1
|
Zhang WW, Li L, Li D, Liu J, Li X, Li W, Xu X, Zhang MJ, Chandler LA, Lin H, Hu A, Xu W, Lam DMK. The First Approved Gene Therapy Product for Cancer Ad-p53 (Gendicine): 12 Years in the Clinic. Hum Gene Ther 2019; 29:160-179. [PMID: 29338444 DOI: 10.1089/hum.2017.218] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gendicine (recombinant human p53 adenovirus), developed by Shenzhen SiBiono GeneTech Co. Ltd., was approved in 2003 by the China Food and Drug Administration (CFDA) as a first-in-class gene therapy product to treat head and neck cancer, and entered the commercial market in 2004. Gendicine is a biological therapy that is delivered via minimally invasive intratumoral injection, as well as by intracavity or intravascular infusion. The wild-type (wt) p53 protein expressed by Gendicine-transduced cells is a tumor suppressor that is activated by cellular stress, and mediates cell-cycle arrest and DNA repair, or induces apoptosis, senescence, and/or autophagy, depending upon cellular stress conditions. Based on 12 years of commercial use in >30,000 patients, and >30 published clinical studies, Gendicine has exhibited an exemplary safety record, and when combined with chemotherapy and radiotherapy has demonstrated significantly higher response rates than for standard therapies alone. In addition to head and neck cancer, Gendicine has been successfully applied to treat various other cancer types and different stages of disease. Thirteen published studies that include long-term survival data showed that Gendicine combination regimens yield progression-free survival times that are significantly longer than standard therapies alone. Although the p53 gene is mutated in >50% of all human cancers, p53 mutation status did not significantly influence efficacy outcomes and long-term survival rate for Ad-p53-treated patients. To date, Shenzhen SiBiono GeneTech has manufactured 41 batches of Gendicine in compliance with CFDA QC/QA requirements, and 169,571 vials (1.0 × 1012 vector particles per vial) have been used to treat patients. No serious adverse events have been reported, except for vector-associated transient fever, which occurred in 50-60% of patients and persisted for only a few hours. The manufacturing accomplishments and clinical experience with Gendicine, as well as the understanding of its cellular mechanisms of action and implications, could provide valuable insights for the international gene therapy community and add valuable data to promote further developments and advancements in the gene therapy field.
Collapse
Affiliation(s)
- Wei-Wei Zhang
- 1 LifeTech Biosciences Group, Hong Kong .,2 Angionetics, Inc., San Diego, California
| | - Longjiang Li
- 3 State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dinggang Li
- 4 Beijing Haidian Hospital Center for Cancer Gene Therapy, Beijing, China
| | - Jiliang Liu
- 5 Shenzhen Hengsheng Hospital Cancer Center, Shenzhen, China
| | - Xiuqin Li
- 6 China Medical University Shengjing Hospital Department of Obstetrics and Gynecology, Shenyang, China
| | - Wei Li
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Xiaolong Xu
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Michael J Zhang
- 8 Department of Medicine University of Minnesota Medical School, Minneapolis, Minnesota
| | | | - Hong Lin
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Aiguo Hu
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Wei Xu
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | | |
Collapse
|
2
|
Buduru S, Zimta AA, Ciocan C, Braicu C, Dudea D, Irimie AI, Berindan-Neagoe I. RNA interference: new mechanistic and biochemical insights with application in oral cancer therapy. Int J Nanomedicine 2018; 13:3397-3409. [PMID: 29922059 PMCID: PMC5997132 DOI: 10.2147/ijn.s167383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Over the last few decades, the incidence of oral cancer has gradually increased, due to the negative influence of environmental factors and also abnormalities within the genome. The main issues in oral cancer treatment consist in surpassing resistance and recurrence. However, continuous discovery of altered signaling pathways in these tumors provides valuable information for the identification of novel gene candidates targeted in personalized therapy. RNA interference (RNAi) is a natural mechanism that involves small interfering RNA (siRNA); this can be exploited in biomedical research by using natural or synthetic constructs for activation of the mechanism. Synthetic siRNA transcripts were developed as a versatile class of molecular tools that have a diverse range of programmable roles, being involved in the regulation of several biological processes, thereby providing the perspective of an alternative option to classical treatment. In this review, we summarize the latest information related to the application of siRNA in oral malignancy together with molecular aspects of the technology and also the perspective upon the delivery system. Also, the emergence of newer technologies such as clustered regularly interspaced short palindromic repeats/Cas9 or transcription activator-like effector nucleases in comparison with the RNAi approach is discussed in this paper.
Collapse
Affiliation(s)
- Smaranda Buduru
- Department of Prosthetics and Dental Materials, Faculty of Dental Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- MEDFUTURE – Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Ciocan
- MEDFUTURE – Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Dudea
- Department of Prosthetic Dentistry and Dental Materials, Division Dental Propaedeutic, Aesthetic, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Iulia Irimie
- Department of Prosthetic Dentistry and Dental Materials, Division Dental Propaedeutic, Aesthetic, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- MEDFUTURE – Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Research Center for Functional Genomics and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof Dr Ion Chiricuta”, Cluj-Napoca, Romania
| |
Collapse
|
3
|
Combination of p53-DC vaccine and rAd-p53 gene therapy induced CTLs cytotoxic against p53-deleted human prostate cancer cells in vitro. Cancer Gene Ther 2017. [PMID: 28621316 DOI: 10.1038/cgt.2017.21] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recently, the US FDA approved sipuleucel-T, which is composed of autologous DCs stimulated with a recombinant fusion protein of prostatic acid phosphatase (PAP) and granulocyte-macrophage colony-stimulating factor (GM-CSF), as the first immunotherapeutic agent for metastatic castration resistant prostate cancer (mCRPC). However, sipuleucel-T demonstrated only modest efficacy in mCPRC patients. Researchers are now investigating the potential of p53 protein as a tumor-associated antigen (TAA) loaded in DC-based cancer vaccine. Approximately half of all tumors overexpress p53, and up to 20% of prostate cancer cells overexpresses p53. In this study, we evaluated the feasibility of combining p53-DC vaccine and rAd-p53 gene therapy, using the p53-overexpressing and non-expressing prostate cancer cells in vitro. We successfully generated the p53-DC vaccine by culturing autologous DCs infected with rAd-p53. This p53-DC vaccine can differentiate CTLs specifically cytotoxic to p53-overexpressing prostate cancer cells. In addition, rAd-p53 infection can induce overexpression of p53 and thus the cytotoxicity of CTLs differentiated by the p53-DC vaccine in p53 non-expressing prostate cancer cells. These findings suggest that this combination therapy using p53-DC vaccine and rAd-p53 gene therapy together may represent a new paradigm for the treatment of mCRPC.
Collapse
|
4
|
Chakroun RW, Zhang P, Lin R, Schiapparelli P, Quinones-Hinojosa A, Cui H. Nanotherapeutic systems for local treatment of brain tumors. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [PMID: 28544801 DOI: 10.1002/wnan.1479] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 04/14/2017] [Accepted: 04/18/2017] [Indexed: 12/31/2022]
Abstract
Malignant brain tumor, including the most common type glioblastoma, are histologically heterogeneous and invasive tumors known as the most devastating neoplasms with high morbidity and mortality. Despite multimodal treatment including surgery, radiotherapy, chemotherapy, and immunotherapy, the disease inevitably recurs and is fatal. This lack of curative options has motivated researchers to explore new treatment strategies and to develop new drug delivery systems (DDSs); however, the unique anatomical, physiological, and pathological features of brain tumors greatly limit the effectiveness of conventional chemotherapy. In this context, we review the recent progress in the development of nanoparticle-based DDSs aiming to address the key challenges in transporting sufficient amount of therapeutic agents into the brain tumor areas while minimizing the potential side effects. We first provide an overview of the standard treatments currently used in the clinic for the management of brain cancers, discussing the effectiveness and limitations of each therapy. We then provide an in-depth review of nanotherapeutic systems that are intended to bypass the blood-brain barrier, overcome multidrug resistance, infiltrate larger tumorous tissue areas, and/or release therapeutic agents in a controlled manner. WIREs Nanomed Nanobiotechnol 2018, 10:e1479. doi: 10.1002/wnan.1479 This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Rami Walid Chakroun
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Pengcheng Zhang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ran Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | | | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
5
|
Moving oncolytic viruses into the clinic: clinical-grade production, purification, and characterization of diverse oncolytic viruses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16018. [PMID: 27088104 PMCID: PMC4822647 DOI: 10.1038/mtm.2016.18] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/07/2016] [Accepted: 01/12/2016] [Indexed: 12/13/2022]
Abstract
Oncolytic viruses (OVs) are unique anticancer agents based on their pleotropic modes of action, which include, besides viral tumor cell lysis, activation of antitumor immunity. A panel of diverse viruses, often genetically engineered, has advanced to clinical investigation, including phase 3 studies. This diversity of virotherapeutics not only offers interesting opportunities for the implementation of different therapeutic regimens but also poses challenges for clinical translation. Thus, manufacturing processes and regulatory approval paths need to be established for each OV individually. This review provides an overview of clinical-grade manufacturing procedures for OVs using six virus families as examples, and key challenges are discussed individually. For example, different virus features with respect to particle size, presence/absence of an envelope, and host species imply specific requirements for measures to ensure sterility, for handling, and for determination of appropriate animal models for toxicity testing, respectively. On the other hand, optimization of serum-free culture conditions, increasing virus yields, development of scalable purification strategies, and formulations guaranteeing long-term stability are challenges common to several if not all OVs. In light of the recent marketing approval of the first OV in the Western world, strategies for further upscaling OV manufacturing and optimizing product characterization will receive increasing attention.
Collapse
|
6
|
Zhang S, Gera L, Mamouni K, Li X, Chen Z, Kucuk O, Wu D. Inhibition of skeletal growth of human prostate cancer by the combination of docetaxel and BKM1644: an aminobisphosphonate derivative. Oncotarget 2016; 7:27489-98. [PMID: 27050371 PMCID: PMC5053666 DOI: 10.18632/oncotarget.8481] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/18/2016] [Indexed: 12/25/2022] Open
Abstract
Bone metastasis is a major cause of prostate cancer (PCa) morbidity and mortality. Despite some success in transiently controlling clinical symptoms with docetaxel-based therapy, PCa patients become docetaxel-resistant and inevitably progress with no cure. We synthesized an acyl-tyrosine bisphosphonate amide derivative, BKM1644, with the intent of targeting bone metastatic PCa and enhancing docetaxel's efficacy. BKM1644 exhibits potent anti-cancer activity in the NCI-60 panel and effectively inhibits the proliferation of metastatic, castration-resistant PCa (mCRPC) cells, with IC50 ranging between 2.1 μM and 6.3 μM. Significantly, BKM1644 sensitizes mCRPC cells to docetaxel treatment. Mice with pre-established C4-2 tumors in the tibia show a marked decrease in serum prostate-specific antigen (control: 173.72 ± 37.52 ng/ml, combined treatment: 64.45 ± 22.19 ng/ml; p < 0.0001) and much improved bone architecture after treatment with the combined regimen. Mechanistic studies found that docetaxel temporarily but significantly increases survivin, an anti-apoptotic protein whose overexpression has been correlated with PCa bone metastasis and therapeutic resistance. Intriguingly, BKM1644 effectively inhibits survivin expression, which may antagonize docetaxel-induced survivin in bone metastatic PCa cells. Signal transducer and activator of transcription 3 (Stat3) may be involved in the suppression of survivin transcription by BKM1644, as confirmed by a survivin reporter assay. Collectively, these data indicate that BKM1644 could be a promising small-molecule agent to improve docetaxel efficacy and retard the bone metastatic growth of PCa.
Collapse
Affiliation(s)
- Shumin Zhang
- 1 Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lajos Gera
- 2 Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, School of Medicine, Aurora, CO, USA
| | - Kenza Mamouni
- 3 Department of Biochemistry and Molecular Biology, Medical College of Georgia and GRU Cancer Center, Augusta University, Augusta, GA, USA
| | - Xin Li
- 3 Department of Biochemistry and Molecular Biology, Medical College of Georgia and GRU Cancer Center, Augusta University, Augusta, GA, USA
| | - Zhengjia Chen
- 4 Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- 5 Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Daqing Wu
- 1 Department of Urology, Emory University School of Medicine, Atlanta, GA, USA,3 Department of Biochemistry and Molecular Biology, Medical College of Georgia and GRU Cancer Center, Augusta University, Augusta, GA, USA,6 MetCure Therapeutics LLC, Atlanta, GA, USA
| |
Collapse
|
7
|
A patient with a large intrathoracic malignant schwannoma who showed a complete clinical response to rAd-p53-combined with radiotherapy. Anticancer Drugs 2015; 26:902-6. [DOI: 10.1097/cad.0000000000000264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Yue PJ, He L, Shen QY, Qiu SW, Rong XM, Gong HX, Peng Y. Long-term toxicity study of rAd5-hTERTC27 in SD rats and Cynomolgus monkeys by intravenous injection. Regul Toxicol Pharmacol 2015; 71:285-94. [PMID: 25592546 DOI: 10.1016/j.yrtph.2014.12.022] [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: 07/07/2014] [Revised: 12/27/2014] [Accepted: 12/29/2014] [Indexed: 11/19/2022]
Abstract
rAd5-hTERTC27, a replication-defective adenovirus vector carrying hTERTC27, has been proposed for possible use against hepatocellular carcinoma (HCC). In this study, we investigated the long-term toxicity of rAd5-hTERTC27 in SD rats and Cynomolgus monkeys. rAd5-hTERTC27 was administered intravenously once a week for 13 weeks followed by a one-month recovery period. As of 4 months, all animals displayed overall good health. Anti-adenoviral antibodies emerged in a dose-independent manner. The levels of complement components, C3 and C4, in the rAd5-hTERTC27 middle-dose and high-dose groups and C4 in the rAd5-EGFP group increased significantly after the 2nd treatment in monkeys. Slight-mild pathological changes of the liver occurred only in the rAd5-hTERTC27 high-dose group (2/16) in rats and not in any other group in either rats or monkeys. With the increase of the dose, the incidence of lymphocyte depletion in the spleen of rats and reactive hyperplasia of the splenic corpuscle in monkeys increased. However, the changes in the liver and spleen were reversible. Given the above data, intravenous administration of rAd5-hTERTC27 (up to 4×10(10)VP/kg in rats and 0.9×10(10)VP/kg in monkeys) appears to be well-tolerated, providing support for its potentially safe use in clinical trials for the treatment of HCC.
Collapse
Affiliation(s)
- Pei-jian Yue
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China
| | - Lei He
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China
| | - Qing-yu Shen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China
| | - Shu-wei Qiu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China
| | - Xiao-ming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China
| | - Han-xian Gong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, No. 107, Yan Jiang Xi Road, Guangzhou 510120, China.
| |
Collapse
|
9
|
Guerrero-Cázares H, Tzeng SY, Young NP, Abutaleb AO, Quiñones-Hinojosa A, Green JJ. Biodegradable polymeric nanoparticles show high efficacy and specificity at DNA delivery to human glioblastoma in vitro and in vivo. ACS NANO 2014; 8:5141-53. [PMID: 24766032 PMCID: PMC4046784 DOI: 10.1021/nn501197v] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/26/2014] [Indexed: 05/18/2023]
Abstract
Current glioblastoma therapies are insufficient to prevent tumor recurrence and eventual death. Here, we describe a method to treat malignant glioma by nonviral DNA delivery using biodegradable poly(β-amino ester)s (PBAEs), with a focus on the brain tumor initiating cells (BTICs), the tumor cell population believed to be responsible for the formation of new tumors and resistance to many conventional therapies. We show transfection efficacy of >60% and low biomaterial-mediated cytotoxicity in primary human BTICs in vitro even when the BTICs are grown as 3-D oncospheres. Intriguingly, we find that these polymeric nanoparticles show intrinsic specificity for nonviral transfection of primary human BTICs over primary healthy human neural progenitor cells and that this specificity is not due to differences in cellular growth rate or total cellular uptake of nanoparticles. Moreover, we demonstrate that biodegradable PBAE/DNA nanoparticles can be fabricated, lyophilized, and then stored for at least 2 years without losing efficacy, increasing the translational relevance of this technology. Using lyophilized nanoparticles, we show transgene expression by tumor cells after intratumoral injection into an orthotopic murine model of human glioblastoma. PBAE/DNA nanoparticles were more effective than naked DNA at exogenous gene expression in vivo, and tumor cells were transfected more effectively than noninvaded brain parenchyma in vivo. This work shows the potential of nonviral gene delivery tools to target human brain tumors.
Collapse
Affiliation(s)
- Hugo Guerrero-Cázares
- Department of Neurosurgery, Department of Biomedical Engineering, Translational Tissue Engineering Center, Institute for Nanobiotechnology, and Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
| | - Stephany Y. Tzeng
- Department of Neurosurgery, Department of Biomedical Engineering, Translational Tissue Engineering Center, Institute for Nanobiotechnology, and Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
| | - Noah P. Young
- Department of Neurosurgery, Department of Biomedical Engineering, Translational Tissue Engineering Center, Institute for Nanobiotechnology, and Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
| | - Ameer O. Abutaleb
- Department of Neurosurgery, Department of Biomedical Engineering, Translational Tissue Engineering Center, Institute for Nanobiotechnology, and Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Department of Biomedical Engineering, Translational Tissue Engineering Center, Institute for Nanobiotechnology, and Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
- Address correspondence to ,
| | - Jordan J. Green
- Department of Neurosurgery, Department of Biomedical Engineering, Translational Tissue Engineering Center, Institute for Nanobiotechnology, and Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
- Address correspondence to ,
| |
Collapse
|
10
|
Abstract
Malignant brain cancer treatment is limited by a number of barriers, including the blood-brain barrier, transport within the brain interstitium, difficulties in delivering therapeutics specifically to tumor cells, the highly invasive quality of gliomas and drug resistance. As a result, the prognosis for patients with high-grade gliomas is poor and has improved little in recent years. Nanomedicine approaches have been developed in the laboratory, with some technologies being translated to the clinic, in order to address these needs. This review discusses the obstacles to effective treatment that are currently faced in the field, as well as various nanomedicine techniques that have been used or are being explored to overcome them, with a focus on liposomal and polymeric nanoparticles.
Collapse
|
11
|
Stancevic B, Varda-Bloom N, Cheng J, Fuller JD, Rotolo JA, García-Barros M, Feldman R, Rao S, Weichselbaum RR, Harats D, Haimovitz-Friedman A, Fuks Z, Sadelain M, Kolesnick R. Adenoviral transduction of human acid sphingomyelinase into neo-angiogenic endothelium radiosensitizes tumor cure. PLoS One 2013; 8:e69025. [PMID: 23936314 PMCID: PMC3732255 DOI: 10.1371/journal.pone.0069025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/03/2013] [Indexed: 12/31/2022] Open
Abstract
These studies define a new mechanism-based approach to radiosensitize tumor cure by single dose radiotherapy (SDRT). Published evidence indicates that SDRT induces acute microvascular endothelial apoptosis initiated via acid sphingomyelinase (ASMase) translocation to the external plasma membrane. Ensuing microvascular damage regulates radiation lethality of tumor stem cell clonogens to effect tumor cure. Based on this biology, we engineered an ASMase-producing vector consisting of a modified pre-proendothelin-1 promoter, PPE1(3x), and a hypoxia-inducible dual-binding HIF-2α-Ets-1 enhancer element upstream of the asmase gene, inserted into a replication-deficient adenovirus yielding the vector Ad5H2E-PPE1(3x)-ASMase. This vector confers ASMase over-expression in cycling angiogenic endothelium in vitro and within tumors in vivo, with no detectable enhancement in endothelium of normal tissues that exhibit a minute fraction of cycling cells or in non-endothelial tumor or normal tissue cells. Intravenous pretreatment with Ad5H2E-PPE1(3x)-ASMase markedly increases SDRT cure of inherently radiosensitive MCA/129 fibrosarcomas, and converts radiation-incurable B16 melanomas into biopsy-proven tumor cures. In contrast, Ad5H2E-PPE1(3x)-ASMase treatment did not impact radiation damage to small intestinal crypts as non-dividing small intestinal microvessels did not overexpress ASMase and were not radiosensitized. We posit that combination of genetic up-regulation of tumor microvascular ASMase and SDRT provides therapeutic options for currently radiation-incurable human tumors.
