120 days. None of the 14 placebo-treated animals survived >54 days. Tumors also showed volumetric response to infusion of 8H9scFv-PE38 by magnetic resonance imaging. Interstitial infusion of 8H9scFv-PE38 shows potential for the treatment of hemispherical and brain stem glioma. Mol Cancer Ther; 9(4); 1039-46. (c)2010 AACR.
Collapse
Affiliation(s)
- Neal Luther
- Department of Neurologic Surgery, Weill Medical College of Cornell University, Room A-969, 1300 York Avenue, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The constituents of the blood-brain barrier, including its efflux transporter system, can efficiently limit brain penetration of potential CNS therapeutics. Effective extrusion from the brain by transporters is a frequent reason for the pharmaceutical industry to exclude novel compounds from further development for CNS therapeutics. Moreover, high transporter expression levels that are present in individual patients or may be generally associated with the pathophysiology seem to be a major cause of therapeutic failure in a variety of CNS diseases including brain tumors, epilepsy, brain HIV infection, and psychiatric disorders. Increasing knowledge of the structure and function of the blood-brain barrier creates a basis for the development of strategies which aim to enhance brain uptake of beneficial pharmaceutical compounds. The different strategies discussed in this review aim to modulate blood-brain barrier function or to bypass constituents of the blood-brain barrier.
Collapse
|
31
|
BOZINOV O, KALK JM, KRAYENBÜHL N, WOERNLE CM, SURE U, BERTALANFFY H. Decreasing Expression of the Interleukin-13 Receptor IL-13R.ALPHA.2 in Treated Recurrent Malignant Gliomas. Neurol Med Chir (Tokyo) 2010; 50:617-21. [DOI: 10.2176/nmc.50.617] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | | | | | | | - Ulrich SURE
- Department of Neurosurgery, University Hospital Essen
| | | |
Collapse
|
32
|
Grauer OM, Wesseling P, Adema GJ. Immunotherapy of diffuse gliomas: biological background, current status and future developments. Brain Pathol 2009; 19:674-93. [PMID: 19744040 DOI: 10.1111/j.1750-3639.2009.00315.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Despite aggressive multimodal treatment approaches, the prognosis for patients with diffuse gliomas remains disappointing. Glioma cells often extensively infiltrate in the surrounding brain parenchyma, a phenomenon that helps them to escape surgical removal, radiation exposure and chemotherapy. Moreover, conventional therapy is often associated with considerable local and systemic side effects. Therefore, the development of novel therapeutic approaches is essential to improve the outcome of these patients. Immunotherapy offers the opportunity to specifically target residual radio-and chemoresistant tumor cells without damaging healthy neighboring brain tissue. Significant progress has been made in recent years both in understanding the mechanisms of immune regulation in the central nervous system (CNS) as well as tumor-induced and host-mediated immunosuppression elicited by gliomas. In this review, after discussing the special requirements needed for the initiation and control of immune responses in the CNS, we focus on immunological phenomena observed in glioma patients, discuss different immunological approaches to attack glioma-associated target structures and touch on further strategies to improve the efficacy of immunotherapy of gliomas.
Collapse
Affiliation(s)
- Oliver M Grauer
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | | | | |
Collapse
|
33
|
Valles F, Fiandaca MS, Bringas J, Dickinson P, LeCouteur R, Higgins R, Berger M, Forsayeth J, Bankiewicz KS. Anatomic compression caused by high-volume convection-enhanced delivery to the brain. Neurosurgery 2009; 65:579-85; discussion 585-6. [PMID: 19687704 DOI: 10.1227/01.neu.0000350229.77462.2f] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Our group has pioneered the use of gadoteridol-loaded liposomes (GDLs) in convection-enhanced delivery (CED) using real-time magnetic resonance imaging (MRI) to visualize the distribution of therapeutic agents in nonhuman primate and canine brains. We have shown that this procedure is highly predictable and safe. In the course of recent studies, however, we noted that infusion of large volumes caused local anatomic alterations, such as ventricular compression, to occur. This article reports our analysis of CED infusions into normal brains and those compromised by tumors and how monitoring the CED infusion with MRI may be helpful in preventing some complications. METHODS A total of 54 CED infusions using GDLs were performed in 7 canines and 10 nonhuman primates and monitored using real-time MRI. The canines, having brain tumors, received infusions of GDLs as well as a chemotherapeutic agent via CED. The nonhuman primates were normal and received GDL infusions alone. Real-time analysis of the CED infusion was performed, looking for correct catheter position and infusion reflux, leakage, and mass effect. Retrospective analysis allowed assessment of CED volume of distribution versus volume of infusion. RESULTS Approximately 10% of these infusions caused anatomic compression of the ventricles, especially in the canines with tumors. Reflux along the cannula and leakage of infusate into the ventricular cerebrospinal fluid or subarachnoid space were seen. Animal behavior, however, did not appear to be affected acutely or during the course of the study, and no ventricular compression was noted 2 weeks after the CED infusion on further brain imaging studies. CONCLUSION These findings illustrate the value of being able to monitor infusions with real-time MRI to identify phenomena such as reflux along the cannula, leakage of infusate, and ventricular compression. Especially in tumor patients, the latter could be associated with morbidity.
