1
|
Sperring CP, Argenziano MG, Savage WM, Teasley DE, Upadhyayula PS, Winans NJ, Canoll P, Bruce JN. Convection-enhanced delivery of immunomodulatory therapy for high-grade glioma. Neurooncol Adv 2023; 5:vdad044. [PMID: 37215957 PMCID: PMC10195574 DOI: 10.1093/noajnl/vdad044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
The prognosis for glioblastoma has remained poor despite multimodal standard of care treatment, including temozolomide, radiation, and surgical resection. Further, the addition of immunotherapies, while promising in a number of other solid tumors, has overwhelmingly failed in the treatment of gliomas, in part due to the immunosuppressive microenvironment and poor drug penetrance to the brain. Local delivery of immunomodulatory therapies circumvents some of these challenges and has led to long-term remission in select patients. Many of these approaches utilize convection-enhanced delivery (CED) for immunological drug delivery, allowing high doses to be delivered directly to the brain parenchyma, avoiding systemic toxicity. Here, we review the literature encompassing immunotherapies delivered via CED-from preclinical model systems to clinical trials-and explore how their unique combination elicits an antitumor response by the immune system, decreases toxicity, and improves survival among select high-grade glioma patients.
Collapse
Affiliation(s)
- Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - William M Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Damian E Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Pavan S Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Nathan J Winans
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
2
|
Quader S, Kataoka K, Cabral H. Nanomedicine for brain cancer. Adv Drug Deliv Rev 2022; 182:114115. [PMID: 35077821 DOI: 10.1016/j.addr.2022.114115] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/18/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
CNS tumors remain among the deadliest forms of cancer, resisting conventional and new treatment approaches, with mortality rates staying practically unchanged over the past 30 years. One of the primary hurdles for treating these cancers is delivering drugs to the brain tumor site in therapeutic concentration, evading the blood-brain (tumor) barrier (BBB/BBTB). Supramolecular nanomedicines (NMs) are increasingly demonstrating noteworthy prospects for addressing these challenges utilizing their unique characteristics, such as improving the bioavailability of the payloadsviacontrolled pharmacokinetics and pharmacodynamics, BBB/BBTB crossing functions, superior distribution in the brain tumor site, and tumor-specific drug activation profiles. Here, we review NM-based brain tumor targeting approaches to demonstrate their applicability and translation potential from different perspectives. To this end, we provide a general overview of brain tumor and their treatments, the incidence of the BBB and BBTB, and their role on NM targeting, as well as the potential of NMs for promoting superior therapeutic effects. Additionally, we discuss critical issues of NMs and their clinical trials, aiming to bolster the potential clinical applications of NMs in treating these life-threatening diseases.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
3
|
Choi HJ, Lee HB, Jung S, Park HK, Jo W, Cho SM, Kim WJ, Son WC. Development of a Mouse Model of Prostate Cancer Using the Sleeping Beauty Transposon and Electroporation. Molecules 2018; 23:molecules23061360. [PMID: 29874846 PMCID: PMC6100630 DOI: 10.3390/molecules23061360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/20/2018] [Accepted: 06/01/2018] [Indexed: 01/12/2023] Open
Abstract
The Sleeping Beauty (SB) transposon system is non-viral and uses insertional mutagenesis, resulting in the permanent expression of transferred genes. Although the SB transposon is a useful method for establishing a mouse tumor model, there has been difficulty in using this method to generate tumors in the prostate. In the present study, electroporation was used to enhance the transfection efficiency of the SB transposon. To generate tumors, three constructs (a c-Myc expression cassette, a HRAS (HRas proto-oncogene, GTPase) expression cassette and a shRNA against p53) contained within the SB transposon plasmids were directly injected into the prostate. Electroporation was conducted on the injection site after the injection of the DNA plasmid. Following the tumorigenesis, the tumors were monitored by animal PET imaging and identified by gross observation. After this, the tumors were characterized by using histological and immunohistochemical techniques. The expression of the targeted genes was analyzed by Real-Time qRT-PCR. All mice subjected to the injection were found to have prostate tumors, which was supported by PSA immunohistochemistry. To our knowledge, this is the first demonstration of tumor induction in the mouse prostate using the electroporation-enhanced SB transposon system in combination with c-Myc, HRAS and p53. This model serves as a valuable resource for the future development of SB-induced mouse models of cancer.
Collapse
Affiliation(s)
- Hyun-Ji Choi
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Han-Byul Lee
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Sunyoung Jung
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Hyun-Kyu Park
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Woori Jo
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Sung-Min Cho
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Woo-Jin Kim
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| | - Woo-Chan Son
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, 05505 Seoul, Korea.
- Department of Pathology, University of Ulsan College of Medicine, Songpa-gu, 05505 Seoul, Korea.
| |
Collapse
|
4
|
Preclinical and clinical advances in transposon-based gene therapy. Biosci Rep 2017; 37:BSR20160614. [PMID: 29089466 PMCID: PMC5715130 DOI: 10.1042/bsr20160614] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023] Open
Abstract
Transposons derived from Sleeping Beauty (SB), piggyBac (PB), or Tol2 typically require cotransfection of transposon DNA with a transposase either as an expression plasmid or mRNA. Consequently, this results in genomic integration of the potentially therapeutic gene into chromosomes of the desired target cells, and thus conferring stable expression. Non-viral transfection methods are typically preferred to deliver the transposon components into the target cells. However, these methods do not match the efficacy typically attained with viral vectors and are sometimes associated with cellular toxicity evoked by the DNA itself. In recent years, the overall transposition efficacy has gradually increased by codon optimization of the transposase, generation of hyperactive transposases, and/or introduction of specific mutations in the transposon terminal repeats. Their versatility enabled the stable genetic engineering in many different primary cell types, including stem/progenitor cells and differentiated cell types. This prompted numerous preclinical proof-of-concept studies in disease models that demonstrated the potential of DNA transposons for ex vivo and in vivo gene therapy. One of the merits of transposon systems relates to their ability to deliver relatively large therapeutic transgenes that cannot readily be accommodated in viral vectors such as full-length dystrophin cDNA. These emerging insights paved the way toward the first transposon-based phase I/II clinical trials to treat hematologic cancer and other diseases. Though encouraging results were obtained, controlled pivotal clinical trials are needed to corroborate the efficacy and safety of transposon-based therapies.
Collapse
|
5
|
Tipanee J, VandenDriessche T, Chuah MK. Transposons: Moving Forward from Preclinical Studies to Clinical Trials. Hum Gene Ther 2017; 28:1087-1104. [DOI: 10.1089/hum.2017.128] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Jaitip Tipanee
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium
| | - Thierry VandenDriessche
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Marinee K. Chuah
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Hou X, Du Y, Deng Y, Wu J, Cao G. Sleeping Beauty transposon system for genetic etiological research and gene therapy of cancers. Cancer Biol Ther 2015; 16:8-16. [PMID: 25455252 DOI: 10.4161/15384047.2014.986944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Carcinogenesis is etiologically associated with somatic mutations of critical genes. Recently, a number of somatic mutations and key molecules have been found to be involved in functional networks affecting cancer progression. Suitable animal models are required to validate cancer-promoting or -inhibiting capacities of these mutants and molecules. Sleeping Beauty transposon system consists of a transposon that carries gene(s) of interest and a transposase that recognizes, excises, and reinserts genes in given location of the genome. It can create both gain-of-function and loss-of-function mutations, thus being frequently chosen to investigate the etiological mechanisms and gene therapy for cancers in animal models. In this review, we summarized current advances of Sleeping Beauty transposon system in revealing molecular mechanism of cancers and improving gene therapy. Understanding molecular mechanisms by which driver mutations contribute to carcinogenesis and metastasis may pave the way for the development of innovative prophylactic and therapeutic strategies against malignant diseases.
Collapse
Key Words
- 7, 12-dimethylbenzanthracene/12-O-tetradecanoylphorbol-13-acetate
- Alb-Cre, Albumin promoter-Cre
- CAG promoter, CMV enhancer/chicken β-actin promoter
- CAR, chimeric antigen receptor
- CIS, common insertion site
- CMV, chimeric cytomegalovirus
- CRC, colorectal cancer
- Cre, cyclization recombination enzyme
- DDE, Asp, Asp, Glu
- DMBA/TPA
- DR, direct orientation
- Fah, fumarylacetoacetate hydrolase gene
- GWAS, gnome wide analysis study
- HBV, Hepatitis B Virus
- HBx, HBV X protein
- HCC, hepatocellular carcinoma
- IRs, inverted repeat sequences
- LsL, loxP-stop-loxP
- MPNSTs, malignant peripheral nerve sheath tumor
- MSCV, murine stem cell virus
- PAI, Pro, Ala, Ile
- PBMCs, peripheral blood mononuclear cells
- RED, Arg, Glu, Asp
- RosaSBaseLsL, Cre-inducible SBase allele
- Rtl1, Retrotransposon-like 1
- SB, Sleeping Beauty
- SBase, Sleeping Beauty transposase
- Sleeping Beauty transposon system
- StatinAE, angiostatin-endostatin fusion gene
- Trp53, transformation related protein 53
- animal model
- driver
- gene function
- gene therapy
- malignant diseases
- sgRNA, single guide RNA
- shp53, short hairpin RNA against the Trp53 gene
- somatic mutation
Collapse
Affiliation(s)
- Xiaomei Hou
- a Department of Epidemiology ; Second Military Medical University ; Shanghai , China
| | | | | | | | | |
Collapse
|
7
|
Becker CM, Oberoi RK, McFarren SJ, Muldoon DM, Pafundi DH, Pokorny JL, Brinkmann DH, Ohlfest JR, Sarkaria JN, Largaespada DA, Elmquist WF. Decreased affinity for efflux transporters increases brain penetrance and molecular targeting of a PI3K/mTOR inhibitor in a mouse model of glioblastoma. Neuro Oncol 2015; 17:1210-9. [PMID: 25972455 DOI: 10.1093/neuonc/nov081] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/08/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Targeting drug delivery to invasive glioma cells is a particularly difficult challenge because these cells lie behind an intact blood-brain barrier (BBB) that can be observed using multimodality imaging. BBB-associated efflux transporters such as P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) influence drug distribution to these cells and may negatively impact efficacy. To test the hypothesis that efflux transporters influence brain pharmacokinetics/pharmacodynamics of molecularly targeted agents in glioma treatment, we assessed region-specific penetrance and molecular-targeting capacity for a PI3K/mTOR kinase inhibitor that has high substrate affinity for efflux transporters (GDC-0980) and an analog (GNE-317) that was purposely designed to have reduced efflux. METHODS Brain tumor penetrance of GDC-0980 and GNE-317 was compared between FVB/n wild-type mice and Mdr1a/b(-/-)Bcrp(-/-) triple-knockout mice lacking P-gp and BCRP. C57B6/J mice bearing intracranial GL261 tumors were treated with GDC-0980, GNE-317, or vehicle to assess the targeted pharmacokinetic/pharmacodynamic effects in a glioblastoma model. RESULTS Animals treated with GNE-317 demonstrated 3-fold greater penetrance in tumor core, rim, and normal brain compared with animals dosed with GDC-0980. Increased brain penetrance correlated with decreased staining of activated p-Akt, p-S6, and p-4EBP1 effector proteins downstream of PI3K and mTOR. CONCLUSIONS GDC-0980 is subject to active efflux by P-gp and BCRP at the BBB, while brain penetrance of GNE-317 is independent of efflux, which translates into enhanced inhibition of PI3K/mTOR signaling. These data show that BBB efflux by P-gp and BCRP is therefore an important determinant in both brain penetrance and molecular targeting efficacy in the treatment of invasive glioma cells.
