1
|
Eggers R, Philippi A, Altmeyer MO, Breinig F, Schmitt MJ. Primary T cells for mRNA-mediated immunotoxin delivery. Gene Ther 2018; 25:47-53. [PMID: 28937681 DOI: 10.1038/gt.2017.87] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 12/30/2022]
Abstract
Immune cells become increasingly attractive as delivery system for immunotoxins in cancer therapy to reduce the intrinsic toxicity and severe side effects of chimeric protein toxins. In this study, we investigated the potential of human primary T cells to deliver a secreted immunotoxin through transient messenger RNA (mRNA) transfection. The chimeric protein toxin was directed toward the neovasculature of cancer cells by fusing a truncated version of Pseudomonas exotoxin A (PE38) to human vascular endothelial growth factor (VEGF) and to the single chain variable fragment (scFv) of anti-Her2/neu. Protocols for the transient transfection of human embryonic kidney cells (HEK293) as well as activated primary human T cells were established. Transient transfection with mRNA coding for the immunotoxins e23-PE38, VEGF-PE38 and its attenuated variant VEGF-PE38D yielded efficient expression and secretion. Mass spectrometry analysis endorsed that a fraction of VEGF-PE38D was properly translocated into the endoplasmic reticulum. Furthermore, cytotoxic activity of immunotoxin secreting T cells toward cancer cells was confirmed in co-culture with ovarian adenocarcinoma cells in the presence of a bispecific antibody (bsAb), highlighting the potential of primary T cells for mRNA-mediated immunotoxin delivery.
Collapse
Affiliation(s)
- R Eggers
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - A Philippi
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - M O Altmeyer
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - F Breinig
- Department of Biosciences, Molecular and Cell Biology and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - M J Schmitt
- Department of Biosciences, Molecular and Cell Biology and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| |
Collapse
|
2
|
Barar J, Omidi Y. Personalized cell-mediated immunotherapy and vaccination: combating detrimental uprisings of malignancies. ACTA ACUST UNITED AC 2015; 5:65-9. [PMID: 26191499 PMCID: PMC4492186 DOI: 10.15171/bi.2015.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/24/2015] [Indexed: 12/17/2022]
Abstract
A large number of researchers worldwide have conducted various investigations to advance the cell-based immunotherapies and to examine their clinical benefits as an ultimate prevention and/or treatment modalities against life-threatening malignancies. This dominion needs integration of science and technology to change the face of treatment of diseases towards much more personalized medicines. It is now plausible to reprogram the human cells for the prevention and treatment of diseases through various mechanisms such as modulation of immune system, nonetheless we should understand the complexity of biological functions of the cells in a holistic way to be able to manipulate the central dogma of the life to prevent any inadvertent mistake. We should, if not must, comprehend the interrelations of the cellular components (e.g., transport machineries) in the developmental processes of diseases. Still, we do not have a complete image of life, perhaps as expressive barcodes, and many pieces are missing. While completing this puzzle to picture the whole image and examine new treatment modalities, we should take extra caution upon unknown/little-known biological phenomena because trifling modulation/ alteration in the complex systems of the life may result in tremendous impacts. In short, it seems we need to consider malignancies as complex systems and treat them in a holistic manner by targeting its hallmarks. Taken all, the immune system reinforcement would be one of the main foundations in combating detrimental malignancy uprising.
Collapse
Affiliation(s)
- Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Shen Y, Liu Q, Luo Y, Zhang P, Bai F, Cheng S, Lou S. Enhancement of the cytotoxic activity of cytokine-induced killer cells transfected with IL3PE38KDEL gene against acute myeloid leukemia cells. Ann Hematol 2014; 93:2019-28. [PMID: 25029985 DOI: 10.1007/s00277-014-2146-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
Cytokine-induced killer (CIK) cells, one of the feasible and effective methods of adoptive immunotherapy, have shown anti-leukemia activity in vivo and in vitro. But the strategy exhibits limited cytotoxic activity in clinical studies. In this study, CIK cells were transfected with an interleukin-3/Pseudomonas exotoxin gene (IL3PE38KDEL). RT-PCR and ELISA were used to verify the expression of IL3PE38KDEL in the transfected CIK cells. These cells released 1,186.7 ± 149.6 pg IL3PE38KDEL/10(4) cells over 48 h into the medium and the culture supernatant selectively killed IL3 receptor(IL3R)-positive HL60 cells, but not IL3R-negative K562 cells. Moreover, IL3PE38KDEL transfection did not influence phenotypes and cytokine production of CIK cells. Co-cultured with leukemia cells, IL3PE38KDEL transfected CIK cells showed enhanced cytotoxicity against IL3R-positive HL60 cells at all effector-to-target (E:T) ratios, but exerted a basal anti-leukemia activity against IL3R-negative K562 cells. Our findings demonstrate that IL3PE38KDEL gene transfection may be a novel strategy for improving anti-leukemia activity of CIK cells.
Collapse
Affiliation(s)
- Yan Shen
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Linjiang Road 76, Chongqing, 400010, China
| | | | | | | | | | | | | |
Collapse
|
4
|
Zhou BG, Liu MY, Qiu XC, Xu YM, Fan QY, Yang AG, Zhang Y, Xia H. A novel recombinant immunocasp-6 fusion gene specifically and efficiently suppresses HER2-overexpressing osteosarcoma. Oncol Rep 2013; 29:276-82. [PMID: 23135254 DOI: 10.3892/or.2012.2122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 09/25/2012] [Indexed: 11/06/2022] Open
Abstract
Osteosarcoma is the most common primary malignant tumor of bone for adolescent or children. The poor prognosis of patients, due to its remote metastasis, has led to the exploration of more effective and less toxic treatments. Immunotherapy is a promising strategy for the treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing tumors. Herein, we describe experiments conducted with a fusion gene, immunocasp-6, which was generated by fusing a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A and an active caspase-6 which can directly cleave lamin A leading to nucleus damage inducing programmed cell death. We demonstrated that immunocasp-6 can specifically and efficiently recognize and induce apoptosis in HER2-overexpressing osteosarcoma cells in vitro. The immunocasp-6 was transferred into BALB/c athymic mice bearing human osteosarcoma by i.m. injection of liposome-encapsulated pCMV-immunocap-6. Expression of immunocasp-6 not only strongly inhibited tumor growth and significantly prolonged animal survival, but also greatly prevented tumor metastasis. Our data showed that the immuno-casp-6 can specifically recognize HER2-overexpressing osteosarcoma cells, can also promptly attack their nucleus and induce apoptotic death, suggesting the potential of this strategy for the treatment of human HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Ben-Gen Zhou
- Graduate School of Southern Medical University, Department of Orthopaedics, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong, PR China
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Multi-Compartmental Vaccine Delivery System for Enhanced Immune Response to gp100 Peptide Antigen in Melanoma Immunotherapy. Pharm Res 2012; 29:3393-403. [DOI: 10.1007/s11095-012-0834-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 07/09/2012] [Indexed: 12/28/2022]
|
6
|
Jia LT, Chen SY, Yang AG. Cancer gene therapy targeting cellular apoptosis machinery. Cancer Treat Rev 2012; 38:868-76. [PMID: 22800735 DOI: 10.1016/j.ctrv.2012.06.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/21/2012] [Accepted: 06/22/2012] [Indexed: 01/14/2023]
Abstract
The unraveling of cellular apoptosis machinery provides novel targets for cancer treatment, and gene therapy targeting this suicidal system has been corroborated to cause inflammation-free autonomous elimination of neoplastic cells. The apoptotic machinery can be targeted by introduction of a gene encoding an inducer, mediator or executioner of apoptotic cell death or by inhibition of anti-apoptotic gene expression. Strategies targeting cancer cells, which are achieved by selective gene delivery, specific gene expression or secretion of target proteins via genetic modification of autologous cells, dictate the outcome of apoptosis-based cancer gene therapy. Despite so far limited clinical success, gene therapy targeting the apoptotic machinery has great potential to benefit patients with threatening malignancies provided the availability of efficient and specific gene delivery and administration systems.
Collapse
Affiliation(s)
- Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| | | | | |
Collapse
|
7
|
Naeem A, Fazili NA. Defective protein folding and aggregation as the basis of neurodegenerative diseases: the darker aspect of proteins. Cell Biochem Biophys 2012; 61:237-50. [PMID: 21573992 DOI: 10.1007/s12013-011-9200-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The ability of a polypeptide to fold into a unique, functional, and three-dimensional structure depends on the intrinsic properties of the amino acid sequence, function of the molecular chaperones, proteins, and enzymes. Every polypeptide has a finite tendency to misfold and this forms the darker side of the protein world. Partially folded and misfolded proteins that escape the cellular quality control mechanism have the high tendency to form inter-molecular hydrogen bonding between the same protein molecules resulting in aggregation. This review summarizes the underlying and universal mechanism of protein folding. It also deals with the factors responsible for protein misfolding and aggregation. This article describes some of the consequences of such behavior particularly in the context of neurodegenerative conformational diseases such as Alzheimer's, Parkinson's, Huntington's, amyotrophic lateral sclerosis and other non-neurodegenerative conformational diseases such as cancer and cystic fibrosis etc. This will encourage a more proactive approach to the early diagnosis of conformational diseases and nutritional counseling for patients.