Collapse
Affiliation(s)
- Francisco Valles
- Movement Disorder Research Program, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94103, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lai EW, Joshi BH, Martiniova L, Dogra R, Fujisawa T, Leland P, de Krijger RR, Lubensky IA, Elkahloun AG, Morris JC, Puri RK, Pacak K. Overexpression of interleukin-13 receptor-alpha2 in neuroendocrine malignant pheochromocytoma: a novel target for receptor directed anti-cancer therapy. J Clin Endocrinol Metab 2009; 94:2952-7. [PMID: 19491224 PMCID: PMC2730867 DOI: 10.1210/jc.2009-0309] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT Pheochromocytomas and paragangliomas are rare catecholamine-secreting neuroendocrine tumors arising from the adrenal medulla and sympathetic tissues. When complete surgical resection is not an option, the treatment of pheochromocytoma is limited. OBJECTIVE The objective of the study was to identify and characterize overexpression of IL-13 receptor-alpha2 (IL-13Ralpha2) gene expression in human and murine tumors and verify xenograft mouse pheochromocytoma cell (MPC)-derived tumor's response to a selective cytotoxin. DESIGN/SETTING/PATIENTS Expression of IL-13Ralpha2 was evaluated in a panel of 25 human pheochromocytoma clinical samples by RT-PCR and eight MPC tumors by indirect immunofluorescence assay and RT-PCR. INTERVENTION The function of IL-13Ralpha2 in these tumor cells was examined by evaluating tumor sensitivity to a recombinant IL-13-Pseudomonas exotoxin (IL-13PE). Subcutaneous small and large MPC tumors in athymic nude mice (n = 10) were treated intratumorally with IL-13PE (100 m icrog/kg). MAIN OUTCOME MEASURES IC(50) and tumor size were measured. RESULTS IL-13PE immunotoxin was highly cytotoxic to IL-13Ralpha2-overexpressing MPC cells (IC(50) <2.5 ng/ml) in vitro. Furthermore, IL-13PE was highly cytotoxic to sc tumors. Our results showed a statistically significant decrease in tumor size as early as 3 d after initial treatment and further suppressed growth of MPC tumors. All tumors displayed a histological evidence of necrosis in response to IL-13 immunotoxin without any adverse effects in host at this dose. CONCLUSIONS Human and murine neuroendocrine pheochromocytoma overexpress the IL-13Ralpha2 chain, and an IL-13PE-based receptor-directed anticancer approach may prove useful in treatment for metastatic pheochromocytoma patients.
Collapse
Affiliation(s)
- Edwin W Lai
- Section on Medical Neuroendocrinology, Reproductive and Adult Endocrinology Program, National Institute of Child Health and Human Development, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1109, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Oh S, Ohlfest JR, Todhunter DA, Vallera VD, Hall WA, Chen H, Vallera DA. Intracranial elimination of human glioblastoma brain tumors in nude rats using the bispecific ligand-directed toxin, DTEGF13 and convection enhanced delivery. J Neurooncol 2009; 95:331-342. [PMID: 19517064 DOI: 10.1007/s11060-009-9932-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 05/25/2009] [Indexed: 10/20/2022]
Abstract
A bispecific ligand-directed toxin (BLT) consisting of human interleukin-13, epithelial growth factor, and the first 389 amino acids of diphtheria toxin was assembled in order to target human glioblastoma. In vitro, DTEGF13 selectively killed the human glioblastoma cell line U87-luc as well as other human glioblastomas. DTEGF13 fulfilled the requirement of a successful BLT by having greater activity than either of its monospecific counterparts or their mixture proving it necessary to have both ligands on the same single chain molecule. Aggressive brain tumors established intracranially (IC) in nude rats with U87 glioma genetically marked with a firefly luciferase reporter gene were treated with two injections of DTEGF13 using convection enhanced delivery resulting in tumor eradication in 50% of the rats which survived with tumor free status at least 110 days post tumor inoculation. An irrelevant BLT control did not protect establishing specificity. The bispecific DTEGF13 MTD dose was measured at 2 microg/injection or 0.5 microg/kg and toxicity studies indicated safety in this dose. Combination of monospecific DTEGF and DTIL13 did not inhibit tumor growth. ELISA assay indicated that anti-DT antibodies were not generated in normal immunocompetent rats given identical intracranial DTEGF13 therapy. Thus, DTEGF13 is safe and efficacious as an alternative drug for glioblastoma therapy and warrants further study.