Collapse
Affiliation(s)
- Chani M Becker
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Rajneet K Oberoi
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Stephan J McFarren
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Daniel M Muldoon
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Deanna H Pafundi
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Jenny L Pokorny
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Debra H Brinkmann
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - John R Ohlfest
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - Jann N Sarkaria
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| | - William F Elmquist
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota (C.M.B., S.J.M., D.M.M., J.R.O., W.F.E); Brain Tumor Program, University of Minnesota, Minneapolis, Minnesota (C.M.B., R.K.O., S.J.M., D.M.M., J.R.O., D.A.L., W.F.E); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (R.K.O., W.F.E); Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota (J.R.O., D.A.L); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.H.P., J.L.P., D.H.B., J.N.S); Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
8
|
Hexum JK, Becker CM, Kempema AM, Ohlfest JR, Largaespada DA, Harki DA. Parthenolide prodrug LC-1 slows growth of intracranial glioma. Bioorg Med Chem Lett 2015; 25:2493-5. [PMID: 25978958 DOI: 10.1016/j.bmcl.2015.04.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 11/25/2022]
Abstract
LC-1 (also known as DMAPT or dimethylamino-parthenolide), a prodrug of parthenolide, was tested for anti-proliferative activity against glioma. LC-1 was found to have low micromolar cytotoxic activity against three glioma cell lines and was also found to be brain penetrant in healthy mice (2.1-3.0 brain-to-plasma ratio). In a syngeneic GL261 murine glioma model, LC-1 slowed tumor growth kinetics and extended the survival time of tumor-bearing mice in comparison to the vehicle control. Consequently, LC-1 represents a promising lead compound for further development as a glioma therapy.
Collapse
Affiliation(s)
- Joseph K Hexum
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chani M Becker
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aaron M Kempema
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Hybrid nonviral/viral vector systems for improved piggyBac DNA transposon in vivo delivery. Mol Ther 2015; 23:667-74. [PMID: 25557623 DOI: 10.1038/mt.2014.254] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 12/19/2014] [Indexed: 12/17/2022] Open
Abstract
The DNA transposon piggyBac is a potential therapeutic agent for multiple genetic diseases such as cystic fibrosis (CF). Recombinant piggyBac transposon and transposase are typically codelivered by plasmid transfection; however, plasmid delivery is inefficient in somatic cells in vivo and is a barrier to the therapeutic application of transposon-based vector systems. Here, we investigate the potential for hybrid piggyBac/viral vectors to transduce cells and support transposase-mediated genomic integration of the transposon. We tested both adenovirus (Ad) and adeno-associated virus (AAV) as transposon delivery vehicles. An Ad vector expressing hyperactive insect piggyBac transposase (iPB7) was codelivered. We show transposase-dependent transposition activity and mapped integrations in mammalian cells in vitro and in vivo from each viral vector platform. We also demonstrate efficient and persistent transgene expression following nasal delivery of piggyBac/viral vectors to mice. Furthermore, using piggyBac/Ad expressing Cystic Fibrosis transmembrane Conductance Regulator (CFTR), we show persistent correction of chloride current in well-differentiated primary cultures of human airway epithelial cells derived from CF patients. Combining the emerging technologies of DNA transposon-based vectors with well-studied adenoviral and AAV delivery provides new tools for in vivo gene transfer and presents an exciting opportunity to increase the delivery efficiency for therapeutic genes such as CFTR.
Collapse
|
10
|
ERG induces taxane resistance in castration-resistant prostate cancer. Nat Commun 2014; 5:5548. [PMID: 25420520 PMCID: PMC4244604 DOI: 10.1038/ncomms6548] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/09/2014] [Indexed: 12/14/2022] Open
Abstract
Taxanes are the only chemotherapies used to treat patients with metastatic castration-resistant prostate cancer (CRPC). Despite the initial efficacy of taxanes in treating CRPC, all patients ultimately fail due to the development of drug resistance. In this study, we show that ERG overexpression in in vitro and in vivo models of CRPC is associated with decreased sensitivity to taxanes. ERG affects several parameters of microtubule dynamics and inhibits effective drug-target engagement of docetaxel or cabazitaxel with tubulin. Finally, analysis of a cohort of 34 men with metastatic CRPC treated with docetaxel chemotherapy reveals that ERG-overexpressing prostate cancers have twice the chance of docetaxel resistance than ERG-negative cancers. Our data suggest that ERG plays a role beyond regulating gene expression and functions outside the nucleus to cooperate with tubulin towards taxane insensitivity. Determining ERG rearrangement status may aid in patient selection for docetaxel or cabazitaxel therapy and/or influence co-targeting approaches. Metastatic castration-resistant prostate cancer is treated with the microtubule-stabilizing drugs taxanes, but resistance ultimately develops. Here Galletti et al. show that ERG, a transcription factor commonly overexpressed in prostate cancers, confers taxane resistance by binding to soluble tubulin.
Collapse
|
11
|
Sharma N, Cai Y, Bak RO, Jakobsen MR, Schrøder LD, Mikkelsen JG. Efficient sleeping beauty DNA transposition from DNA minicircles. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e74. [PMID: 23443502 PMCID: PMC3586802 DOI: 10.1038/mtna.2013.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
DNA transposon-based vectors have emerged as new potential delivery tools in therapeutic gene transfer. Such vectors are now showing promise in hematopoietic stem cells and primary human T cells, and clinical trials with transposon-engineered cells are on the way. However, the use of plasmid DNA as a carrier of the vector raises safety concerns due to the undesirable administration of bacterial sequences. To optimize vectors based on the Sleeping Beauty (SB) DNA transposon for clinical use, we examine here SB transposition from DNA minicircles (MCs) devoid of the bacterial plasmid backbone. Potent DNA transposition, directed by the hyperactive SB100X transposase, is demonstrated from MC donors, and the stable transfection rate is significantly enhanced by expressing the SB100X transposase from MCs. The stable transfection rate is inversely related to the size of circular donor, suggesting that a MC-based SB transposition system benefits primarily from an increased cellular uptake and/or enhanced expression which can be observed with DNA MCs. DNA transposon and transposase MCs are easily produced, are favorable in size, do not carry irrelevant DNA, and are robust substrates for DNA transposition. In accordance, DNA MCs should become a standard source of DNA transposons not only in therapeutic settings but also in the daily use of the SB system.