Collapse
Affiliation(s)
- Aabgeena Naeem
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India.
| | | |
Collapse
|
8
|
Hu Y, Zhang L, Wu R, Han R, Jia Y, Jiang Z, Cheng M, Gan J, Tao X, Zhang Q. Specific killing of CCR9 high-expressing acute T lymphocytic leukemia cells by CCL25 fused with PE38 toxin. Leuk Res 2011; 35:1254-60. [PMID: 21295855 DOI: 10.1016/j.leukres.2011.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/14/2011] [Accepted: 01/15/2011] [Indexed: 12/18/2022]
Abstract
We have previously demonstrated that CCR9 plays a pivotal role in drug resistance and invasion in human acute T-lymphocytic leukemia (T-ALL). In this study, we investigated whether the MOLT4 cells, which naturally express CCR9 at high levels, can be successfully killed by the specific ligand, CCL25 fused to Pseudomonas exotoxin 38 (PE38) toxin. Our results demonstrated that CCL25-PE38 was able to specifically kill MOLT4 cells via apoptosis induction, and suppress the growth of CCR9(+) tumors. This work shows that CCR9 high-expressing human T-ALL cells can be successfully killed by delivering PE38 toxin fused to the ligand CCL25.
Collapse
Affiliation(s)
- Yi Hu
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuchang, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Sun XL, Xu ZM, Ke YQ, Hu CC, Wang SY, Ling GQ, Yan ZJ, Liu YJ, Song ZH, Jiang XD, Xu RX. Molecular targeting of malignant glioma cells with an EphA2-specific immunotoxin delivered by human bone marrow-derived mesenchymal stem cells. Cancer Lett 2011; 312:168-77. [PMID: 21924825 DOI: 10.1016/j.canlet.2011.07.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/28/2011] [Accepted: 07/30/2011] [Indexed: 12/17/2022]
Abstract
Immunotoxins have shown great promise as an alternative treatment for brain malignancies such as gliomas, but their failure to penetrate into the tumor mass remains a major problem. Mesenchymal stem cells exhibit tropism to tumor tissue and may serve as a cellular vehicle for the delivery and local production of antitumor agents. In this study, we used human bone marrow-derived mesenchymal stem cells (hMSCs) as a vehicle for the targeted delivery of EphrinA1-PE38, a very specific immunotoxin against the EphA2 receptor that is overexpressed in gliomas. hMSCs were transduced with adenovirus to express secretable EphrinA1-PE38. Our invitro assays confirmed the expression, release and selective killing effect of the immunotoxin produced by hMSCs. Furthermore, the intratumoral injection of engineered hMSCs was effective at inhibiting tumor growth in a malignant glioma tumor model. These results indicate that gene therapy utilizing EphrinA1-PE38-secreting hMSCs may provide a novel approach for the local treatment of malignant gliomas.
Collapse
Affiliation(s)
- Xin-Lin Sun
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Xue H, Liang F, Liu N, Song X, Yuan F, Luo Y, Zhao X, Long J, Sun Y, Xi Y. Potent antirheumatic activity of a new DNA vaccine targeted to B7-2/CD28 costimulatory signaling pathway in autoimmune arthritis. Hum Gene Ther 2011; 22:65-76. [PMID: 20695769 DOI: 10.1089/hum.2010.110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis is a proinflammatory autoimmune disease attributed to failure of both CD4(+)CD25(+) regulatory T (Tr) and CD8(+)CD28(-) suppressor T (Ts) cells to control autoreactive CD4(+)CD28(+) Th1 (Th1) and autoantibody-producing B cells. Here we show a single intramuscular injection of our novel targeted DNA vaccine encoding Pseudomonas exotoxin A and costimulatory molecule B7-2 without autoantigens in a collagen-induced arthritis model simultaneously increased Tr and Ts cells and selectively decreased autoreactive Th1 cells. The vaccine induced a shift from Th1 to Th2 and Th3 cellular and cytokine profiles and a decrease in CD4(+)/CD8(+) cell ratios. Importantly, the vaccine showed potent antirheumatic activity by clinical and other examinations such as X-ray, histopathology, and anti-type II collagen IgG levels and was comparable to methotrexate, the current "gold standard" treatment. As an effective stimulator of both Tr and Ts cells and a specific suppressor of autoreactive Th1 cells, this vaccine is a promising therapeutic approach for rheumatoid arthritis.
Collapse
Affiliation(s)
- Hong Xue
- Department of Immunology, Beijing 307 Hospital, Academy of Medical Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Hu CC, Ji HM, Chen SL, Zhang HW, Wang BQ, Zhou LY, Zhang ZP, Sun XL, Chen ZZ, Cai YQ, Qin LS, Lu L, Jiang XD, Xu RX, Ke YQ. Investigation of a plasmid containing a novel immunotoxin VEGF165-PE38 gene for antiangiogenic therapy in a malignant glioma model. Int J Cancer 2010; 127:2222-9. [PMID: 20127864 DOI: 10.1002/ijc.25217] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inhibition of tumor neovascularization has profound effects on the growth of solid tumors. Our previous studies have shown the effect of VEGF165-PE38 recombinant immunotoxin on proliferation and apoptosis in human umbilical vein endothelial cells in vitro. In this study, we explored the direct inhibition of angiogenesis in chick chorioallantoic membrane and antiangiogenic therapy in a malignant glioma model. HEK293 cells were transfected with the pVEGF165PE38-IRES2-EGFP plasmid. ELISA was used to confirm the expression of VEGF165-PE38 in the transfected cells. These cells released 1396 + or - 131.9 pg VEGF165-PE38/1x10(4) cells/48 h into the culture medium and the supernatant was capable of inhibiting the growth of capillary-like structures in chick chorioallantoic membrane assay. In a murine malignant glioma model, plasmid was directly administered via multiple local intratumoral delivery. After day 16 the tumor volume in mice treated with pVEGF165PE38-IRES2-EGFP was significantly lower than that in mice in the control groups. Immunohistochemistry studies showed that the treated group had decreased expression of CD31. Quantitative analysis of microvessel density in the treated group was 1.99 + or - 0.69/0.74 mm(2), and was significantly lower than that in the control groups (9.33 + or - 1.99/0.74 mm(2), 8.09 + or - 1.39/0.74 mm(2) and 8.49 + or - 1.69/0.74 mm(2)). Immunohistochemistry analysis indicated that immunotoxin VEGF165-PE38 was distributed in the treated group in malignant glioma tissue. Our findings provide evidence that the in vivo production of VEGF165-PE38 through gene therapy using a eukaryotic expression plasmid had potential antiangiogenic activity in malignant glioma in vivo.
Collapse
Affiliation(s)
- Chang-chen Hu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wang F, Ren J, Qiu XC, Wang LF, Zhu Q, Zhang YQ, Huan Y, Meng YL, Yao LB, Chen SY, Xu YM, Yang AG. Selective Cytotoxicity to HER2-Positive Tumor Cells by a Recombinant e23sFv-TD-tBID Protein Containing a Furin Cleavage Sequence. Clin Cancer Res 2010; 16:2284-94. [DOI: 10.1158/1078-0432.ccr-09-2367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Liu X, Wu J, Zhang S, Li C, Huang Q. Novel strategies to augment genetically delivered immunotoxin molecular therapy for cancer therapy. Cancer Gene Ther 2009; 16:861-72. [PMID: 19461676 DOI: 10.1038/cgt.2009.30] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2008] [Revised: 01/01/2009] [Accepted: 02/19/2009] [Indexed: 11/09/2022]
Abstract
Immunotoxin therapy is a promising molecular cancer treatment strategy. Its main advantage is seletive cytotoxicity towards tumor cells and minimal toxicity in normal tissues. However, a short half-life and rapid clearance severely hampers its clinical application. We report here a novel genetic approach in which a recombinant adenovirus vector was used to deliver an immunotoxin gene e23(scFv)-PE40 targeted to the oncogene c-erbB-2 (also known as Her2/neu). This vector, when combined with a low dose of a conditionally replicative adenovirus vector (CRAd), has enhanced tumor-killing ability either alone or in combination with the chemotherapeutic agent etoposide. Our data show that low-dose CRAd facilitated the replication of replication-deficient Ad-e23(scFv)-PE40 up to 6-20 times and the transcription of e23(scFv)-PE40 gene up to 12 times. Moreover, etoposide increased the e23(scFv)-PE40 transcription up to 8.5 times. Furthermore, we show that systemic application of Ad-e23(scFv)-PE40 and enhanced expression of the immunotoxin gene was well tolerated as determined by serum biochemical markers and histological examination of most vital organs. Taken together, our data support a novel genetic immunotoxin delivery approach that may yield enhanced efficacy against a variety of Her2/neu-expressing tumors.