Collapse
Affiliation(s)
- Seunguk Oh
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Deborah A Todhunter
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - Vincent D Vallera
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - Walter A Hall
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Hua Chen
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA
| | - Daniel A Vallera
- Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Masonic Cancer Center, MMC: 367, Minneapolis, MN, 55455, USA.
| |
Collapse
|
36
|
Mercer RW, Tyler MA, Ulasov IV, Lesniak MS. Targeted therapies for malignant glioma: progress and potential. BioDrugs 2009; 23:25-35. [PMID: 19344189 DOI: 10.2165/00063030-200923010-00003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malignant gliomas represent one of the most aggressive forms of brain cancer. Recent advances in the understanding of the deregulated molecular pathways of gliomas have brought about targeted therapies that have the ability to increase therapeutic efficacy in tumors while decreasing toxicity. Multi-targeted kinase inhibitors, novel monoclonal antibodies, and new vaccines have been developed. Standard treatments and current development of new therapies for malignant gliomas are reviewed, focusing specifically on growth factors and their receptors (e.g. epidermal growth factor receptor, vascular endothelial growth factor receptor, and platelet-derived growth factor receptor), as well as the intracellular effector molecules that are downstream of these growth factors (e.g. Ras/Raf/mitogen-activated protein kinase, phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin, and protein kinase C). The efficacies of other novel targeted inhibitors such as deacetylase inhibitors and heat shock protein 90 inhibitors in the treatment of gliomas are also discussed, as well as new combination therapies. In order for new agents to increase treatment efficacy, new targets need to be developed, drug delivery efficiency needs to be improved, and new biomarkers need to be discovered. All of these goals can be accomplished with time through innovative experimental designs.
Collapse
Affiliation(s)
- Ronald W Mercer
- University of Chicago Brain Tumor Center, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
37
|
Djedid R, Kiss R, Lefranc F. Targeted therapy of glioblastomas: a 5-year view. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/thy.09.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
38
|
Lefranc F, Rynkowski M, DeWitte O, Kiss R. Present and potential future adjuvant issues in high-grade astrocytic glioma treatment. Adv Tech Stand Neurosurg 2009; 34:3-35. [PMID: 19368079 DOI: 10.1007/978-3-211-78741-0_1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Despite major advances in the management of malignant gliomas of which glioblastomas represent the ultimate grade of malignancy, they remain characterized by dismal prognoses. Glioblastoma patients have a median survival expectancy of only 14 months on the current standard treatment of surgical resection to the extent feasible, followed by adjuvant radiotherapy plus temozolomide, given concomitantly with and after radiotherapy. Malignant gliomas are associated with such dismal prognoses because glioma cells can actively migrate through the narrow extra-cellular spaces in the brain, often travelling relatively long distances, making them elusive targets for effective surgical management. Clinical and experimental data have demonstrated that invasive malignant glioma cells show a decrease in their proliferation rates and a relative resistance to apoptosis (type I programmed cell death) as compared to the highly cellular centre of the tumor, and this may contribute to their resistance to conventional pro-apoptotic chemotherapy and radiotherapy. Resistance to apoptosis results from changes at the genomic, transcriptional and post-transcriptional level of proteins, protein kinases and their transcriptional factor effectors. The PTEN/ PI3K/Akt/mTOR/NF-kappaB and the Ras/Raf/MEK/ERK signaling cascades play critical roles in the regulation of gene expression and prevention of apoptosis. Components of these pathways are mutated or aberrantly expressed in human cancer, notably glioblastomas. Monoclonal antibodies and low molecular-weight kinase inhibitors of these pathways are the most common classes of agents in targeted cancer treatment. However, most clinical trials of these agents as monotherapies have failed to demonstrate survival benefit. Despite resistance to apoptosis being closely linked to tumorigenesis, tumor cells can still be induced to die by non-apoptotic mechanisms such as necrosis, senescence, autophagy (type II programmed cell death) and mitotic catastrophe. Temozolomide brings significant therapeutic benefits in glioblastoma treatment. Part of temozolomide cytotoxic activity is exerted through pro-autophagic processes and also through the induction of late apoptosis. Autophagy, type II programmed cell death, represents an alternative mechanism to overcome, at least partly, the dramatic resistance of many cancers to pro-apoptotic-related therapies. Another way to potentially overcome apoptosis resistance is to decrease the migration of malignant glioma cells in the brain, which then should restore a level of sensitivity to pro-apoptotic drugs. Recent series of studies have supported the concept that malignant gliomas might be seen as an orchestration of cross-talks between cancer cells, microenvironment, vasculature and cancer stem cells. The present chapter focuses on (i) the major signaling pathways making glioblastomas resistant to apoptosis, (ii) the signaling pathways distinctly activated by pro-autophagic drugs as compared to pro-apoptotic ones, (iii) autophagic cell death as an alternative to combat malignant gliomas, (iv) the major scientific data already obtained by researchers to prove that temozolomide is actually a pro-autophagic and pro-apoptotic drug, (v) the molecular and cellular therapies and local drug delivery which could be used to complement conventional treatments, and a review of some of the currently ongoing clinical trials, (vi) the fact that reducing the levels of malignant glioma cell motility can restore pro-apoptotic drug sensitivity, (vii) the observation that inhibiting the sodium pump activity reduces both glioma cell proliferation and migration, (viii) the brain tumor stem cells as a target to complement conventional treatment.
Collapse
Affiliation(s)
- F Lefranc
- Department of Neurosurgery, Erasme University Hospital, Free University of Brussels, Brussels, Belgium
| | | | | | | |
Collapse
|
39
|
Shimamura T, Fujisawa T, Husain SR, Kioi M, Nakajima A, Puri RK. Novel role of IL-13 in fibrosis induced by nonalcoholic steatohepatitis and its amelioration by IL-13R-directed cytotoxin in a rat model. THE JOURNAL OF IMMUNOLOGY 2008; 181:4656-65. [PMID: 18802068 DOI: 10.4049/jimmunol.181.7.4656] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NASH), the most common cause of chronic liver fibrosis, progresses to cirrhosis in up to 20% of patients. We report that hepatic stellate cells (HSC) in sinusoidal lesions of liver of patients with NASH express high levels of high-affinity IL-13R (IL-13Ralpha2), which is colocalized with smooth muscle actin, whereas fatty liver and normal liver specimens do not express IL-13Ralpha2. HSCs engineered to overexpress IL-13Ralpha2 respond to IL-13 and induce TGFB1 promoter activity and TGF-beta1 production. We also developed NASH in rats by feeding a choline-deficient l-amino acid diet. These rats developed liver fibrosis as assessed by H&E staining, Masson's trichrome and Sirius red staining, and hydroxyproline assays. Treatment of these rats with IL-13R-directed cytotoxin caused a substantial decline in fibrosis and liver enzymes without organ toxicity. These studies demonstrate that functional IL-13Ralpha2 are overexpressed in activated HSCs involved in NASH and that IL-13 cytotoxin ameliorates pathological features of NASH in rat liver, indicating a novel role of this cytotoxin in potential therapy.
Collapse
Affiliation(s)
- Takeshi Shimamura
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
40
|
Lazovic J, Jensen MC, Ferkassian E, Aguilar B, Raubitschek A, Jacobs RE. Imaging immune response in vivo: cytolytic action of genetically altered T cells directed to glioblastoma multiforme. Clin Cancer Res 2008; 14:3832-9. [PMID: 18559603 DOI: 10.1158/1078-0432.ccr-07-5067] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Clinical trials have commenced to evaluate the feasibility of targeting malignant gliomas with genetically engineered CTLs delivered directly to the tumor bed in the central nervous system. The objective of this study is to determine a suite of magnetic resonance imaging (MRI) measurements using an orthotopic xenograft murine model that can noninvasively monitor immunologically mediated tumor regression and reactive changes in the surrounding brain parenchyma. EXPERIMENTAL DESIGN Our preclinical therapeutic platform is based on CTL genetic modification to express a membrane tethered interleukin-13 (IL-13) cytokine chimeric T-cell antigen receptor. This enables selective binding and signal transduction on encountering the glioma-restricted IL-13 alpha2 receptor (IL-13Ralpha2). We used MRI to visualize immune responses following adoptive transfer of IL-13Ralpha2-specific CD8+ CTL clones. RESULTS Based on MRI measurements, several phases following IL-13Ralpha2-specific T-cell adoptive transfer could be distinguished, all of which correlated well with glioblastoma regression confirmed on histology. The first detectable changes, 24 hours post-treatment, were significantly increased T2 relaxation times and strongly enhanced signal on T1-weighted postcontrast images. In the next phase, the apparent diffusion coefficient was significantly increased at 2 and 3 days post-treatment. In the last phase, at day 3 after IL-13Ralpha2-specific T-cell injection, the volume of hyperintense signal on T1-weighted postcontrast image was significantly decreased, whereas apparent diffusion coefficient remained elevated. CONCLUSIONS The present study indicates the feasibility of MRI to visualize different phases of immune response when IL-13Ralpha2-specific CTLs are administered directly to the glioma tumor bed. This will further the aim of better predicting clinical outcome following immunotherapy.
