Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jean-Marc Balloul is active.

Publication


Featured researches published by Jean-Marc Balloul.


Journal of Immunotherapy | 2000

Recombinant vaccinia virus encoding human MUC1 and IL2 as immunotherapy in patients with breast cancer

Susy Scholl; Jean-Marc Balloul; Gwenaelle Le Goc; Nadine Bizouarne; Christian Schatz; Marie Paule Kieny; Sylvia Von Mensdorff-Pouilly; Anne Vincent-Salomon; Laurent Deneux; Eric Tartour; Wolf H. Fridman; P. Pouillart; Bruce Acres

Polymorphic epithelial mucin, encoded by the MUC1 gene, is present at the apical surface of glandular epithelial cells. It is over-expressed and aberrantly glycosylated in most breast tumors, resulting in an antigenically distinct molecule and a potential target for immunotherapy. This transmembrane protein, when produced by tumor cells, is often cleaved into the circulation, where it is detectable as a tumor marker (CA 15.3) by various antibodies, allowing for early detection of recurrences and evaluation of treatment efficacy. The objective of the current study was to examine the clinical and environmental safety and immunogenicity of a live recombinant vaccinia virus expressing the human MUC1 and IL2 genes (VV TG5058), referred to here as TG1031. The study was an open-label phase 1 and 2 trial in nine patients with advanced inoperable breast cancer recurrences to the chest wall. The patients were vaccinated intramuscularly with a single dose of TG1031; three patients were treated at each of three progressive dose levels ranging from 5×105 to 5×107 plaque-forming units. A boost injection at their original dose level was administered in patients responding immunologically, clinically, or both. Vaccination resulted in no significant clinical adverse effects, and there was no environmental contamination by live TG1031. All patients had been vaccinated as children, and patients treated at the highest dose level mounted a significant anti-vaccinia antibody response. None of the nine patients had a significant increase in MUC1-specific antibody titers after one single injection, whereas five patients had a detectable increase in vaccinia virus antibody titers. Peripheral blood mononuclear cells of one patient at the intermediate dose level showed a proliferative response to in vitro culture with vaccinia virus, with a stimulation index of 6. A second patient treated at the intermediate dose level had a stimulation index of 7 to MUC1 peptide and of 14 after a boost injection. This patient had a concomitant decrease in carcinoembryonic antigen serum levels and remained clinically stable for 10 weeks. Evidence of MUC1-specific cytotoxic T lymphocytes was detected in two patients. Immunohistochemical analysis revealed an increase in T memory cells (CD45RO) in tumor biopsies after vaccination. The absence of serious adverse events, together with the documentation of immune stimulations in vivo, warrant the further use of TG1031 in immunotherapy trials of breast cancer.


Cancer Immunology, Immunotherapy | 2000

MUC1-specific immune responses in human MUC1 transgenic mice immunized with various human MUC1 vaccines

Bruce Acres; Vasso Apostolopoulos; Jean-Marc Balloul; Danny Wreschner; Pei-Xiang Xing; Dahlila Ali-Hadji; Nadine Bizouarne; Marie Paule Kieny; Ian F. C. McKenzie

Abstract Analyses of MUC1-specific cytotoxic T cell precursor (CTLp) frequencies were performed in mice immunized with three different MUC1 vaccine immunotherapeutic agents. Mice were immunized with either a fusion protein comprising MUC1 and glutathione S-transferase (MUC1-GST), MUC1-GST fusion protein coupled to mannan (MFP) or with a recombinant vaccinia virus expressing both MUC1 and interleukin-2. Mouse strain variations in immune responsiveness have been observed with these vaccines. We have constructed mice transgenic for the human MUC1 gene to study MUC1-specific immune responses and the risk of auto-immunity following MUC1 immunization. Transgenic mice immunized with MUC1 were observed to be partially tolerant in that the MUC1-specific antibody response is lower than that observed in syngeneic but non-transgenic mice. However, a significant MUC1-specific CTLp response to all three vaccines was observed, indicating the ability to overcome T cell, but to a lesser extent B cell, tolerance to MUC1 in these mice. Histological analysis indicates no evidence of auto-immunity to the cells expressing the human MUC1 molecule. These results suggest that it is possible to generate an immune response to a cancer-related antigen without damage to normal tissues expressing the antigen.