Collapse
Affiliation(s)
- X Liu
- Experimental Center, First People's Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | | | | | | | | |
Collapse
|
14
|
Hu CC, Ke YQ, Sun XL, Jiang XD, Xu RX, Lv J, Wang YS, Cai YQ, Qin LS, Zou YX. Human mesenchymal stem cells-like cells as cellular vehicles for delivery of immunotoxin in vitro. Biotechnol Lett 2008; 31:181-9. [DOI: 10.1007/s10529-008-9860-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Revised: 09/10/2008] [Accepted: 09/19/2008] [Indexed: 11/30/2022]
|
15
|
Wang T, Zhao J, Ren JL, Zhang L, Wen WH, Zhang R, Qin WW, Jia LT, Yao LB, Zhang YQ, Chen SY, Yang AG. Recombinant Immunoproapoptotic Proteins with Furin Site Can Translocate and Kill HER2-Positive Cancer Cells. Cancer Res 2007; 67:11830-9. [DOI: 10.1158/0008-5472.can-07-1160] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Yang Q, Tian Y, Liu S, Zeine R, Chlenski A, Salwen HR, Henkin J, Cohn SL. Thrombospondin-1 Peptide ABT-510 Combined with Valproic Acid Is an Effective Antiangiogenesis Strategy in Neuroblastoma. Cancer Res 2007; 67:1716-24. [PMID: 17308113 DOI: 10.1158/0008-5472.can-06-2595] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the pediatric cancer neuroblastoma, clinically aggressive disease is associated with increased levels of angiogenesis stimulators and high vascular index. We and others have hypothesized that blocking angiogenesis may be effective treatment for this pediatric malignancy. However, little is known about the efficacy of antiangiogenic agents in pediatric malignancies. Recently, promising results have been reported in an adult phase I study of ABT-510, a peptide derivative of the natural angiogenic inhibitor thrombospondin-1. Histone deacetylase inhibitors, such as valproic acid (VPA), have also been shown to have antiangiogenic activity in several cancer models. In this study, we evaluated the effects of ABT-510 and VPA on neuroblastoma tumor growth and angiogenesis. Although only VPA was capable of blocking the proliferation of neuroblastoma cells and inducing neuroblastoma cell apoptosis in vitro, treatment with VPA or ABT-510 alone significantly suppressed the growth of neuroblastoma xenografts established from two different MYCN-amplified cell lines. Combination therapy more effectively inhibited the growth of small neuroblastoma xenografts than single-agent treatment, and in animals with large xenografts, total cessation of tumor growth was achieved with this treatment approach. The microvascular density was significantly reduced in the xenografts treated with combination therapy compared with controls or tumors treated with single agents. In addition, the number of structurally abnormal vessels was reduced, suggesting that these agents may "normalize" the tumor vasculature. Our results indicate that ABT-510 combined with VPA may be an effective antiangiogenic treatment strategy for children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Qiwei Yang
- The Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, 5841 Maryland Avenue, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Nguyen van Binh P, Duc HT. Analyses and perspectives in cancer immunotherapy. Biomed Pharmacother 2006; 60:621-8. [PMID: 16978826 DOI: 10.1016/j.biopha.2006.07.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 07/28/2006] [Indexed: 01/15/2023] Open
Abstract
Since the last two decades, rapid progress has been made in the field of cancer immunotherapy relevant to manipulation of adaptative cytotoxic T lymphocytes (CTLs) and innate immunity natural killer (NK) cells as well as antibodies. Many possibilities are now offered for therapeutic purposes contributing to better approaches in treatment of cancer.
Collapse
Affiliation(s)
- P Nguyen van Binh
- Inserm, U602, Micro-environnement et Physiopathologie de la Différenciation, Hôpital Paul-Brousse, 16, avenue Paul-Vaillant-Couturier, 94807 Villejuif cedex, France
| | | |
Collapse
|
18
|
Wang L, Du F, Cao Q, Sheng H, Shen B, Zhang Y, Diao Y, Zhang J, Li N. Immunization with autologous T cells enhances in vivo anti-tumor immune responses accompanied by up-regulation of GADD45β. Cell Res 2006; 16:702-12. [PMID: 16826163 DOI: 10.1038/sj.cr.7310083] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Immunization with inactivated autoreactive T cells may induce idiotype anti-idiotypic reactions to deplete autoreactive T cells, which are involved in autoimmune diseases. However, it is unknown whether attenuated activated healthy autologous T-cell immunization could increase anti-tumor immune responses. To this end, C57Bl/6 mice were immunized with attenuated activated autologous T cells. The splenocytes from immunized mice showed a higher proliferative ability than that from naive mice. The special phenotype analysis showed that there were more CD8+ T cells and CD62L+ T cells in immunized mice after 24 h of culture with 10% fetal calf serum complete medium in vitro (P<0.01). These results demonstrated that this immunization may activate T cells in vivo. Furthermore, the splenocytes from immunized mice revealed resistance to activation-induced cell death (AICD) in vitro. To further study the relative genes that are responsible for the higher proliferation and resistance to AICD, the expression of Fas/Fas ligand (FasL) and GADD45b was measured by real-time PCR. The results indicated that GADD45beta transcription was higher in the splenocytes from immunized mice than that in the naive mice. In addition, the Fas expression showed a parallel higher, but FasL did not change obviously. To investigate the biologic functions induced by immunization in vivo, a tumor model was established by EL-4 tumor cell inoculation in C57/Bl mice. Mice receiving autologous T-cell immunization had significantly inhibited tumor growth in vivo (P<0.01). This study implicated that immunization with attenuated activated autologous T cells enhances anti-tumor immune responses that participate in tumor growth inhibition.
Collapse
Affiliation(s)
- Li Wang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Spronk HMH. Vitamin K epoxide reductase complex and vascular calcification: is this the important link between vitamin K and the arterial vessel wall? Circulation 2006; 113:1550-2. [PMID: 16567578 DOI: 10.1161/circulationaha.105.617167] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Yu CJ, Jia LT, Meng YL, Zhao J, Zhang Y, Qiu XC, Xu YM, Wen WH, Yao LB, Fan DM, Jin BQ, Chen SY, Yang AG. Selective proapoptotic activity of a secreted recombinant antibody/AIF fusion protein in carcinomas overexpressing HER2. Gene Ther 2006; 13:313-20. [PMID: 16267568 DOI: 10.1038/sj.gt.3302672] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Revised: 07/19/2005] [Accepted: 08/25/2005] [Indexed: 11/09/2022]
Abstract
Apoptosis-inducing factor (AIF) represents a caspase-independent apoptotic pathway in the cell, and a mitochondrial localization sequence-truncated AIF (AIFDelta1-120) can be relocated from the cytoplasm to the nucleus and exhibit a constitutive proapoptotic activity. Here, we generated a chimeric immuno-AIF protein, which comprised an HER2 antibody, a Pseudomonas exotoxin translocation domain and AIFDelta1-120. Human Jurkat cells transfected with the immuno-AIF gene could express and secrete the chimeric protein, which selectively recognized HER2-overexpressing tumor cells and was endocytosed. Subsequent cleavage of truncated AIF from immuno-AIF and its release from the internalized vesicles resulted in apoptosis of tumor cells. Intramuscular injection of the immuno-AIF gene caused significant suppression of tumors and substantially prolonged mice survival in an HER2-overexpressing xenograft tumor model. Our study demonstrates the feasibility of the immuno-AIF gene as a novel approach to treating cancers that overexpress HER2.
Collapse
Affiliation(s)
- C-J Yu
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Schurgers LJ, Teunissen KJF, Knapen MHJ, Kwaijtaal M, van Diest R, Appels A, Reutelingsperger CP, Cleutjens JPM, Vermeer C. Novel conformation-specific antibodies against matrix gamma-carboxyglutamic acid (Gla) protein: undercarboxylated matrix Gla protein as marker for vascular calcification. Arterioscler Thromb Vasc Biol 2005; 25:1629-33. [PMID: 15961706 DOI: 10.1161/01.atv.0000173313.46222.43] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Matrix gamma-carboxyglutamic acid (Gla) protein (MGP), a vitamin K-dependent protein, is a potent in vivo inhibitor of arterial calcification. We hypothesized that low endogenous production of MGP and impaired carboxylation of MGP may contribute to the development or the progression of vascular disease. METHODS AND RESULTS Novel conformation-specific antibodies against MGP were used for immunohistochemistry of healthy and sclerotic arteries. In healthy arteries, MGP was mainly displayed around the elastin fibers in the tunica media. The staining colocalized with that for carboxylated MGP, whereas undercarboxylated MGP (ucMGP) was not detected. In atherosclerotic arteries, ucMGP was found in the intima, where it was associated with vesicular structures. In Mönckeberg's sclerosis of the media, ucMGP was localized around all areas of calcification. The results indicate that ucMGP is strongly associated with vascular calcification of different etiologies. In a separate study, serum MGP concentrations in a cohort of 172 subjects who had undergone percutaneous coronary intervention were significantly reduced compared with an apparently healthy population. CONCLUSIONS These data show that impaired carboxylation of MGP is associated with intimal and medial vascular calcification and suggest the essentiality of the vitamin K modification to the function of MGP as an inhibitor of ectopic calcification.