Collapse
Affiliation(s)
- Jelena Lazovic
- Biological Imaging Center, Beckman Institute, Division of Biology, California Institute of Technology, Pasadena, California, USA
| | | | | | | | | | | |
Collapse
|
41
|
Allen C, Paraskevakou G, Iankov I, Giannini C, Schroeder M, Sarkaria J, Schroeder M, Puri RK, Russell SJ, Galanis E. Interleukin-13 displaying retargeted oncolytic measles virus strains have significant activity against gliomas with improved specificity. Mol Ther 2008; 16:1556-1564. [PMID: 18665158 DOI: 10.1038/mt.2008.152] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Accepted: 06/20/2008] [Indexed: 11/09/2022] Open
Abstract
The majority of glioblastoma multiforme (GBM) tumors (80%) overexpress interleukin-13 receptor alpha2 (IL-13Ralpha2), but there is no expression of IL-13Ralpha2 in normal brain. Vaccine strains of measles virus have significant antitumor activity against gliomas. We tested the hypothesis that measles virus entry could be retargeted via the IL-13Ralpha2. MV-GFP-H(AA)-IL-13 was generated from the Edmonston-NSe vaccine strain, by displaying human IL-13 at the C-terminus of the H protein, and introducing CD46 and signaling lymphocyte activation molecule (SLAM)-ablating mutations in H. The IL-13 retargeted virus showed significant cytopathic effect (CPE) against IL-13Ralpha2 overexpressing glioma lines, and lack of CPE/viral replication in normal human astrocytes and normal human fibroblasts not expressing IL-13Ralpha2. In vivo treatment of orthotopically implanted GBM12 xenografts demonstrated significant prolongation of survival in mice treated with the retargeted strain (P < 0.0001), and comparable activity between the IL-13R retargeted strain and MV-GFP (P = 0.6377). In contrast to MV-GFP-treated mice, administration of the retargeted strain in the central nervous system of measles replication-permissive Ifnar(ko) CD46 Ge mice resulted in lack of neurotoxicity. Strains of measles virus retargeted against the glioma-specific IL-13Ralpha2 receptor have comparable therapeutic efficacy, and improved specificity as compared with the unmodified measles virus strain MV-GFP in vitro and in vivo.
Collapse
Affiliation(s)
- Cory Allen
- Molecular Medicine Department, Mayo Clinic College Of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mut M, Sherman JH, Shaffrey ME, Schiff D. Cintredekin besudotox in treatment of malignant glioma. Expert Opin Biol Ther 2008; 8:805-12. [PMID: 18476792 DOI: 10.1517/14712598.8.6.805] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Interleukin-13 (IL-13) receptors are overexpressed in glioblastoma multiforme (GBM). The presence of IL-13 binding sites in GBM and their absence in normal brain tissue validates IL-13 receptor as an important target in human GBM. OBJECTIVE This review discusses the bench-to-bedside experience with a recombinant cytotoxin composed of human IL-13 and a truncated form of Pseudomonas exotoxin A (PE38QQR), delivered via convection-enhanced delivery (CED), in GBM treatment. METHODS The authors review publications regarding the laboratory research and clinical development of IL-13-directed therapies and summarize the future of IL-13-targeted cytotoxin. CONCLUSION The IL-13 receptor remains an important potential target in GBM, and preliminary experience with the IL-13-PE38QQR cytotoxin (also called cintredekin besudotox) has helped to pave the way for study of CED as an important means of drug delivery to malignant gliomas. Ongoing analysis of recently completed clinical trials will determine the future of this agent and its potential therapeutic targets.
Collapse
Affiliation(s)
- Melike Mut
- Hacettepe Hastanesi Beyin Cerrahisi, Bolum 71 Sihhiye, Ankara 06100, Turkey.
| | | | | | | |
Collapse
|
43
|
Joshi BH, Puri RA, Leland P, Varricchio F, Gupta G, Kocak M, Gilbertson RJ, Puri RK. Identification of interleukin-13 receptor alpha2 chain overexpression in situ in high-grade diffusely infiltrative pediatric brainstem glioma. Neuro Oncol 2008; 10:265-74. [PMID: 18430795 DOI: 10.1215/15228517-2007-066] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Human malignant glioma cell lines and adult brain tumors overexpress high levels of interleukin-13 receptor alpha2 chain (IL-13Ralpha2). Because the IL-13Ralpha2 chain is an important target for cancer therapy and prognosis for patients with brainstem glioma (BSG) remains dismal, we investigated the expression of this receptor in specimens of diffusely infiltrative pediatric BSG relative to normal brain tissue. Twenty-eight BSG specimens and 15 normal brain specimens were investigated for IL-13Ralpha2 protein expression by immunohistochemical analysis (IHC) using two different antibodies in two different laboratories. Highly sensitive Q-dot-based IHC and in situ hybridization (ISH) assays were also developed to identify IL-13Ralpha2 protein and RNA in these specimens. The results were evaluated independently in two laboratories in a blinded fashion. By Q-dot IHC or a standard IHC assay, 17 of 28 (61%) tumor specimens showed modest to strong staining for IL-13Ralpha2, while 15 normal brain tissue samples showed weak expression for IL-13Ralpha2 protein. Significant interrater agreement between the two laboratories was seen in the assessment of IL-13Ralpha2 intensity. High-level IL-13Ralpha2 RNA expression was detected in tumor samples by Q-dot ISH, but only weak RNA expression was observed in normal brain. Significant agreement between ISH and IHC assays was observed (simple kappa [kappa] estimate=0.358, weighted kappa=0.89, p=0.001). IL-13Ralpha2 protein and mRNA are expressed to significantly higher levels in BSG than in normal brain tissue. Both IHC and ISH represent robust methods to detect expression of the IL-13Ralpha2 receptor in BSG that could represent an important new drug target for treatment of this disease.
Collapse
Affiliation(s)
- Bharat H Joshi
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Office of Cellular, Tissue, and Gene Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ferguson SD, Foster K, Yamini B. Convection-enhanced delivery for treatment of brain tumors. Expert Rev Anticancer Ther 2008; 7:S79-85. [PMID: 18076322 DOI: 10.1586/14737140.7.12s.s79] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Recently, innovative therapies have been developed for the treatment of malignant gliomas. Unfortunately, adequate delivery of these therapies has been a major obstacle to clinical success. Intravenous administration is restricted by the presence of the blood-brain barrier while local delivery, such as with drug-impregnated wafers, results in limited parenchyma penetration. Convection-enhanced delivery is a promising method for the delivery of macromolecules to the CNS. Convection-enhanced delivery involves the infusion of therapeutic agents via surgically implanted catheters and uses a pressure gradient to achieve a greater volume of distribution compared with that seen with diffusion alone. This article will review the development of convection-enhanced delivery and its use in the treatment of malignant gliomas.
Collapse
Affiliation(s)
- Sherise D Ferguson
- The University of Chicago Hospitals, Section of Neurosurgery, MC4066, 5841 South Maryland Avenue, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
45
|
Anti-glioblastoma effect of a recombinant bispecific cytotoxin cotargeting human IL-13 and EGF receptors in a mouse xenograft model. J Neurooncol 2007; 87:51-61. [PMID: 18084721 DOI: 10.1007/s11060-007-9499-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 11/20/2007] [Indexed: 10/22/2022]
Abstract
To improve activity of a recombinant IL-13 cytotoxin (CT) comprised of IL-13 spliced to truncated diphtheria toxin (DT(390)), epidermal growth factor (EGF) was added to the same single chain protein. This new recombinant bispecific CT, called DTEGF13, enhanced the killing potency against the human glioblastoma lines, U87MG (0.015 nM) and U118MG (0.02 nM). A similar enhancement was observed against the lung carcinoma cell line, Calu-3 (0.0018 nM). Enhanced activity could not be explained by an increased number of cytokines available for binding since a combination of monospecific DTEGF and DTIL13 did not cause the same enhanced activity. Enhanced activity was dependent on the presence of both cytokines on the same single chain molecule and killing was receptor specific since target receptor negative leukemia cells were unaffected by the highly selective DTEGF13 and cytotoxicity could be blocked with anti-EGFR and anti-IL-13 antibodies. In a xenograft flank tumor model, intratumoral injection of DTEGF13, but not monospecific DTEGF or DTIL13, significantly inhibited the growth of established U87 tumors in nude mice (P < 0.04). In this model, the human EGF and IL-13 components of DTEGF13 are reactive with mouse EGFR and IL-13R, respectively. These studies show that a new co-targeting agent that simultaneously recognizes EGFR and IL-13R is more effective than its monospecific counterparts and that DTEGF13 has therapeutic advantages for glioblastoma.
Collapse
|
46
|
Xiaobing J, Xiaoling L, Ruen L, Fangcheng Z, Hongyang Z. Induction of cytotoxic T-lymphocytes specific for malignant glioma by HLA dimer-based artificial antigen-presenting cells. Cancer Biother Radiopharm 2007; 22:826-35. [PMID: 18158774 DOI: 10.1089/cbr.2007.0406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIM The aim of this study was to investigate the novel approach for targeting malignant glioma. METHODS Interleukin-13 receptor alpha2 (IL-13Ralpha2)-specific cytotoxic T-cells (CTLs) were induced from the peripheral blood lymphocytes (PBLs) of human leukocyte antigen (HLA)-A2 positive healthy donors by multiple stimulations with artificial antigen-presenting cells (aAPCs) made by coating HLA-A2-Ig/pIL-13Ralpha2(345-354) dimeric complexes, anti-CD28 antibody, and CD83 molecules to cell-sized latex beads. RESULTS The induced CTLs exhibited a specific lysis against T2 cells pulsed with the peptide pIL-13Ralpha2(345-354) and HLA-A2(+) glioma cells expressing IL-13Ralpha2(345-354), while HLA-A2(-) glioma cell lines that express IL-13Ralpha2(345-354) could not be recognized by the CTLs. The peptide-specific activity was inhibited by the anti-HLA class I monoclonal antibody. CONCLUSIONS The induced CTLs specific for the IL-13Ralpha2(345-354) peptide could be a potential target of specific immunotherapy for HLA-A2(+) patients with malignant glioma.
Collapse
MESH Headings
- Antibodies/chemistry
- Antibodies/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigen Presentation/immunology
- Antigens, CD/chemistry
- Antigens, CD/immunology
- CD28 Antigens/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Gene Expression
- Glioma/immunology
- Glioma/pathology
- Glioma/therapy
- HLA-A2 Antigen/immunology
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Testing
- Humans
- Immunoglobulins/chemistry
- Immunoglobulins/immunology
- Interferon-gamma/metabolism
- Interleukin-13 Receptor alpha2 Subunit/genetics
- Interleukin-13 Receptor alpha2 Subunit/immunology
- Leukocytes, Mononuclear/immunology
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/immunology
- Microspheres
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- CD83 Antigen
Collapse
Affiliation(s)
- Jiang Xiaobing
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | |
Collapse
|
47
|
Vogelbaum MA, Sampson JH, Kunwar S, Chang SM, Shaffrey M, Asher AL, Lang FF, Croteau D, Parker K, Grahn AY, Sherman JW, Husain SR, Puri RK. CONVECTION-ENHANCED DELIVERY OF CINTREDEKIN BESUDOTOX (INTERLEUKIN-13-PE38QQR) FOLLOWED BY RADIATION THERAPY WITH AND WITHOUT TEMOZOLOMIDE IN NEWLY DIAGNOSED MALIGNANT GLIOMAS. Neurosurgery 2007; 61:1031-7; discussion 1037-8. [DOI: 10.1227/01.neu.0000303199.77370.9e] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Abstract
OBJECTIVE
Cintredekin besudotox (CB), a recombinant cytotoxin consisting of interleukin-13 and truncated Pseudomonas exotoxin, binds selectively to interleukin-13Rα2 receptors overexpressed by malignant gliomas. This study assessed the safety of CB administered by convection-enhanced delivery followed by standard external beam radiation therapy (EBRT) with or without temozolomide (Temodar; Schering-Plough, Kenilworth, NJ) in patients with newly diagnosed malignant gliomas.
METHODS
After gross total resection of the tumor, two to four intraparenchymal catheters were stereotactically placed and CB (0.25 or 0.5 μg/mL) was infused for 96 hours. This was followed, 10 to 14 days later, by EBRT (5940–6100 cGy, 5 d/wk for 6–7 wk) with or without temozolomide (75 mg/m2/d, 7 d/wk during EBRT). Safety was assessed during an 11-week observation period after catheter placement
RESULTS
Twenty-two patients (12 men, 10 women; median age, 55 yr; 21 with glioblastoma multiforme and one with an anaplastic mixed oligoastrocytoma) were enrolled. None of the patients experienced dose-limiting toxicities in the first two cohorts (0.25 μg/mL CB + EBRT [n = 3] and 0.25 μg/mL CB + EBRT + temozolomide [n = 3]). One patient experienced a dose-limiting toxicity (Grade 4 seizure) in the third cohort (0.5 μg/mL CB + EBRT [n = 6]). Six patients in the final cohort (0.5 μg/mL CB + EBRT + temozolomide [n = 10]) completed treatment, and one patient experienced a dose-limiting toxicity (Grade 3 aphasia and confusion). Four patients were not considered evaluable for a dose decision and were replaced. CB related adverse events occurring in more than one patient were fatigue, gait disturbance, nystagmus, and confusion. No Grade 3 to 4 hematological toxicities were observed.
CONCLUSION
CB (0.5 μg/mL) administered via convection-enhanced delivery before standard radiochemotherapy seems to be well tolerated in adults with newly diagnosed malignant gliomas. Further clinical study assessment is warranted.
Collapse
Affiliation(s)
- Michael A. Vogelbaum
- Brain Tumor and Neuro-Oncology Center, The Cleveland Clinic Foundation, Cleveland, Ohio
| | - John H. Sampson
- Department of Neurosurgery, Duke University, Durham, North Carolina
| | - Sandeep Kunwar
- Department of Neurosurgery, University of California, San Francisco, San Francisco, California
| | - Susan M. Chang
- Department of Neurosurgery, University of California, San Francisco, San Francisco, California
| | - Mark Shaffrey
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | - Anthony L. Asher
- Carolina Neurosurgery and Spine Associates, Charlotte, North Carolina
| | - Frederick F. Lang
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | - S. Rafat Husain
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland
| | - Raj K. Puri
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland
| |
Collapse
|
48
|
Jiang X, Lu X, Liu R, Zhang F, Zhao H. Induction of cytotoxic T lymphocytes specific to malignant glioma using T2 cells pulsed with HLA-A2-restricted interleukin-13 receptor alpha 2 peptide in vitro. Acta Biochim Biophys Sin (Shanghai) 2007; 39:641-8. [PMID: 17805458 DOI: 10.1111/j.1745-7270.2007.00331.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Interleukin-13 receptor alpha2 (IL-13Ralpha2) is a glioma-restricted cell-surface epitope not otherwise detected within the central nervous system. The present study is a report of a novel approach of targeting malignant glioma with IL-13Ralpha2-specific cytotoxic T lymphocyte (CTL) induced from the peripheral blood mononuclear cells of healthy donors by multiple stimulations with human leukocyte antigen (HLA)-A2-restricted IL-13Ralpha2(345-353) peptide-pulsed T2 cells. The induced CTL showed specific lysis against T2 cells pulsed with the peptide and HLA-A2+ glioma cells expressing IL-13R2(345-353), while HLA-A2 glioma cell lines that express IL-13Ralpha2(345-353) could not be recognized by CTL. The peptide-specific activity was inhibited by anti-HLA class I monoclonal antibody. These results suggest that the induced CTL specific for IL-13Ralpha2(345-353) peptide could be a potential target of specific immunotherapy for HLA-A2 patients with malignant glioma.
Collapse
Affiliation(s)
- Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | | | | | | | | |
Collapse
|
49
|
Lu XL, Jiang XB, Liu RE, Zhang FC, Zhao HY. Generation of allo-restricted cytotoxic T lymphocytes against malignant glioma by artificial antigen-presenting cells. Cancer Lett 2007; 256:128-35. [PMID: 17719173 DOI: 10.1016/j.canlet.2007.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 07/15/2007] [Accepted: 07/17/2007] [Indexed: 10/22/2022]
Abstract
The interleukin (IL) 13 receptor alpha2 (IL-13Ralpha2) is a glioma-restricted cell-surface epitope not otherwise detected within the central nervous system. This study reported a novel approach for targeting malignant glioma with IL-13Ralpha2-specific allo-restricted cytotoxic T cells (CTLs) induced from the peripheral blood lymphocytes (PBLs) of HLA-A2-negative healthy donors by multiple stimulations with artificial antigen-presenting cells (aAPCs) made by coating HLA-A2/pIL-13Ralpha2(345-354) tetrameric complexes, anti-CD28 antibody and CD83 molecules to cell-sized latex beads. The induced allo-restricted CTLs exhibited specific lysis against T2 cells pulsed with the peptide pIL-13Ralpha2(345-354) and HLA-A2+ glioma cells expressing IL-13Ralpha2(345-354), while HLA-A2- glioma cell lines that express IL-13Ralpha2(345-354) could not be recognized by the CTLs. The peptide-specific activity was inhibited by anti-HLA class I monoclonal antibody. These results suggested the induced allo-restricted CTLs specific for IL-13Ralpha2(345-354) peptide could be a potential target of specific immunotherapy for HLA-A2+ patients with malignant glioma.
Collapse
Affiliation(s)
- Xiao-Ling Lu
- Department of Immunology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | | | | | | |
Collapse
|
50
|
Kunwar S, Prados MD, Chang SM, Berger MS, Lang FF, Piepmeier JM, Sampson JH, Ram Z, Gutin PH, Gibbons RD, Aldape KD, Croteau DJ, Sherman JW, Puri RK. Direct Intracerebral Delivery of Cintredekin Besudotox (IL13-PE38QQR) in Recurrent Malignant Glioma: A Report by the Cintredekin Besudotox Intraparenchymal Study Group. J Clin Oncol 2007; 25:837-44. [PMID: 17327604 DOI: 10.1200/jco.2006.08.1117] [Citation(s) in RCA: 242] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Glioblastoma multiforme (GBM) is a devastating brain tumor with a median survival of 6 months after recurrence. Cintredekin besudotox (CB) is a recombinant protein consisting of interleukin-13 (IL-13) and a truncated form of Pseudomonas exotoxin (PE38QQR). Convection-enhanced delivery (CED) is a locoregional-administration method leading to high-tissue concentrations with large volume of distributions. We assessed the use of intracerebral CED to deliver CB in patients with recurrent malignant glioma (MG). Patients and Methods Three phase I clinical studies evaluated intracerebral CED of CB along with tumor resection. The main objectives were to assess the tolerability of various concentrations and infusion durations; tissue distribution; and methods for optimizing delivery. All patients underwent tumor resection followed by a single intraparenchymal infusion (in addition to the intraparenchymal one following resection), with a portion of patients who had a preresection intratumoral infusion. Results A total of 51 patients with MG were treated including 46 patients with GBM. The maximum tolerated intraparenchymal concentration was 0.5 μg/mL and tumor necrosis was observed at this concentration. Infusion durations of up to 6 days were well tolerated. Postoperative catheter placement appears to be important for optimal drug distribution. CB- and procedure-related adverse events were primarily limited to the CNS. Overall median survival for GBM patients is 42.7 weeks and 55.6 weeks for patients with optimally positioned catheters with patient follow-up extending beyond 5 years. Conclusion CB appears to have a favorable risk-benefit profile. CED is a complex delivery method requiring catheter placement via a second procedure to achieve accurate catheter positioning, better drug distribution, and better outcome.
Collapse
Affiliation(s)
- Sandeep Kunwar
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143-0350, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|