Collapse
Affiliation(s)
- Branka Stancevic
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Nira Varda-Bloom
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jin Cheng
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - John D. Fuller
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jimmy A. Rotolo
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Mónica García-Barros
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Regina Feldman
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Shyam Rao
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, and the Ludwig Center for Metastasis Research, Chicago, Illinois, United States of America
| | | | - Adriana Haimovitz-Friedman
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Zvi Fuks
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Richard Kolesnick
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
12
|
Yue PJ, He L, Li Y, Shen QY, Li M, Huang DQ, Huang JJ, Peng Y. Preclinical safety evaluation of rAd5-hTERTC27 by intravenous injection. Regul Toxicol Pharmacol 2013; 67:53-62. [PMID: 23827714 DOI: 10.1016/j.yrtph.2013.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 10/26/2022]
Abstract
The safety of rAd5-hTERTC27, a replication defective adenovirus vector carrying hTERTC27 for possible use against hepatocellular carcinoma (HCC) was assessed. In single-dose evaluations, intravenous dose levels of up to 2×10(11)VP/kg in rats and 9×10(10)VP/kg in monkeys were well tolerated with no abnormal changes in general signs, body weight and food consumption, and no significant differences in biochemical parameters, urinalysis, ECG, and systemic necropsy observations between the rAd5 groups and solvent control group except that slight hematological change was observed. No hemolytic effect using rabbit blood, local perivasculitis following intravenous injection in rabbits or systemic anaphylaxis in guinea pigs following intravenous dosing was seen. No effects on the central nervous system of mice occurred following intravenous dosing with the exception of an increase in sleep duration at the dose of 1.2×10(11)VP/kg (p<0.05) but not at lower doses of 2×10(10) and 6×10(10)VP/kg in the hypnotic synergism test. These results demonstrate that administration of rAd5-hTERTC27 was well tolerated in an initial set of safety studies as part of an evaluation to allow human trials for the treatment of HCC.
Collapse
Affiliation(s)
- Pei-jian Yue
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107#, Yan Jiang Xi Road of Guangzhou, Guangzhou City 510120, China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Tuppurainen L, Sallinen H, Kokki E, Koponen J, Anttila M, Pulkkinen K, Heikura T, Toivanen P, Hämäläinen K, Kosma VM, Heinonen S, Alitalo K, Ylä-Herttuala S. Preclinical safety, toxicology, and biodistribution study of adenoviral gene therapy with sVEGFR-2 and sVEGFR-3 combined with chemotherapy for ovarian cancer. HUM GENE THER CL DEV 2013; 24:29-37. [PMID: 23692381 DOI: 10.1089/humc.2013.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Abstract Antiangiogenic and antilymphangiogenic gene therapy with soluble vascular endothelial growth factor receptor-2 (VEGFR-2) and soluble VEGFR-3 in combination with chemotherapy is a potential new treatment for ovarian carcinoma. We evaluated the safety, toxicology, and biodistribution of intravenous AdsVEGFR-2 and AdsVEGFR-3 combined with chemotherapy in healthy rats (n=90) before entering a clinical setting. The study groups were: AdLacZ and AdLacZ with chemotherapy as control groups, low dose AdsVEGFR-2 and AdsVEGFR-3, high dose AdsVEGFR-2 and AdsVEGFR-3, combination of low dose AdsVEGFR-2 and AdsVEGFR-3 with chemotherapy, combination of high dose AdsVEGFR-2 and AdVEGFR-3 with chemotherapy, and chemotherapy only. The follow-up time was 4 weeks. Safety and toxicology were assessed by monitoring the clinical status of the animals and by histological, hematological, and clinical chemistry parameters. For the biodistribution studies, quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used. Low dose (2×10(10) vp) AdsVEGFR-2 and AdsVEGFR-3 gene therapy was well tolerated, even when gene therapy was combined with chemotherapy. Notably, only transient elevation of liver enzymes and mild regenerative changes were seen in liver after the gene transfer in the groups that received high doses (2×10(11) vp) of AdsVEGFR-2 and AdsVEGFR-3 with or without chemotherapy. No life-threatening adverse effects were noticed in any of the treatment groups. The highest protein concentration of soluble VEGFR-2 (sVEGFR-2) in circulation was seen 1 week after the gene transfer. The combination of chemotherapy to gene therapy seemed to prolong the time of detectable transgene protein at least 1 week in the circulation. The expression of AdsVEGFR-2 and AdsVEGFR-3 transgenes was mainly seen in the liver and spleen as detected by qRT-PCR. According to these results, AdsVEGFR-2 and AdsVEGFR-3 gene therapy combined with chemotherapy is safe and can be brought to clinical testing in ovarian cancer patients.
Collapse
Affiliation(s)
- Laura Tuppurainen
- Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, FIN-70211, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Schmitz S, Machiels JP. Molecular biology of squamous cell carcinoma of the head and neck: relevance and therapeutic implications. Expert Rev Anticancer Ther 2011; 10:1471-84. [PMID: 20836682 DOI: 10.1586/era.10.115] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
More than 90% of all head and neck cancers are squamous cell carcinoma. Despite advances in the management of patients with this disease, the survival rate has not been significantly improved. Several mechanisms of carcinogenesis have been elucidated and molecular targeted agents seem to be promising therapeutic tools. Cetuximab, a monoclonal antibody inhibitor of the EGF receptor, improves survival rates in association with radiotherapy in advanced squamous cell carcinoma of the head and neck (SCCHN) or in palliative disease, and is nowadays the only targeted agent approved in this indication. Other targeted agents are also clinically relevant to the treatment of different malignancies, including SCCHN. This article focuses on the major molecular pathways implicated in SCCHN carcinogenesis and provides an overview of their therapeutic implications.