Collapse
Affiliation(s)
- Leon J Schurgers
- Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yousefi M, Mattu R, Gao C, Man YG. Mammary ducts with and without focal myoepithelial cell layer disruptions show a different frequency of white blood cell infiltration and growth pattern: implications for tumor progression and invasion. Appl Immunohistochem Mol Morphol 2005; 13:30-7. [PMID: 15722791 DOI: 10.1097/00129039-200503000-00006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The authors' previous studies revealed that a subset of ductal carcinoma in situ (DCIS) contained focally disrupted myoepithelial (ME) cell layers and basement membrane (BM). As the disruption of these two structures is a prerequisite for tumor invasion, and white blood cells (WBCs) contain digestive enzymes capable of degrading both the BM and damaged host cells, this study was designed to assess the possible roles of WBC in ME cell layer disruptions and tumor invasion. A total of 23 DCIS containing ducts with focally disrupted ME cell layers were selected from 94 such cases identified in the authors' previous studies. Two consecutive sections from each case were double immunostained, one with leukocyte common antigen (LCA) plus smooth muscle actin (SMA) and the other with Ki-67 plus SMA. Ducts lined by at least 50 epithelial cells and distinct ME cell layers were examined. A total of 191 duct cross-sections were found to contain focal ME cell layer disruptions; of these, 186 (97.4%) were with and 5 (2.6%) were without WBC infiltration. Of 207 morphologically similar sections without ME disruptions, 46 (22.2%) were with and 161 (77.8%) were without WBC infiltration. Ki-67-positive cells in ducts with focally disrupted ME cell layers were generally subjacent to ME cell layers, and more than 30 clusters of multiple proliferating cells were seen directly overlying or near focally disrupted ME cell layers. In contrast, Ki-67-positive cells in ducts without ME disruptions were scattered over the entire epithelial compartment. The significantly different frequency of WBC infiltration and clusters of multiple proliferating cells in ducts with and without ME disruptions suggests that WBCs might play important roles in ME disruption and tumor invasion.
Collapse
Affiliation(s)
- Morvarid Yousefi
- Department of Gynecologic and Breast Pathology, Armed Forces Institute of Pathology and American Registry of Pathology, Washington, DC 20306-6000, USA
| | | | | | | |
Collapse
|
23
|
Abstract
Cancer is a difficult target for any therapeutic strategy; therefore, there is a continuous search for new therapeutic modalities, for application either alone or in combination. In this regard, gene-based therapy is a new approach that offers hope of improved control of tumors. Intensive research to apply gene therapy for cancer treatment has led to identification of the most important technical and theoretical barriers that need to be overcome for clinical success. One of the central unresolved challenges remains the issue of specific and efficient delivery of genes to target cells or tissues, emphasizing the importance of the gene carrier. Along with different viral and non-viral vector systems, mammalian cells have also been considered as vehicles for delivery of anti-cancer therapeutics. The cell-based delivery approach was introduced as the first attempt to apply gene therapy to cancer treatment, and in general, has followed most of the ups and downs of gene therapy applications, progressing alongside new knowledge gained in this field. As a result, significant progress has been made in some aspects of the cell-based approach, while the development of other essential issues is only just gaining speed. It appears that the initial phase of development of cell-based protocols - the achievement of efficient ex vivo cell loading with therapeutics - has largely been fulfilled. However, the desired efficacy of cell-based strategies in general has not yet been reached, and specificity of tumor homing needs to be improved considerably. There is hope that advances in related scientific fields will promote the utilization of cells as powerful and versatile vehicles for cancer gene therapy.
Collapse
Affiliation(s)
- Larisa Pereboeva
- Division of Human Gene Therapy, Department of Medicine, The Gene Therapy Center, BMRII-572, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL 35294, USA.
| | | |
Collapse
|
24
|
Xu YM, Wang LF, Jia LT, Qiu XC, Zhao J, Yu CJ, Zhang R, Zhu F, Wang CJ, Jin BQ, Chen SY, Yang AG. A caspase-6 and anti-human epidermal growth factor receptor-2 (HER2) antibody chimeric molecule suppresses the growth of HER2-overexpressing tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2004; 173:61-7. [PMID: 15210759 DOI: 10.4049/jimmunol.173.1.61] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Clinical studies have suggested that human epidermal growth factor receptor-2 (HER2) provide a useful target for antitumor therapy. We previously described the generation of a chimeric HER2-targeted immunocasp-3 protein. In this study, we extend the repertoire of chimeric proapoptotic proteins with immunocasp-6, a construct that comprises a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A, and an active caspase-6, which can directly cleave lamin A leading to nucleus damage and inducing programmed cell death. We demonstrate that the secreted immunocasp-6 molecule selectively recognizes and induces apoptosis in HER2-overexpressing tumor cells in vitro, but not in cells with undetectable HER2. The immunocasp-6 gene was next transferred into BALB/c athymic mice bearing human breast SK-BR-3 tumors by i.m. injection of liposome-encapsulated vectors, by intratumor injection of adenoviral vectors, or by i.v. injection of PBMC modified by retroviral infection. Regardless of the method used, expression of immunocasp-6 suppressed tumor growth and prolonged animal survival significantly. Our data show that the chimeric immunocasp-6 molecule can recognize HER2-positive tumor cells, promptly attack their nucleus, and induce their apoptotic death, suggesting the potential of this strategy for the treatment of human cancers that overexpress HER2.
Collapse
Affiliation(s)
- Yan-Ming Xu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 17 Changle West Road, 710-032 Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhao J, Zhang LH, Jia LT, Zhang L, Xu YM, Wang Z, Yu CJ, Peng WD, Wen WH, Wang CJ, Chen SY, Yang AG. Secreted Antibody/Granzyme B Fusion Protein Stimulates Selective Killing of HER2-overexpressing Tumor Cells. J Biol Chem 2004; 279:21343-8. [PMID: 15004021 DOI: 10.1074/jbc.m312648200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Targeted cell killing is required for effective treatment of cancers. We previously described the generation of a chimeric immunocasp-3 protein and its potent selective antitumor activity (Jia, L. T., Zhang, L. H., Yu, C. J., Zhao, J., Xu, Y. M., Gui, J. H., Jin, M., Ji, Z. L., Wen, W. H., Wang, C. J., Chen, S. Y., and Yang, A. G. (2003) Cancer Res. 63, 3257-3262). Here we extend the repertoire of another chimeric pro-apoptotic protein immunoGrB, which comprises an anti-HER2 single-chain antibody, a Pseudomonas exotoxin A translocation domain and active granzyme B. Human lymphoma Jurkat cells transfected with the immunoGrB gene expression vector were able to produce and secrete the chimeric protein. The immunoGrB molecule selectively recognized and destroyed HER2-overexpressing tumor cells both in vitro and in nude mouse after intramuscular injection of the immunoGrB expression plasmid. Further in vivo study showed that intravenous administration of immunoGrB gene-modified lymphocytes led to suppression of HER2-overexpressing tumor growth and prolonged animal survival because of continuous secretion of immunoGrB molecules into blood and lymph fluid. These results demonstrate that the chimeric immunoGrB molecule, which is capable of antibody-directed targeting and granzyme B-mediated killing, has therapeutic potential against HER2 tumors, especially in cases in which caspase-dependent apoptosis is inhibited.
Collapse
Affiliation(s)
- Jing Zhao
- Departments of Biochemistry and Molecular Biology and Immunology, the Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Vallera DA, Jin N, Shu Y, Panoskaltsis-Mortari A, Kelekar A, Chen W. Retroviral Immunotoxin Gene Therapy of Leukemia in Mice Using Leukemia-Specific T Cells Transduced with an Interleukin-3/Bax Fusion Protein Gene. Hum Gene Ther 2003; 14:1787-98. [PMID: 14670129 DOI: 10.1089/104303403322611791] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In past studies, we showed that T cells transduced with retroviral diphtheria immunotoxin (IT) target genes could serve as vehicles for delivering IT to tumors in vivo. We took advantage of the observation that antigen-specific T cells are able to penetrate tumors to design an approach delivering combined cellular and humoral therapy directly to the tumor site. To improve tumor specificity, we selected interleukin (IL)-3 as a ligand because its receptor is selectively overexpressed on myeloid leukemia progenitors. Because Bcl-2 family proteins show structural similarity to diphtheria toxin (DT), we constructed a unique retroviral IT using Bax, a proapoptotic member of the Bcl-2 family, in place of DT. Bax was chosen because several studies showed that its transduction induces lethal apoptosis in different cancers. The retroviral construct for gene therapy included IL-3 positioned downstream of its 80 amino acid leader, and permitted cotranslational protein synthesis of hybrid IL-3/human Bax fusion protein. Other vectors were constructed with IL-3 fused to DT or Pseudomonas exotoxin. Retroviral vectors were used to transiently transduce C8, a CD4(+) T cell clone that specifically recognized FBL-3, a lethal myeloid leukemia. Supernatants collected from transduced cells showed proapoptotic activity and selectively inhibited FBL-3 cells in vitro. Intraperitoneal injection of transduced but not nontransduced C8 into mice with subcutaneous tumors or systemic cancer significantly inhibited tumor growth. These results indicate that retroviral IT made with IL-3 and various toxic proteins may be useful in patients with acute myelogenous leukemia (AML). Furthermore, the Bax construct may be particularly useful as a nonimmunogenic substitute for bacterial toxins in retIT.
Collapse
Affiliation(s)
- Daniel A Vallera
- University of Minnesota Cancer Center, Department of Therapeutic Radiology, Section on Experimental Cancer Immunology, Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Cheng H, Liu YF, Zhang HZ, Shen WA, Zhang J, Zhang J. In vivo antitumour activity of PBMCs via genetic modification of single-chain immunotoxin. Shijie Huaren Xiaohua Zazhi 2003; 11:708-711. [DOI: 10.11569/wcjd.v11.i6.708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate In vivo antitumour activity of single-chain immunotoxin (sFv-TNF-α fusion protein).
METHODS HCC-specific killer cells were generated by transducing the recombinant retroviral virus in supernatant of the virus producing cells (C22) into human peripheral blood mononuclear cells (PBMCs). SMMC-7721 xenograft nude mice were given iv either 1×106 (0.2 mL) transduced or mock-transduced PBMCs once five days for three weeks and tumour growth was detected.
RESULTS Tumour growth were (20.8±4.9) mg/d in PBMCs/PST group and (28.5±6.7)mg/d in PBMCs/ pLXSN group, with a significant difference (P<0.05).
CONCLUSION Genetic modification of PBMCs by single-chain immunotoxin has antitumour activity In vivo.
Collapse
Affiliation(s)
- Hong Cheng
- Department of Pathology, Xijing Hospita Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Yan-Fang Liu
- Department of Pathology, Xijing Hospita Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Hui-Zhong Zhang
- Orthopeadics Oncology Institute of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, Shaanxi Province, China
| | - Wan-An Shen
- Orthopeadics Oncology Institute of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, Shaanxi Province, China
| | - Ju Zhang
- Department of Biochemistry, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Jing Zhang
- Department of Pathology, Xijing Hospita Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| |
Collapse
|
28
|
Rampaul RS, Pinder SE, Gullick WJ, Robertson JFR, Ellis IO. HER-2 in breast cancer--methods of detection, clinical significance and future prospects for treatment. Crit Rev Oncol Hematol 2003; 43:231-44. [PMID: 12270780 DOI: 10.1016/s1040-8428(01)00207-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The Human Epidermal Growth Factor (HER-2) oncogene encodes a transmembrane tyrosine kinase receptor with extensive homology to the Epidermal Growth Factor Receptor (EGFR) which is the prototypal member of this family of receptor tyrosine kinases. HER-2 gene amplification is found in 20-30% of breast cancers. Various methods such as immunohistochemistry, southern and slot blotting, enzyme immunoassays and fluorescence in situ hybridization have all been employed to evaluate HER-2 gene and protein abnormalities. Of these immunohistochemistry is the most frequently employed but there are valid indications for the other avaliable methods. However, it is prudent that whichever methods employed are standardized, especially those that possess may have a degree of subjectivity in their assesment.
Collapse
Affiliation(s)
- R S Rampaul
- Department of Surgery, Nottingham City Hospital, Nottingham, UK
| | | | | | | | | |
Collapse
|
29
|
Stöcker M, Tur MK, Sasse S, Krüssmann A, Barth S, Engert A. Secretion of functional anti-CD30-angiogenin immunotoxins into the supernatant of transfected 293T-cells. Protein Expr Purif 2003; 28:211-9. [PMID: 12699683 DOI: 10.1016/s1046-5928(02)00709-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Immunotoxins consist of a target-cell-specific binding moiety, chemically or recombinantly linked to a cytotoxic component. A number of different immunotoxins (IT) have increasingly been evaluated for immunotherapy. Since these foreign proteins are highly immunogenic in human, we have developed recombinant IT using the human ribonuclease angiogenin. Due to their potential toxic effects on eucaryotic cells, these IT are usually expressed in bacteria. Depending on the structure, size, and sequence of the desired IT, bacterial expression can be limited and the yield after purification be unsatisfactory. Therefore, the expression of IT in eucaryotic cells could provide a promising alternative. For this purpose we genetically fused the anti-CD30 single-chain variable fragment (scFv) Ki4 to the N- and C-termini of recombinant angiogenin. Both IT possess leader sequences, which mediate their secretion into the cell culture supernatant. Using a bicistronic mRNA the IT were simultaneously expressed together with enhanced green fluorescent protein (EGFP). This allows direct monitoring of transfected cells. An additional plasmid encoded Zeocin resistance enhances the cultivation of transfected cells under selection pressure. Three days after transfection of 293T-cells, unpurified IT were analyzed by flow cytometry and competitive cell proliferation assays. This is the first report on the use of eucaryotic cells for the secretion of functionally active IT with a human effector domain.
Collapse
Affiliation(s)
- Michael Stöcker
- Department of Pharmaceutical Product Development, Fraunhofer Institute for Molecular Biology and Applied Ecology, Worringerweg 1, 52074 Aachen, Germany.
| | | | | | | | | | | |
Collapse
|
30
|
Cheng H, Liu YF, Zhang HZ, Shen WA, Zhang J, Zhang J. In vitro cytotoxicity of PBMCs via genetic modification of single-chain immunotoxin. Shijie Huaren Xiaohua Zazhi 2003; 11:281-284. [DOI: 10.11569/wcjd.v11.i3.281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the selective cytotoxicity of single-chain immunotoxin (sFv-TNF-α fusion proteins) in cell line SMMC-7721.
METHODS: HCC-specific killer cells were generated by transducing the recombinant retroviral virus in supernatant of the virus producing cells into human peripheral blood mononuclear cells (PBMCs). PCR and RT-PCR were used to detect integration and transcription of the sFv-TNF-α gene in transduced PBMCs (PBMCs/PST). MTT method was used to detect antitumour activity of the sFv-TNF-α fusion proteins.
RESULTS: There was integrated sFv-TNF-α gene in the genome of PBMCs/PST, and PBMCs/PST were able to express the fusion sFv-TNF-α proteins. Cell killing was significant in HCC cells co-cultivated with PBMCs/PST, whereas the PBMCs/pLXSN control cells had no significant cytotoxic effects on HCC cells.
CONCLUSION: Expression of sFv-TNF-α fusion proteins in PBMCs/PST has cytotoxicity to HCC cells in vitro.
Collapse
|
31
|
Jin N, Chen W, Blazar BR, Ramakrishnan S, Vallera DA. Gene therapy of murine solid tumors with T cells transduced with a retroviral vascular endothelial growth factor--immunotoxin target gene. Hum Gene Ther 2002; 13:497-508. [PMID: 11874628 DOI: 10.1089/10430340252809793] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Solid tumor growth can be inhibited by targeting its neovasculature with vascular endothelial growth factor (VEGF)-toxin fusion proteins (FPs), but these agents have been limited by their inability to localize at the tumor site. In this study, we devised a gene therapy approach intended to deliver VEGF-toxin directly to tumor. Antigen-specific cytotoxic T lymphocytes (CTLs) served as vehicles to deliver a retroviral VEGF-toxin fusion protein to its specific leukemia cell target in vivo. A retroviral vector was constructed for gene therapy with VEGF positioned downstream of its 27-amino acid leader sequence, which promoted secretion of a catalytic immunotoxin containing either truncated diphtheria toxin or Pseudomonas exotoxin A. VEGF was chosen on the basis of the expression of VEGF receptor on endothelial cells in the tumor neovasculature. The VEGF FP was first expressed and secreted by mammalian NIH 3T3 cells. Intracellular expression of both VEGF and toxin was verified by immunofluorescence. In vitro, supernatants collected from transfected cells specifically inhibited the growth of VEGF receptor-expressing human umbilical vein endothelial cells (HUVECs), but not a control cell line. In vivo findings correlated with in vitro findings. A retroviral vector containing the target gene and a nerve growth factor receptor (NGFR) reporter gene was used to transiently transduce T15, a CD8(+) CTL line that specifically recognizes C1498, a lethal C57BL/6 myeloid tumor. Transduced T15 cells injected intravenously significantly inhibited the growth of subcutaneous tumor, whereas nontransduced controls did not. Together, these data indicate that gene therapy of T cells with retrovirus containing a VEGF-immunotoxin target gene may be a valid means of inhibiting a broad range of solid tumors dependent on angiogenesis.
