Network


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

Hotspot


Dive into the research topics where Geoff Symonds is active.

Publication


Featured researches published by Geoff Symonds.


Nature Medicine | 2009

Phase 2 gene therapy trial of an anti-HIV ribozyme in autologous CD34+ cells

Ronald T. Mitsuyasu; Thomas C. Merigan; Andrew Carr; Jerome A. Zack; Mark A. Winters; Cassy Workman; Mark Bloch; Jacob Lalezari; Stephen Becker; Lorna Thornton; Bisher Akil; Homayoon Khanlou; Robert Finlayson; R. McFarlane; Don Smith; Roger Garsia; David H.K. Ma; Matthew Law; John M. Murray; Christof von Kalle; Julie A. Ely; Sharon M Patino; Alison Knop; Philip Wong; Alison Velyian Todd; Margaret A. Haughton; Caroline J. Fuery; Janet L. Macpherson; Geoff Symonds; Louise Evans

Gene transfer has potential as a once-only treatment that reduces viral load, preserves the immune system and avoids lifetime highly active antiretroviral therapy. This study, which is to our knowledge the first randomized, double-blind, placebo-controlled, phase 2 cell-delivered gene transfer clinical trial, was conducted in 74 HIV-1-infected adults who received a tat-vpr-specific anti-HIV ribozyme (OZ1) or placebo delivered in autologous CD34+ hematopoietic progenitor cells. There were no OZ1-related adverse events. There was no statistically significant difference in viral load between the OZ1 and placebo group at the primary end point (average at weeks 47 and 48), but time-weighted areas under the curve from weeks 40-48 and 40-100 were significantly lower in the OZ1 group. Throughout the 100 weeks, CD4+ lymphocyte counts were higher in the OZ1 group. This study indicates that cell-delivered gene transfer is safe and biologically active in individuals with HIV and can be developed as a conventional therapeutic product.


Cell | 1984

Subcellular localization of proteins encoded by oncogenes of avian myeloblastosis virus and avian leukemia virus E26 and by the chicken c-myb gene

Karl-Heinz Klempnauer; Geoff Symonds; Gerard I. Evan; J. Michael Bishop

Analysis of the subcellular location of the proteins encoded by the oncogenes of avian myeloblastosis virus and avian leukemia virus E26 ( p45v -myb and p135gag -myb-ets, respectively) and by the chicken c-myb gene ( p75c -myb) shows that all three proteins are located in the nucleus. In AMV-infected (but not transformed) chicken fibroblasts p45v -myb also resides in the nucleus, indicating that a nuclear location of p45v -myb in these cells is not sufficient to achieve transformation. In AMV-transformed myeloblasts a small fraction of p45v -myb occupies an additional site in the perinuclear region of the cytoplasm. If the myeloblasts are caused to differentiate to macrophages, most of p45v -myb is found in the cytoplasm. This redistribution of p45v -myb within the cell may be responsible for reversion of the transformed phenotype.


Human Gene Therapy | 2004

Anti-Human Immunodeficiency Virus Hematopoietic Progenitor Cell-Delivered Ribozyme in a Phase I Study: Myeloid and Lymphoid Reconstitution in Human Immunodeficiency Virus Type-1–Infected Patients

Rafael G. Amado; Ronald T. Mitsuyasu; Joseph D. Rosenblatt; Frances K. Ngok; Andreas Bakker; Steve W. Cole; Nathalie Chorn; Lii Shin Lin; Gregory Bristol; Maureen Boyd; Janet L. Macpherson; Gregory Fanning; Alison Velyian Todd; Julie A. Ely; Jerome A. Zack; Geoff Symonds

A phase I gene transfer clinical study was undertaken to examine the ability to introduce a potential anti-human immunodeficiency virus (HIV) gene therapeutic into hematopoietic progenitor cells (HPC), thereby contributing to multilineage engraftment. The potential therapeutic effect of genetically modifying HPC with protective genes in HIV-infected adults depends in part on the presence of adult thymic activity and myeloid capacity in the setting of HIV replication. Herein we report the presence and expression of a retroviral vector encoding an anti-HIV-1 ribozyme in mature hematopoietic cells of different lineages, and de novo T-lymphocyte development ensuing from genetically engineered CD34(+) HPC. Sustained output of vector-containing mature myeloid and T-lymphoid cells was detected even in patients with multidrug-resistant infection. In addition, the study showed that the degree of persistence of gene-containing cells was dependent on transduced HPC dose. These novel findings support the concept of gene therapy as a modality to effect immune reconstitution with cells engineered to inhibit HIV replication and this report represents the first demonstration of long-term maintenance of a potential therapeutic transgene in HIV disease.


Journal of Gene Medicine | 2005

Long-term survival and concomitant gene expression of ribozyme-transduced CD4+ T-lymphocytes in HIV-infected patients

Janet L. Macpherson; Maureen Boyd; Allison J Arndt; Alison Velyian Todd; Gregory Fanning; Julie A. Ely; Fiona Elliott; Alison Knop; Mitch Raponi; John M. Murray; Wayne Gerlach; Lun-Quan Sun; Ronald Penny; Geoff Symonds; Andrew Carr; David A. Cooper

An anti‐HIV‐1 tat ribozyme, termed Rz2, has been shown to inhibit HIV‐1 infection/replication and to decrease HIV‐1‐induced pathogenicity in T‐lymphocyte cell lines and normal peripheral blood T‐lymphocytes. We report here the results of a phase I gene transfer clinical trial using Rz2.


Stem Cells | 2008

Glycogen Synthase Kinase-3β Inhibition Preserves Hematopoietic Stem Cell Activity and Inhibits Leukemic Cell Growth

Tiffany Holmes; Tracey O'Brien; Robert Knight; Robert Lindeman; Sylvie Shen; Emma Song; Geoff Symonds; Alla Dolnikov

Ex vivo expansion of cord blood cells generally results in reduced stem cell activity in vivo. Glycogen synthase kinase‐3β (GSK‐3β) regulates the degradation of β‐catenin, a critical regulator of hematopoietic stem cells (HSCs). Here we show that GSK‐3β inhibition activates β‐catenin in cord blood CD34+ cells and upregulates β‐catenin transcriptional targets c‐myc and HoxB4, both known to regulate HSC self‐renewal. GSK‐3β inhibition resulted in delayed ex vivo expansion of CD34+ cells, yet enhanced the preservation of stem cell activity as tested in long‐term culture with bone marrow stroma. Delayed cell cycling, reduced apoptosis, and increased adherence of hematopoietic progenitor cells to bone marrow stroma were observed in these long‐term cultures treated with GSK‐3β inhibitor. This improved adherence to stroma was mediated via upregulation of CXCR4. In addition, GSK‐3β inhibition preserved severe combined immunodeficiency (SCID) repopulating cells as tested in the nonobese diabetic/SCID mouse model. Our data suggest the involvement of GSK‐3β inhibition in the preservation of HSC and their interaction with the bone marrow environment. Methods for the inhibition of GSK‐3β may be developed for clinical ex vivo expansion of HSC for transplantation. In addition, GSK‐3β inhibition suppressed leukemic cell growth via the induction of apoptosis mediated by the downregulation of survivin. Modulators of GSK‐3β may increase the range of novel drugs that specifically kill leukemic cells while sparing normal stem cells.


PLOS ONE | 2012

Engineering HIV-1-resistant T-cells from short-hairpin RNA-expressing hematopoietic stem/progenitor cells in humanized BLT mice.

Gene-Errol Ringpis; Saki Shimizu; Hubert Arokium; Joanna Camba-Colón; Maria V. Carroll; Ruth Cortado; Yiming Xie; Patrick Y. Kim; Anna Sahakyan; Emily L. Lowe; Munetoshi Narukawa; Fadi Kandarian; Bryan P. Burke; Geoff Symonds; Dong Sung An; Irvin S. Y. Chen; Masakazu Kamata

Down-regulation of the HIV-1 coreceptor CCR5 holds significant potential for long-term protection against HIV-1 in patients. Using the humanized bone marrow/liver/thymus (hu-BLT) mouse model which allows investigation of human hematopoietic stem/progenitor cell (HSPC) transplant and immune system reconstitution as well as HIV-1 infection, we previously demonstrated stable inhibition of CCR5 expression in systemic lymphoid tissues via transplantation of HSPCs genetically modified by lentiviral vector transduction to express short hairpin RNA (shRNA). However, CCR5 down-regulation will not be effective against existing CXCR4-tropic HIV-1 and emergence of resistant viral strains. As such, combination approaches targeting additional steps in the virus lifecycle are required. We screened a panel of previously published shRNAs targeting highly conserved regions and identified a potent shRNA targeting the R-region of the HIV-1 long terminal repeat (LTR). Here, we report that human CD4+ T-cells derived from transplanted HSPC engineered to co-express shRNAs targeting CCR5 and HIV-1 LTR are resistant to CCR5- and CXCR4- tropic HIV-1-mediated depletion in vivo. Transduction with the combination vector suppressed CXCR4- and CCR5- tropic viral replication in cell lines and peripheral blood mononuclear cells in vitro. No obvious cytotoxicity or interferon response was observed. Transplantation of combination vector-transduced HSPC into hu-BLT mice resulted in efficient engraftment and subsequent stable gene marking and CCR5 down-regulation in human CD4+ T-cells within peripheral blood and systemic lymphoid tissues, including gut-associated lymphoid tissue, a major site of robust viral replication, for over twelve weeks. CXCR4- and CCR5- tropic HIV-1 infection was effectively inhibited in hu-BLT mouse spleen-derived human CD4+ T-cells ex vivo. Furthermore, levels of gene-marked CD4+ T-cells in peripheral blood increased despite systemic infection with either CXCR4- or CCR5- tropic HIV-1 in vivo. These results demonstrate that transplantation of HSPCs engineered with our combination shRNA vector may be a potential therapy against HIV disease.


Viruses | 2015

CCR5 Targeted Cell Therapy for HIV and Prevention of Viral Escape

Gero Hütter; Josef Bodor; Scott Ledger; Maureen Boyd; Michelle Millington; Marlene Tsie; Geoff Symonds

Allogeneic transplantation with CCR5-delta 32 (CCR5-d32) homozygous stem cells in an HIV infected individual in 2008, led to a sustained virus control and probably eradication of HIV. Since then there has been a high degree of interest to translate this approach to a wider population. There are two cellular ways to do this. The first one is to use a CCR5 negative cell source e.g., hematopoietic stem cells (HSC) to copy the initial finding. However, a recent case of a second allogeneic transplantation with CCR5-d32 homozygous stem cells suffered from viral escape of CXCR4 quasi-species. The second way is to knock down CCR5 expression by gene therapy. Currently, there are five promising techniques, three of which are presently being tested clinically. These techniques include zinc finger nucleases (ZFN), clustered regularly interspaced palindromic repeats/CRISPR-associated protein 9 nuclease (CRISPR/Cas9), transcription activator-like effectors nuclease (TALEN), short hairpin RNA (shRNA), and a ribozyme. While there are multiple gene therapy strategies being tested, in this review we reflect on our current knowledge of inhibition of CCR5 specifically and whether this approach allows for consequent viral escape.


Molecular therapy. Nucleic acids | 2015

Engineering Cellular Resistance to HIV-1 Infection In Vivo Using a Dual Therapeutic Lentiviral Vector

Bryan P. Burke; Bernard Levin; Jane Zhang; Anna Sahakyan; Joshua Boyer; Maria V. Carroll; Joanna Camba Colón; Naomi Keech; Valerie Rezek; Gregory Bristol; Erica Eggers; Ruth Cortado; Maureen Boyd; Helen Impey; Saki Shimizu; Emily L. Lowe; Gene-Errol Ringpis; Sohn G. Kim; Dimitrios N. Vatakis; Louis Breton; Jeffrey S. Bartlett; Irvin S. Y. Chen; Scott G. Kitchen; Dong Sung An; Geoff Symonds

We described earlier a dual-combination anti-HIV type 1 (HIV-1) lentiviral vector (LVsh5/C46) that downregulates CCR5 expression of transduced cells via RNAi and inhibits HIV-1 fusion via cell surface expression of cell membrane-anchored C46 antiviral peptide. This combinatorial approach has two points of inhibition for R5-tropic HIV-1 and is also active against X4-tropic HIV-1. Here, we utilize the humanized bone marrow, liver, thymus (BLT) mouse model to characterize the in vivo efficacy of LVsh5/C46 (Cal-1) vector to engineer cellular resistance to HIV-1 pathogenesis. Human CD34+ hematopoietic stem/progenitor cells (HSPC) either nonmodified or transduced with LVsh5/C46 vector were transplanted to generate control and treatment groups, respectively. Control and experimental groups displayed similar engraftment and multilineage hematopoietic differentiation that included robust CD4+ T-cell development. Splenocytes isolated from the treatment group were resistant to both R5- and X4-tropic HIV-1 during ex vivo challenge experiments. Treatment group animals challenged with R5-tropic HIV-1 displayed significant protection of CD4+ T-cells and reduced viral load within peripheral blood and lymphoid tissues up to 14 weeks postinfection. Gene-marking and transgene expression were confirmed stable at 26 weeks post-transplantation. These data strongly support the use of LVsh5/C46 lentiviral vector in gene and cell therapeutic applications for inhibition of HIV-1 infection.


Molecular therapy. Methods & clinical development | 2014

Preclinical safety and efficacy of an anti-HIV-1 lentiviral vector containing a short hairpin RNA to CCR5 and the C46 fusion inhibitor

Orit Wolstein; Maureen Boyd; Michelle Millington; Helen Impey; Joshua Boyer; Annett Howe; Frederic Delebecque; Kenneth Cornetta; Michael Rothe; Christopher Baum; Tamara Nicolson; Rachel Koldej; Jane Zhang; Naomi Keech; Joanna Camba Colón; Louis Breton; Jeffrey S. Bartlett; Dong Sung An; Irvin S. Y. Chen; Bryan P. Burke; Geoff Symonds

Gene transfer has therapeutic potential for treating HIV-1 infection by generating cells that are resistant to the virus. We have engineered a novel self-inactivating lentiviral vector, LVsh5/C46, using two viral-entry inhibitors to block early steps of HIV-1 cycle. The LVsh5/C46 vector encodes a short hairpin RNA (shRNA) for downregulation of CCR5, in combination with the HIV-1 fusion inhibitor, C46. We demonstrate here the effective delivery of LVsh5/C46 to human T cell lines, peripheral blood mononuclear cells, primary CD4+ T lymphocytes, and CD34+ hematopoietic stem/progenitor cells (HSPC). CCR5-targeted shRNA (sh5) and C46 peptide were stably expressed in the target cells and were able to effectively protect gene-modified cells against infection with CCR5- and CXCR4-tropic strains of HIV-1. LVsh5/C46 treatment was nontoxic as assessed by cell growth and viability, was noninflammatory, and had no adverse effect on HSPC differentiation. LVsh5/C46 could be produced at a scale sufficient for clinical development and resulted in active viral particles with very low mutagenic potential and the absence of replication-competent lentivirus. Based on these in vitro results, plus additional in vivo safety and efficacy data, LVsh5/C46 is now being tested in a phase 1/2 clinical trial for the treatment of HIV-1 disease.


Molecular therapy. Nucleic acids | 2013

Promoter Targeting shRNA Suppresses HIV-1 Infection In vivo Through Transcriptional Gene Silencing

Kazuo Suzuki; Shinichiro Hattori; Katherine Marks; Chantelle Ahlenstiel; Yosuke Maeda; Takaomi Ishida; Michelle Millington; Maureen Boyd; Geoff Symonds; David A. Cooper; Seiji Okada; Anthony D. Kelleher

Despite prolonged and intensive application, combined antiretroviral therapy cannot eradicate human immunodeficiency virus (HIV)-1 because it is harbored as a latent infection, surviving for long periods of time. Alternative approaches are required to overcome the limitations of current therapy. We have been developing a short interfering RNA (siRNA) gene silencing approach. Certain siRNAs targeting promoter regions of genes induce transcriptional gene silencing. We previously reported substantial transcriptional gene silencing of HIV-1 replication by an siRNA targeting the HIV-1 promoter in vitro. In this study, we show that this siRNA, expressed as a short hairpin RNA (shRNA) (shPromA-JRFL) delivered by lentiviral transduction of human peripheral blood mononuclear cells (PBMCs), which are then used to reconstitute NOJ mice, is able to inhibit HIV-1 replication in vivo, whereas a three-base mismatched variant (shPromA-M2) does not. In shPromA-JRFL–treated mice, HIV-1 RNA in serum is significantly reduced, and the ratio of CD4+/CD8+ T cells is significantly elevated. Expression levels of the antisense RNA strand inversely correlates with HIV-1 RNA in serum. The silenced HIV-1 can be reactivated by T-cell activation in ex vivo cultures. HIV-1 suppression is not due to offtarget effects of shPromA-JRFL. These data provide “proof-of principle” that an shRNA targeting the HIV-1 promoter is able to suppress HIV-1 replication in vivo.

Collaboration


Dive into the Geoff Symonds's collaboration.

Top Co-Authors

Avatar

Alla Dolnikov

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Janet L. Macpherson

Royal Prince Alfred Hospital

View shared research outputs
Top Co-Authors

Avatar

Michelle Millington

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Sylvie Shen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

John M. Murray

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bryan P. Burke

University of California

View shared research outputs
Top Co-Authors

Avatar

Karen L. MacKenzie

University of New South Wales

View shared research outputs
Researchain Logo
Decentralizing Knowledge