Network


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

Hotspot


Dive into the research topics where Douglas V. Dolfi is active.

Publication


Featured researches published by Douglas V. Dolfi.


Science | 2012

Progenitor and Terminal Subsets of CD8+ T Cells Cooperate to Contain Chronic Viral Infection

Michael A. Paley; Daniela C. Kroy; Pamela M. Odorizzi; Jonathan B. Johnnidis; Douglas V. Dolfi; Burton E. Barnett; Elizabeth K. Bikoff; Elizabeth J. Robertson; Georg M. Lauer; Steven L. Reiner; E. John Wherry

Chronic infections strain the regenerative capacity of antiviral T lymphocyte populations, leading to failure in long-term immunity. The cellular and molecular events controlling this regenerative capacity, however, are unknown. We found that two distinct states of virus-specific CD8+ T cells exist in chronically infected mice and humans. Differential expression of the T-box transcription factors T-bet and Eomesodermin (Eomes) facilitated the cooperative maintenance of the pool of antiviral CD8+ T cells during chronic viral infection. T-bethi cells displayed low intrinsic turnover but proliferated in response to persisting antigen, giving rise to Eomeshi terminal progeny. Genetic elimination of either subset resulted in failure to control chronic infection, which suggests that an imbalance in differentiation and renewal could underlie the collapse of immunity in humans with chronic infections. Chronic viral infections like HIV are kept in check by two functionally distinct types of T lymphocyte. Chronic infections like hepatitis C virus (HCV) or HIV are hard on the immune system. In the face of a constant threat, some immune cells like T lymphocytes become “exhausted”; although present, they can no longer mount responses that are effective enough to eliminate the virus. These responses, however, are still important because in many cases they do keep the virus relatively controlled. The mechanisms underlying the population dynamics of T cell responses during chronic viral infection, however, are not well understood. Paley et al. (p. 1220) now demonstrate that the T-box transcription factors T-bet and Eomesodermin differentially regulate two phenotypically and functionally distinct subsets of antiviral CD8+ T cells in mice. The cooperation of these subsets may be important for antiviral immunity during chronic viral infections in humans.


Clinical Cancer Research | 2014

Multifactorial T-cell Hypofunction That Is Reversible Can Limit the Efficacy of Chimeric Antigen Receptor–Transduced Human T cells in Solid Tumors

Edmund Moon; Liang-Chuan Wang; Douglas V. Dolfi; Caleph B. Wilson; Raghuveer Ranganathan; Jing Sun; Veena Kapoor; John Scholler; Ellen Puré; Michael C. Milone; Carl H. June; James L. Riley; E. John Wherry; Steven M. Albelda

Purpose: Immunotherapy using vaccines or adoptively transferred tumor-infiltrating lymphocytes (TIL) is limited by T-cell functional inactivation within the solid tumor microenvironment. The purpose of this study was to determine whether a similar tumor-induced inhibition occurred with genetically modified cytotoxic T cells expressing chimeric antigen receptors (CAR) targeting tumor-associated antigens. Experimental Design: Human T cells expressing CAR targeting mesothelin or fibroblast activation protein and containing CD3ζ and 4–1BB cytoplasmic domains were intravenously injected into immunodeficient mice bearing large, established human mesothelin-expressing flank tumors. CAR TILs were isolated from tumors at various time points and evaluated for effector functions and status of inhibitory pathways. Results: CAR T cells were able to traffic into tumors with varying efficiency and proliferate. They were able to slow tumor growth, but did not cause regressions or cures. The CAR TILs underwent rapid loss of functional activity that limited their therapeutic efficacy. This hypofunction was reversible when the T cells were isolated away from the tumor. The cause of the hypofunction seemed to be multifactorial and was associated with upregulation of intrinsic T-cell inhibitory enzymes (diacylglycerol kinase and SHP-1) and the expression of surface inhibitory receptors (PD1, LAG3, TIM3, and 2B4). Conclusions: Advanced-generation human CAR T cells are reversibly inactivated within the solid tumor microenvironment of some tumors by multiple mechanisms. The model described here will be an important tool for testing T cell–based strategies or systemic approaches to overcome this tumor-induced inhibition. Our results suggest that PD1 pathway antagonism may augment human CAR T-cell function. Clin Cancer Res; 20(16); 4262–73. ©2014 AACR.


