1
|
Lv T, Zhang H. Mitophagy-related gene signature for predicting the prognosis of multiple myeloma. Heliyon 2024; 10:e24520. [PMID: 38317923 PMCID: PMC10838706 DOI: 10.1016/j.heliyon.2024.e24520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/26/2023] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Aims The aims of this study were to explore the molecular mechanism of mitophagy in multiple myeloma (MM) and to develop an effective prognostic signature for the disease based on mitophagy-related genes (MRGs). Methods Three gene sets from the Reactome database were used to explore MRGs, following which those that were differentially expressed between MM and normal samples were investigated using the data from the Genomic Data Commons-Multiple Myeloma Research Foundation-CoMMpass Study. Mitophagy-related molecular subtypes of MM were identified and their immune infiltration, associated patient survival rates, immune checkpoint genes, and mitophagy scores were compared. Prognostic genes for MM were identified, and a prognostic model was constructed. Additionally, a nomogram was constructed using the prognostic model and prognosis-related clinical features. Finally, the drug sensitivity and correlation analyses of the subtypes were performed between the two risk groups. Results We identified two MM molecular subtypes that exhibited significant differences in mitophagy scores, associated patient survival rates, immune infiltration, and immune checkpoint genes. An MRG-based prognostic signature was constructed using six genes (TRIP13, KIF7, GPR63, CRIP2, DNTT, and HSPB8), which had high predictive prognostic value. A nomogram was constructed by screening five indicators (risk score, subtype, age, sex, and stage) that could predict the 1-, 3-, and 5-year survival probabilities of patients with MM. The two risk groups displayed significant differences in their IC50 values of 33 drugs, such as bleomycin. Patients in the high-risk group tended to fall within Mitophagy_cluster_A. Conclusion Our MRG-based signature is a promising prognostic biomarker for MM.
Collapse
Affiliation(s)
- Tiange Lv
- Cadre's Ward, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110015, China
| | - Haocong Zhang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110015, China
| |
Collapse
|
2
|
Blalock LT, Landsberg J, Messmer M, Shi J, Pardee AD, Haskell R, Vujanovic L, Kirkwood JM, Butterfield LH. Human dendritic cells adenovirally-engineered to express three defined tumor antigens promote broad adaptive and innate immunity. Oncoimmunology 2021; 1:287-357. [PMID: 22737604 PMCID: PMC3382861 DOI: 10.4161/onci.18628] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dendritic cell (DC) immunotherapy has shown a promising ability to promote anti-tumor immunity in vitro and in vivo. Many trials have tested single epitopes and single antigens to activate single T cell specificities, and often CD8(+) T cells only. We previously found that determinant spreading and breadth of antitumor immunity correlates with improved clinical response. Therefore, to promote activation and expansion of polyclonal, multiple antigen-specific CD8(+) T cells, as well as provide cognate help from antigen-specific CD4(+) T cells, we have created an adenovirus encoding three full length melanoma tumor antigens (tyrosinase, MART-1 and MAGE-A6, "AdVTMM"). We previously showed that adenovirus (AdV)-mediated antigen engineering of human DC is superior to peptide pulsing for T cell activation, and has positive biological effects on the DC, allowing for efficient activation of not only antigen-specific CD8(+) and CD4(+) T cells, but also NK cells. Here we describe the cloning and testing of "AdVTMM2," an E1/E3-deleted AdV encoding the three melanoma antigens. This novel three-antigen virus expresses mRNA and protein for all antigens, and AdVTMM-transduced DC activate both CD8(+) and CD4(+) T cells which recognize melanoma tumor cells more efficiently than single antigen AdV. Addition of physiological levels of interferon-α (IFNα) further amplifies melanoma antigen-specific T cell activation. NK cells are also activated, and show cytotoxic activity. Vaccination with multi-antigen engineered DC may provide for superior adaptive and innate immunity and ultimately, improved antitumor responses.