Collapse
Affiliation(s)
- Nynne Sharma
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
12
|
Filipovic R, Santhosh Kumar S, Fiondella C, Loturco J. Increasing doublecortin expression promotes migration of human embryonic stem cell-derived neurons. Stem Cells 2013; 30:1852-62. [PMID: 22753232 DOI: 10.1002/stem.1162] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Human embryonic stem cell-derived neuronal progenitors (hNPs) provide a potential source for cellular replacement following neurodegenerative diseases. One of the greatest challenges for future neuron replacement therapies will be to control extensive cell proliferation and stimulate cell migration of transplanted cells. The doublecortin (DCX) gene encodes the protein DCX, a microtubule-associated protein essential for the migration of neurons in the human brain. In this study, we tested whether increasing the expression of DCX in hNPs would favorably alter their proliferation and migration. Migration and proliferation of hNPs was compared between hNPs expressing a bicistronic DCX/IRES-GFP transgene and those expressing a green fluorescent protein (GFP) transgene introduced by piggyBac-mediated transposition. The DCX-transfected hNPs showed a significant decrease in their proliferation and migrated significantly further on two different substrates, Matrigel and brain slices. Additionally, a dense network of nestin-positive (+) and vimentin+ fibers were found to extend from neurospheres transplanted onto brain slices, and this fiber growth was increased from neurospheres containing DCX-transfected hNPs. In summary, our results show that increased DCX expression inhibits proliferation and promotes migration of hNPs.
Collapse
Affiliation(s)
- Radmila Filipovic
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06268, USA.
| | | | | | | |
Collapse
|
13
|
Ohlfest JR, Andersen BM, Litterman AJ, Xia J, Pennell CA, Swier LE, Salazar AM, Olin MR. Vaccine injection site matters: qualitative and quantitative defects in CD8 T cells primed as a function of proximity to the tumor in a murine glioma model. THE JOURNAL OF IMMUNOLOGY 2012; 190:613-20. [PMID: 23248259 DOI: 10.4049/jimmunol.1201557] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Malignant gliomas are lethal brain tumors for which novel therapies are urgently needed. In animal models, vaccination with tumor-associated Ags efficiently primes T cells to clear gliomas. In clinical trials, cancer vaccines have been less effective at priming T cells and extending survival. Generalized immune suppression in the tumor draining lymph nodes has been documented in multiple cancers. However, a systematic analysis of how vaccination at various distances from the tumor (closest to farthest) has not been reported. We investigated how the injection site chosen for vaccination dictates CD8 T cell priming and survival in an OVA-transfected murine glioma model. Glioma-bearing mice were vaccinated with Poly:ICLC plus OVA protein in the neck, hind leg, or foreleg for drainage into the cervical, inguinal, or axillary lymph nodes, respectively. OVA-specific CD8 T cell number, TCR affinity, effector function, and infiltration into the brain decreased as the vaccination site approached the tumor. These effects were dependent on the presence of the tumor, because injection site did not appreciably affect CD8 T cell priming in tumor-free mice. Our data suggest the site of vaccination can greatly impact the effectiveness of cancer vaccines. Considering that previous and ongoing clinical trials have used a variety of injection sites, vaccination site is potentially a critical aspect of study design that is being overlooked.
Collapse
Affiliation(s)
- John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Di Matteo M, Belay E, Chuah MK, Vandendriessche T. Recent developments in transposon-mediated gene therapy. Expert Opin Biol Ther 2012; 12:841-58. [PMID: 22679910 DOI: 10.1517/14712598.2012.684875] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The continuous improvement of gene transfer technologies has broad implications for stem cell biology, gene discovery, and gene therapy. Although viral vectors are efficient gene delivery vehicles, their safety, immunogenicity and manufacturing challenges hamper clinical progress. In contrast, non-viral gene delivery systems are less immunogenic and easier to manufacture. AREAS COVERED In this review, we explore the emerging potential of transposons in gene and cell therapy. The safety, efficiency, and biology of novel hyperactive Sleeping Beauty (SB) and piggyBac (PB) transposon systems will be highlighted for ex vivo gene therapy in clinically relevant adult stem/progenitor cells, particularly hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), myoblasts, and induced pluripotent stem (iPS) cells. Moreover, efforts toward in vivo transposon-based gene therapy will be discussed. EXPERT OPINION The latest generation SB and PB transposons currently represent some of the most attractive systems for stable non-viral genetic modification of primary cells, particularly adult stem cells. This paves the way toward the use of transposons as a non-viral gene therapy approach to correct hereditary disorders including those that affect the hematopoietic system. The development of targeted integration into "safe harbor" genetic loci may further improve their safety profile.
Collapse
Affiliation(s)
- Mario Di Matteo
- Free University of Brussels, Division of Gene Therapy & Regenerative Medicine, Laarbeeklaan 103, B-1090 Brussels, Belgium
| | | | | | | |
Collapse
|
15
|
Olin MR, Oh S, Roy S, Peterson PK, Molitor T. Morphine induces splenocyte trafficking into the CNS. J Neuroimmune Pharmacol 2012; 7:436-43. [PMID: 21858458 PMCID: PMC3570027 DOI: 10.1007/s11481-011-9307-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 08/03/2011] [Indexed: 10/17/2022]
Abstract
Opioids significantly alter functional responses of lymphocytes following activation. The opiate Morphine, alters the Th1 to Th2 response and modulates functional responses such as cytolytic activity and T-cell proliferation. Although there has been extensive research involving morphine's effects on lymphocytes, little is known about the effects morphine has on lymphocyte trafficking. The objective of the study was to use in vivo bioluminescent imaging to determine morphine's effect on the trafficking pattern of splenocytes systemically and into the CNS either in a naïve state or following a neuroinflammatory stimulus. A neuroinflammatory response was induced by intracerebrally administering a DNA IFN-γ DNA plasmid into morphine-dependent or placebo wildtype mice. Mice with or without a neurostimulus received adoptively transferred firefly luciferase transgenic splenocytes and imaged. Morphine dependence significantly altered the inherent ability of splenocytes to traffic into the spleen, and lead to non-directed chaotic trafficking throughout the animal, including into the CNS. The morphine-mediated effects on trafficking were blocked by the antagonist naltrexone. Morphine dependence intensified splenocyte infiltration into the CNS following neuroinflammation induced by IFN-γ gene transfer. The study precented determined that morphine severely altered the ability of non-activated splenocytes to home to the spleen, inducing extrasplenic trafficking thoughout the animal. In addition to altering the ability of naive splenocyte to traffic to the spleen, this study demonstrated that morphine profoundly exacerbated lymphocyte infiltration into the CNS following a neurostimulus.
Collapse
Affiliation(s)
- Michael R Olin
- University of Minnesota. Department of pediatrics, Minneapolis, MN 55455, 612-616-2246
| | - Seunguk Oh
- University of Minnesota. McGuire Translational Research Facility, Minneapolis, MN 55455, 612-624-1195
| | - Sabita Roy
- University of Minnesota, Department of Pharmacology and Surgery, Minneapolis, MN 55455, 612-624-4615
| | - Phillip K Peterson
- University of Minnesota Medical School. Center for Infectious Diseases and Translational Research, Minneapolis, Minnesota 55415, 612 626-9923
| | - Thomas Molitor
- University of Minnesota, Department of Veterinary Population Medicine, 225 Veterinary Teaching Hospital, 1365 Gortner Ave., St. Paul, MN 55108, 612-625-5295
| |
Collapse
|
16
|
Ivics Z, Izsvák Z. Nonviral gene delivery with the sleeping beauty transposon system. Hum Gene Ther 2012; 22:1043-51. [PMID: 21867398 DOI: 10.1089/hum.2011.143] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Effective gene therapy requires robust delivery of therapeutic genes into relevant target cells, long-term gene expression, and minimal risks of secondary effects. Nonviral gene transfer approaches typically result in only short-lived transgene expression in primary cells, because of the lack of nuclear maintenance of the vector over several rounds of cell division. The development of efficient and safe nonviral vectors armed with an integrating feature would thus greatly facilitate clinical gene therapy studies. The latest generation transposon technology based on the Sleeping Beauty (SB) transposon may potentially overcome some of these limitations. SB was shown to provide efficient stable gene transfer and sustained transgene expression in primary cell types, including human hematopoietic progenitors, mesenchymal stem cells, muscle stem/progenitor cells (myoblasts), induced pluripotent stem cells, and T cells. These cells are relevant targets for stem cell biology, regenerative medicine, and gene- and cell-based therapies of complex genetic diseases. Moreover, the first-in-human clinical trial has been launched to use redirected T cells engineered with SB for gene therapy of B cell lymphoma. We discuss aspects of cellular delivery of the SB transposon system, transgene expression provided by integrated transposon vectors, target site selection of the transposon vectors, and potential risks associated with random genomic insertion.
Collapse
Affiliation(s)
- Zoltán Ivics
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.
| | | |
Collapse
|
17
|
Swierczek M, Izsvák Z, Ivics Z. The Sleeping Beauty transposon system for clinical applications. Expert Opin Biol Ther 2011; 12:139-53. [PMID: 22176302 DOI: 10.1517/14712598.2012.642358] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Extensive efforts have been made to establish efficient and safe gene delivery protocols that could meet demanding expectations of a successful gene therapy. The Sleeping Beauty (SB) transposon system combines simplicity and inexpensive manufacture offered by plasmid-based vector formulation with integrative features exhibited by some viral vectors. Activated after over ten million years of silent genomic existence, the SB transposable element entered the 21st century as a potent technology for a broad range of applications in genome engineering, including gene therapy. Beneficially for gene therapy purposes, the SB system has been demonstrated to enable persistent expression of therapeutic genes followed by restoration of homeostasis in a variety of disease models. Importantly, this non-viral gene delivery vehicle is postulated to constitute a relatively safe vector system, because it lacks a preference for inserting into transcription units and their upstream regulatory regions, thereby minimizing genotoxic risks that might be associated with vector integration. Further evolution and wide, comprehensive preclinical testing of the SB transposon system in the context of several disease models is expected to further refine this valuable technology matched by enhanced biosafety towards disease treatment.
Collapse
Affiliation(s)
- Marta Swierczek
- Paul Ehrlich Institute, Paul Ehrlich Strasse 51-59, D-63225 Langen, Germany
| | | | | |
Collapse
|
18
|
Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AKM, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2011; 11:155-80. [PMID: 21453286 DOI: 10.2174/156652311795684722] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Maria G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xiong Z, Ohlfest JR. Topical imiquimod has therapeutic and immunomodulatory effects against intracranial tumors. J Immunother 2011; 34:264-9. [PMID: 21389872 DOI: 10.1097/cji.0b013e318209eed4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Topical imiquimod cream (trade name: Aldara) is a Toll-like receptor (TLR) 7 agonist that is approved for the treatment of cutaneous tumors. Aldara is also used as a vaccine adjuvant in clinical trials in patients with glioma and other tumors. The main mechanism of action ascribed to Aldara has been the local activation of TLR7(+) cells near the application site. Here we report the unexpected finding that Aldara has therapeutic and immunomodulatory activity as a single agent in mice bearing intracranial tumors. Repeated administration of Aldara onto the skin significantly increased the survival of mice bearing intracranial GL261 glioma and EMT6 breast carcinoma. Aldara treatment was associated with a reduction in the number CD4(+)Foxp3(+) regulatory T cells in the blood and brain tumor site. Mice treated with Aldara exhibited a generalized lymphopenia in the blood amidst an increase in brain tumor infiltrating CD4(+) and CD8(+) T cells and dendritic cells. Brain-infiltrating CD8(+) T cells were tumor reactive as demonstrated by degranulation and interferon-γ secretion in a GL261-dependent manner. In addition, soluble imiquimod directly inhibited the proliferation of GL261 cells in a TLR7-independent manner. This is the first report demonstrating that topical application of imiquimod can enhance T-cell responses to intracranial tumors as a single agent. The results must be interpreted with caution considering anatomical and biological differences between mice and humans. Nevertheless, Aldara that is being used as a vaccine adjuvant in clinical trials may have direct antitumor effects that are independent of exogenous antigen. Further studies in humans are warranted.
Collapse
Affiliation(s)
- Zhengming Xiong
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
20
|
Belur LR, Podetz-Pedersen KM, Sorenson BS, Hsu AH, Parker JB, Carlson CS, Saltzman DA, Ramakrishnan S, McIvor RS. Inhibition of angiogenesis and suppression of colorectal cancer metastatic to the liver using the Sleeping Beauty Transposon System. Mol Cancer 2011; 10:14. [PMID: 21310067 PMCID: PMC3045363 DOI: 10.1186/1476-4598-10-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 02/10/2011] [Indexed: 12/18/2022] Open
Abstract
Background Metastatic colon cancer is one of the leading causes of cancer-related death worldwide, with disease progression and metastatic spread being closely associated with angiogenesis. We investigated whether an antiangiogenic gene transfer approach using the Sleeping Beauty (SB) transposon system could be used to inhibit growth of colorectal tumors metastatic to the liver. Results Liver CT26 tumor-bearing mice were hydrodynamically injected with different doses of a plasmid containing a transposon encoding an angiostatin-endostatin fusion gene (Statin AE) along with varying amounts of SB transposase-encoding plasmid. Animals that were injected with a low dose (10 μg) of Statin AE transposon plasmid showed a significant decrease in tumor formation only when co-injected with SB transposase-encoding plasmid, while for animals injected with a higher dose (25 μg) of Statin AE transposon, co-injection of SB transposase-encoding plasmid did not significantly affect tumor load. For animals injected with 10 μg Statin AE transposon plasmid, the number of tumor nodules was inversely proportional to the amount of co-injected SB plasmid. Suppression of metastases was further evident in histological analyses, in which untreated animals showed higher levels of tumor cell proliferation and tumor vascularization than animals treated with low dose transposon plasmid. Conclusion These results demonstrate that hepatic colorectal metastases can be reduced using antiangiogenic transposons, and provide evidence for the importance of the transposition process in mediating suppression of these tumors.
Collapse
Affiliation(s)
- Lalitha R Belur
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Maes W, Van Gool SW. Experimental immunotherapy for malignant glioma: lessons from two decades of research in the GL261 model. Cancer Immunol Immunother 2011; 60:153-60. [PMID: 21120655 PMCID: PMC11028904 DOI: 10.1007/s00262-010-0946-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 11/05/2010] [Indexed: 02/04/2023]
Abstract
Nearly twenty years of experimental immunotherapy for malignant glioma yielded important insights in the mechanisms governing glioma immunology. Still considered promising, it is clear that immunotherapy does not on its own represent the magic bullet in glioma therapy. In this review, we summarize the major immunotherapeutic achievements in the mouse GL261 glioma model, which has emerged as the gold standard syngeneic model for experimental glioma therapy. Gene therapy, monoclonal antibody treatment, cytokine therapy, cell transfer strategies and dendritic cell therapy were hereby considered. Apart from the considerable progress made in understanding glioma immunology in this model, we also addressed its most pertinent issues and shortcomings. Despite these, the GL261 model will remain indispensable in glioma research since it is a fast, highly reproducible and easy-to-establish model system.
Collapse
Affiliation(s)
- Wim Maes
- Laboratory of Experimental Immunology (WM and SWVG), Pediatric Hemato-oncology (SWVG), University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Stefaan W. Van Gool
- Laboratory of Experimental Immunology (WM and SWVG), Pediatric Hemato-oncology (SWVG), University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
22
|
Izsvák Z, Hackett PB, Cooper LJN, Ivics Z. Translating Sleeping Beauty transposition into cellular therapies: victories and challenges. Bioessays 2010; 32:756-67. [PMID: 20652893 DOI: 10.1002/bies.201000027] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent results confirm that long-term expression of therapeutic transgenes can be achieved by using a transposon-based system in primary stem cells and in vivo. Transposable elements are natural DNA transfer vehicles that are capable of efficient genomic insertion. The latest generation, Sleeping Beauty transposon-based hyperactive vector (SB100X), is able to address the basic problem of non-viral approaches - that is, low efficiency of stable gene transfer. The combination of transposon-based non-viral gene transfer with the latest improvements of non-viral delivery techniques could provide a long-term therapeutic effect without compromising biosafety. The new challenges of pre-clinical research will focus on further refinement of the technology in large animal models and improving the safety profile of SB vectors by target-selected transgene integration into genomic "safe harbors." The first clinical application of the SB system will help to validate the safety of this approach.
Collapse
Affiliation(s)
- Zsuzsanna Izsvák
- Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.
| | | | | | | |
Collapse
|
23
|
Olin MR, Andersen BM, Zellmer DM, Grogan PT, Popescu FE, Xiong Z, Forster CL, Seiler C, SantaCruz KS, Chen W, Blazar BR, Ohlfest JR. Superior efficacy of tumor cell vaccines grown in physiologic oxygen. Clin Cancer Res 2010; 16:4800-8. [PMID: 20858839 DOI: 10.1158/1078-0432.ccr-10-1572] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Atmospheric oxygen (∼20% O(2)) has been the universal condition employed to culture tumor cells used as vaccine antigen. We tested the hypothesis that reducing oxygen tension would increase the efficacy of tumor cell lysate vaccines. EXPERIMENTAL DESIGN GL261 glioma cells and EMT6 breast carcinoma cells were grown in 5% or 20% O(2). Syngeneic tumor-bearing mice were vaccinated with these tumor cell lysates mixed with CpG oligodeoxynucleotides as an adjuvant. Tumor infiltrating T cells and apoptotic GL261 cells were quantified by immunohistochemistry. Tumor-reactive immunoglobulin was detected by Western blot. Ovalbumin and gp100-derived peptides were mixed with GL261 lysates as marker antigens to detect changes in presentation of exogenous antigen on MHC class I in vitro, and in vivo following adoptive transfer of gp100-specific CD8(+) T cells. RESULTS Mice bearing orthotopic glioma and breast carcinoma survived significantly longer when vaccinated with 5% O(2) lysates. Antigen-specific CTL activation was significantly enhanced following stimulation with lysates derived from GL261 cells grown in 5% O(2) versus 20% O(2) through a mechanism that involved enhanced cross-presentation of exogenous antigen on MHC I. Vaccination with 5% O(2) GL261 cell lysates caused a significant increase in CTL proliferation, tumoricidal function, and trafficking into brain tumor sites, whereas 20% O(2) lysate vaccines predominantly evoked an antibody response. CONCLUSIONS Tissue culture oxygen functions as an "immunologic switch" by dictating the cellular and humoral immune responses elicited by tumor cell lysates. These results have profound implications for cancer vaccines that utilize tumor cells as the source of antigen.
Collapse
Affiliation(s)
- Michael R Olin
- Departments of Pediatrics, University of Minnesota, Minneapolis, 55455, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pluhar GE, Grogan PT, Seiler C, Goulart M, Santacruz KS, Carlson C, Chen W, Olin MR, Lowenstein PR, Castro MG, Haines SJ, Ohlfest JR. Anti-tumor immune response correlates with neurological symptoms in a dog with spontaneous astrocytoma treated by gene and vaccine therapy. Vaccine 2010; 28:3371-8. [PMID: 20197146 DOI: 10.1016/j.vaccine.2010.02.082] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 01/28/2010] [Accepted: 02/15/2010] [Indexed: 11/29/2022]
Abstract
Gene therapy and vaccination have been tested in malignant glioma patients with modest, albeit encouraging results. The combination of these therapies has demonstrated synergistic efficacy in murine models but has not been reported in large animals. Gemistocytic astrocytoma (GemA) is a low-grade glioma that typically progresses to lethal malignancy despite conventional therapies. Until now there has been no useful animal model of GemA. Here we report the treatment of a dog with spontaneous GemA using the combination of surgery, intracavitary adenoviral interferon gamma (IFNgamma) gene transfer, and vaccination with glioma cell lysates mixed with CpG oligodeoxynucleotides. Surgical tumor debulking and delivery of Ad-IFNgamma into the resection cavity were performed. Autologous tumor cells grew slowly in culture, necessitating vaccination with allogeneic tumor lysate in four of the five vaccinations. Transient left-sided blindness and hemiparesis occurred following the fourth and fifth vaccinations. These neurological symptoms correlated with a peak in the levels of tumor-reactive IgG and CD8(+) T cells measured in the blood. All symptoms resolved and this dog remains tumor-free over 450 days following surgery. This case report preliminarily demonstrates the feasibility of treating dogs with spontaneous glioma using immune-based therapy and warrants further study using this therapeutic approach.
Collapse
Affiliation(s)
- G Elizabeth Pluhar
- Department of Veterinary Animal Clinical Sciences, University of Minnesota, St. Paul, MN 55108, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
NAKANISHI H, HIGUCHI Y, KAWAKAMI S, YAMASHITA F, HASHIDA M. Development and Therapeutic Application of Transposon-based Vectors. YAKUGAKU ZASSHI 2009; 129:1433-43. [DOI: 10.1248/yakushi.129.1433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hideyuki NAKANISHI
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Yuriko HIGUCHI
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Shigeru KAWAKAMI
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Fumiyoshi YAMASHITA
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Mitsuru HASHIDA
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
- Institute for Integrated Cell-Material Sciences, Kyoto University (iCeMS)
| |
Collapse
|
27
|
Wiesner SM, Decker SA, Larson JD, Ericson K, Forster C, Gallardo JL, Long C, Demorest ZL, Zamora EA, Low WC, SantaCruz K, Largaespada DA, Ohlfest JR. De novo induction of genetically engineered brain tumors in mice using plasmid DNA. Cancer Res 2009; 69:431-9. [PMID: 19147555 DOI: 10.1158/0008-5472.can-08-1800] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Spontaneous mouse models of cancer show promise to more accurately recapitulate human disease and predict clinical efficacy. Transgenic mice or viral vectors have been required to generate spontaneous models of glioma, a lethal brain tumor, because nonviral gene transfer is typically transient. To overcome this constraint, we used the Sleeping Beauty transposable element to achieve chromosomal integration of human oncogenes into endogenous brain cells of immunocompetent mice. Genetically engineered, spontaneous brain tumors were induced with plasmid DNA in a matter of weeks in three separate mouse strains. The phenotype of tumors was influenced by the combination of oncogenes delivered, resembling human astrocytoma or glioblastoma in the majority of cases. At least five different genes can be cotransfected simultaneously including reporters, allowing measurement of tumor viability by in vivo imaging. This model can accelerate brain tumor research in a variety of ways such as generation of "humanized" models for high throughput drug screening and candidate gene validation with exceptional speed and flexibility.
Collapse
Affiliation(s)
- Stephen M Wiesner
- Departments of Pediatrics, Center for Allied Health Programs, Laboratory Medicine and Pathology, and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ohlfest JR, Ivics Z, Izsvák Z. Transposable elements as plasmid-based vectors for long-term gene transfer into tumors. Methods Mol Biol 2009; 542:105-116. [PMID: 19565898 DOI: 10.1007/978-1-59745-561-9_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A primary limitation to using nonviral vectors for cancer gene therapy is transient expression of the therapeutic gene. Even when the ultimate goal is tumor cell death, a minimum threshold of gene expression is required to kill tumor cells by direct or indirect mechanisms. It has been shown that transposable elements can significantly enhance the duration of gene expression when plasmid DNA vectors are used to transfect tumor or tumor-associated stroma. Much like a retrovirus, transposon-based plasmid vectors achieve integration into the genome, and thereby sustain transgene expression, which is especially important in actively mitotic cells such as tumor cells. Herein we briefly discuss the different transposons available for gene therapy applications, and provide a detailed protocol for nonviral transposon-based gene delivery to solid experimental tumors in mice.
Collapse
Affiliation(s)
- John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | | | |
Collapse
|
29
|
Xiong Z, Gharagozlou S, Vengco I, Chen W, Ohlfest JR. Effective CpG immunotherapy of breast carcinoma prevents but fails to eradicate established brain metastasis. Clin Cancer Res 2008; 14:5484-93. [PMID: 18765540 DOI: 10.1158/1078-0432.ccr-07-4139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Breast cancer patients with brain metastasis have a dismal prognosis. We determined the ability of immunostimulatory CpG oligodeoxynucleotides (ODN) to treat or prevent brain metastasis in a mouse model. EXPERIMENTAL DESIGN Mice bearing orthotopic breast carcinoma with or without concurrent i.c. tumors were treated by injections of CpG ODN at the primary tumor. Immunologic memory was tested by tumor rechallenge and immune responses were assessed by flow cytometry, delayed-type hypersensitivity, and CTL assays. RESULTS Orthotopic tumors regressed in treated mice regardless of whether concurrent i.c. disease was present. In mice bearing only orthotopic tumors, CpG ODN rendered 50% tumor-free and they rejected tumor rechallenge in breast and brain. In mice with concurrent i.c. disease, there was no difference in brain tumor growth compared with saline controls, despite regression of the primary tumor. Flow cytometry revealed that treated mice that died from i.c. disease exhibited a significant increase in brain-infiltrating T and natural killer cells relative to saline controls. CTLs from these mice were able to kill tumor in vitro and extend survival of naive mice bearing less-established brain tumors by adoptive transfer. CONCLUSIONS The lack of survival benefit in mice with appreciable brain metastasis was not explained by a deficit in lymphocyte trafficking or function because CTLs from these mice killed tumor and inhibited microscopic brain metastasis by adoptive transfer. These results indicate that CpG ODN might be beneficial as a preventative adjuvant to initial therapy preceding brain metastasis or to inhibit progression of microscopic brain metastases.
Collapse
Affiliation(s)
- Zhengming Xiong
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
30
|
Wu A, Oh S, Wiesner SM, Ericson K, Chen L, Hall WA, Champoux PE, Low WC, Ohlfest JR. Persistence of CD133+ cells in human and mouse glioma cell lines: detailed characterization of GL261 glioma cells with cancer stem cell-like properties. Stem Cells Dev 2008; 17:173-84. [PMID: 18271701 DOI: 10.1089/scd.2007.0133] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The concept of cancer stem cells suggests that there are malignant stem-like cells within a tumor that are responsible for tumor renewal and resistance to cytotoxic therapies. Studies have identified glioma stem-like cells that extrude Hoechst 33342 dye, representing a double-negative "side population" (SP) thought to be selectively resistant to drug therapy. A CD133+ stem cell-like subpopulation has been isolated from a human glioma that was enriched for tumor-initiating cells. It is unknown whether CD133+ cells with similar phenotype persist in established glioma cell lines, or if CD133 is a marker of glioma stem-like cells in rodents. We investigated whether CD133+ and SP cells existed in the GL261 cell line, a syngeneic mouse glioma model that is widely used for preclinical and translational research. Intracerebral injection of less than 100 CD133+ GL261 cells formed tumors, whereas it required 10,000 CD133(-) cells to initiate a tumor. CD133+ GL261 cells expressed nestin, formed tumor spheres with high frequency, and differentiated into glial and neuronal-like cells. Similar to GL261, seven human glioma cell lines analyzed also contained a rare CD133+ population. Surprisingly, we found that CD133+ GL261 cells did not reside in the SP, nor did the majority ( approximately 94%) of CD133+ human glioma cells. These results demonstrate that the expression of CD133 in murine glioma cells is associated with enhanced tumorigenicity and a stem-like phenotype. This study also reveals a previously unrecognized level of heterogeneity in glioma cell lines, exposing several populations of cells that have characteristics of cancer stem cells.
Collapse
Affiliation(s)
- Anhua Wu
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
In vivo vaccination with tumor cell lysate plus CpG oligodeoxynucleotides eradicates murine glioblastoma. J Immunother 2008; 30:789-97. [PMID: 18049330 DOI: 10.1097/cji.0b013e318155a0f6] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dendritic cell (DC) vaccines have shown antitumor activity in experimental glioma models and in human glioma patients. The typical approach has been to generate the vaccine ex vivo, by pulsing DCs with tumor lysate or peptides, then administering the DCs back into the patient. This process requires significant expertise and expenses in DC generation. Immature DCs which present antigens to T cells in the absence of appropriate costimulatory signals can lead to induction of immune tolerance. Recent studies have shown that coadministration of toll-like receptor 9 agonists, CpG oligodeoxynucleotides, can promote DC vaccines to break immune tolerance to tumor antigens. We investigated the therapeutic efficacy of in vivo DC activation, by directly administering glioma cell lysate with CpG oligodeoxynucleotides (CpG/lysate), in glioma-bearing mice. Subcutaneous vaccination with CpG/lysate induced a significant increase (P<0.05) in the number of total T cells and activated DCs in lymph nodes draining the vaccination site as compared to mice treated with CpG or tumor lysate alone. Mice vaccinated with CpG/lysate exhibited over 2 times greater median survival than mice in the control groups (P<0.05). Up to 55% of mice vaccinated with CpG/lysate were rendered tumor-free as assessed by survival and bioluminescent imaging. Splenocytes taken from mice vaccinated with CpG/lysate elaborated significantly more IFN-gamma production and displayed greater tumor cell lysis activity compared with the control groups (P<0.05). These results suggest direct vaccination with CpG/lysate provides an alternative and effective approach to induce host antitumor immunity and warrants clinical investigation in the immunotherapy of cancer.
Collapse
|