Journal of Immunotherapy | 1993

Vaccinia virus MUC1 immunization of mice : immune response and protection against the growth of murine tumors bearing the MUC1 antigen

R. B. Acres; M. Hareuveni; Jean-Marc Balloul; Marie Paule Kieny

MUC1 is a mucin found on the apical surfaces of some normal mammalian mucin-secreting cells. It is characterized by heavy glycosylation and a 20-amino-acid tandem repeat segment. In most cases of human breast adenocarcinoma, this antigen is overexpressed. Moreover, abnormal glycosylation exposes a novel peptide epitope within the tandem repeat, such that antibodies to this epitope can distinguish normal from malignant adenocarcinomatous breast tissue. We have constructed a vaccinia virus (VV) that carries the cDNA for the MUC1 antigen. Murine and human cells infected with this virus express the MUC1 molecule, with three to four tandem repeats per molecule and with the tumor-associated epitopes exposed. Mice immunized with this virus produce antibodies that recognize MUC1 outside the tandem repeat, within the tandem repeat, and within the tumor-associated protein core epitope. Tumorigenic P815 (DBA) and 3T3 (BALB/c) cells have been transfected with MUC1. Thirty percent of DBA mice immunized with VV-MUC1 are protected from growth of P815-MUC1 tumors when implanted with 10(5) cells. Immunized BALB/c mice show a late development of transfected 3T3 tumor cells. Immunized mice show a moderate MUC1-specific IgG titer, but it cannot be correlated with subsequent tumor rejection. No evidence for a MUC1-specific cytotoxic T lymphocyte response has been found after immunization with VV-MUC1.


Molecular and Biochemical Parasitology | 1990

Molecular cloning and tissue distribution of a 26-kilodalton Schistosoma mansoni glutathione S-transferase.

François Trottein; Marie Paule Kieny; Claudie Verwaerde; Gérard Torpier; Raymond J. Pierce; Jean-Marc Balloul; D. Schmitt; Jean-Pierre Lecocq; André Capron

A Schistosoma mansoni cDNA library was constructed from the mRNA of adult worms in the expression vector lambda gt11 and screened with a rabbit antiserum raised against the 26-kDa S. mansoni glutathione S-transferase isoforms (Sm GST 26). Two clones were selected and the nucleotide sequences deduced. The predicted amino acid sequence, specified by these cDNAs, shows strong homology with a Schistosoma japonicum 26 kDa glutathione S-transferase and a lower level of homology with mammalian glutathione S-transferase class mu isoenzymes (EC 2.5.1.18). No significant homology score was found with a 28-kDa S. mansoni glutathione S-transferase (Sm GST 28). A study of the tissue distribution of the cloned Sm GST 26 by immunoelectron microscopy shows similarities to Sm GST 28 in that they are present in the tegument and in subtegumentary parenchymal cells. However, a major difference exists in the protonephridial region in which Sm GST 26 is present in the cytoplasmic digitations localized in the apical chamber delineated by the flame cell body, suggesting that Sm GST 26 may be actively excreted by adult worms.


Human Gene Therapy | 2000

Targeted macrophage cytotoxicity using a nonreplicative live vector expressing a tumor-specific single-chain variable region fragment

Stephane Paul; David Snary; Johan Hoebeke; Deborah Allen; Jean-Marc Balloul; Nadine Bizouarne; Karine Dott; Michel Geist; Joseph Hilgers; Marie Paule Kieny; Joy Burchell; Joyce Taylor-Papadimitriou; R. Bruce Acres

Antigen-specific recognition and subsequent destruction of tumor cells is the goal of vaccine-based immunotherapy of cancer. Often, however, tumor antigen-specific cytotoxic T lymphocytes (CTLs) are either not available or in a state of anergy. In addition, MHCI expression on tumor cells is often downregulated. Either or both of these situations can allow tumor growth to proceed unchecked by CTL control. We have shown previously that tumor antigen-specific monoclonal antibodies can be expressed in vaccinia virus and that activated macrophages infected with this virus acquire the ability to kill tumor cells expressing that antigen. Here we show that a membrane-anchored form of the scFv portion of the MUC1 tumor antigen-specific monoclonal antibody, SM3, can be expressed on activated macrophages with the highly attenuated poxvirus, modified vaccinia Ankara (MVA), as a gene transfer vector. Cells infected with the MVA-scFv construct were shown to express the membrane-bound scFv by Western blot and FACS analysis. That cells expressing the membrane-anchored scFv specifically bind antigen was shown by FACS and by BIAcore analysis. GM-CSF-activated macrophages were infected with the construct and shown to recognize specifically MUC1-expressing tumor cells as measured by IL-12 release. Furthermore, activated macrophages expressing the membrane-bound scFv specifically lyse target cells expressing the MUC1 antigen but not cells that do not express MUC1.


Cancer Gene Therapy | 2002

Tumor gene therapy by MVA-mediated expression of T-cell–stimulating antibodies

Stephane Paul; Etienne Régulier; Ronald Rooke; Fabienne Stoeckel; Michel Geist; Horst Homann; Jean-Marc Balloul; Dominique Villeval; Yves Poitevin; Marie-Paule Kieny; R. Bruce Acres