Collapse
Affiliation(s)
- Ni Jin
- Section on Experimental Cancer Immunology, Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Cancer Center, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- N Bitton
- Laboratoire d'Immunologie Cellulaire, CERVI, INSERM U543, Hopital Pitie-Salpetriere, 83 Bvd de l'Hopital, 75013 Paris, France
| | | | | | | |
Collapse
|
33
|
Selbo PK, Sivam G, Fodstad O, Sandvig K, Berg K. In vivo documentation of photochemical internalization, a novel approach to site specific cancer therapy. Int J Cancer 2001; 92:761-6. [PMID: 11340584 DOI: 10.1002/1097-0215(20010601)92:5<761::aid-ijc1238>3.0.co;2-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Photochemical internalization (PCI) is a unique procedure for site-specific delivery of several types of membrane-impermeable molecules to the cytosol of target cells. The technology is based on photochemical-induced release of endocytosed macromolecules from endosomes and lysosomes into the cytosol. The purpose of this study was to evaluate the therapeutic potential of PCI of the type I ribosomal-inactivating protein gelonin in an animal model. The photosensitizer aluminum phthalocyanine disulfonate (AlPcS(2a)) was injected intraperitoneally (10 mg/kg) into athymic female BALB/c (nu/nu) nude mice (8-9 mice per group) with subcutaneously growing human adenocarcinoma (WiDr) tumors 48 hr before exposure to 135 J/cm(2) of red light focused on the tumor. Six hours before light exposure a single dose of 50 microg gelonin was administrated intratumorally. Tumor growth was measured at least twice a week. After immunomagnetic separation of in vivo growing tumor cells the subcellular localization of the photosensitizer was evaluated by fluorescence microscopy. The photosensitizer localized in endocytic vesicles in in vivo growing WiDr cells. Furthermore, it was found that in vitro gelonin treatment of WiDr cells isolated from photosensitizer-treated mice potentiated a light-induced decrease of clonal survival. Complete remission in 6 of 9 (67%) of the treated mice were induced. Our findings indicate that photochemical treatment with the photosensitizer AlPcS(2a) activates the cytotoxic potential of gelonin in vivo. These results demonstrate that the synergistic effect of combining photoactivation of photosensitizer located in endocytic vesicles and gelonin is indeed a result of PCI of gelonin.
Collapse
Affiliation(s)
- P K Selbo
- Department of Biophysics, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, N-0310 Oslo, Norway.
| | | | | | | | | |
Collapse
|
34
|
Abstract
Cardiovascular disease and osteoporosis together account for most of the morbidity and mortality in our aging population despite significant improvements in treatment. Recently, converging lines of evidence suggest that these 2 diseases share an etiologic factor--that hyperlipidemia contributes not only to atherosclerotic plaque formation, but also to osteoporosis, following a similar biologic mechanism involving lipid oxidation. In vitro studies indicate that lipid products of oxidation promote osteoblastic differentiation of vascular cells and inhibit such differentiation in bone cells. Ex vivo, in vivo, and clinical studies further suggest that lipid-lowering agents reduce both atherosclerotic calcification and osteoporosis. Whether lipid-lowering agents reduce osteoporosis directly or indirectly through lipid reduction remains controversial.
Collapse
Affiliation(s)
- F Parhami
- Division of Cardiology, Department of Medicine, UCLA School of Medicine, Los Angeles, California, USA
| | | | | |
Collapse
|
35
|
You Z, Huang XF, Hester J, Rollins L, Rooney C, Chen SY. Induction of vigorous helper and cytotoxic T cell as well as B cell responses by dendritic cells expressing a modified antigen targeting receptor-mediated internalization pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4581-91. [PMID: 11035100 DOI: 10.4049/jimmunol.165.8.4581] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Efficient Ag presentation is essential to induce effective cellular and humoral immune responses. Thus, one central goal of current immunotherapy and vaccine development is to enhance Ag presentation to induce potent and broad immune responses. Here, a novel Ag presentation strategy is developed by transducing dendritic cells (DCs) to produce an Ag for presentation as an exogenous Ag to efficiently induce both humoral and cellular immunity. The principle of this strategy is illustrated by genetically modifying DCs to secrete a model hepatitis B virus Ag fused with a cell-binding domain and to process the fusion Ag as an exogenous Ag after receptor-mediated internalization for MHC class I and II presentation. Vigorous Ag-specific CD4(+) helper and CD8(+) cytotoxic T cell, as well as B cell, responses were induced by the transduced DCs in mouse models. Thus, this novel strategy uses a receptor-mediated internalization process to efficiently induce all arms of the adaptive immunity and may provide a powerful means to develop potent vaccines and immunotherapies.
Collapse
Affiliation(s)
- Z You
- Center for Cell and Gene Therapy, Department of Molecular and Human Genetics, and Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
36
|
Liu YY, Gordienko I, Mathias A, Ma S, Thompson J, Woo JH, Neville DM. Expression of an anti-CD3 single-chain immunotoxin with a truncated diphtheria toxin in a mutant CHO cell line. Protein Expr Purif 2000; 19:304-11. [PMID: 10873546 DOI: 10.1006/prep.2000.1255] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ADP-ribosylating immunotoxins are generally expressed in Escherichia coli and then refolded in vitro. Because the efficiency of the in vitro refolding process decreases with the number of protein domains and internal disulfide bonds, these immunotoxins have been generally limited to single-chain monovalent structures. We now show that using the hamster cell line CHO K1 RE1.22c (J. M. Moehring and T. J. Moehring, 1979, Somat. Cell Genet. 5, 453-468) that has been mutated to ADP-ribosylation insensitivity, a level of 4 microg/ml of a truncated anti-T cell immunotoxin, DT390-scFvUCHT1, can be secreted into the medium. This immunotoxin is glycosylated at the two potential N-linked glycosylation sites in the toxin moiety: positions 16-18 in the A chain and residues 235-237 in the B chain. The glycosylated immunotoxin is relatively nontoxic (IC(50) 4.8 x 10(-10) M). Removal of the N-linked oligosaccharides by N-glycosidase F treatment or mutations at the two N-linked glycosylation sites results in a highly active immunotoxin with an IC(50) of 4 x 10(-12) M toward CD3(+) Jurkat cells. This is a 12-fold increase in toxicity over the same immunotoxin harvested from E. coli periplasm without refolding. A single Asn(235) Ala mutation that removed the B chain glycosylation was nearly as toxic as the double mutant. This suggests that B chain glycosylation is the major cause for the loss of toxicity.
Collapse
Affiliation(s)
- Y Y Liu
- Section on Biophysical Chemistry, National Institute of Mental Health, Bethesda, Maryland, 28092-4034, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Fabbrini MS, Carpani D, Soria MR, Ceriotti A. Cytosolic immunization allows the expression of preATF-saporin chimeric toxin in eukaryotic cells. FASEB J 2000; 14:391-8. [PMID: 10657995 DOI: 10.1096/fasebj.14.2.391] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In this work, we have devised an intracellular immunization strategy for the expression in high amounts of ATF-saporin, a targeted chimeric toxin constituted by the ATF receptor binding domain of human urokinase and the plant ribosome-inactivating protein saporin, which has been shown to be highly cytotoxic to target cells. This strategy may allow the production of highly toxic secretory proteins in eukaryotic cells, avoiding cell suicide caused by autointoxication. The procedure consists of equipping host cells with cytosolic neutralizing antibodies directed toward the toxic domain of the heterologous polypeptide. We show that this intracellular immunization is essential for the synthesis of correctly folded, biologically active ATF-SAP in the high amounts needed to investigate its in vivo anti-metastatic potential. Such a strategy should be generally useful for the production of toxic molecules of therapeutic value whose folding and maturation require transit through the eukaryotic secretory pathway. Fabbrini, M. S., Carpani, D., Soria, M. R., Ceriotti, A. Cytosolic immunization allows the expression of preATF-saporin chimeric toxin in eukaryotic cells.
Collapse
Affiliation(s)
- M S Fabbrini
- Department of Biological and Technological Research-Dibit, San Raffaele Scientific Institute, 20132 Milano, Italy.
| | | | | | | |
Collapse
|
38
|
Matthews IT. Antibodies for neoplastic disease : solid tumors. METHODS IN MOLECULAR MEDICINE 2000; 40:73-84. [PMID: 21337084 DOI: 10.1385/1-59259-076-4:73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The ease with which polyclonal, monoclonal, and engineered antibody fragments can be prepared allows access to a series of reagents with high selectivity and affinity. These reagents therefore have long held promise as a means of influencing the growth and spread of malignant disease. Over a number of decades therapeutic antibodies have been developed either as single entities or conjugated to a variety of potential disease-ameliorating agents.
Collapse
|
39
|
Castro MG, Davis JR, Xiong W, Lowenstein PR. Recent developments in gene therapy: applications for the treatment of pituitary tumours. BAILLIERE'S BEST PRACTICE & RESEARCH. CLINICAL ENDOCRINOLOGY & METABOLISM 1999; 13:431-49. [PMID: 10909434 DOI: 10.1053/beem.1999.0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pituitary tumours are normally benign, highly differentiated and slow growing neoplasms. Nevertheless, as many as half of them will show evidence of local invasion into the surrounding structures. Despite their benign growth characteristics and slow clinical progression, pituitary tumours commonly cause serious morbidity. The mass effects of large tumours, including headache and visual failure from optic chiasm compression, may cause lifelong disability. Hormone hypersecretion or deficiency causes major clinical problems that often require expensive and long-term medical therapy. Major advances have been made in the therapy of pituitary tumours over the past 20-30 years, but despite this, their treatment often remains an unsatisfactory compromise in practice. There is, therefore, a place for improvements in therapy, and to this end, gene therapy may come to hold a significant place in the future treatment of human pituitary tumours. With the development of new gene delivery vehicles, this concept can now be explored with a view to treating specific types of pituitary tumours.
Collapse
Affiliation(s)
- M G Castro
- Molecular Medicine and Gene Therapy Unit, School of Medicine, University of Manchester, UK
| | | | | | | |
Collapse
|
40
|
Vallera DA, Seo SY, Panoskaltsis-Mortari A, Griffin JD, Blazar BR. Targeting myeloid leukemia with a DT(390)-mIL-3 fusion immunotoxin: ex vivo and in vivo studies in mice. PROTEIN ENGINEERING 1999; 12:779-85. [PMID: 10506288 DOI: 10.1093/protein/12.9.779] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The IL-3 receptor was expressed on a high frequency of myeloid leukemia cells and also on hematopoietic and vascular cells. We previously showed that a recombinant IL-3 fusion immunotoxin (DT(390)IL-3) expressed by splicing the murine IL-3 gene to a truncated diphtheria toxin (DT(390)) gene selectively killed IL-3R(+) expressing cells and was not uniformly toxic to uncommitted BM progenitor cells (Chan,C.-H., Blazar,B.R., Greenfield,L., Kreitman,R.J. and Vallera,D.A., 1996, Blood, 88, 1445-1456). Thus, we explored the feasibility of using DT(390)IL-3 as an anti-leukemia agent. DT(390)IL-3 was toxic when administered to mice at doses as low as 0.1 microg/day. The dose limiting toxicity appeared to be related to platelet and bleeding effects of the fusion toxin. Because of these effects, DT(390)IL-3 was studied ex vivo as a means of purging contaminating leukemia cells from BM grafts in a murine autologous BM transplantation. In this setting, as few as 1000 IL-3R-expressing, bcr/abl transformed myeloid 32Dp210 leukemia cells were lethal. An optimal purging interval of 10 nM/l for 8 h eliminated leukemia cells from 32Dp210/BM mixtures given to lethally irradiated (8 Gy) C3H/HeJ syngeneic mice. Mice given treated grafts containing BM and a lethal dose of 32Dp210 cells survived over 100 days while mice given untreated grafts did not survive (P < 0.00001). DT(390)IL-3 may prove highly useful for ex vivo purging of lethal malignant leukemia cells from autologous BM grafts.
Collapse
Affiliation(s)
- D A Vallera
- University of Minnesota Cancer Center, Department of Therapeutic Radiology (Section on Experimental Cancer Immunology) Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
41
|
|
42
|
Klapper LN, Kirschbaum MH, Seta M, Yarden Y. Biochemical and Clinical Implications of the ErbB/HER Signaling Network of Growth Factor Receptors. Adv Cancer Res 1999. [DOI: 10.1016/s0065-230x(08)60784-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
43
|
Watson KE, Parhami F, Shin V, Demer LL. Fibronectin and collagen I matrixes promote calcification of vascular cells in vitro, whereas collagen IV matrix is inhibitory. Arterioscler Thromb Vasc Biol 1998; 18:1964-71. [PMID: 9848891 DOI: 10.1161/01.atv.18.12.1964] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular calcification is a frequent component of atherosclerosis, yet the pathological mechanisms that regulate its formation are poorly understood. Calcification of the vessel wall may represent a process by which cells that normally exhibit a smooth muscle phenotype differentiate into cells that exhibit an osteoblast-like phenotype. One of the determinants of cellular phenotype is extracellular matrix; thus, we undertook the current study to evaluate the influence of extracellular matrix on calcification of vascular cells in vitro. Cell lines derived from bovine aortic media were divided into 1 of 3 groups: those that did not mineralize, those that mineralized slowly, or those that mineralized rapidly. When slowly mineralizing cells were plated onto matrix produced by rapidly mineralizing cells, the time required for mineralization decreased from 33+/-3.0 days to 7.8+/-1.3 days. Matrix produced by rapidly mineralizing cells was found to contain 3 times the amount of collagen I and fibronectin but 70% less collagen IV than nonmineralizing clones. When slowly mineralizing cells were cultured on purified collagen I or fibronectin, mineralized nodule formation, calcium incorporation, von Kossa staining, and alkaline phosphatase activity increased. In contrast, culturing slowly mineralizing cells on purified collagen IV inhibited these mineralization parameters. Furthermore, blocking antibodies to alpha5 integrins significantly inhibited the fibronectin-mediated increases in alkaline phosphatase activity, indicating that integrin-based signaling may be involved. These data suggest that matrix composition can regulate development of arterial calcification and that a subpopulation of vascular cells preferentially produces positively regulating matrix components.
Collapse
Affiliation(s)
- K E Watson
- Division of Cardiology, Departments of Medicine and Physiology, UCLA School of Medicine, Los Angeles, CA, USA.
| | | | | | | |
Collapse
|
44
|
Vogt U, Bielawski K, Schlotter CM, Bosse U, Falkiewicz B, Podhajska AJ. Amplification of erbB-4 oncogene occurs less frequently than that of erbB-2 in primary human breast cancer. Gene 1998; 223:375-80. [PMID: 9858771 DOI: 10.1016/s0378-1119(98)00454-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
ErbB-4 protein is a recently discovered member of the ErbB family. The role of ErbB-4 protein in mammary-gland tissue has not been definitively established. To date, the expression of erbB-4 in breast tissue has been determined in only a few cases and, to the best of our knowledge, its amplification has not been examined. We therefore used the double differential polymerase chain reaction (ddPCR) for determination of the amplification profile of erbB-4 and erbB-2, another gene from the ErbB family, in human primary breast cancer specimens. We examined the amplification of the genes in 20 normal breasts and 176 invasive breast cancer samples. Amplification of erbB-2 was detected in 19% and erbB-4 in 13% of the samples studied. Co-amplification of the two oncogenes was found in only five out of 176 samples. Human breast cancer-derived cell lines in most cases overexpress both erbB-2 and erbB-4 (Beerli et al., 1995. Mol. Cell Biol. 15, 6496-6505; Han et al., 1995. Proc. Natl. Acad. Sci. USA 92, 9747-9751), but data on separate erbB-2 overexpression, without overexpression of erbB-4, were also reported (Wosikowski et al., 1997. Clin. Cancer Res. 3, 2405-2414). At the gene level, we found that co-amplification of the genes in the case of human breast cancer is rare. Moreover, an inverse association of the erbB-4 amplification with estrogen receptor activity and direct correlation with the tumor size were found. Due to these correlations, erbB-4 oncogene amplification can be assumed to be of prognostic or predictive value in the diagnosis of breast cancer.
Collapse
Affiliation(s)
- U Vogt
- European Laboratory Association Section Ibbenbüren, Roggenkampstr. 10, 49477, Ibbenbüren,
| | | | | | | | | | | |
Collapse
|
45
|
Cao GW, Qi ZT, Pan X, Zhang XQ, Miao XH, Feng Y, Lu XH, Kuriyama S, Du P. Gene therapy for human colorectal carcinoma using human CEA promoter controled bacterial ADP-ribosylating toxin genes: PEA and DTA gene transfer. World J Gastroenterol 1998; 4:388-391. [PMID: 11819328 PMCID: PMC4767734 DOI: 10.3748/wjg.v4.i5.388] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a tissue-specific gene therapy for colorectal carcinoma using bacterial ADP-ribosylating toxin genes.
METHODS: Pseudomonas exotoxin A domain II +III (PEA) was cloned from genomic DNA of Pseudomonas aeruginosa. PEA and diphtheria toxin A chain gene (DTA) were modified to express eukaryotically. After sequencing, the toxin genes under the control of human carcinoembryonic antigen (CEA) promoter were cloned into retroviral vectors to construct CEAPEA and CEADTA respectively. In vitro cotransfection of the constructs with luciferase vectors and in vivo gene transfer in nude mice were subsequently carried out.
RESULTS: Both CEAPEA and CEADTA specifically inhibited the reporter gene expression in the CEA positive human colorectal carcinoma (CRC) cells in vitro. Direct injection of CEAPEA and CEADTA constructs into the established human tumors in BALB/c nude mice led to significant and selective reductions in CRC tumor size as compared with that in control groups.
CONCLUSION: The toxin genes, working as therapeutic genes, are suitable for the tissue-specific gene therapy for colorectal carcinoma.
Collapse
|
46
|
Gorski JP. Is all bone the same? Distinctive distributions and properties of non-collagenous matrix proteins in lamellar vs. woven bone imply the existence of different underlying osteogenic mechanisms. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 1998; 9:201-23. [PMID: 9603236 DOI: 10.1177/10454411980090020401] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The purpose of this review is to summarize recent functional and structural findings regarding non-collagenous matrix proteins in bone and teeth, to compare gene locations for bone and tooth matrix proteins with loci for hereditary skeletal diseases, and to present several provocative hypotheses which integrate this new information into a physiological context. Hypothesis I proposes that the molecular composition of rapidly deposited and mineralized woven bone, as well as the responsiveness of cells synthesizing woven bone to stimuli, is different from that for more slowly synthesized lamellar bone, implying the existence of distinctive osteogenic mechanisms. This review of recent research strongly supports this proposal. Briefly, the protein composition of woven bone matrix is enriched in acidic phosphoproteins BAG-75 and BSP, which are not expressed in lamellar bone, which is itself enriched in osteocalcin. De novo deposition and mineralization of woven bone occurs faster than in lamellar bone by means of a matrix-vesicle-assisted mechanism. Deposition of woven bone occurs at sites experiencing biomechanical strains higher than those experienced by lamellar bone. In addition, woven bone in metaphyseal regions is more susceptible to osteoclastic resorption after space flight, ovariectomy, and loss of weightbearing than is lamellar bone. Finally, osteoprogenitor cells responsive to parathyroid hormone reside in the metaphyseal region of long bones. Taken together, these findings suggest that Hypothesis I represents a useful paradigm for future studies. Specific functions mediated by most individual bone and tooth matrix proteins remain uncertain. A review of current literature suggests that the functionality of skeletal matrix proteins is expressed through specific binding sites composed of particular species-conserved structural motifs (Hypothesis 2). Examples include the previously recognized Asp-Ser-Ser motif of dentin phosphophoryns and the gamma-carboxyglutamic acid motif of matrix GLA protein and osteocalcin. A new polyacidic amino acid motif composed of consecutive Asp and Glu residues (n > 7) was defined in extracellular matrix components osteopontin, bone sialoprotein, and bone acidic glycoprotein-75 on the basis of strong functional analogies with similar polyacidic stretches in divalent metal storage proteins of the endoplasmic reticulum and sarcoplasmic reticulum. These structural motifs represent prime targets for future structure-function studies in vivo and in vitro.
Collapse
Affiliation(s)
- J P Gorski
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, 64110, USA
| |
Collapse
|
47
|
Alami M, Taupiac MP, Reggio H, Bienvenüe A, Beaumelle B. Involvement of ATP-dependent Pseudomonas exotoxin translocation from a late recycling compartment in lymphocyte intoxication procedure. Mol Biol Cell 1998; 9:387-402. [PMID: 9450963 PMCID: PMC25269 DOI: 10.1091/mbc.9.2.387] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/1997] [Accepted: 11/24/1997] [Indexed: 02/06/2023] Open
Abstract
Pseudomonas exotoxin (PE) is a cytotoxin which, after endocytosis, is delivered to the cytosol where it inactivates protein synthesis. Using diaminobenzidine cytochemistry, we found over 94% of internalized PE in transferrin (Tf) -positive endosomes of lymphocytes. When PE translocation was examined in a cell-free assay using purified endocytic vesicles, more than 40% of endosomal 125I-labeled PE was transported after 2 h at 37 degrees C, whereas a toxin inactivated by point mutation in its translocation domain was not translocated. Sorting of endosomes did not allow cell-free PE translocation, whereas active PE transmembrane transport was observed after > 10 min of endocytosis when PE and fluorescent-Tf were localized by confocal immunofluorescence microscopy within a rab5-positive and rab4- and rab7-negative recycling compartment in the pericentriolar region of the cell. Accordingly, when PE delivery to this structure was inhibited using a 20 degrees C endocytosis temperature, subsequent translocation from purified endosomes was impaired. Translocation was also inhibited when endosomes were obtained from cells labeled with PE in the presence of brefeldin A, which caused fusion of translocation-competent recycling endosomes with translocation-incompetent sorting elements. No PE processing was observed in lymphocyte endosomes, the full-sized toxin was translocated and recovered in an enzymatically active form. ATP hydrolysis was found to directly provide the energy required for PE translocation. Inhibitors of endosome acidification (weak bases, protonophores, or bafilomycin A1) when added to the assay did not significantly affect 125I-labeled PE translocation, demonstrating that this transport is independent of the endosome-cytosol pH gradient. Nevertheless, when 125I-labeled PE endocytosis was performed in the presence of one of these molecules, translocation from endosomes was strongly inhibited, indicating that exposure to acidic pH is a prerequisite for PE membrane traversal. When applied during endocytosis, treatments that protect cells against PE intoxication (low temperatures, inhibitors of endosome acidification, and brefeldin A) impaired 125I-labeled PE translocation from purified endosomes. We conclude that PE translocation from a late receptor recycling compartment is implicated in the lymphocyte intoxication procedure.
Collapse
Affiliation(s)
- M Alami
- UMR 5539 Centre National de la Recherche Scientifique, Département Biologie-Santé, Université Montpellier II, Montpellier, France
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Juhl H, Downing SG, Wellstein A, Czubayko F. HER-2/neu is rate-limiting for ovarian cancer growth. Conditional depletion of HER-2/neu by ribozyme targeting. J Biol Chem 1997; 272:29482-6. [PMID: 9368008 DOI: 10.1074/jbc.272.47.29482] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Amplification and overexpression of the HER-2/neu proto-oncogene frequently coincide with an aggressive clinical course of certain human adenocarcinomas. To assess whether HER-2/neu plays a rate-limiting role in ovarian cancer, we used human SK-OV-3 ovarian cancer cells as a model. We applied a conditional mRNA depletion strategy of HER-2/neu with anti-HER-2/neu-targeted hammerhead ribozymes expressed under the control of a tetracycline-regulated promoter system. In these ovarian cancer cells, we reduced HER-2/neu mRNA, protein expression, and tumor growth in nude mice by transfection with HER-2/neu-targeted ribozymes and generated cell lines expressing different levels of HER-2/neu. Expression of the most effective ribozyme (Rz3) quenched HER-2/neu mRNA levels by >90%. Concomitantly, fluorescence-activated cell sorting analysis revealed that expression of the HER-2/neu-encoded surface glycoprotein was almost completely abrogated. In nude mice, tumor growth was dramatically inhibited in the HER-2/neu-depleted Rz3-expressing SK-OV-3 cells. Furthermore, already established tumors started to regress when Rz3 expression was activated midstream by withdrawal of the tetracycline treatment. This study supports the thesis that HER-2/neu can be rate-limiting for the malignant phenotype of ovarian cancer in a gene dose-dependent manner.
Collapse
Affiliation(s)
- H Juhl
- Lombardi Cancer Center and Department of Pharmacology, Georgetown University, Washington, D. C. 20007, USA
| | | | | | | |
Collapse
|
50
|
Yang AG, Bai X, Huang XF, Yao C, Chen S. Phenotypic knockout of HIV type 1 chemokine coreceptor CCR-5 by intrakines as potential therapeutic approach for HIV-1 infection. Proc Natl Acad Sci U S A 1997; 94:11567-72. [PMID: 9326650 PMCID: PMC23540 DOI: 10.1073/pnas.94.21.11567] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/1997] [Indexed: 02/05/2023] Open
Abstract
A genetic defect in a CC-chemokine receptor (CCR)-5, the principal coreceptor for the macrophage-tropic HIV type 1 (HIV-1), recently was found to naturally protect CCR-5-defective, but healthy, individuals from HIV-1 infection. In this study, we mimic the natural resistance of the CCR-5-defective individuals by designing a strategy to phenotypically knock out CCR-5. The inactivation of the CCR-5 coreceptor is accomplished by targeting a modified CC-chemokine to the endoplasmic reticulum to block the surface expression of newly synthesized CCR-5. The lymphocytes transduced to express the intracellular chemokine, termed "intrakine," were found to be viable and resistant to macrophage-tropic HIV-1 infection. Thus, this gene-based intrakine strategy targeted at the conserved cellular receptor for the prevention of HIV-1 entry should have significant advantages over currently described approaches for HIV-1 therapy.
Collapse
Affiliation(s)
- A G Yang
- Department of Cancer Biology, Comprehensive Cancer Center, Bowman Gray School of Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|