Collapse
Affiliation(s)
- Leeann T Blalock
- Department of Medicine; University of Pittsburgh; Pittsburgh, PA USA
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Domingo-Musibay E, Yamamoto M. Gene and virotherapy for hematological malignancies. Int J Hematol 2016; 104:29-41. [PMID: 27289361 PMCID: PMC5089843 DOI: 10.1007/s12185-016-2031-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 05/22/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022]
Abstract
Recent years have seen a transformation in the treatment of hematological malignancies. Advances in gene therapy and molecular techniques and significant gains in computational abilities have supported the rapid development of safer and better tolerated therapies for many patients with hematologic cancers. In this review, we discuss novel applications of gene therapy, including immunomodulation and gene silencing, and report on the rise of oncolytic viruses for use in the treatment of malignancies arising in cells of the blood, lymph, and marrow. We discuss the relationship of the tropism of wildtype viruses and their oncolytic behavior as well as the tumoricidal and immunostimulatory properties of a number of attenuated and recombinant viruses currently in clinical development in countries around the world. While we have focused on promising virotherapy applications for future development, we also present a historical perspective and identify areas of potential clinical and regulatory practice change. We outline several of the virus systems being developed for applications in hematology, and summarize efficacy data in the context of ongoing or future human clinical testing. We also present the advantages and limitations of gene and virus therapy, including challenges and opportunities for improved treatment tolerability and outcomes for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Evidio Domingo-Musibay
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Masato Yamamoto
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota, MoosT 11-210, MMC195, 515 Delaware St SE, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
4
|
Patel JM, Vartabedian VF, Bozeman EN, Caoyonan BE, Srivatsan S, Pack CD, Dey P, D'Souza MJ, Yang L, Selvaraj P. Plasma membrane vesicles decorated with glycolipid-anchored antigens and adjuvants via protein transfer as an antigen delivery platform for inhibition of tumor growth. Biomaterials 2015; 74:231-44. [PMID: 26461116 DOI: 10.1016/j.biomaterials.2015.09.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 09/19/2015] [Accepted: 09/23/2015] [Indexed: 02/05/2023]
Abstract
Antigen delivered within particulate materials leads to enhanced antigen-specific immunity compared to soluble administration of antigen. However, current delivery approaches for antigen encapsulated in synthetic particulate materials are limited by the complexity of particle production that affects stability and immunogenicity of the antigen. Herein, we describe a protein delivery system that utilizes plasma membrane vesicles (PMVs) derived from biological materials such as cultured cells or isolated tissues and a simple protein transfer technology. We show that these particulate PMVs can be easily modified within 4 h by a protein transfer process to stably incorporate a glycosylphosphatidylinositol (GPI)-anchored form of the breast cancer antigen HER-2 onto the PMV surface. Immunization of mice with GPI-HER-2-modified-PMVs induced strong HER-2-specific antibody responses and protection from tumor challenge in two different breast cancer models. Further incorporation of the immunostimulatory molecules IL-12 and B7-1 onto the PMVs by protein transfer enhanced tumor protection and induced beneficial Th1 and Th2-type HER-2-specific immune responses. Since protein antigens can be easily converted to GPI-anchored forms, these results demonstrate that isolated plasma membrane vesicles can be modified with desired antigens along with immunostimulatory molecules by protein transfer and used as a vaccine delivery vehicle to elicit potent antigen-specific immunity.
Collapse
Affiliation(s)
- Jaina M Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vincent F Vartabedian
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Erica N Bozeman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brianne E Caoyonan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sanjay Srivatsan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher D Pack
- Metaclipse Therapeutics Corporation, 3175 Presidential Drive, Atlanta, GA 30340, USA
| | - Paulami Dey
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Martin J D'Souza
- Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
5
|
Bozeman EN, Cimino-Mathews A, Machiah DK, Patel JM, Krishnamoorthy A, Tien L, Shashidharamurthy R, Selvaraj P. Expression of membrane anchored cytokines and B7-1 alters tumor microenvironment and induces protective antitumor immunity in a murine breast cancer model. Vaccine 2013; 31:2449-56. [PMID: 23541884 DOI: 10.1016/j.vaccine.2013.03.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/25/2013] [Accepted: 03/15/2013] [Indexed: 01/08/2023]
Abstract
Many studies have shown that the systemic administration of cytokines or vaccination with cytokine-secreting tumors augments an antitumor immune response that can result in eradication of tumors. However, these approaches are hampered by the risk of systemic toxicity induced by soluble cytokines. In this study, we have evaluated the efficacy of 4TO7, a highly tumorigenic murine mammary tumor cell line, expressing glycosyl phosphatidylinositol (GPI)-anchored form of cytokine molecules alone or in combination with the costimulatory molecule B7-1 as a model for potential cell or membrane-based breast cancer vaccines. We observed that the GPI-anchored cytokines expressed on the surface of tumor cells greatly reduced the overall tumorigenicity of the 4TO7 tumor cells following direct live cell challenge as evidenced by transient tumor growth and complete regression within 30 days post challenge. Tumors co-expressing B7-1 and GPI-IL-12 grew the least and for the shortest duration, suggesting that this combination of immunostimulatory molecules is most potent. Protective immune responses were also observed following secondary tumor challenge. Further, the 4TO7-B7-1/GPI-IL-2 and 4TO7-B7-1/GPI-IL-12 transfectants were capable of inducing regression of a wild-type tumor growing at a distant site in a concomitant tumor challenge model, suggesting the tumor immunity elicited by the transfectants can act systemically and inhibit the tumor growth at a distant site. Additionally, when used as irradiated whole cell vaccines, 4TO7-B7-1/GPI-IL-12 led to a significant inhibition in tumor growth of day 7 established tumors. Lastly, we observed a significant decrease in the prevalence of myeloid-derived suppressor cells and regulatory T-cells in the tumor microenvironment on day 7 post challenge with 4TO7-B7-1/GPI-IL-12 cells, which provides mechanistic insight into antitumor efficacy of the tumor-cell membrane expressed IL-12. These studies have implications in designing membrane-based therapeutic vaccines with GPI-anchored cytokines for breast cancer.
Collapse
Affiliation(s)
- Erica N Bozeman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, United States.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Cerullo V, Koski A, Vähä-Koskela M, Hemminki A. Chapter eight--Oncolytic adenoviruses for cancer immunotherapy: data from mice, hamsters, and humans. Adv Cancer Res 2013; 115:265-318. [PMID: 23021247 DOI: 10.1016/b978-0-12-398342-8.00008-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adenovirus is one of the most commonly used vectors for gene therapy and two products have already been approved for treatment of cancer in China (Gendicine(R) and Oncorine(R)). An intriguing aspect of oncolytic adenoviruses is that by their very nature they potently stimulate multiple arms of the immune system. Thus, combined tumor killing via oncolysis and inherent immunostimulatory properties in fact make these viruses in situ tumor vaccines. When further engineered to express cytokines, chemokines, tumor-associated antigens, or other immunomodulatory elements, they have been shown in various preclinical models to induce antigen-specific effector and memory responses, resulting both in full therapeutic cures and even induction of life-long tumor immunity. Here, we review the state of the art of oncolytic adenovirus, in the context of their capability to stimulate innate and adaptive arms of the immune system and finally how we can modify these viruses to direct the immune response toward cancer.
Collapse
Affiliation(s)
- Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Division of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | | | | | | |
Collapse
|
7
|
Optimization of the PiggyBac transposon system for the sustained genetic modification of human T lymphocytes. J Immunother 2010; 32:826-36. [PMID: 19752751 DOI: 10.1097/cji.0b013e3181ad762b] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Optimal implementation of adoptive T-cell therapy for cancer will likely require multiple and maintained genetic modifications of the infused T cells and their progeny so that they home to tumor sites and recognize tumor cells, overcome tumor immune evasion strategies, and remain safe. Retroviral vectors readily transduce T cells and integrate into the host cell genome, but have a limited capacity for multigene insertion and cotransduction and are prohibitively expensive to produce at clinical grade. Genetic modification of T cells using transposons as integrating plasmids is an attractive alternative because of the increased simplicity and cost of production. Of available transposons, piggyBac has the higher transposase activity and larger cargo capacity, and we now evaluate piggyBac for potential adoptive therapies with primary T cells. PiggyBac transposons mediated stable gene expression in approximately 20% of primary T cells without selection. Treatment and maintenance of T cells with interleukin-15 increased stable transgene expression up to approximately 40% and expression was sustained through multiple logs of expansion for over 9 weeks in culture. We demonstrate simultaneous integration of 2 independent transposons in 20% of T cells, a frequency that could be increased to over 85% by selection of a transgenic surface marker (truncated CD19). PiggyBac could also deliver transposons of up to 13 kb with 10,000-fold expansion of transduced T cells in culture and finally we demonstrate delivery of a functional suicide gene (iCasp9). PiggyBac transposons may thus be used to express the multiple integrated transgenes that will likely be necessary for the broader success of T-cell therapy.
Collapse
|
8
|
Li ZH, Wen XY, Mandelbaum S, Falcioni N, Hawley TS, Hawley RG, Stewart AK. Improved Therapeutic Outcome Following Combination Immunogene Vaccination Therapy in Murine Myeloma. Leuk Lymphoma 2009; 44:1775-84. [PMID: 14692533 DOI: 10.1080/1042819031000119208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Increasing evidence suggests a role for immunologic vaccination and therapy in the management of minimal residual myeloma. We have previously demonstrated a synergistic effect of combining the Th1 stimulating cytokine IL-12 with the co-stimulatory molecule CD80 in murine myeloma vaccination therapy. We reasoned that the efficacy of such treatment might be further improved by incorporating additional gene products which enhance the function of antigen presenting cells. Studies were therefore conducted with murine myeloma BM1 cells expressing Flt3L (membrane bound or soluble forms) or GM-CSF and the IL-12 x CD80 combination. Single agent and combined therapeutic approaches were explored. All gene-modified BM1 cells, except BM1/IL-12 x CD80, developed tumors when subcutaneously injected into BALB/c mice. As prophylactic tumor vaccines, the combined use of gene-modified BM1/sFlt3L+GM-CSF+IL-12 x CD80 was most effective, providing 100% protection against subsequent parental BM1 tumor challenge. By comparison, only partial protection was observed with any single gene-engineered tumor vaccine. Notably, IL-12 x CD80 coexpressing BM1 cell vaccines were the most effective therapeutic vaccine in a minimal disease model. Such protective vaccination was achieved by stimulation of lymphocyte proliferation and enhancement of cytotoxic lymphocyte activity.
Collapse
Affiliation(s)
- Z H Li
- The Toronto General Hospital Research Institute and Medical Oncology, The Princess Margaret Hospital, McLaughlin Center for Molecular Medicine, Toronto, Ont., Canada
| | | | | | | | | | | | | |
Collapse
|
9
|
Mir MA, Agrewala JN. Signaling through CD80: an approach for treating lymphomas. Expert Opin Ther Targets 2008; 12:969-79. [DOI: 10.1517/14728222.12.8.969] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
10
|
Abstract
BACKGROUND Current therapies for multiple myeloma (MM) are not curative, thus novel targeted therapeutics are being developed. One such targeted therapy is oncolytic virotherapy, wherein viruses specifically infect and kill the malignant plasma cells, leaving normal cells intact. OBJECTIVE This review provides an overview of the mechanisms and results of the oncolytic viruses being used to date and discusses the recent advances in the field of virotherapy for MM. METHODS All papers using viruses to treat MM were identified and screened. Only papers describing replicating, oncolytic viruses were reviewed. RESULTS/CONCLUSIONS Several viruses are currently being developed preclinically and clinically to treat MM, including measles virus, vesicular stomatitis virus, coxsackievirus A21 and vaccinia virus. Other viruses are being used preclinically to purge myeloma cells from autologous bone marrow transplants. Efforts to improve myeloma-specific targeting, avoid the antiviral immune response and evaluate combination therapies for MM are ongoing.
Collapse
Affiliation(s)
- Amaalia E Stief
- Experimental Therapeutics, Toronto General Research Institute, 67 College Street, Room 4-408, Toronto, ON, M5G 2M1, Canada
| | | |
Collapse
|
11
|
Belousova N, Harris R, Zinn K, Rhodes-Selser MA, Kotov A, Kotova O, Wang M, Aurigemma R, Zhu ZB, Curiel DT, Alvarez RD. Circumventing recombination events encountered with production of a clinical-grade adenoviral vector with a double-expression cassette. Mol Pharmacol 2006; 70:1488-93. [PMID: 16896072 DOI: 10.1124/mol.106.025619] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Delivery of multiple exogenous genes into target cells is important for a broad range of gene therapy applications, including combined therapeutic gene expression and noninvasive imaging. Previous studies ( Mol Ther 4: 223-231, 2001 ) have described the adenoviral vector RGDTKSSTR with a double-expression cassette that encodes herpes simplex virus thymidine kinase (HSVtk) for molecular chemotherapy and human somatostatin receptor subtype-2 (hSSTR2) for indirect imaging. In this vector, both genes are inserted in place of the E1 region of the adenoviral genome and expressed independently from two cytomegalovirus (CMV) promoters. During production of clinical-grade RGDTKSSTR, we found that the CMV promoters and simian virus 40 (SV40) poly(A) regions located in both expression cassettes provoked homologous recombination and deletion of one of the cassettes. To resolve this problem, we designed a strategy for substituting the duplicate promoters and poly(A) regions. We placed the hSSTR2 gene in the new Ad5.SSTR/TK.RGD vector under the control of a CMV promoter with a bovine growth hormone poly(A) region, whereas the SV40 promoter, enhancer, and poly(A) signal controlled HSVtk expression. This use of different regulatory sequences allowed independent expression of both transgenes from a single adenoviral vector and circumvented the recombination problem. Reconstruction of the vector with a double-expression cassette enables its use in human clinical trials.
Collapse
Affiliation(s)
- Natalya Belousova
- Division of Human Gene Therapy, Gene Therapy Center, The University of Alabama at Birmingham, 901-19th Street, South, BMR2-502, 35249-2184, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Szymczak AL, Vignali DAA. Development of 2A peptide-based strategies in the design of multicistronic vectors. Expert Opin Biol Ther 2005; 5:627-38. [PMID: 15934839 DOI: 10.1517/14712598.5.5.627] [Citation(s) in RCA: 209] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
As science progresses in its understanding of diseases and their treatment, advances have been made in the biotechnology used in disease therapy. Most gene therapy approaches utilise viral vectors to deliver genes of interest. However, multiple proteins are often involved in disease processes and there is often a need to efficiently deliver more than one gene. Researchers have employed several strategies to accomplish this goal. When designing vectors to express multiple genes, there are several factors that need to be taken into account, including cell type, the activity of the protein of interest and subcellular protein localisation. In most cases, it is ideal for each protein to be expressed at comparable levels, a leading issue with traditional strategies for multigene expression. This review describes some of the techniques that have been used to express multiple genes, and will focus on the use of 2A peptides or 2A peptide-like sequences in the design of multicistronic vectors that may alleviate some of these issues.
Collapse
Affiliation(s)
- Andrea L Szymczak
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
13
|
Triozzi PL, Allen KO, Carlisle RR, Craig M, LoBuglio AF, Conry RM. Phase I study of the intratumoral administration of recombinant canarypox viruses expressing B7.1 and interleukin 12 in patients with metastatic melanoma. Clin Cancer Res 2005; 11:4168-75. [PMID: 15930353 DOI: 10.1158/1078-0432.ccr-04-2283] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The objective of this study was to evaluate the safety and activity of the intratumoral administration of the immune costimulatory molecule, B7.1, encoded by a vector derived from the canarypox virus, ALVAC (ALVAC-B7.1), alone and with the intratumoral injection of ALVAC encoding the immune-stimulatory cytokine, interleukin 12 (ALVAC-IL-12). Fourteen patients with metastatic melanoma who had s.c. nodules received intratumoral injections on days 1, 4, 8, and 11. Nine patients were given escalating doses of up to 25 x 10(8) plaque-forming units of ALVAC-B7.1. Five patients were given 25 x 10(8) plaque-forming units of ALVAC-B7.1 combined with ALVAC-IL-12 50% tissue culture infective dose of 2 x 10(6). Toxicity was mild to moderate and consisted of inflammatory reactions at the injection site and fever, chills, myalgia, and fatigue. Higher levels of B7.1 mRNA were observed in ALVAC-B7.1-injected tumors compared with saline-injected control tumors. Higher levels of intratumoral vascular endothelial growth factor and IL-10, cytokines with immune suppressive activities, were also observed in ALVAC-B7.1- and ALVAC-IL-12-injected tumors compared with saline-injected controls. Serum levels of vascular endothelial growth factor increased at day 18 and returned to baseline at day 43. All patients developed antibody to ALVAC. Intratumoral IL-12 and IFN-gamma mRNA decreased. Changes in serum IL-12 and IFN-gamma levels were not observed. Tumor regressions were not observed. The intratumoral injections of ALVAC-B7.1 and ALVAC-IL-12 were well tolerated at these dose levels and at this schedule and resulted in measurable biological response. This response included the production of factors that may suppress the antitumor immunologic activity of these vectors.
Collapse
Affiliation(s)
- Pierre L Triozzi
- The University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, Alabama 35294-3300, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Noblitt LW, Bangari DS, Shukla S, Knapp DW, Mohammed S, Kinch MS, Mittal SK. Decreased tumorigenic potential of EphA2-overexpressing breast cancer cells following treatment with adenoviral vectors that express EphrinA1. Cancer Gene Ther 2005; 11:757-66. [PMID: 15359289 DOI: 10.1038/sj.cgt.7700761] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The EphA2 receptor tyrosine kinase is frequently overexpressed in invasive breast cancer cells. Moreover, these malignant cells have unstable cell-cell contacts, which preclude EphA2 from interacting with its ligand, EphrinA1, which is anchored to the membrane of adjacent cells. This defect is important because ligand binding causes EphA2 to transmit signals that negatively regulate tumor cell growth and survival, whereas the absence of ligand binding favors these same behaviors. In our present study, human adenoviral type 5 (HAd) vectors were engineered to express secreted-forms of EphrinA1. These vectors were used to infect MDA-MB-231 human breast cancer cells, or MCF-10A human breast epithelial cells providing matched controls. Infection with HAd-EphrinA1-Fc (HAd vector expressing extracellular domain of human EphrinA1 attached to Fc portion of human IgG1 heavy chain) caused increased EphA2 activation and turnover and consequently decreased tumor cell viability in soft agar assays. Consistent with this observation, infection of MDA-MB-231 cells with HAd-EphrinA1-Fc prevented tumor formation in xenograft models. Furthermore, therapeutic modeling via intratumoral inoculation revealed that HAd-EphrinA1-Fc significantly inhibited subsequent tumor growth as compared to matched controls. These results suggest that targeting of EphA2 with adenoviral vectors may have therapeutic value.
Collapse
Affiliation(s)
- Loren W Noblitt
- Laboratory of Gene Therapy, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Boutonnet C, Boijoux O, Bernat S, Kharrat A, Favre G, Faye JC, Vagner S. Pharmacological-based translational induction of transgene expression in mammalian cells. EMBO Rep 2004; 5:721-7. [PMID: 15192697 PMCID: PMC1299091 DOI: 10.1038/sj.embor.7400170] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 04/20/2004] [Accepted: 04/20/2004] [Indexed: 11/09/2022] Open
Abstract
In the quest for the development of pharmacological switches that control gene expression, no system has been reported that regulates at the translational level. To permit small-molecule control of transgene translation, we have constructed a farnesyl transferase inhibitor-responsive translation initiation factor. This artificial protein is a three-component chimaera consisting of the ribosome recruitment core of the eIF4G1 eukaryotic translation initiation factor, the RNA-binding domain of the R17 bacteriophage coat protein and the plasma membrane localization CAAX motif of farnesylated H-Ras. This membrane-delocalized translation factor is inactive unless liberated in the cytosol. Farnesyl transferase inhibitor FTI-277 prevents the membrane association of the CAAX motif and thus increases the cytoplasmic levels of the eIF4G fusion protein, which is then capable of inducing translation of the second cistron of a bicistronic messenger RNA containing an R17-binding site in its intercistronic space. Such direct translational control by farnesyl transferase inhibitors provides a system for fast, graded and reversible regulation of transgene expression.
Collapse
Affiliation(s)
- Christel Boutonnet
- INSERM U589, Institut Louis Bugnard, CHU Rangueil, 31054 Toulouse, France
- MILLEGEN SA, Rue Pierre et Marie Curie BP 28262, 31682 Labège, France
| | - Olivier Boijoux
- INSERM U563, Centre Claudius Régaud, Rue du Pont Saint-Pierre, 31052 Toulouse, France
| | - Sandra Bernat
- MILLEGEN SA, Rue Pierre et Marie Curie BP 28262, 31682 Labège, France
| | | | - Gilles Favre
- INSERM U563, Centre Claudius Régaud, Rue du Pont Saint-Pierre, 31052 Toulouse, France
| | - Jean-Charles Faye
- INSERM U563, Centre Claudius Régaud, Rue du Pont Saint-Pierre, 31052 Toulouse, France
| | - Stéphan Vagner
- INSERM U589, Institut Louis Bugnard, CHU Rangueil, 31054 Toulouse, France
- Tel: +33 561 32 31 28; Fax: +33 561 32 21 41; E-mail:
| |
Collapse
|
16
|
Kolb HJ, Schmid C, Chen X, Woiciechowski A, Roskrow M, Weber M, Guenther W, Ledderose G, Schleuning M. Adoptive immunotherapy in chimeras with donor lymphocytes. Acta Haematol 2003; 110:110-20. [PMID: 14583671 DOI: 10.1159/000072460] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Allogeneic stem cell transplantation has a well-defined indication in the treatment of hematological malignancies. The beneficial immune effect of allogeneic marrow transplantation has long been known, but only recently have methods been developed to separate the graft-versus-leukemia (GVL) effect from graft-versus-host disease (GVHD). Animal experiments have shown that lymphocytes from the marrow donor can be transfused without causing severe GVHD if stable chimerism and tolerance is established. First clinical studies have been preformed in patients with recurrent chronic myelogenous leukemia. In these patients complete molecular remissions were induced that persist without further maintenance treatment. These results have been confirmed in larger multicenter studies in Europe and the USA. The best results were obtained in chronic myelogenous leukemia (CML); repeated successes have been reported in relapsing acute myeloid leukemia (AML), myelodysplastic syndromes and multiple myeloma (MMY), and rare responses were reported for acute lymphoid leukemia. Contrary to animal experiments GVHD has been observed in human patients although to a lesser extent than expected in transplants not given immunosuppression. Secondly myelosuppression has been observed in patients treated with relapsing CML. In CML the incidence of GVHD could be reduced by depleting CD8+ T cells from the donor lymphocyte concentrate. Alternatively only small numbers of T lymphocytes can be transfused and in the case of failing responses, the numbers of donor lymphocytes may be increased. Results in recurrent AML have been improved by the use of low-dose cytosine arabinoside, granulocyte-macrophage colony-stimulating factor and granulocyte colony-stimulating factor mobilized blood cells as compared to lymphocytes only. In MMY the response rate is higher than in AML, but the remissions are of limited duration in most patients. Several protocols have been designed to include preemptive donor lymphocyte transfusion in patients with a high relapse risk after transplantation. Problems remain to avoid chronic GVHD and to circumvent the immune escape mechanisms of leukemia.
Collapse
Affiliation(s)
- Hans-Jochem Kolb
- Department of Medicine III, Klinikum University of Munich-Grosshadern, and GSF-National Research Center for Environment and Health, Munich, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wu Q, Moraes MP, Grubman MJ. Recombinant adenovirus co-expressing capsid proteins of two serotypes of foot-and-mouth disease virus (FMDV): in vitro characterization and induction of neutralizing antibodies against FMDV in swine. Virus Res 2003; 93:211-9. [PMID: 12782369 DOI: 10.1016/s0168-1702(03)00116-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human adenovirus type 5 (Ad5) has been evaluated as a novel gene delivery vector for the development of live-viral vaccines for foot-and-mouth disease (FMD). In this study, we constructed an Ad5 vector co-expressing the capsid precursor proteins, P1, of FMD virus (FMDV) field strains A24 Cruzeiro and O1 Campos and examined the neutralizing antibody responses in swine after inoculation with the vector. To construct the Ad5 vector, a bicistronic expression cassette containing a cytomegalovirus promoter, the P1 coding sequence of FMDV A24, the internal ribosomal entry site (IRES) of FMDV A12, the P1 coding sequence of FMDV O1 Campos and the coding region of A12 3C protease was inserted into the E1 region of an E1/E3-deleted Ad5. The recombinant adenovirus, Ad5A24+O1, was generated by transfection of 293 cells with full-length pAd5A24+O1 recombinant plasmid DNA. The recombinant Ad5 co-expressed P1 of both A24 and O1 in infected 293 cells and P1 of both serotypes was processed to produce VP0, VP3, and VP1. We further demonstrated the formation of capsid protein complexes by co-precipitation of VP0, VP3, and VP1 with monoclonal antibodies against viral capsid proteins. Swine inoculated with Ad5A24+O1 generated neutralizing antibodies against both A24 and O1. However, the overall neutralizing antibody response was considerably lower than that induced by a commercial FMD vaccine or a monovalent Ad5-A24 vaccine.
Collapse
Affiliation(s)
- Qiaohua Wu
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, US Department of Agriculture, PO Box 848, Greenport, NY 11944-0848, USA
| | | | | |
Collapse
|
18
|
Block A, Puls F, Müller J, Milasinovic D, Igelmann D, Schäfer P, Kupfermann N, Schmoldt A, Ameis D, Greten H. Highly suppressible expression of single-chain interleukin-12 by doxycycline following adenoviral infection with a single-vector Tet-regulatory system. J Gene Med 2003; 5:190-200. [PMID: 12666185 DOI: 10.1002/jgm.334] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Adenoviral vectors have been shown to efficiently transfer DNA into a wide variety of eukaryotic cells in vitro and in vivo. However, the therapeutic benefit of this approach is limited by severe side effects as a result of uncontrolled transgene expression. METHODS A bi-directional promoter that controls the desired transgene as well as a tetracycline-suppressible transactivator (tTA) was cloned into the E1-region of E1-deleted recombinant adenoviral vectors. Autoregulation within this construct was obtained by tTA expression under control of the operator, to which tTA binds in the absence of tetracycline. Consequently, binding of tetracycline to tTA results in downregulation of tTA as well as the co-expressed transgene in the infected cell. RESULTS We were able to suppress luciferase-reporter gene expression by up to 16 000-fold in the presence of doxycycline (dox, 2 micro g/ml). Under control of this tetracycline-regulated system, single-chain interleukin-12 (scIL12) was expressed. Adenovirally mediated expression of this potentially lethal cytokine with strong activation of antitumoral immune response was downregulated by up to 6000-fold in the presence of dox. Subsequently, this downregulation also resulted in a highly significant reduction of interferon-gamma secretion by stimulated splenocytes. These mainly contribute to the toxicity of this immunotherapeutic approach. CONCLUSIONS With expression levels exceeding those of the cytomegalovirus (CMV) promoter in almost all cell lines tested, these new vectors will also contribute to the safety of adenoviral approaches by controlled expression without compromising on maximum expression levels.
Collapse
Affiliation(s)
- Andreas Block
- Department of Medicine, University Hospital Hamburg-Eppendorf, D-20246 Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
|