Collapse
Affiliation(s)
- Sandra Schmitz
- Centre du Cancer, Department of Medical Oncology, Clinique de Cancérologie Cervico-Maxillo-Faciale, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium.
| | | |
Collapse
|
15
|
Mathis JM, Bhatia S, Khandelwal A, Kovesdi I, Lokitz SJ, Odaka Y, Takalkar AM, Terry T, Curiel DT. Genetic incorporation of human metallothionein into the adenovirus protein IX for non-invasive SPECT imaging. PLoS One 2011; 6:e16792. [PMID: 21347423 PMCID: PMC3036658 DOI: 10.1371/journal.pone.0016792] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 01/14/2011] [Indexed: 11/18/2022] Open
Abstract
As the limits of existing treatments for cancer are recognized, clearly novel therapies must be considered for successful treatment; cancer therapy using adenovirus vectors is a promising strategy. However tracking the biodistribution of adenovirus vectors in vivo is limited to invasive procedures such as biopsies, which are error prone, non-quantitative, and do not give a full representation of the pharmacokinetics involved. Current non-invasive imaging strategies using reporter gene expression have been applied to analyze adenoviral vectors. The major drawback to approaches that tag viruses with reporter genes is that these systems require initial viral infection and subsequent cellular expression of a reporter gene to allow non-invasive imaging. As an alternative to conventional vector detection techniques, we developed a specific genetic labeling system whereby an adenoviral vector incorporates a fusion between capsid protein IX and human metallothionein. Our study herein clearly demonstrates our ability to rescue viable adenoviral particles that display functional metallothionein (MT) as a component of their capsid surface. We demonstrate the feasibility of 99mTc binding in vitro to the pIX-MT fusion on the capsid of adenovirus virions using a simple transchelation reaction. SPECT imaging of a mouse after administration of a 99mTc-radiolabeled virus showed clear localization of radioactivity to the liver. This result strongly supports imaging using pIX-MT, visualizing the normal biodistribution of Ad primarily to the liver upon injection into mice. The ability we have developed to view real-time biodistribution in their physiological milieu represents a significant tool to study adenovirus biology in vivo.
Collapse
Affiliation(s)
- J Michael Mathis
- Gene Therapy Program, Department of Cellular Biology, LSU Health Sciences Center, Shreveport, Louisiana, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Touchefeu Y, Harrington KJ, Galmiche JP, Vassaux G. Review article: gene therapy, recent developments and future prospects in gastrointestinal oncology. Aliment Pharmacol Ther 2010; 32:953-68. [PMID: 20937041 DOI: 10.1111/j.1365-2036.2010.04424.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gene therapy consists of the introduction of genetic material into cells for a therapeutic purpose. A wide range of gene therapy vectors have been developed and used for applications in gastrointestinal oncology. AIM To review recent developments and published clinical trials concerning the application of gene therapy in the treatment of liver, colon and pancreatic cancers. METHODS Search of the literature published in English using the PubMed database. RESULTS A large variety of therapeutic genes are under investigation, such as tumour suppressor, suicide, antiangiogenesis, inflammatory cytokine and micro-RNA genes. Recent progress concerns new vectors, such as oncolytic viruses, and the synergy between viral gene therapy, chemotherapy and radiation therapy. As evidence of these basic developments, recently published phase I and II clinical trials, using both single agents and combination strategies, in adjuvant or advanced disease settings, have shown encouraging results and good safety records. CONCLUSIONS Cancer gene therapy is not yet indicated in clinical practice. However, basic and clinical advances have been reported and gene therapy is a promising, new therapeutic approach for the treatment of gastrointestinal tumours.
Collapse
Affiliation(s)
- Y Touchefeu
- Institut des Maladies de l'Appareil Digestif, INSERM U, University Hospital, Nantes, France.
| | | | | | | |
Collapse
|
17
|
Lavilla-Alonso S, Bauerschmitz G, Abo-Ramadan U, Halavaara J, Escutenaire S, Diaconu I, Tatlisumak T, Kanerva A, Hemminki A, Pesonen S. Adenoviruses with an αvβ integrin targeting moiety in the fiber shaft or the HI-loop increase tumor specificity without compromising antitumor efficacy in magnetic resonance imaging of colorectal cancer metastases. J Transl Med 2010; 8:80. [PMID: 20727221 PMCID: PMC2936307 DOI: 10.1186/1479-5876-8-80] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 08/23/2010] [Indexed: 12/22/2022] Open
Abstract
Background Colorectal cancer is often a deadly disease and cannot be cured at metastatic stage. Oncolytic adenoviruses have been considered as a new therapeutic option for treatment of refractory disseminated cancers, including colorectal cancer. The safety data has been excellent but tumor transduction and antitumor efficacy especially in systemic administration needs to be improved. Methods Here, the utility of αvβ integrin targeting moiety Arg-Gly-Asp (RGD) in the Lys-Lys-Thr-Lys (KKTK) domain of the fiber shaft or in the HI-loop of adenovirus serotype 5 for increased tumor targeting and antitumor efficacy was evaluated. To this end, novel spleen-to-liver metastatic colorectal cancer mouse model was used and the antitumor efficacy was evaluated with magnetic resonance imaging (MRI). Results Both modifications (RGD in the HI-loop or in the fiber shaft) increased gene transfer efficacy in colorectal cancer cell lines and improved tumor-to-normal ratio in systemic administration of the vector. Conclusions Antitumor potency was not compromised with RGD modified viruses suggesting increased safety profile and tumor specificity.
Collapse
Affiliation(s)
- Sergio Lavilla-Alonso
- Transplantation Laboratory, Haartman Institute and Finnish Institute of Molecular Medicine, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
A phase I trial of repeated intrapleural adenoviral-mediated interferon-beta gene transfer for mesothelioma and metastatic pleural effusions. Mol Ther 2010; 18:852-60. [PMID: 20068553 DOI: 10.1038/mt.2009.309] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We previously showed that a single intrapleural dose of an adenoviral vector expressing interferon-beta (Ad.IFN-beta) in patients with malignant pleural mesothelioma (MPM) or malignant pleural effusions (MPE) resulted in gene transfer, humoral antitumor immune responses, and anecdotal clinical responses manifested by modified Response Evaluation Criteria in Solid Tumors (RECIST) disease stability in 3 of 10 patients at 2 months and an additional patient with significant metabolic response on positron emission tomography (PET) imaging. This phase I trial was conducted to determine whether using two doses of Ad.IFN-beta vector would be superior. Ten patients with MPM and seven with MPE received two doses of Ad.IFN-beta through an indwelling pleural catheter. Repeated doses were generally well tolerated. High levels of IFN-beta were detected in pleural fluid after the first dose; however, only minimal levels were seen after the second dose of vector. Lack of expression correlated with the rapid induction of neutralizing Ad antibodies (Nabs). Antibody responses against tumor antigens were induced in most patients. At 2 months, modified RECIST responses were as follows: one partial response, two stable disease, nine progressive disease, and two nonmeasurable disease. One patient died after 1 month. By PET scanning, 2 patients had mixed responses and 11 had stable disease. There were seven patients with survival times longer than 18 months. This approach was safe, induced immune responses and disease stability. However, rapid development of Nabs prevented effective gene transfer after the second dose, even with a dose interval as short as 7 days.
Collapse
|
19
|
Multiple hepatic arterial injections of recombinant adenovirus p53 and 5-fluorouracil after transcatheter arterial chemoembolization for unresectable hepatocellular carcinoma: a pilot phase II trial. Anticancer Drugs 2009; 20:389-95. [PMID: 19287305 DOI: 10.1097/cad.0b013e32832a2df9] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This pilot phase II study was designed to determine the efficacy, toxicities, and biological activity of multiple hepatic arterial injections of recombinant adenovirus p53 (rAd-p53) and 5-fluorouracil (5-FU) after transcatheter arterial chemoembolization (TACE) when compared with TACE alone in patients with unresectable hepatocellular carcinoma (HCC). Forty-six patients with unresectable HCC were randomized in either group 1 [23 patients, multiple hepatic arterial injections of Ad-p53 (1x10 viral particles) and 5-FU (500-750 mg), after TACE] or group 2 (23 patients, TACE alone). In group 1, the number of Ad-p53/5-FU courses administered was 166 (median 7, range 3-12). In group 2, the number of TACE courses administered was 47 (median 2, range 1-3). Partial response and stable disease were 69.5% in group 1 and 65.2% in group 2. Times to progression were 9.6 months (range 2.1-21.7) in group 1 and 8.3 months (range 2.1-16.8) in group 2. Overall survivals were 12.8 months (range 2.7-26.2) in group 1 and 10.4 months (range 2.7-22.5) in group 2. Toxicities in both groups were generally mild and reversible. The most common Ad-p53-related toxicity was a transient fever. Specific p53 transgene expression was detected using reverse-transcriptase polymerase chain reaction in biopsied tumor tissues. Distribution studies revealed that the vector was detected in the plasma, but rarely in the gargle and urine. This study shows that multiple hepatic arterial injections of Ad-p53 and 5-FU after TACE can be active and safe as a treatment for patients with unresectable HCC.
Collapse
|
20
|
A patient with huge hepatocellular carcinoma who had a complete clinical response to p53 gene combined with chemotherapy and transcatheter arterial chemoembolization. Anticancer Drugs 2009; 20:403-7. [PMID: 19318914 DOI: 10.1097/cad.0b013e328323d6b8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The prognosis of hepatocellular carcinoma (HCC) is poor and current therapies are largely ineffective. Transcatheter arterial chemoembolization is a standard treatment option for patients with unresectable HCC, especially when combined with other therapies. We report a 62-year-old male with huge HCC. The patient was first treated with adenovirus-mediated wild-type p53 gene (Ad-p53, gendicine) combined with oxaliplatin (200 mg) and transcatheter arterial chemoembolization or transcatheter arterial chemotherapy for two cycles. Review showed tumor shrinkage and a decrease in alpha-fetoprotein. Oxaliplatin was stopped because of side effects. Then the patient was treated with a tumor feeding arterial injection of Ad-p53 (1 x 10 viral particles) twice and Ad-p53 (1 x 10 viral particles) followed by 5-fluorouracil (500-750 mg) six times through port-catheter system. We observed marked tumor shrinkage and sustained normal alpha-fetoprotein and liver function during a 614-day follow-up period.
Collapse
|
21
|
Terashima T, Oka K, Kritz AB, Kojima H, Baker AH, Chan L. DRG-targeted helper-dependent adenoviruses mediate selective gene delivery for therapeutic rescue of sensory neuronopathies in mice. J Clin Invest 2009; 119:2100-112. [PMID: 19603551 PMCID: PMC2701884 DOI: 10.1172/jci39038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 04/29/2009] [Indexed: 12/11/2022] Open
Abstract
Dorsal root ganglion (DRG) neuron dysfunction occurs in a variety of sensory neuronopathies for which there are currently no satisfactory treatments. Here we describe the development of a strategy to target therapeutic genes to DRG neurons for the treatment of these disorders. We genetically modified an adenovirus (Ad) to generate a helper virus (HV) that was detargeted for native adenoviral tropism and contained DRG homing peptides in the adenoviral capsid fiber protein; we used this HV to generate DRG-targeted helper-dependent Ad (HDAd). In mice, intrathecal injection of this HDAd produced a 100-fold higher transduction of DRG neurons and a markedly attenuated inflammatory response compared with unmodified HDAd. We also injected HDAd encoding the beta subunit of beta-hexosaminidase (Hexb) into Hexb-deficient mice, a model of the neuronopathy Sandhoff disease. Delivery of the DRG-targeted HDAd reinstated neuron-specific Hexb production, reversed gangliosidosis, and ameliorated peripheral sensory dysfunction. The development of DRG neuron-targeted HDAd with proven efficacy in a preclinical model may have implications for the treatment of sensory neuronopathies of diverse etiologies.
Collapse
Affiliation(s)
- Tomoya Terashima
- Department of Medicine and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Kazuhiro Oka
- Department of Medicine and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Angelika B. Kritz
- Department of Medicine and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Hideto Kojima
- Department of Medicine and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Andrew H. Baker
- Department of Medicine and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Lawrence Chan
- Department of Medicine and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
22
|
Kelley T, Beck R, Absi A, Jin T, Pohlman B, Hsi E. Biologic predictors in follicular lymphoma: Importance of markers of immune response. Leuk Lymphoma 2009; 48:2403-11. [DOI: 10.1080/10428190701665954] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
23
|
Moretto P, Hotte SJ. Targeting apoptosis: preclinical and early clinical experience with mapatumumab, an agonist monoclonal antibody targeting TRAIL-R1. Expert Opin Investig Drugs 2009; 18:311-25. [DOI: 10.1517/13543780902752463] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
24
|
Huang PI, Chang JF, Kirn DH, Liu TC. Targeted genetic and viral therapy for advanced head and neck cancers. Drug Discov Today 2009; 14:570-8. [PMID: 19508919 DOI: 10.1016/j.drudis.2009.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Revised: 03/07/2009] [Accepted: 03/11/2009] [Indexed: 11/24/2022]
Abstract
Head and neck cancers usually present with advanced disease and novel therapies are urgently needed. Genetic therapy aims at restoring malfunctioned tumor suppressor gene(s) or introducing proapoptotic genes. Oncolytic virotherapeutics induce multiple cycles of cancer-specific virus replication, followed by oncolysis, virus spreading and infection of adjacent cancer cells. Oncolytic viruses can also be armed to express therapeutic transgene(s). Recent advances in preclinical and clinical studies are revealing the potential of both therapeutic classes for advanced head and neck cancers, including the approval of two products (Gendicine and H101) by a governmental agency. This review summarizes the available clinical data to date and discusses the challenges and future directions.
Collapse
Affiliation(s)
- Pin-I Huang
- Cancer Center, Taipei Veterans General Hospital, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | |
Collapse
|
25
|
Jaboin JJ, Han Z, Hallahan DE. Using in vivo biopanning for the development of radiation-guided drug delivery systems. Methods Mol Biol 2009; 542:285-300. [PMID: 19565908 DOI: 10.1007/978-1-59745-561-9_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
This chapter illustrates our protocol for in vivo biopanning using T7 bacteriophage libraries for the purpose of selecting recombinant peptides for the tumor-specific delivery of radiosensitizers to radiation-inducible antigens within tumor neovasculature. Our goal is to discover peptides binding within tumor vascular endothelium of irradiated tumors. We have previously demonstrated that tumor irradiation increases the spectrum of antigenic targets for drug delivery. To identify candidate peptides with the ability to bind radiation-induced antigens, we inject the phage peptide library intravenously into mice bearing irradiated GL261 and Lewis lung carcinoma (LLC) hind limb tumors. Phage are recovered from excised tumors, amplified, and readministered to mouse-bearing tumors for six total rounds. At least 50 bacterial colonies are selected from each of the tumor types, and prioritized. This prioritization is based on their relative concentrations in tumor versus normal tissues, and then assessment of dominant phage present in both tumor types. These phage are amplified, and the gene sequences determined to deduce the recombinant peptide product. Further prioritization is performed by fluorescence labeling of the selected phage, and injection into irradiated and mock-irradiated tumor-bearing mice for evaluation of in vivo targeting of the candidate phage/peptides.
Collapse
Affiliation(s)
- Jerry J Jaboin
- Department of Radiation Oncology, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
26
|
Plati J, Bucur O, Khosravi-Far R. Dysregulation of apoptotic signaling in cancer: molecular mechanisms and therapeutic opportunities. J Cell Biochem 2008; 104:1124-49. [PMID: 18459149 DOI: 10.1002/jcb.21707] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Apoptosis is a tightly regulated cell suicide program that plays an essential role in the maintenance of tissue homeostasis by eliminating unnecessary or harmful cells. Defects in this native defense mechanism promote malignant transformation and frequently confer chemoresistance to transformed cells. Indeed, the evasion of apoptosis has been recognized as a hallmark of cancer. Given that multiple mechanisms function at many levels to orchestrate the regulation of apoptosis, a multitude of opportunities for apoptotic dysregulation are present within the intricate signaling network of cell. Several of the molecular mechanisms by which cancer cells are protected from apoptosis have been elucidated. These advances have facilitated the development of novel apoptosis-inducing agents that have demonstrated single-agent activity against various types of cancers cells and/or sensitized resistant cancer cells to conventional cytotoxic therapies. Herein, we will highlight several of the central modes of apoptotic dysregulation found in cancer. We will also discuss several therapeutic strategies that aim to reestablish the apoptotic response, and thereby eradicate cancer cells, including those that demonstrate resistance to traditional therapies.
Collapse
Affiliation(s)
- Jessica Plati
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
27
|
Curtin JF, Candolfi M, Xiong W, Lowenstein PR, Castro MG. Turning the gene tap off; implications of regulating gene expression for cancer therapeutics. Mol Cancer Ther 2008; 7:439-48. [PMID: 18347132 DOI: 10.1158/1535-7163.mct-07-2328] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer poses a tremendous therapeutic challenge worldwide, highlighting the critical need for developing novel therapeutics. A promising cancer treatment modality is gene therapy, which is a form of molecular medicine designed to introduce into target cells genetic material with therapeutic intent. Anticancer gene therapy strategies currently used in preclinical models, and in some cases in the clinic, include proapoptotic genes, oncolytic/replicative vectors, conditional cytotoxic approaches, inhibition of angiogenesis, inhibition of growth factor signaling, inactivation of oncogenes, inhibition of tumor invasion and stimulation of the immune system. The translation of these novel therapeutic modalities from the preclinical setting to the clinic has been driven by encouraging preclinical efficacy data and advances in gene delivery technologies. One area of intense research involves the ability to accurately regulate the levels of therapeutic gene expression to achieve enhanced efficacy and provide the capability to switch gene expression off completely if adverse side effects should arise. This feature could also be implemented to switch gene expression off when a successful therapeutic outcome ensues. Here, we will review recent developments related to the engineering of transcriptional switches within gene delivery systems, which could be implemented in clinical gene therapy applications directed at the treatment of cancer.
Collapse
Affiliation(s)
- James F Curtin
- University of California-Los Angeles and Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Although there is no shortage of potential targets for cancer therapeutics, we know of only a handful of molecules that are differentially expressed in cancer and intersect multiple pathways required for tumour maintenance. Survivin embodies these properties, and orchestrates integrated cellular networks that are essential for tumour cell proliferation and viability. Pursuing the nodal functions of survivin in cancer might lead to the development of global pathway inhibitors with unique therapeutic potential.