Immune responses to tumor-associated antigens are often dampened by a tumor-induced state of immune anergy. Previous work has attempted to overcome tumor-induced T-cell anergy by the direct injection of vectors carrying the genes encoding one of a variety of cytokines. We hypothesised that the polyclonal stimulation of T cells, preferably through the TCR complex, would result in a cascade of cytokines associated with T-cell activation and would be best able to overcome T-cell anergy. Here we use the highly attenuated MVA poxvirus to express on tumor cells, in vitro and in vivo, either of three membrane-bound monoclonal antibodies specific for murine TCR complex. Using this system, we have expressed antibodies specific for the CD3ɛ chain (KT3), TCRα/β complex (H57-597), and Vβ7 chain (TR310). Tumor cells bristling with these antibodies are capable of inducing murine T-cell proliferation and cytokine production. When injected into growing tumors (P815, RenCa, and B16F10), these constructs induce the activation of immune effector cells and result in the rejection of the tumor. Histological and FACS analysis of tumor-infiltrating leukocytes reveal that the injection of recombinant virus-expressing antibodies specific for the TCR complex attracts and activates (CD25+, CD69+) CD4 and CD8 lymphocytes. This approach represents a novel strategy to overcome T-cell anergy in tumors and allow the stimulation of tumor-specific T cells.


Molecular Therapy | 2016

Polymeric Cups for Cavitation-mediated Delivery of Oncolytic Vaccinia Virus

Rachel Myers; Christian Coviello; Philippe Erbs; Johann Foloppe; Cliff Rowe; James J. Kwan; Calum Crake; Seán Finn; Edward Jackson; Jean-Marc Balloul; Colin Story; Constantin Coussios; Robert Carlisle

Oncolytic viruses (OV) could become the most powerful and selective cancer therapies. However, the limited transport of OV into and throughout tumors following intravenous injection means their clinical administration is often restricted to direct intratumoral dosing. Application of physical stimuli, such as focused ultrasound, offers a means of achieving enhanced mass transport. In particular, shockwaves and microstreaming resulting from the instigation of an ultrasound-induced event known as inertial cavitation can propel OV hundreds of microns. We have recently developed a polymeric cup formulation which, when delivered intravenously, provides the nuclei for instigation of sustained inertial cavitation events within tumors. Here we report that exposure of tumors to focused ultrasound after intravenous coinjection of cups and oncolytic vaccinia virus , leads to substantial and significant increases in activity. When cavitation was instigated within SKOV-3 or HepG2 xenografts, reporter gene expression from vaccinia virus was enhanced 1,000-fold (P < 0.0001) or 10,000-fold (P < 0.001), respectively. Similar increases in the number of vaccinia virus genomes recovered from tumors were also observed. In survival studies, the application of cup mediated cavitation to a vaccinia virus expressing a prodrug converting enzyme provided significant (P < 0.05) retardation of tumor growth. This technology could improve the clinical utility of all biological therapeutics including OV.


International Journal of Nanomedicine | 2018

Ultrasound-mediated cavitation does not decrease the activity of small molecule, antibody or viral-based medicines

Rachel Myers; Megan Grundy; Cliff Rowe; Christian Coviello; Luca Baù; Philippe Erbs; Johann Foloppe; Jean-Marc Balloul; Colin Story; Constantin C. Coussios; Robert Carlisle

The treatment of cancer using nanomedicines is limited by the poor penetration of these potentially powerful agents into and throughout solid tumors. Externally controlled mechanical stimuli, such as the generation of cavitation-induced microstreaming using ultrasound (US), can provide a means of improving nanomedicine delivery. Notably, it has been demonstrated that by focusing, monitoring and controlling the US exposure, delivery can be achieved without damage to surrounding tissue or vasculature. However, there is a risk that such stimuli may disrupt the structure and thereby diminish the activity of the delivered drugs, especially complex antibody and viral-based nanomedicines. In this study, we characterize the impact of cavitation on four different agents, doxorubicin (Dox), cetuximab, adenovirus (Ad) and vaccinia virus (VV), representing a scale of sophistication from a simple small-molecule drug to complex biological agents. To achieve tight regulation of the level and duration of cavitation exposure, a “cavitation test rig” was designed and built. The activity of each agent was assessed with and without exposure to a defined cavitation regime which has previously been shown to provide effective and safe delivery of agents to tumors in preclinical studies. The fluorescence profile of Dox remained unchanged after exposure to cavitation, and the efficacy of this drug in killing a cancer cell line remained the same. Similarly, the ability of cetuximab to bind its epidermal growth factor receptor target was not diminished following exposure to cavitation. The encoding of the reporter gene luciferase within the Ad and VV constructs tested here allowed the infectivity of these viruses to be easily quantified. Exposure to cavitation did not impact on the activity of either virus. These data provide compelling evidence that the US parameters used to safely and successfully delivery nanomedicines to tumors in preclinical models do not detrimentally impact on the structure or activity of these nanomedicines.


Proceedings of the National Academy of Sciences of the United States of America | 2004

Gene-based vaccines and immunotherapeutics

Margaret A. Liu; Bruce Acres; Jean-Marc Balloul; Nadine Bizouarne; Stephane Paul; Philippe Slos; Patrick Squiban


Archive | 1998

Antitumoral composition based on immunogenic polypeptide with modified cell location

Marie-Paule Kieny; Jean-Marc Balloul; Nadine Bizouarne

Collaboration


Dive into the Jean-Marc Balloul's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge