Network


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

Hotspot


Dive into the research topics where Patrick J. Hanley is active.

Publication


Featured researches published by Patrick J. Hanley.


Blood | 2009

Functionally active virus-specific T cells that target CMV, adenovirus, and EBV can be expanded from naive T-cell populations in cord blood and will target a range of viral epitopes

Patrick J. Hanley; Conrad Russell Y. Cruz; Barbara Savoldo; Ann M. Leen; Maja Stanojevic; Mariam Khalil; William K. Decker; Jeffrey J. Molldrem; Hao Liu; Adrian P. Gee; Cliona M. Rooney; Helen E. Heslop; Gianpietro Dotti; Malcolm K. Brenner; Elizabeth J. Shpall; Catherine M. Bollard

The naive phenotype of cord blood (CB) T cells may reduce graft-versus-host disease after umbilical cord blood transplantation, but this naivety and their low absolute numbers also delays immune reconstitution, producing higher infection-related mortality that is predominantly related to CMV, adenovirus (Adv), and EBV. Adoptive immunotherapy with peripheral blood-derived virus-specific cytotoxic T lymphocytes (CTLs) can effectively prevent viral disease after conventional stem cell transplantation, and we now describe the generation of single cultures of CTLs from CB that are specific for multiple viruses. Using EBV-infected B cells transduced with a clinical-grade Ad5f35CMVpp65 adenoviral vector as sources of EBV, Adv, and CMV antigens, we expanded virus-specific T cells even from CB T cells with a naive phenotype. After expansion, each CTL culture contained both CD8(+) and CD4(+) T-cell subsets, predominantly of effector memory phenotype. Each CTL culture also had HLA-restricted virus-specific cytotoxic effector function against EBV, CMV, and Adv targets. The CB CTLs recognized multiple viral epitopes, including CD4-restricted Adv-hexon epitopes and immunosubdominant CD4- and CD8-restricted CMVpp65 epitopes. Notwithstanding their naive phenotype, it is therefore possible to generate trivirus-specific CTLs in a single culture of CB, which may be of value to prevent or treat viral disease in CB transplant recipients. This study is registered at www.clinicaltrials.gov as NCT00078533.


Blood | 2010

Derivation of human T-lymphocytes from cord blood and peripheral blood with antiviral and antileukemic specificity from a single culture as protection against infection and relapse after stem cell transplantation

Kenneth P. Micklethwaite; Barbara Savoldo; Patrick J. Hanley; Ann M. Leen; Gail J. Demmler-Harrison; Laurence J.N. Cooper; Hao Liu; Adrian P. Gee; Elizabeth J. Shpall; Cliona M. Rooney; Helen E. Heslop; Malcolm K. Brenner; Catherine M. Bollard; Gianpietro Dotti

Viral infections and leukemic relapse account for the majority of treatment failures in patients with B-cell acute lymphoblastic leukemia (B-ALL) receiving allogeneic hematopoietic stem cell (HSC) or cord blood (CB) transplants. Adoptive transfer of virus-specific cytotoxic T lymphocytes (CTLs) provides protection against common viruses causing serious infections after HSC transplantation without concomitant graft-versus-host disease. We have now generated CTL lines from peripheral blood (PB) or CB units that recognize multiple common viruses and provide antileukemic activity by transgenic expression of a chimeric antigen receptor (CAR) targeting CD19 expressed on B-ALL. PB-derived CAR(+) CTLs produced interferon-gamma (IFNgamma) in response to cytomegalovirus-pp65, adenovirus-hexon, and Epstein-Barr virus pepmixes (from 205 +/- 104 to 1034 +/- 304 spot-forming cells [SFCs]/10(5) T cells) and lysed primary B-ALL blasts in (51)Cr-release assays (mean, 66% +/- 5% specific lysis; effector-target [E/T] ratio, 40:1) and the CD19(+) Raji cell line (mean, 78% +/- 17%) in contrast to nontransduced controls (8% +/- 8% and 3% +/- 2%). CB-derived CAR(+) CTLs showed similar antiviral and antitumor function and both PB and CB CAR(+) CTLs completely eliminated B-ALL blasts over 5 days of coculture. This approach may prove beneficial for patients with high-risk B-ALL who have recently received an HSC or CB transplant and are at risk of infection and relapse.


Cytotherapy | 2014

Efficient manufacturing of therapeutic mesenchymal stromal cells with the use of the Quantum Cell Expansion System

Patrick J. Hanley; Zhuyong Mei; April G. Durett; Marie da Graca Cabreira-Harrison; Mariola Klis; Wei Li; Yali Zhao; Bing Yang; Kaushik Parsha; Osman Mir; Farhaan Vahidy; Debra D. Bloom; R. Brent Rice; Peiman Hematti; Sean I. Savitz; Adrian P. Gee

BACKGROUND The use of bone marrow-derived mesenchymal stromal cells (MSCs) as a cellular therapy for various diseases, such as graft-versus-host disease, diabetes, ischemic cardiomyopathy and Crohns disease, has produced promising results in early-phase clinical trials. However, for widespread application and use in later phase studies, manufacture of these cells must be cost-effective, safe and reproducible. Current methods of manufacturing in flasks or cell factories are labor-intensive, involve a large number of open procedures and require prolonged culture times. METHODS We evaluated the Quantum Cell Expansion System for the expansion of large numbers of MSCs from unprocessed bone marrow in a functionally closed system and compared the results with a flask-based method currently in clinical trials. RESULTS After only two passages, we were able to expand a mean of 6.6 × 10(8) MSCs from 25 mL of bone marrow reproducibly. The mean expansion time was 21 days, and cells obtained were able to differentiate into all three lineages: chondrocytes, osteoblasts and adipocytes. The Quantum was able to generate the target cell number of 2.0 × 10(8) cells in an average of 9 fewer days and in half the number of passages required during flask-based expansion. We estimated that the Quantum would involve 133 open procedures versus 54,400 in flasks when manufacturing for a clinical trial. Quantum-expanded MSCs infused into an ischemic stroke rat model were therapeutically active. CONCLUSIONS The Quantum is a novel method of generating high numbers of MSCs in less time and at lower passages when compared with flasks. In the Quantum, the risk of contamination is substantially reduced because of the substantial decrease in open procedures.


Science Translational Medicine | 2015

CMV-specific T cells generated from naïve T cells recognize atypical epitopes and may be protective in vivo

Patrick J. Hanley; J. Joseph Melenhorst; Sarah Nikiforow; Phillip Scheinberg; James W. Blaney; Gail J. Demmler-Harrison; C. Russell Cruz; Sharon Lam; Robert A. Krance; Kathryn Leung; Caridad Martinez; Hao Liu; Helen E. Heslop; Cliona M. Rooney; Elizabeth J. Shpall; A. John Barrett; Catherine M. Bollard

CMV-specific T cells, derived from the naïve population, recognize different epitopes of CMV than do memory-derived T cells and may be functional and protective in vivo. Sourcing CMV immunotherapy Immunotherapy—such as adoptive T cell therapy—is making headway in treating cancer, autoimmunity, and infectious disease. Indeed, adoptive transfer of cytomegalovirus (CMV)–specific T cells can restore immunity to the virus. However, these cells have been primarily derived from memory cells from CMV-seropositive individuals, which limits the donor pool. Now, Hanley et al. demonstrate that CMV-specific T cells can be derived from naïve T cells taken from CMV-seronegative people. These cells react to different epitopes than the memory cell–derived T cells but with similar avidity. What’s more, these cells were associated with periods of CMV-free survival when transplanted into patients. These data support expanded trials of adoptive therapy of CMV-restricted T cells from seronegative donors. Adoptive transfer of cytomegalovirus (CMV)–specific T cells derived from adult seropositive donors can effectively restore antiviral immunity after transplantation. However, CMV-seronegative donors lack CMV-specific memory T cells, which restricts the availability of virus-specific T cells for immunoprophylaxis. We demonstrate the feasibility of deriving CMV-specific T cells from naïve cells for T cell therapy. Naïve T cells primed to recognize CMV were restricted to different, atypical epitopes than T cells derived from CMV-seropositive individuals; however, these two cell populations had similar avidities. CMV-seropositive individuals also had T cells recognizing these atypical epitopes, but these cells had a lower avidity than those derived from the seronegative subjects, which suggests that high-avidity T cells to these epitopes may be lost over time. Indeed, recipients of cord blood (CB) grafts who did not develop CMV were found by clonotypic analysis to have T cells recognizing atypical CMVpp65 epitopes. Therefore, we examined unmanipulated CB units and found that T cells with T cell receptors restricted by atypical epitopes were the most common, which may explain why these T cells expanded. When infused to recipients, naïve donor–derived virus-specific T cells that recognized atypical epitopes were associated with prolonged periods of CMV-free survival and complete remission. These data suggest that naïve-derived T cells from seronegative patients may be an additional source of cells for CMV immunoprophylaxis.


Journal of Immunotherapy | 2012

Generation of polyclonal CMV-specific T cells for the adoptive immunotherapy of glioblastoma.

Alexia Ghazi; Aidin Ashoori; Patrick J. Hanley; Vita S. Brawley; Donald R. Shaffer; Yvonne Kew; Suzanne Z. Powell; Robert G. Grossman; Zakaria Grada; Michael E. Scheurer; Meenakshi Hegde; Ann M. Leen; Catherine M. Bollard; Cliona M. Rooney; Helen E. Heslop; Stephen Gottschalk; Nabil Ahmed

Glioblastoma (GBM) is the most common primary brain cancer in adults and is virtually incurable. Recent studies have shown that cytomegalovirus (CMV) is present in majority of GBMs. To evaluate whether the CMV antigens pp65 and IE1, which are expressed in GBMs, could be targeted by CMV-specific T cells, we measured the frequency of T cells targeting pp65 and IE1 in the peripheral blood of a cohort of 11 sequentially diagnosed CMV-seropositive GBM patients, and evaluated whether it was feasible to expand autologous CMV-specific T cells for future clinical studies. All 11 CMV-seropositive GBM patients had T cells specific for pp65 and IE1 in their peripheral blood assessed by IFN&ggr; enzyme-linked immunospot assay. However, the precursor frequency of pp65-specific T cells was decreased in comparison with healthy donors (P=0.001). We successfully reactivated and expanded CMV-specific T cells from 6 out of 6 GBM patients using antigen-presenting cells transduced with an adenoviral vector encoding pp65 and IE1. CMV-specific T-cell lines contained CD4+ as well as CD8+ T cells, recognized pp65+ and IE1+ targets and killed CMV-infected autologous GBM cells. Infusion of such CMV-specific T-cell lines may extend the benefits of T-cell therapy to patients with CMV+ GBMs.


Cytotherapy | 2014

The time is now: moving toward virus-specific T cells after allogeneic hematopoietic stem cell transplantation as the standard of care

Francesco Saglio; Patrick J. Hanley; Catherine M. Bollard

Abstract Adoptive immunotherapy—in particular, T-cell therapy—has recently emerged as a useful strategy with the potential to overcome many of the limitations of antiviral drugs for the treatment of viral complications after hematopietic stem cell transplantation. In this review, we briefly summarize the current methods for virus-specific T-cell isolation or selection and we report results from clinical trials that have used these techniques, focusing specifically on the strategies aimed to broaden the application of this technology.


Viruses | 2014

Controlling cytomegalovirus: helping the immune system take the lead.

Patrick J. Hanley; Catherine M. Bollard

Cytomegalovirus, of the Herpesviridae family, has evolved alongside humans for thousands of years with an intricate balance of latency, immune evasion, and transmission. While upwards of 70% of humans have evidence of CMV infection, the majority of healthy people show little to no clinical symptoms of primary infection and CMV disease is rarely observed during persistent infection in immunocompetent hosts. Despite the fact that the majority of infected individuals are asymptomatic, immunologically, CMV hijacks the immune system by infecting and remaining latent in antigen-presenting cells that occasionally reactivate subclinically and present antigen to T cells, eventually causing the inflation of CMV-specific T cells until they can compromise up to 10% of the entire T cell repertoire. Because of this impact on the immune system, as well as its importance in fields such as stem cell and organ transplant, the relationship between CMV and the immune response has been studied in depth. Here we provide a review of many of these studies and insights into how CMV-specific T cells are currently being used therapeutically.


Cytotherapy | 2013

Manufacturing mesenchymal stromal cells for phase I clinical trials

Patrick J. Hanley; Zhuyong Mei; Maria da Graça Cabreira-Hansen; Mariola Klis; Wei Li; Yali Zhao; April G. Durett; Xingwu Zheng; Yongping Wang; Adrian P. Gee; Edwin M. Horwitz

Mesenchymal stromal cells (MSCs) are multipotent progenitor cells capable of differentiating into adipocytes, osteoblasts and chondroblasts as well as secreting a vast array of soluble mediators. This potentially makes MSCs important mediators of a variety of therapeutic applications. They are actively under evaluation for immunomodulatory purposes such as graft-versus-host disease and Crohns disease as well as regenerative applications such as stroke and congestive heart failure. We report our method of generating clinical-grade MSCs together with suggestions gathered from manufacturing experience in our Good Manufacturing Practices facility.


Cytotherapy | 2011

Expansion of T cells targeting multiple antigens of cytomegalovirus, Epstein–Barr virus and adenovirus to provide broad antiviral specificity after stem cell transplantation

Patrick J. Hanley; Donald R. Shaffer; Conrad Russell Y. Cruz; Stephanie Ku; Benjamin Tzou; Hao Liu; Gail J. Demmler-Harrison; Helen E. Heslop; C.M. Rooney; Stephen Gottschalk; Catherine M. Bollard

BACKGROUND AIMS Hematopoietic stem cell transplant (HSCT) is the treatment of choice for a proportion of patients with hematologic malignancies as well as for non-malignant diseases. However, viral infections, particularly Epstein-Barr virus (EBV), cytomegalovirus (CMV) and adenovirus (Ad), remain problematic after transplant despite the use of antiviral drugs. We have shown that cytotoxic T lymphocytes (CTL) generated against CMV-pp65, EBV and Ad antigens in a single culture are capable of controlling infections with all three viruses after HSCT. Although pp65-specific CTL have proved efficacious for the control of CMV infection, several reports highlight the importance of targeting additional CMV antigens. METHODS To expand multivirus-specific T cells with activity against both CMV-pp65 and CMV-IE-1, peripheral blood mononuclear cells (PBMC) were transduced with the adenoviral vector (Ad5f35-IE-1-I-pp65). After 9-12 days the CTL were restimulated with autologous EBV-transformed B cells transduced with the same Ad vector. RESULTS After 18 days in culture nine CTL lines expanded from less than 1.5 × 10(7) PBMC to a mean of 6.1 × 10(7) T cells that recognized CMV antigens pp65 [median 273 spot-forming cells (SFC), range 47-995] and IE-1 (median 154 SFC, range 11-505), the Ad antigens hexon (median 153 SFC, range 26-465) and penton (median 37 SFC, range 1-353), as well as EBV lymphoblastoid cell lines (median 55 SFC, range 9-301). Importantly, the T cells recognized at least two antigens per virus and lysed virus peptide-pulsed targets. CONCLUSIONS CTL that target at least two antigens each of CMV, EBV and Ad should have clinical benefit with broad coverage of all three viruses and enhanced control of CMV infections compared with current protocols.


Journal of Immunology | 2011

Activation of Wnt signaling arrests effector differentiation in human peripheral and cord blood-derived T lymphocytes

Sujatha Muralidharan; Patrick J. Hanley; Enli Liu; Rikhia Chakraborty; Catherine M. Bollard; Elizabeth J. Shpall; Cliona M. Rooney; Barbara Savoldo; John R. Rodgers; Gianpietro Dotti

The canonical Wnt/β-catenin signaling pathway plays an important role in thymocyte development and T cell migration, but little is known about its role in naive-to-effector differentiation in human peripheral T cells. We show that activation of Wnt/β-catenin signaling arrests human peripheral blood and cord blood T lymphocytes in the naive stage and blocks their transition into functional T effector cells. Wnt signaling was induced in polyclonally activated human T cells by treatment either with the glycogen synthase kinase 3β inhibitor TWS119 or the physiological Wnt agonist Wnt-3a, and these T cells preserved a naive CD45RA+CD62L+ phenotype compared with control-activated T cells that progressed to a CD45RO+CD62L− effector phenotype, and this occurred in a TWS119 dose-dependent manner. TWS119-induced Wnt signaling reduced T cell expansion, as a result of a block in cell division, and impaired acquisition of T cell effector function, measured by degranulation and IFN-γ production in response to T cell activation. The block in T cell division may be attributed to the reduced IL-2Rα expression in TWS119-treated T cells that lowers their capacity to use autocrine IL-2 for expansion. Collectively, our data suggest that Wnt/β-catenin signaling is a negative regulator of naive-to-effector T cell differentiation in human T lymphocytes. The arrest in T cell differentiation induced by Wnt signaling might have relevant clinical applications such as to preserve the naive T cell compartment in Ag-specific T cells generated ex vivo for adoptive T cell immunotherapy.

Collaboration


Dive into the Patrick J. Hanley's collaboration.

Top Co-Authors

Avatar

Catherine M. Bollard

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Helen E. Heslop

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Elizabeth J. Shpall

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cliona M. Rooney

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Keller

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adrian P. Gee

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Ann M. Leen

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Barbara Savoldo

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Gianpietro Dotti

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge