1
|
Stribbling SM, Beach C, Ryan AJ. Orthotopic and metastatic tumour models in preclinical cancer research. Pharmacol Ther 2024; 257:108631. [PMID: 38467308 PMCID: PMC11781865 DOI: 10.1016/j.pharmthera.2024.108631] [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: 08/17/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Mouse models of disease play a pivotal role at all stages of cancer drug development. Cell-line derived subcutaneous tumour models are predominant in early drug discovery, but there is growing recognition of the importance of the more complex orthotopic and metastatic tumour models for understanding both target biology in the correct tissue context, and the impact of the tumour microenvironment and the immune system in responses to treatment. The aim of this review is to highlight the value that orthotopic and metastatic models bring to the study of tumour biology and drug development while pointing out those models that are most likely to be encountered in the literature. Important developments in orthotopic models, such as the increasing use of early passage patient material (PDXs, organoids) and humanised mouse models are discussed, as these approaches have the potential to increase the predictive value of preclinical studies, and ultimately improve the success rate of anticancer drugs in clinical trials.
Collapse
Affiliation(s)
- Stephen M Stribbling
- Department of Chemistry, University College London, Gower Street, London WC1E 6BT, UK.
| | - Callum Beach
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Anderson J Ryan
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK; Fast Biopharma, Aston Rowant, Oxfordshire, OX49 5SW, UK.
| |
Collapse
|
2
|
Gamat-Huber M, Jeon D, Johnson LE, Moseman JE, Muralidhar A, Potluri HK, Rastogi I, Wargowski E, Zahm CD, McNeel DG. Treatment Combinations with DNA Vaccines for the Treatment of Metastatic Castration-Resistant Prostate Cancer (mCRPC). Cancers (Basel) 2020; 12:cancers12102831. [PMID: 33008010 PMCID: PMC7601088 DOI: 10.3390/cancers12102831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 01/04/2023] Open
Abstract
Simple Summary The only vaccine approved by FDA as a treatment for cancer is sipuleucel-T, a therapy for patients with metastatic castration-resistant prostate cancer (mCRPC). Most investigators studying anti-tumor vaccines believe they will be most effective as parts of combination therapies, rather than used alone. Unfortunately, the cost and complexity of sipuleucel-T makes it difficult to feasibly be used in combination with many other agents. In this review article we discuss the use of DNA vaccines as a simpler vaccine approach that has demonstrated efficacy in several animal species. We discuss the use of DNA vaccines in combination with traditional treatments for mCRPC, and other immune-modulating treatments, in preclinical and early clinical trials for patients with mCRPC. Abstract Metastatic castration-resistant prostate cancer (mCRPC) is a challenging disease to treat, with poor outcomes for patients. One antitumor vaccine, sipuleucel-T, has been approved as a treatment for mCRPC. DNA vaccines are another form of immunotherapy under investigation. DNA immunizations elicit antigen-specific T cells that cause tumor cell lysis, which should translate to meaningful clinical responses. They are easily amenable to design alterations, scalable for large-scale manufacturing, and thermo-stable for easy transport and distribution. Hence, they offer advantages over other vaccine formulations. However, clinical trials with DNA vaccines as a monotherapy have shown only modest clinical effects against tumors. Standard therapies for CRPC including androgen-targeted therapies, radiation therapy and chemotherapy all have immunomodulatory effects, which combined with immunotherapies such as DNA vaccines, could potentially improve treatment. In addition, many investigational drugs are being developed which can augment antitumor immunity, and together with DNA vaccines can further enhance antitumor responses in preclinical models. We reviewed the literature available prior to July 2020 exploring the use of DNA vaccines in the treatment of prostate cancer. We also examined various approved and experimental therapies that could be combined with DNA vaccines to potentially improve their antitumor efficacy as treatments for mCRPC.
Collapse
|
3
|
Michels LR, Fachel FNS, Azambuja JH, Gelsleichter NE, Braganhol E, Teixeira HF. HPLC-UV method for temozolomide determination in complex biological matrices: Application for in vitro, ex vivo and in vivo studies. Biomed Chromatogr 2019; 33:e4615. [PMID: 31166608 DOI: 10.1002/bmc.4615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/17/2019] [Accepted: 05/29/2019] [Indexed: 11/10/2022]
Abstract
A high-performance liquid chromatography method for temozolomide (TMZ) determination in complex biological matrices was developed and validated for application in in vitro, ex vivo and in vivo studies of new nanotechnology-based systems for TMZ nasal delivery. The method was able to quantify TMZ in nanoemulsions, following cellular uptake, in the porcine nasal mucosa and in mouse plasma and brain. Analyses were performed on a C18 column at 35°C, under UV detection at 330 nm. The mobile phase was methanol-acetic acid 0.5% (30:70, v/v), eluted at an isocratic flow rate of 1.1 mL/min. The method was found to be specific, precise, accurate, robust and linear (0.05 to 5 μg/mL) for TMZ determination in all matrices. No interference of TMZ degradation products was found under various stress conditions such as acidic, alkaline, oxidative, light and thermal exposure, demonstrating stability. The method was applied for the quantification of TMZ in different matrices, i.e. the efficiency of nanoemulsions in vitro in increasing TMZ cellular uptake, ex vivo TMZ permeation and retention in the porcine nasal mucosa tissue, and for in vivo TMZ quantification in mouse brain following intranasal nanoemulsion administration compared with free TMZ.
Collapse
Affiliation(s)
- Luana R Michels
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Flávia N S Fachel
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Juliana H Azambuja
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Nicolly E Gelsleichter
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Elizandra Braganhol
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Helder F Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Kumari S, Bhattacharya D, Rangaraj N, Chakarvarty S, Kondapi AK, Rao NM. Aurora kinase B siRNA-loaded lactoferrin nanoparticles potentiate the efficacy of temozolomide in treating glioblastoma. Nanomedicine (Lond) 2018; 13:2579-2596. [DOI: 10.2217/nnm-2018-0110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: To investigate the efficacy of lactoferrin nanoparticles (LfNPs) in delivering siRNA across the blood–brain barrier to treat glioblastoma multiforme (GBM) and with an additional objective of potentiation of conventional temozolomide (TMZ) chemotherapy. Methods: Aurora kinase B (AKB) siRNA-loaded nanoparticles (AKB–LfNPs) were prepared with milk protein, lactoferrin, by water in oil emulsion method. AKB–LfNPs were tested in cell lines and in GBM orthotopic mouse model with and without TMZ treatment. Results: AKB silencing, cytotoxicity and cell cycle arrest by these LfNPs were shown to be effective on GL261 cells. Tumor growth was significantly lower in AKB–LfNPs alone and in combination with TMZ treated mice and increased the survival by 2.5-times. Conclusion: Treatment of AKB–LfNPs to GBM mice improves life expectancy and has potential to combine with conventional chemotherapy.
Collapse
Affiliation(s)
- Sonali Kumari
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad 500 046, Telangana State, India
| | - Dwaipayan Bhattacharya
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Council of Scientific & Industrial Research, Uppal Road, Hyderabad 500 007, Telangana State, India
| | - Nandini Rangaraj
- Centre for Cellular & Molecular Biology (CCMB), Council of Scientific & Industrial Research (CSIR), Uppal Road, Hyderabad 500007, Telangana State, India
| | - Sumana Chakarvarty
- Centre for Cellular & Molecular Biology (CCMB), Council of Scientific & Industrial Research (CSIR), Uppal Road, Hyderabad 500007, Telangana State, India
| | - Anand K Kondapi
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad 500 046, Telangana State, India
| | - Nalam M Rao
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Council of Scientific & Industrial Research, Uppal Road, Hyderabad 500 007, Telangana State, India
| |
Collapse
|
5
|
Lee YE, Choi SA, Kwack PA, Kim HJ, Kim IH, Wang KC, Phi JH, Lee JY, Chong S, Park SH, Park KD, Hwang DW, Joo KM, Kim SK. Repositioning disulfiram as a radiosensitizer against atypical teratoid/rhabdoid tumor. Neuro Oncol 2018; 19:1079-1087. [PMID: 28340172 DOI: 10.1093/neuonc/now300] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Atypical teratoid/rhabdoid tumor (AT/RT) is one of the most common malignant brain tumors in infants. Although cancer stem cells of AT/RT express aldehyde dehydrogenase (ALDH), effective chemotherapies against AT/RT have not been established. Here, we examined radiosensitizing effects of disulfiram (DSF), an irreversible inhibitor of ALDH against AT/RT for a novel therapeutic method. Methods Patient-derived primary cultured AT/RT cells (SNU.AT/RT-5 and SNU.AT/RT-6) and established AT/RT cell lines (BT-12 and BT-16) were used to assess therapeutic effects of combining DSF with radiation treatment (RT). Survival fraction by clonogenic assay, protein expression, immunofluorescence, and autophagy analysis were evaluated in vitro. Antitumor effects of combining DSF with RT were verified by bioluminescence imaging, tumor volume, and survival analysis in vivo. Results The results demonstrated that DSF at low concentration enhanced the radiosensitivity of AT/RT cells with reduction of survival fraction to 1.21‒1.58. DSF increased DNA double-strand break (γ-H2AX, p-DNA-PKcs, and p-ATM), apoptosis (cleaved caspase-3), autophagy (LC3B), and cell cycle arrest (p21) in irradiated AT/RT cells, while it decreased anti-apoptosis (nuclear factor-kappaB, Survivin, and B-cell lymphoma 2 [Bcl2]). In vivo, DSF and RT combined treatment significantly reduced tumor volumes and prolonged the survival of AT/RT mouse models compared with single treatments. The combined treatment also increased γ-H2AX, cleaved caspase-3, and LC3B expression and decreased ALDH1, Survivin, and Bcl2 expression in vivo. Conclusions DSF and RT combination therapy has additive therapeutic effects on AT/RT by potentiating programmed cell death, including apoptosis and autophagy of AT/RT cells. We suggest that DSF can be applied as a radiosensitizer in AT/RT treatment.
Collapse
Affiliation(s)
- Young Eun Lee
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Pil Ae Kwack
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Hak Jae Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Il Han Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Kyu-Chang Wang
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Ji Hoon Phi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Sangjoon Chong
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Sung-Hye Park
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Kyung Duk Park
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Do Won Hwang
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Kyeung Min Joo
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea; Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, South Korea; Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul, South Korea; Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, South Korea; Department of Anatomy and Cell Biology, SungKyunKwan University School of Medicine, Suwon, South Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| |
Collapse
|
6
|
Abstract
Immunotherapy has become standard of care in advanced non-small cell lung cancer (NSCLC) in a number of settings. Radiotherapy remains an important and potentially curative treatment for localized and locally advanced NSCLC not amenable to surgery. While the principal cytotoxic effect of ionizing radiation is via DNA damage, the effect on tumour microenvironment, promoting dendritic cell presentation of tumour-derived antigens to T cells stimulating the host adaptive immune system to mount an immune response against tumours cells, has become of particular interest when combining immunomodulating agents with radiation. The 'abscopal effect' of radiation where non-irradiated metastatic lesions may respond to radiation may be immune-mediated, via radiation primed anti-tumour T cells. Immune priming by radiation offers the potential for increasing the efficacy of immunotherapy and this is subject to on-going clinical trials underpinned by immunological bioassays. Increasing understanding of the interaction between tumour, radiation and immune cells at a molecular level provides a further opportunity for intervention to enhance the potential synergy between radiation and immunotherapy. Applying the potential efficacy of combination therapy to clinical practice requires caution particularly to ensure the safety of the two treatment modalities in early phase clinical trials, many of which are currently underway. We review the biological basis for combining radiation and immunotherapy and examine the existing pre-clinical and clinical evidence and the challenges posed by the new combination of treatments.
Collapse
Affiliation(s)
- Neeraj Bhalla
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside, UK
| | - Rachel Brooker
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside, UK
| | - Michael Brada
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside, UK.,Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
7
|
Kumari S, Ahsan SM, Kumar JM, Kondapi AK, Rao NM. Overcoming blood brain barrier with a dual purpose Temozolomide loaded Lactoferrin nanoparticles for combating glioma (SERP-17-12433). Sci Rep 2017; 7:6602. [PMID: 28747713 PMCID: PMC5529576 DOI: 10.1038/s41598-017-06888-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023] Open
Abstract
Targeted delivery of drugs to the brain is challenging due to the restricted permeability across the blood brain barrier (BBB). Gliomas are devastating cancers and their positive treatment outcome using Temozolomide (TMZ) is limited due to its short plasma half-life, systemic toxicity and limited access through the blood-brain barrier (BBB). Nanoparticles made of Lactoferrin (Lf) protein, have been shown to enhance the pharmacological properties of drugs. Here, we report the specific ability of Lf nanoparticles to cross BBB and target over-expressed Lf receptors on glioma for enhanced TMZ delivery. TMZ-loaded Lf nanoparticles (TMZ-LfNPs) were prepared by our previously reported sol-oil method. While the Lf protein in the NP matrix aids in transcytosis across the BBB and preferential tumor cell uptake, the pH responsiveness leads to TMZ release exclusively in the tumor microenvironment. Delivery through LfNPs results in an enhanced and sustained intracellular concentration of TMZ in GL261 cells in vitro along with improving its in vivo pharmacokinetics and brain accumulation. TMZ-LfNPs treatment results in a significant reduction of tumor volume, higher tumor cell apoptosis and improved median survival in glioma bearing mice. These results demonstrate that LfNPs present an efficient TMZ delivery platform for an effective treatment of gliomas.
Collapse
Affiliation(s)
- Sonali Kumari
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad, 500 046, Telangana State, India
| | - Saad M Ahsan
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, Telangana State, India
| | - Jerald M Kumar
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, Telangana State, India
| | - Anand K Kondapi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad, 500 046, Telangana State, India.
| | - Nalam M Rao
- Centre for Cellular and Molecular Biology (CCMB), Council of Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, Telangana State, India.
| |
Collapse
|
8
|
Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol 2015; 141:575-89. [PMID: 25005789 DOI: 10.1007/s00432-014-1772-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/30/2014] [Indexed: 12/13/2022]
Abstract
Malignant gliomas invariably recur after irradiation, showing radioresistance. Meanwhile, cranial irradiation can bring some risk for developing cognitive dysfunction. There is increasing evidence that cytokines play their peculiar roles in these processes. On the one hand, cytokines directly influence the progression of malignant glioma, promoting or suppressing tumor progression. On the other hand, cytokines indirectly contribute to the immunologic response against gliomas, exhibiting pro-inflammatory or immunosuppressive activities. We propose that cytokines are not simply unregulated products from tumor cells or immune cells, but mediators finely adjust the balance between glioma cells and tumor microenvironment after irradiation. The paper, therefore, focuses on the changes of cytokines after irradiation, analyzing how these mediate the response of tumor cells and normal cells to irradiation. In addition, cytokine-based immunotherapeutic strategies, accompanied with irradiation, for the treatment of gliomas are also discussed.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Radiation Oncology, Cancer Centre, Qilu Hospital, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | | | | | | | | |
Collapse
|
9
|
PD-L1 expression by neurons nearby tumors indicates better prognosis in glioblastoma patients. J Neurosci 2013; 33:14231-45. [PMID: 23986257 DOI: 10.1523/jneurosci.5812-12.2013] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor. In general, tumor growth requires disruption of the tissue microenvironment, yet how this affects glioma progression is unknown. We studied program death-ligand (PD-L)1 in neurons and gliomas in tumors from GBM patients and associated the findings with clinical outcome. Remarkably, we found that upregulation of PD-L1 by neurons in tumor-adjacent brain tissue (TABT) associated positively with GBM patient survival, whereas lack of neuronal PD-L1 expression was associated with high PD-L1 in tumors and unfavorable prognosis. To understand the molecular mechanism of PD-L1 signaling in neurons, we investigated PD-L1 function in cerebellar and cortical neurons and its impact on gliomas. We discovered that neuronal PD-L1-induced caspase-dependent apoptosis of glioma cells. Because interferon (IFN)-β induces PD-L1 expression, we studied the functional consequences of neuronal Ifnb gene deletion on PD-L1 signaling and function. Ifnb-/- neurons lacked PD-L1 and were defective in inducing glioma cell death; this effect was reversed on PD-L1 gene transfection. Ifnb-/- mice with intracerebral isografts survived poorly. Similar to the observations in GBM patients, better survival in wild-type mice was associated with high neuronal PD-L1 in TABT and downregulation of PD-L1 in tumors, which was defective in Ifnb-/- mice. Our data indicated that neuronal PD-L1 signaling in brain cells was important for GBM patient survival. Reciprocal PD-L1 regulation in TABT and tumor tissue could be a prognostic biomarker for GBM. Understanding the complex interactions between tumor and adjacent stromal tissue is important in designing targeted GBM therapies.
Collapse
|
10
|
Abstract
Radiation therapy methods have evolved remarkably in recent years which have resulted in more effective local tumor control with negligible toxicity of surrounding normal tissues. However, local recurrence and distant metastasis often occur following radiation therapy mostly due to the development of radioresistance through the deregulation of the cell cycle, apoptosis, and inhibition of DNA damage repair mechanisms. Over the last decade, extensive progress in radiotherapy and gene therapy combinatorial approaches has been achieved to overcome resistance of tumor cells to radiation. In this review, we summarize the results from experimental cancer therapy studies on the combination of radiation therapy and gene therapy.
Collapse
|
11
|
The effect of ionizing radiation on the homeostasis and functional integrity of murine splenic regulatory T cells. Inflamm Res 2012; 62:201-12. [PMID: 23080082 DOI: 10.1007/s00011-012-0567-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 09/28/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE Radiotherapy affects antitumor immune responses; therefore, it is important to study radiation effects on various compartments of the immune system. Here we report radiation effects on the homeostasis and function of regulatory T (Treg) cells, which are important in down-regulating antitumor immune responses. METHODS C57Bl/6 mice were irradiated with 2 Gy and alterations in splenic lymphocyte fractions analyzed at different intervals. RESULTS Total CD4+ numbers showed stronger decrease after irradiation than CD4+Foxp3+ Tregs. Tregs were less prone to radiation-induced apoptosis than CD4+Foxp3- T cells. The ratio of CD4+Foxp3- and CD4+Foxp3+ fractions within the proliferating CD4+ pool progressively changed from 74:26 in control animals to 59:41 eleven days after irradiation, demonstrating a more dynamic increase in the proliferation and regeneration of the Treg pool. The CD4+Foxp3+ fraction expressing cell-surface CTLA4, an antigen associated with Treg cell activation increased from 5.3 % in unirradiated mice to 10.5 % three days after irradiation. The expression of IL-10 mRNA was moderately upregulated, while TGF-β expression was not affected. On the other hand, irradiation reduced Treg capacity to suppress effector T cell proliferation by 2.5-fold. CONCLUSION Tregs are more radioresistant, less prone to radiation-induced apoptosis, and have faster repopulation kinetics than CD4+Foxp3- cells, but irradiated Tregs are functionally compromised, having a reduced suppressive capacity.
Collapse
|
12
|
Weiss EM, Wunderlich R, Ebel N, Rubner Y, Schlücker E, Meyer-Pittroff R, Ott OJ, Fietkau R, Gaipl US, Frey B. Selected anti-tumor vaccines merit a place in multimodal tumor therapies. Front Oncol 2012; 2:132. [PMID: 23087898 PMCID: PMC3466463 DOI: 10.3389/fonc.2012.00132] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/17/2012] [Indexed: 12/12/2022] Open
Abstract
Multimodal approaches are nowadays successfully applied in cancer therapy. Primary locally acting therapies such as radiotherapy (RT) and surgery are combined with systemic administration of chemotherapeutics. Nevertheless, the therapy of cancer is still a big challenge in medicine. The treatments often fail to induce long-lasting anti-tumor responses. Tumor recurrences and metastases result. Immunotherapies are therefore ideal adjuncts to standard tumor therapies since they aim to activate the patient's immune system against malignant cells even outside the primary treatment areas (abscopal effects). Especially cancer vaccines may have the potential both to train the immune system against cancer cells and to generate an immunological memory, resulting in long-lasting anti-tumor effects. However, despite promising results in phase I and II studies, most of the concepts finally failed. There are some critical aspects in development and application of cancer vaccines that may decide on their efficiency. The time point and frequency of medication, usage of an adequate immune adjuvant, the vaccine's immunogenic potential, and the tumor burden of the patient are crucial. Whole tumor cell vaccines have advantages compared to peptide-based ones since a variety of tumor antigens (TAs) are present. The master requirements of cell-based, therapeutic tumor vaccines are the complete inactivation of the tumor cells and the increase of their immunogenicity. Since the latter is highly connected with the cell death modality, the inactivation procedure of the tumor cell material may significantly influence the vaccine's efficiency. We therefore also introduce high hydrostatic pressure (HHP) as an innovative inactivation technology for tumor cell-based vaccines and outline that HHP efficiently inactivates tumor cells by enhancing their immunogenicity. Finally studies are presented proving that anti-tumor immune responses can be triggered by combining RT with selected immune therapies.
Collapse
Affiliation(s)
- Eva-Maria Weiss
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Activation of a glioma-specific immune response by oncolytic parvovirus Minute Virus of Mice infection. Cancer Gene Ther 2012; 19:468-75. [PMID: 22539062 DOI: 10.1038/cgt.2012.20] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Rodent autonomous parvoviruses (PVs) are endowed with oncotropic properties and represent virotherapeutics with inherent oncolytic features. This work aimed to evaluate the capacity of Minute Virus of Mice (MVMp) to act as an adjuvant stimulating a mouse glioblastoma-specific immune response. MVMp was shown to induce cell death through apoptosis in glioma GL261 cells. Antigen-presenting cells (APCs) provide the initial cue for innate and adaptive immune responses, and thus MVMp-infected GL261 cells were tested for their ability to activate dendritic cells (DCs) and microglia (MG), two distinct cell types that are able to act as APCs. MG and discrete DC subsets were activated after co-culture with MVMp-infected glioma GL261 cells, as evidenced by upregulation of specific activation markers (CD80, CD86) and release of proinflammatory cytokines (tumor necrosis factor-α and interleukin-6). The in vivo analysis of immunodeficient and immunocompetent mice revealed a clear difference in their susceptibility to MVMp-mediated tumor suppression. Immunocompetent mice were fully protected from tumor outgrowth of GL261 cells infected ex vivo with MVMp. In contrast, immunodeficient animals were less competent for MVMp-dependent tumor inhibition, with only 20% of the recipients being protected, arguing for an additional immune component to allow full tumor suppression. In keeping with this conclusion, immunocompetent mice engrafted with MVMp-infected glioma cells developed a level of anti-tumor immunity with isolated splenocytes producing elevated levels of interferon-γ. In rechallenge experiments using uninfected GL261 cells, we could show complete protection against the tumor, arguing for the induction of a T-cell-mediated, tumor-specific, long-term memory response. These findings indicate that the anticancer effect of PVs can be traced back not only for their direct oncolytic effect, but also to their ability to break tumor tolerance.
Collapse
|
14
|
Revoltella RP, Menicagli M, Campani D. Granulocyte-macrophage colony-stimulating factor as an autocrine survival-growth factor in human gliomas. Cytokine 2011; 57:347-59. [PMID: 22200506 DOI: 10.1016/j.cyto.2011.11.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 10/12/2011] [Accepted: 11/20/2011] [Indexed: 11/28/2022]
Abstract
We studied the expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) and its receptors (GM-CSF.R) in 20 human brain gliomas with different tumor gradings and demonstrated constitutive high levels of both mRNA gene expression and protein production exclusively in the highest-grade tumors (WHO, III-IV grade). Five astrocytic cell lines were isolated in vitro from glioma cells, which had selectively adhered to plates pre-coated with rhGM-CSF. These cells were tumorigenic when xenografted to athymic mice, and produced GM-CSF constitutively in culture. Two lines, particularly lines AS1 and PG1, each from a patient with glioblastoma multiforme, constitutively over-expressed both GM-CSF and GM-CSF.R genes and secreted into their culture media biologically active GM-CSF. Different clones of the AS1 line, isolated after subsequent passages in vitro and then transplanted to athymic mice, demonstrated higher tumorigenic capacity with increasing passages in vivo. Cell proliferation was stimulated by rhGM-CSF in late-stage malignant clones, whereas apoptosis occurred at high frequency in the presence of blocking anti-GM-CSF antibodies. In contrast, rhGM-CSF did not induce any apparent effect in early-stage clones expressing neither GM-CSF nor GM-CSF.R. The addition of rhGM-CSF or rhIL-1β, to cultures induced the overproduction of both GM-CSF and its receptors and increased gene activation for several functional proteins (e.g. NGF, VEGF, VEGF.R1, G-CSF, MHC-II), indicating that these cells may undergo dynamic changes in response to environmental stimuli. These findings thus revealed: (1) that the co-expression of both autocrine GM-CSF and GM-CSF.R correlates with the advanced tumor stage; (2) that an important contribution of GM-CSF in malignant glioma cells is the prevention of apoptosis. These results imply that GM-CSF has an effective role in the evolution and pathogenesis of gliomas.
Collapse
Affiliation(s)
- Roberto P Revoltella
- Institute for Chemical-Physical Processes (IPCF), National Research Council of Italy (CNR), Pisa, Italy.
| | | | | |
Collapse
|
15
|
Hua W, Yao Y, Chu Y, Zhong P, Sheng X, Xiao B, Wu J, Yang B, Mao Y, Zhou L. The CD133+ tumor stem-like cell-associated antigen may elicit highly intense immune responses against human malignant glioma. J Neurooncol 2011; 105:149-57. [DOI: 10.1007/s11060-011-0572-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 03/26/2011] [Indexed: 10/18/2022]
|
16
|
Persson BRR, Koch CB, Grafström G, Ceberg C, Rosenschöld PM, Nittby H, Widegren B, Salford LG. Radiation Immunomodulatory Gene Tumor Therapy of Rats with Intracerebral Glioma Tumors. Radiat Res 2010; 173:433-40. [DOI: 10.1667/rr1733.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
17
|
Van Gool S, Maes W, Ardon H, Verschuere T, Van Cauter S, De Vleeschouwer S. Dendritic cell therapy of high-grade gliomas. Brain Pathol 2009; 19:694-712. [PMID: 19744041 DOI: 10.1111/j.1750-3639.2009.00316.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The prognosis of patients with malignant glioma is poor in spite of multimodal treatment approaches consisting of neurosurgery, radiochemotherapy and maintenance chemotherapy. Among innovative treatment strategies like targeted therapy, antiangiogenesis and gene therapy approaches, immunotherapy emerges as a meaningful and feasible treatment approach for inducing long-term survival in at least a subpopulation of these patients. Setting up immunotherapy for an inherent immunosuppressive tumor located in an immune-privileged environment requires integration of a lot of scientific input and knowledge of both tumor immunology and neuro-oncology. The field of immunotherapy is moving into the direction of active specific immunotherapy using autologous dendritic cells (DCs) as vehicle for immunization. In the translational research program of the authors, the whole cascade from bench to bed to bench of active specific immunotherapy for malignant glioma is covered, including proof of principle experiments to demonstrate immunogenicity of patient-derived mature DCs loaded with autologous tumor lysate, preclinical in vivo experiments in a murine orthotopic glioma model, early phase I/II clinical trials for relapsing patients, a phase II trial for patients with newly diagnosed glioblastoma (GBM) for whom immunotherapy is integrated in the current multimodal treatment, and laboratory analyses of patient samples. The strategies and results of this program are discussed in the light of the internationally available scientific literature in this fast-moving field of basic science and translational clinical research.
Collapse
Affiliation(s)
- Stefaan Van Gool
- Laboratory of Experimental Immunology, and Department of Child & Woman, Catholic University of Leuven, Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
18
|
Immunizations With IFNγ Secreting Tumor Cells can Eliminate Fully Established and Invasive Rat Gliomas. J Immunother 2009; 32:593-601. [DOI: 10.1097/cji.0b013e3181a95148] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Maes W, Rosas GG, Verbinnen B, Boon L, De Vleeschouwer S, Ceuppens JL, Van Gool SW. DC vaccination with anti-CD25 treatment leads to long-term immunity against experimental glioma. Neuro Oncol 2009; 11:529-42. [PMID: 19336528 DOI: 10.1215/15228517-2009-004] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We studied the feasibility, efficacy, and mechanisms of dendritic cell (DC) immunotherapy against murine malignant glioma in the experimental GL261 intracranial (IC) tumor model. When administered prophylactically, mature DCs (DCm) ex vivo loaded with GL261 RNA (DCm-GL261-RNA) protected half of the vaccinated mice against IC glioma, whereas treatment with mock-loaded DCm or DCm loaded with irrelevant antigens did not result in tumor protection. In DCm-GL261-RNA-vaccinated mice, a tumor-specific cellular immune response was observed ex vivo in the spleen and tumordraining lymph node cells. Specificity was also shown in vivo on the level of tumor challenge. Depletion of CD8(+) T-cells by anti-CD8 treatment at the time of tumor challenge demonstrated their essential role in vaccine-mediated antitumor immunity. Depletion of CD25(+) regulatory T-cells (Tregs) by anti-CD25 (aCD25) treatment strongly enhanced the efficacy of DC vaccination and was itself also protective, independently of DC vaccination. However, DC vaccination was essential to protect the animals from IC tumor rechallenge. No long-term protection was observed in animals that initially received aCD25 treatment only. In mice that received DC and/or aCD25 treatment, we retrieved tumor-specific brain-infiltrating cytotoxic T-lymphocytes. These data clearly demonstrate the effectiveness of DC vaccination for the induction of long-lasting immunological protection against IC glioma. They also show the beneficial effect of Treg depletion in this kind of glioma immunotherapy, even combined with DC vaccination.
Collapse
Affiliation(s)
- Wim Maes
- Clinical Immunology, Department of Experimental Medicine, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
20
|
Adenoviral vector transduction of the human deoxycytidine kinase gene enhances the cytotoxic and radiosensitizing effect of gemcitabine on experimental gliomas. Cancer Gene Ther 2008; 15:154-64. [DOI: 10.1038/sj.cgt.7701115] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Newcomb EW, Demaria S, Lukyanov Y, Shao Y, Schnee T, Kawashima N, Lan L, Dewyngaert JK, Zagzag D, McBride WH, Formenti SC. The combination of ionizing radiation and peripheral vaccination produces long-term survival of mice bearing established invasive GL261 gliomas. Clin Cancer Res 2007; 12:4730-7. [PMID: 16899624 DOI: 10.1158/1078-0432.ccr-06-0593] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE High-grade glioma treatment includes ionizing radiation therapy. The high invasiveness of glioma cells precludes their eradication and is responsible for the dismal prognosis. Recently, we reported the down-regulation of MHC class I (MHC-I) products in invading tumor cells in human and mouse GL261 gliomas. Here, we tested the hypothesis that whole-brain radiotherapy (WBRT) up-regulates MHC-I expression on GL261 tumors and enhances the effectiveness of immunotherapy. EXPERIMENTAL DESIGN MHC-I molecule expression on GL261 cells was analyzed in vitro and in vivo by flow cytometry and immunohistochemistry, respectively. To test the response of established GL261 gliomas to treatment, mice with measurable (at CT imaging) brain tumors were randomly assigned to four groups receiving (a) no treatment, (b) WBRT in two fractions of 4 Gy, (c) vaccination with irradiated GL261 cells secreting granulocyte-macrophage colony-stimulating factor, or (d) WBRT and vaccination. Endpoints were tumor response and survival. RESULTS An ionizing radiation dose of 4 Gy maximally up-regulated MHC-I molecules on GL261 cells in vitro. In vivo, WBRT induced the expression of the beta2-microglobulin light chain subunit of the MHC class I complex on glioma cells invading normal brain and increased CD4+ and CD8+ T cell infiltration. However, the survival advantage obtained with WBRT or vaccination alone was minimal. In contrast, WBRT in combination with vaccination increased long-term survival to 40% to 80%, compared with 0% to 10% in the other groups (P < 0.002). Surviving animals showed antitumor immunity by rejecting challenge tumors. CONCLUSION Ionizing radiation can be successfully combined with peripheral vaccination for the treatment of established high-grade gliomas.
Collapse
Affiliation(s)
- Elizabeth W Newcomb
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yokouchi H, Chamoto K, Wakita D, Yamazaki K, Shirato H, Takeshima T, Dosaka-Akita H, Nishimura M, Yue Z, Kitamura H, Nishimura T. Combination tumor immunotherapy with radiotherapy and Th1 cell therapy against murine lung carcinoma. Clin Exp Metastasis 2007; 24:533-40. [PMID: 17653821 DOI: 10.1007/s10585-007-9090-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 07/05/2007] [Indexed: 11/25/2022]
Abstract
Mice bearing established Lewis lung carcinoma (LLC) expressing model tumor antigen, ovalbumin (OVA) (LLC-OVA) marginally responded to local radiotherapy, but none of the mice was cured. In contrast, treatment of the tumor-bearing mice with intratumoral injection of tumor-specific T helper type 1 (Th1) cells and tumor antigen (OVA) after radiotherapy dramatically prolonged the survival days and induced complete cure of the mice at high frequency (80%). Radiation therapy combined with Th1 cells or OVA alone showed no significant therapeutic activity against LLC-OVA. Such a strong therapeutic activity was not induced by intratumoral injection of Th1 cells plus OVA. Compared with other treatment, radiation therapy combined with Th1 cells and OVA was superior to induce the generation of OVA/H-2(b) tetramer(+) tumor-specific cytotoxic T lymphocyte (CTL) with a strong cytotoxicity against LLC-OVA in draining lymph node (DLN). Moreover, the combined therapy is demonstrated to inhibit the growth of tumor mass, which grew at contralateral side. These results indicated that radiotherapy combined with Th1 cell/vaccine therapy induced a systemic antitumor immunity. These findings suggested that combination therapy with radiotherapy and Th1 cell/vaccine therapy may become a practical strategy for cancer treatment.
Collapse
Affiliation(s)
- Hiroshi Yokouchi
- Division of Immunoregulation, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, N-15, W-7, Kita-ku, Sapporo 060-0815, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Smith KE, Janelidze S, Visse E, Badn W, Salford L, Siesjö P, Darabi A. Synergism between GM-CSF and IFNgamma: enhanced immunotherapy in mice with glioma. Int J Cancer 2007; 120:75-80. [PMID: 17044023 DOI: 10.1002/ijc.22286] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glioblastoma multiforme is the most common malignant primary brain tumor and also one of the most therapy-resistant tumors. Because of the dismal prognosis, various therapies modulating the immune system have been developed in experimental models. Previously, we have shown a 37-70% cure in a rat glioma model where rats were peripherally immunized with tumor cells producing IFNgamma. On the basis of these results, we wanted to investigate whether a combination of GM-CSF and IFNgamma could improve the therapeutic effect in a mouse glioma model, GL261 (GL-wt). Three biweekly intraperitoneal (i.p.) immunizations with irradiated GM-CSF-transduced GL261 cells (GL-GM) induced a 44% survival in mice with intracranial glioma. While treatment of GL-wt and GL-GM with IFNgamma in vitro induced upregulation of MHC I and MHC II on the tumor cells, it could not enhance survival after immunization. However, immunizations with GL-GM combined with recombinant IFNgamma at the immunization site synergistically enhanced survival with a cure rate of 88%. Tumors from mice receiving only 1 immunization on Day 10 after tumor inoculation were sectioned on Day 20 for analysis of leukocyte infiltration. Tumor volume was reduced and the infiltration of macrophages was denser in mice immunized with GL-GM combined with IFNgamma compared with that of both wildtype and nonimmunized mice. To our knowledge, this is the first study to demonstrate a synergy between GM-CSF and IFNgamma in experimental immunotherapy of tumors, by substantially increasing survival as well as inducing a potent anti-tumor response after only 1 postponed immunization.
Collapse
Affiliation(s)
- Karin E Smith
- Glioma Immunotherapy Group, Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, University of Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
24
|
Lumniczky K, Sáfrány G. Cancer gene therapy: Combination with radiation therapy and the role of bystander cell killing in the anti-tumor effect. Pathol Oncol Res 2006; 12:118-24. [PMID: 16799716 DOI: 10.1007/bf02893457] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 04/20/2006] [Indexed: 11/28/2022]
Abstract
Current anti-cancer modalities such as surgery, chemo- and radiation therapies have only limited success in cancer treatment. Gene therapy is a promising new tool to improve outcomes. In this review, first we summarize the various strategies to kill tumor cells, and then focus on the bystander effect of gene therapy. A variety of strategies, such as gene-directed enzyme pro-drug therapy, activation of an anti-tumor immune attack, application of replication-competent and oncolytic viral vectors, tumor-specific as well as radiation- and hypoxiainduced gene expression, might be applied to target tumor cells. We put special emphasis on the combination of these approaches with local tumor irradiation. Using the available vector systems, only a small portion of cancer cells contains the therapeutic genes under clinical situations. However, cells directly targeted by gene therapy will transfer death signals to neighboring cancer cells. This bystander cell killing improves the efficiency of cancer gene therapy. Death signals are delivered by cell-to-cell communication through gap junction intercellular contacts, release of toxic metabolites into the neighborhood or to larger distances, phagocytosis of apoptotic bodies, and the activation of the immune system. Bystander cell killing can be enhanced by the introduction of gap junction proteins into cells, by further activating the immune system with immune-stimulatory molecules, or by introducing genes that help the transfer of cytotoxic genes and/or metabolites into bystander cells. In conclusion, although bystander cell killing can improve therapeutic effects, there should be additional developments in cancer gene therapy for a more efficient clinical application.
Collapse
Affiliation(s)
- Katalin Lumniczky
- Department of Molecular and Tumor Radiobiology, National Research Institute for Radiobiology and Radiohygiene, Budapest, H-1221, Hungary
| | | |
Collapse
|
25
|
Szatmári T, Lumniczky K, Désaknai S, Trajcevski S, Hídvégi EJ, Hamada H, Sáfrány G. Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci 2006; 97:546-53. [PMID: 16734735 PMCID: PMC11159227 DOI: 10.1111/j.1349-7006.2006.00208.x] [Citation(s) in RCA: 252] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Mouse glioma 261 (Gl261) cells are used frequently in experimental glioblastoma therapy; however, no detailed description of the Gl261 tumor model is available. Here we present that Gl261 cells carry point mutations in the K-ras and p53 genes. Basal major histocompatibility complex (MHC)I, but not MHCII, expression was detected in Gl261 cells. The introduction of interferon-gamma-encoding genes increased expression of both MHCI and MHCII. A low amount of B7-1 and B7-2 RNA was detected in wild-type cells, but cytokine production did not change expression levels. Gl261 cells were transduced efficiently by adenoviral vectors; the infectivity of retroviral vectors was limited. Low numbers of transplanted Gl261 cells formed both subcutaneous and intracranial tumors in C57BL/6 mice. The cells were moderately immunogenic: prevaccination of mice with irradiated tumor cells 7 days before intracranial tumor challenge prevented tumor formation in approximately 90% of mice. When vaccination was carried out on the day or 3 days after tumor challenge, no surviving animals could be found. In vitro-growing cells were radiosensitive: less than 2 Gy was required to achieve 50% cell mortality. Local tumor irradiation with 4 Gy X-rays in brain tumor-bearing mice slowed down tumor progression, but none of the mice were cured off the tumor. In conclusion, the Gl261 brain tumor model might be efficiently used to study the antitumor effects of various therapeutic modalities, but the moderate immunogenicity of the cells should be considered.
Collapse
Affiliation(s)
- Tünde Szatmári
- Department of Molecular and Tumor Radiobiology, Frederic Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest 1221, Hungary
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The long-term survival of children with brain tumor has improved considerably in the last three decades, owing to advances in neuroimaging, neurosurgical, and radiation therapy modalities, coupled with the application of conventional chemotherapy. MRI, MR spectroscopy and diffusion-weighted MRI have contributed to more accurate diagnosis, prognostication and better treatment planning. Neurosurgical treatment has been advanced by the use of functional MRI, and intraoperative image-guided stereotactic techniques and electrophysiologic monitoring. The use of 3-D conformal and intensity-modulated radiation therapy, stereotactic radiosurgery, and radiosensitizing agents has made radiation therapy safer and more effective. Conventional chemotherapy, administered either alone or combined with radiation therapy has improved survival and quality of life of children with brain tumors. These improved outcomes have also occurred, due, in part, to their treatment on collaborative national and international studies. Recent promising diagnostic and therapeutic strategies have resulted from advances in understanding molecular brain tumor biology. Important new approaches include the refinement of drug-delivery strategies, the evaluation of biologic markers to stratify patients for optimal treatment and to exploit these molecular differences using "targeted" therapeutic strategies. These approaches include blocking tumor cell drug resistance mechanisms, immunotherapy, inhibition of molecular signal transduction pathways important in tumorigenesis, anti-angiogenic therapy, and gene therapy. The thrust of such approaches for children with brain tumors is especially directed at reducing the toxicity of therapy and improving quality-of-life, as well as increasing disease-free survival.
Collapse
Affiliation(s)
- Patricia L Robertson
- Department of Pediatrics and Neurology, Division of Pediatric Neurology, University of Michigan Health System, 1500 E. Medical Center Dr., L3215 Women's Hospital, Ann Arbor, 48109-0203, USA.
| |
Collapse
|
27
|
Clavreul A, Delhaye M, Jadaud E, Menei P. Effects of syngeneic cellular vaccinations alone or in combination with GM-CSF on the weakly immunogenic F98 glioma model. J Neurooncol 2006; 79:9-17. [PMID: 16575532 DOI: 10.1007/s11060-005-9115-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Accepted: 12/30/2005] [Indexed: 01/02/2023]
Abstract
Cancer vaccines are one approach for the treatment of brain tumors. Most experimental studies are performed on so-called "immunogenic" brain tumor models such as the rat 9L glioma which does not reflect characteristics of human glioblastoma. In the present study, we tested syngeneic cellular vaccinations alone or in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) on the weakly immunogenic F98 glioma model. Previous studies have shown the efficacy of this treatment on the 9L glioma model. Fisher rats received an intracerebral implantation of F98 cells. Three days later, two subcutaneous vaccinations with irradiated F98 cells were realized in presence or absence of GM-CSF. This scheme of vaccination induced a systemic cellular and humoral immune response capable of in vitro cytolytic activity against F98 cells. However, no significant differences in survival times were noted between vaccinated and untreated animals. Animals vaccinated with GM-CSF or without GM-CSF had respectively a survival time of 26 +/- 2.1 and 25 +/- 4.4 days following tumor challenge versus 26.5 +/- 2.4 days for untreated rats. Fourteen days after the intracerebral tumor implantation, the tumors of vaccinated animals showed a robust infiltration by T lymphocytes, NK cells, dendritic cells, granulocytes and CD11b/c+ myeloid cells. This infiltration was nearly absent in untreated animals except for CD11b/c+ myeloid cells. This study shows that, contrary to the 9L glioma model, the F98 glioma model is resistant to syngeneic cellular vaccinations although a strong peripheral and intratumoral immune response can be induced. These results suggest that the F98 glioma is an attractive model to understand the mechanisms of glioma immunotherapy resistance.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, 49033, Angers Cedex 01, France
| | | | | | | |
Collapse
|
28
|
Horsman MR, Bohm L, Margison GP, Milas L, Rosier JF, Safrany G, Selzer E, Verheij M, Hendry JH. Tumor radiosensitizers--current status of development of various approaches: report of an International Atomic Energy Agency meeting. Int J Radiat Oncol Biol Phys 2006; 64:551-61. [PMID: 16414371 DOI: 10.1016/j.ijrobp.2005.09.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Revised: 08/30/2005] [Accepted: 09/29/2005] [Indexed: 01/02/2023]
Abstract
PURPOSE The International Atomic Energy Agency (IAEA) held a Technical Meeting of Consultants to (1) discuss a selection of relatively new agents, not those well-established in clinical practice, that operated through a variety of mechanisms to sensitize tumors to radiation and (2) to compare and contrast their tumor efficacy, normal tissue toxicity, and status of development regarding clinical application. The aim was to advise the IAEA as to which developing agent or class of agents would be worth promoting further, by supporting additional laboratory research or clinical trials, with the eventual goal of improving cancer control rates using radiotherapy, in developing countries in particular. RESULTS The agents under discussion included a wide, but not complete, range of different types of drugs, and antibodies that interfered with molecules in cell signaling pathways. These were contrasted with new molecular antisense and gene therapy strategies. All the drugs discussed have previously been shown to act as tumor cell radiosensitizers or to kill hypoxic cells present in tumors. CONCLUSION Specific recommendations were made for more preclinical studies with certain of the agents and for clinical trials that would be suitable for industrialized countries, as well as trials that were considered more appropriate for developing countries.
Collapse
Affiliation(s)
- Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gallo P, Dharmapuri S, Cipriani B, Monaci P. Adenovirus as vehicle for anticancer genetic immunotherapy. Gene Ther 2006; 12 Suppl 1:S84-91. [PMID: 16231059 DOI: 10.1038/sj.gt.3302619] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenoviruses (Ads) are in the forefront of genetic immunization methods being developed against cancer. Their ability to elicit an effective immune response against tumor-associated antigens has been demonstrated in many model systems. Several clinical trials, which use Ad as vehicle for immunization, are already in progress. Preclinical studies have also demonstrated the efficacy of combining Ad-mediated immunization with adjuvants such as chemotherapeutic agents and cytokines. Issues related to sero-prevalence and safety of Ads, however, continue to pose a challenge and need to be addressed.
Collapse
Affiliation(s)
- P Gallo
- Department of Molecular and Cell Biology, I.R.B.M.P. Angeletti, Pomezia, Roma, Italy
| | | | | | | |
Collapse
|
30
|
Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys 2005; 63:655-66. [PMID: 16199306 PMCID: PMC1489884 DOI: 10.1016/j.ijrobp.2005.06.032] [Citation(s) in RCA: 276] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Revised: 06/28/2005] [Accepted: 06/30/2005] [Indexed: 02/07/2023]
Abstract
Ionizing radiation therapy (RT) is an important local modality for the treatment of cancer. The current rationale for its use is based largely on the ability of RT to kill the cancer cells by a direct cytotoxic effect. Nevertheless, considerable evidence indicates that RT effects extend beyond the mere elimination of the more radiosensitive fraction of cancer cells present within a tumor at the time of radiation exposure. For instance, a large body of evidence is accumulating on the ability of RT to modify the tumor microenvironment and generate inflammation. This might have far-reaching consequences regarding the response of a patient to treatment, especially if radiation-induced tumor cell kill were to translate into the generation of effective antitumor immunity. Although much remains to be learned about how radiation can impact tumor immunogenicity, data from preclinical studies provide the proof of principle that different immunotherapeutic strategies can be combined with RT to enhance antitumor effects. Conversely, RT could be a useful tool to combine with immunotherapy. This article will briefly summarize what is known about the impact of RT on tumor immunity, including tumor-associated antigens, antigen-presenting cells, and effector mechanisms. In addition, the experimental evidence supporting the contention that RT can be used as a tool to induce antitumor immunity is discussed, and a new approach to radioimmunotherapy of cancer is proposed.
Collapse
Affiliation(s)
- Sandra Demaria
- Departments of Pathology and
- Address correspondence and reprint requests to: Sandra Demaria, M. D., Department of Pathology, MSB-563, New York University School of Medicine, 550 First Avenue, New York, NY 10016. Tel: (212) 263-7308; Fax: (212) 263-8211; e-mail:
| | - Nina Bhardwaj
- NYU Cancer Institute, New York University School of Medicine, New York, NY 10016
| | - William H. McBride
- Department of Radiation Oncology, Experimental Division, University of California at Los Angeles School of Medicine, Los Angeles, CA 90095, USA
| | | |
Collapse
|
31
|
Kuwashima N, Nishimura F, Eguchi J, Sato H, Hatano M, Tsugawa T, Sakaida T, Dusak JE, Fellows-Mayle WK, Papworth GD, Watkins SC, Gambotto A, Pollack IF, Storkus WJ, Okada H. Delivery of dendritic cells engineered to secrete IFN-alpha into central nervous system tumors enhances the efficacy of peripheral tumor cell vaccines: dependence on apoptotic pathways. THE JOURNAL OF IMMUNOLOGY 2005; 175:2730-40. [PMID: 16081851 DOI: 10.4049/jimmunol.175.4.2730] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We tested whether modulation of the CNS-tumor microenvironment by delivery of IFN-alpha-transduced dendritic cells (DCs: DC-IFN-alpha) would enhance the therapeutic efficacy of peripheral vaccinations with cytokine-gene transduced tumor cells. Mice bearing intracranial GL261 glioma or MCA205 sarcoma received peripheral immunizations with corresponding irradiated tumor cells engineered to express IL-4 or GM-CSFs, respectively, as well as intratumoral delivery of DC-IFN-alpha. This regimen prolonged survival of the animals and induced tumor-specific CTLs that expressed TRAIL, which in concert with perforin and Fas ligand (FasL) was involved in the tumor-specific CTL activity of these cells. The in vivo antitumor activity associated with this approach was abrogated by administration of neutralizing mAbs against TRAIL or FasL and was not observed in perforin-/-, IFN-gamma-/-, or FasL-/- mice. Transduction of the tumor cells with antiapoptotic protein cellular FLIP rendered the gene-modified cells resistant to TRAIL- or FasL-mediated apoptosis and to CTL killing activity in vitro. Furthermore, the combination therapeutic regimen was ineffective in an intracranial cellular FLIP-transduced MCA205 brain tumor model. These results suggest that the combination of intratumoral delivery of DC-IFN-alpha and peripheral immunization with cytokine-gene transduced tumor cells may be an effective therapy for brain tumors that are sensitive to apoptotic signaling pathways.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Adoptive Transfer
- Animals
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Apoptosis/immunology
- Brain Neoplasms/immunology
- Brain Neoplasms/pathology
- Brain Neoplasms/prevention & control
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cytokines/biosynthesis
- Cytokines/physiology
- Cytokines/therapeutic use
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Glioma/immunology
- Glioma/pathology
- Glioma/prevention & control
- Injections, Intralesional
- Injections, Intraventricular
- Interferon-alpha/administration & dosage
- Interferon-alpha/genetics
- Interferon-alpha/metabolism
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Signal Transduction/immunology
- Transduction, Genetic
Collapse
Affiliation(s)
- Naruo Kuwashima
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sikorski CW, Lesniak MS. Immunotherapy for malignant glioma: current approaches and future directions. Neurol Res 2005; 27:703-16. [PMID: 16197807 DOI: 10.1179/016164105x49481] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Traditional therapies for the treatment of malignant glioma have failed to make appreciable gains regarding patient outcome in the last decade. Therefore, immunotherapeutic approaches have become increasingly popular in the treatment of this cancer. This article reviews general immunology of the central nervous system and the immunobiology of malignant glioma to provide a foundation for understanding the rationale behind current glioma immunotherapies. A review of currently implemented immunological treatments is then provided with special attention paid to the use of vaccines, gene therapy, cytokines, dendritic cells and viruses. Insights into future and developing avenues of glioma immunotherapy, such as novel delivery systems, are also discussed.
Collapse
Affiliation(s)
- Christian W Sikorski
- Division of Neurosurgery, The University of Chicago Pritzker School of Medicine, 5841 South Maryland Avenue, MC 3026, Chicago, Illinois 60637, USA
| | | |
Collapse
|
33
|
Meng Y, Carpentier AF, Chen L, Boisserie G, Simon JM, Mazeron JJ, Delattre JY. Successful combination of local CpG-ODN and radiotherapy in malignant glioma. Int J Cancer 2005; 116:992-7. [PMID: 15856470 DOI: 10.1002/ijc.21131] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Oligodeoxynucleotides containing CpG motifs (CpG-ODN) display broad immunostimulating activity and are currently under clinical trial in various malignancies, including recurrent glioblastomas. Combining CpG-ODN with another therapy that could induce antigen release might enhance tumor-specific immune response. We investigated whether radiotherapy (RT) could be associated advantageously to intratumoral injections of CpG-ODN. Fisher rats bearing 9L glioma were treated with various combinations of RT and CpG-28, an oligonucleotide with good immunostimulating activity. RT and CpG-28 induced complete tumor remission in one-third of the animals. When both treatments were combined, complete tumor remission was achieved in two-thirds of the animals (p < 0.001 when compared to non-treated rats, p < 0.03 when compared to CpG-28 alone). Such efficacy was not observed in nude mice, underlying the role of T cells in antitumor effects. The combination of both treatments appeared optimal when the delay between RT and CpG-28 administration was <3 days (from 100% survival for a 3 days delay, to 57% survival for a 21 days delay, p < 0.05). Tumor infiltration by immune cells and expression within tumors of the CpG receptor, TLR9, were not modified by irradiation. These results support an attractive strategy of sequential radiotherapy and immunotherapy by CpG-ODN and have potential implications for future clinical trials with CpG-ODN.
Collapse
Affiliation(s)
- Yuxia Meng
- Fédération de neurologie Mazarin and Institut National de la Santé et de la Recherche Médicale, UMR-495, Hôpital de la Salpêtrière, Paris, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Gene therapy is a potentially useful approach in the treatment of human brain tumors, which are notoriously refractory to conventional approaches. Most human clinical trials to date have been unsuccessful in terms of improving patient outcome. Recent improvements in viral vectors, the development of stem cell technology, and increased understanding of the mechanism of action of therapeutic transgenes provide hope that the next generation of gene therapeutics may show increased efficacy in treatment of this devastating disease.
Collapse
Affiliation(s)
- S E Lawler
- Department of Neurological Surgery, The Dardinger Family Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Medical Center, Columbus, 43210, USA
| | | | | |
Collapse
|
35
|
Quatan N, Spicer J, Plunkett T, Pandha H. Cellular immunotherapy for cancer: current concepts and clinical perspectives. Clin Oncol (R Coll Radiol) 2004; 16:356-65. [PMID: 15341440 DOI: 10.1016/j.clon.2004.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- N Quatan
- Department of Urology, St George's Hospital Medical School, London, UK
| | | | | | | |
Collapse
|
36
|
Ishikawa E, Tsuboi K, Takano S, Uchimura E, Nose T, Ohno T. Intratumoral injection of IL-2-activated NK cells enhances the antitumor effect of intradermally injected paraformaldehyde-fixed tumor vaccine in a rat intracranial brain tumor model. Cancer Sci 2004; 95:98-103. [PMID: 14720334 PMCID: PMC11158400 DOI: 10.1111/j.1349-7006.2004.tb03177.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Revised: 11/18/2003] [Accepted: 11/18/2003] [Indexed: 11/29/2022] Open
Abstract
Combined therapy with a fixed-tumor cell vaccine and intratumoral injection of NK cells induced strong tumor regression of rat glioma. Rat 9L glioma cells were inoculated into syngeneic male rats at the flank (subcutaneous tumor model) or at the basal ganglia of the right hemisphere (intracranial tumor model). Rats were intradermally injected three times with vaccine comprising fixed 9L cells, IL-2- and GMCSF-microparticles, and tuberculin prior to (protective studies) or after (therapeutic studies) challenge with live 9L cells. In the protective studies, the vaccine alone achieved significant tumor growth inhibition and elongation of mean life span in both the subcutaneous and intracranial tumor models. No therapeutic effect was observed in the intracranial tumor model with the vaccine alone. However, intratumoral injection of rat NK cells strongly assisted the therapeutic effect of the vaccine in the brain tumor model and resulted in a statistically significant elongation of life span. We propose that intratumoral injection of NK cells may not only kill brain tumor cells directly, but also trigger a strong immune response in the focal lesion of the brain after vaccination.
Collapse
Affiliation(s)
- Eiichi Ishikawa
- Department of Neurosurgery, Institute of Clinical Medicine, University of Tsukuba, Tsukuba Science City, Ibaraki 305-8575, Japan
| | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Walker PR, Calzascia T, de Tribolet N, Dietrich PY. T-cell immune responses in the brain and their relevance for cerebral malignancies. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2003; 42:97-122. [PMID: 12738053 DOI: 10.1016/s0165-0173(03)00141-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In order that cellular immune responses afford protection without risk to sensitive normal tissue, they must be adapted to individual tissues of the body. Nowhere is this more critical than for the brain, where various passive and active mechanisms maintain a state of immune privilege that can limit high magnitude immune responses. Nevertheless, it is now clear that immune responses are induced to antigens in the brain, including those expressed by cerebral malignancies. We discuss hypotheses of how this can occur, although details such as which antigen presenting cells are involved remain to be clarified. Antitumor responses induced spontaneously are insufficient to eradicate malignant astrocytomas; many studies suggest that this can be explained by a combination of low level immune response induction and tumor mediated immunosuppression. A clinical objective currently pursued is to use immunotherapy to ameliorate antitumour immunity. This will necessitate a high level immune response to ensure sufficient effector cells reach the tumor bed, focused cytotoxicity to eradicate malignant cells with little collateral damage to critical normal cells, and minimal inflammation. To achieve these aims, priority should be given to identifying more target antigens in astrocytoma and defining those cells present in the brain parenchyma that are essential to maintain antitumour effector function without exacerbating inflammation. If we are armed with better understanding of immune interactions with brain tumor cells, we can realistically envisage that immunotherapy will one day offer hope to patients with currently untreatable neoplastic diseases of the CNS.
Collapse
Affiliation(s)
- Paul R Walker
- Laboratory of Tumour Immunology, Division of Oncology, Geneva University Hospital, 24 rue Micheli-du-Crest, 1211 Geneva 14, Switzerland.
| | | | | | | |
Collapse
|
39
|
Desaknai S, Lumniczky K, Esik O, Hamada H, Safrany G. Local tumour irradiation enhances the anti-tumour effect of a double-suicide gene therapy system in a murine glioma model. J Gene Med 2003; 5:377-85. [PMID: 12731086 DOI: 10.1002/jgm.357] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Gliomas are invasive malignant tumours with poor prognosis. Combination of gene directed enzyme pro-drug therapy with existing treatment modalities might open new therapeutic potentials. METHODS Murine glioma 261 (Gl261) cells were transduced with an adenoviral vector (Adex-CAUPTK) encoding both uracil phosphoribosyltransferase and thymidine kinase genes which sensitise cells to 5-fluorouracil (5-FU) and ganciclovir (GC), respectively. Subcutaneous or intracranial tumours were established in C57Bl mice by transplanting drug-sensitising gene containing Gl261 cells. In vitro growing cells or established tumours were treated with 5-FU, GC and ionising radiation either alone or in combinations. Finally, subcutaneous tumours were established with non-transduced cells, directly injected with Adex-CAUPTK, and mice were treated with combinations of 5-FU, GC and tumour irradiation. RESULTS In vitro treatment of transduced Gl261 cells with both 5-FU and GC showed enhanced cytotoxic effect compared with single agents. Combination of drug treatments with irradiation greatly increased cytotoxicity. In subcutaneous and intracranial tumours double-agent treatment was more effective than a single drug. Combination with local irradiation highly improved the anti-tumour effect (90-100% survival) even when only part of the tumour cells carried drug-sensitising genes (40-50% survival at 10% rate). Treatment of established tumours with direct intra-tumour Adex-CAUPTK inoculations and intraperitoneal 5-FU, GC injections slowed down tumour progression that was further enhanced by local irradiation. CONCLUSION The combination of double-suicide gene therapy with local irradiation is a promising tool to eradicate small, residual tumours.
Collapse
Affiliation(s)
- Szilvia Desaknai
- Department of Molecular and Tumour Radiobiology, National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | | | | | | | | |
Collapse
|