Collapse
Affiliation(s)
- Dario C Altieri
- Department of Cancer Biology and the Cancer Center, Lazare Research Building 428, University of Massachusetts Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, USA.
| |
Collapse
|
29
|
Schenk-Braat EAM, van Mierlo MMKB, Wagemaker G, Bangma CH, Kaptein LCM. An inventory of shedding data from clinical gene therapy trials. J Gene Med 2007; 9:910-21. [PMID: 17880045 DOI: 10.1002/jgm.1096] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Viruses are the most commonly used vectors for clinical gene therapy. The risk of dissemination of a viral vector into the environment via excreta from the treated patient, a phenomenon called shedding, is a major safety concern for the environment. Despite the significant number of clinical gene therapy trials that have been conducted worldwide, there is currently no overview of actual shedding data available. In this article, an inventory of shedding data obtained from a total of 100 publications on clinical gene therapy trials using retroviral, adenoviral, adeno-associated viral and pox viral vectors is presented. In addition, the experimental set-up for shedding analysis including the assays used and biological materials tested is summarized. The collected data based on the analysis of 1619 patients in total demonstrate that shedding of these vectors occurs in practice, mainly determined by the type of vector and the route of vector administration. Due to the use of non-quantitative assays, the lack of information on assay sensitivity in most publications, and the fact that assay sensitivity is expressed in various ways, general conclusions cannot be made as to the level of vector shedding. The evaluation of the potential impact and consequences of the observations is complicated by the high degree of variety in the experimental design of shedding analysis between trials. This inventory can be supportive to clinical gene therapy investigators for the establishment of an evidence-based risk assessment to be included in a clinical protocol application, as well as to national regulatory authorities for the ongoing development of regulatory guidelines regarding gene therapy.
Collapse
|
30
|
Intranasal administration of a recombinant adenovirus expressing the norovirus capsid protein stimulates specific humoral, mucosal, and cellular immune responses in mice. Vaccine 2007; 26:460-8. [PMID: 18160189 DOI: 10.1016/j.vaccine.2007.11.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 10/19/2007] [Accepted: 11/18/2007] [Indexed: 11/23/2022]
Abstract
Norovirus (NV) is a major cause of acute, epidemic nonbacterial gastroenteritis in individuals of all ages. The immunological mechanism of NV infection and the approaches used to prevent infection remain to be elucidated. In this study, the specific immune responses of BALB/c mice were assessed following intranasal immunization with a recombinant adenovirus vector expressing the genogroup II4 (GGII/4) norovirus capsid protein. Analysis of IgM, IgG, and IgA antibodies specific for the recombinant virus-like particles (VLPs) of NV demonstrated that a high level of humoral immunity developed following immunization. Mucosal immune responses were also detectable in stool, intestinal homogenates, lung homogenates, and lung lavage samples. Specific cellular immune responses were observed in NV VLPs-restimulated splenocytes by ELISPOT and Th1/Th2 cytokine cytometric array (CBA). Serum IgG subclass analysis showed that a balanced Th1- and Th2-like cellular immune response was induced in BALB/c mice following immunization with recombinant adenovirus. These findings demonstrate that the intranasal immunization of a recombinant adenovirus expressing the NV capsid protein is an efficient strategy to stimulate systemic, mucosal, and cellular Th1/Th2 immune responses in mice, and could serve as a novel approach for designing NV vaccines.
Collapse
|
31
|
Chng WJ, Price-Troska T, Gonzalez-Paz N, Van Wier S, Jacobus S, Blood E, Henderson K, Oken M, Van Ness B, Greipp P, Rajkumar SV, Fonseca R. Clinical significance of TP53 mutation in myeloma. Leukemia 2007; 21:582-4. [PMID: 17215851 DOI: 10.1038/sj.leu.2404524] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Abstract
Defects in programmed cell death or apoptosis are major hallmarks of cancer contributing to tumorigenesis, tumor progression, and therapy resistance. In the past decade, many of the pathways leading to apoptosis, as well as the molecular mechanisms blocking the death of tumor cells, have been elucidated. This detailed knowledge of the core apoptosis machinery is now being exploited for translation into novel cancer therapies in order to restore apoptosis induction in tumor cells. Strategies include activation of proapoptotic mediators such as death receptors, tumor protein p53, and second mitochondria-derived activator of caspases (SMAC)/DIABLO as well as inhibition of endogenous apoptosis inhibitors such as IAPs (inhibitor of apoptosis proteins) and BCL-2 (B-cell chronic lymphoid leukemia/lymphoma) proteins. Several approaches employing gene therapy and antisense strategies, recombinant biologics, or classic organic and combinatorial chemistry, have advanced into clinical trials or are already approved. This review looks at recent developments in apoptosis-based cancer therapies and highlights some very promising advances in drug design.
Collapse
Affiliation(s)
- Ute Fischer
- Institute of Molecular Medicine, Heinrich-Heine University, Düsseldorf, Germany.
| | | | | |
Collapse
|
33
|
Woo CY, Osada T, Clay TM, Lyerly HK, Morse MA. Recent clinical progress in virus-based therapies for cancer. Expert Opin Biol Ther 2006; 6:1123-34. [PMID: 17049011 DOI: 10.1517/14712598.6.11.1123] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As our knowledge of the molecular basis of cancer expands, viral vectors have been increasingly studied as potential antitumour therapeutic agents. With their ability to invade and replicate within target cells, viruses have been utilised as oncolytic agents to directly lyse tumour cells. Viruses can also deliver their genetic payload into infected cells, allowing for the repair of defective tumour suppressor genes, disruption of oncogenic pathways, and production of cytokines that activate the immune system. Finally, viruses encoding tumour-associated antigens can infect dendritic cells, triggering the development of a tumour-specific immune response. The ability to engineer viruses with high levels of tumour specificity and efficient rates of infection has enhanced the safety profile of these agents, allowing for the development of viable therapeutic options that have been examined in the clinic, either alone or in conjunction with more conventional therapies. This review highlights the principles underlying virus-based therapies for cancer, with an emphasis on recent developments from the clinic.
Collapse
Affiliation(s)
- Christopher Y Woo
- Duke University Medical Center, Department of Medicine, Programme in Molecular Therapeutics, Comprehensive Cancer Center, 401 MSRB, Research Drive, Durham, NC 27710, USA.
| | | | | | | | | |
Collapse
|
34
|
Abstract
Mutations in the p53 gene are the most frequent genetic alterations in human tumours, occurring in approximately 50% of all cancers. The p53 protein is pivotal in maintaining genetic integrity after DNA damage, and alterations in the p53 pathway, including mutations in the p53 gene, greatly increase the probability of tumour formation. Gene therapy using adenoviral p53 has emerged as a novel treatment option, with the potential to be safe and effective in a wide range of cancer types. INGN 201 (Ad5CMV-p53, Advexin), a replication-impaired adenoviral vector that carries the p53 gene, has been evaluated in both preclinical and clinical trials. Results show that Advexin is a well-tolerated and efficacious treatment for numerous cancers, both as monotherapy and in combination with radiation and/or chemotherapy agents. In addition, there is now data to support the use of Advexin in cancer immunotherapy.
Collapse
Affiliation(s)
- Dmitry I Gabrilovich
- University of South Florida, H. Lee Moffitt Cancer Center and the Department of Interdisciplinary Oncology, MRC 2067, 12902 Magnolia Dr., Tampa, FL 33612, USA.
| |
Collapse
|