PLOS Pathogens | 2010

Perforin and IL-2 Upregulation Define Qualitative Differences among Highly Functional Virus-Specific Human CD8(+) T Cells

George Makedonas; Natalie A. Hutnick; Danielle Haney; Alexandra C. Amick; Jay Gardner; Gabriela L. Cosma; Adam R. Hersperger; Douglas V. Dolfi; E. John Wherry; Guido Ferrari; Michael R. Betts

The prevailing paradigm of T lymphocyte control of viral replication is that the protective capacity of virus-specific CD8+ T cells is directly proportional to the number of functions they can perform, with IL-2 production capacity considered critical. Having recently defined rapid perforin upregulation as a novel effector function of antigen-specific CD8+ T cells, here we sought to determine whether new perforin production is a component of polyfunctional CD8+ T cell responses that contributes to the control of several human viral infections: cytomegalovirus (CMV), Epstein-Barr virus (EBV), influenza (flu), and adenovirus (Ad). We stimulated normal human donor PBMC with synthetic peptides whose amino acid sequences correspond to defined CTL epitopes in the aforementioned viruses, and then used polychromatic flow cytometry to measure the functional capacity and the phenotype of the responding CD8+ T cells. While EBV and flu-specific CD8+ T cells rarely upregulate perforin, CMV-specific cells often do and Ad stimulates an exceptionally strong perforin response. The differential propensity of CD8+ T cells to produce either IL-2 or perforin is in part related to levels of CD28 and the transcription factor T-bet, as CD8+ T cells that rapidly upregulate perforin harbor high levels of T-bet and those producing IL-2 express high amounts of CD28. Thus, “polyfunctional” profiling of antigen-specific CD8+ T cells must not be limited to simply the number of functions the cell can perform, or one particular memory phenotype, but should actually define which combinations of memory markers and functions are relevant in each pathogenic context.


Journal of Immunology | 2008

Late Signals from CD27 Prevent Fas-Dependent Apoptosis of Primary CD8+ T Cells

Douglas V. Dolfi; Alina C. Boesteanu; Constantinos Petrovas; Dong Xia; Eric A. Butz; Peter D. Katsikis

The role of costimulation has previously been confined to the very early stages of the CD8+ T cell response. In this study, we demonstrate the requirement for CD27 costimulation during the later phase, but not programming of the primary CD8+ T cell response to influenza virus and reveal a novel mechanism of action for CD27 costimulation. CD27 signals, during the later phase of the primary CD8+ T cell response, prevent apoptosis of Ag-specific CD8+ T cells. Blocking CD27L (CD70) on days 6 and 8 after infection reduces the number of NP(366–374)-specific CD8+ T cells, increases their sensitivity to CD95/Fas-mediated apoptosis, and up-regulates FasL on CD4+ T cells. This reduction of NP(366–374)-specific CD8+ T cells requires the presence of CD4+ T cells and Fas signaling. Lack of CD27 signals also decreases the quality of memory CD8+ T cell responses. Memory CD8+ T cells, which express surface CD27 similar to naive cells, however, do not require CD27 costimulation during a secondary response. Thus, CD27 acts indirectly to regulate primary Ag-specific CD8+ T cell responses by preventing apoptosis of CD8+ T cells during the later phase of the primary response and is required for optimal quality of memory cells, but is not required during normally primed secondary CD8+ T cell responses.


Journal of Immunology | 2011

Dendritic Cells and CD28 Costimulation Are Required To Sustain Virus-Specific CD8+ T Cell Responses during the Effector Phase In Vivo

Douglas V. Dolfi; Priyanka A. Duttagupta; Alina C. Boesteanu; Yvonne M. Mueller; Caspian H. Oliai; Annie B. Borowski; Peter D. Katsikis

Although much is known about the initiation of immune responses, much less is known about what controls the effector phase. CD8+ T cell responses are believed to be programmed in lymph nodes during priming without any further contribution by dendritic cells (DCs) and Ag. In this study, we report the requirement for DCs, Ag, and CD28 costimulation during the effector phase of the CD8+ T cell response. Depleting DCs or blocking CD28 after day 6 of primary influenza A virus infection decreases the virus-specific CD8+ T cell response by inducing apoptosis, and this results in decreased viral clearance. Furthermore, effector CD8+ T cells adoptively transferred during the effector phase fail to expand without DC, CD28 costimulation, and cognate Ag. The absence of costimulation also leads to reduced survival of virus-specific effector cells as they undergo apoptosis mediated by the proapoptotic molecule Bim. Finally, IL-2 treatment restored the effector response in the absence of CD28 costimulation. Thus, in contrast to naive CD8+ T cells, which undergo an initial Ag-independent proliferation, effector CD8+ T cells expanding in the lungs during the effector phase require Ag, CD28 costimulation, and DCs for survival and expansion. These requirements would greatly impair effector responses against viruses and tumors that are known to inhibit DC maturation and in chronic infections and aging where CD28−/− CD8+ T cells accumulate.


Immunity | 2014

Bystander Chronic Infection Negatively Impacts Development of CD8+ T Cell Memory

Erietta Stelekati; Haina Shin; Travis A. Doering; Douglas V. Dolfi; Carly G.K. Ziegler; Daniel P. Beiting; Lucas Dawson; Jennifer Liboon; David Wolski; Mohammed-Alkhatim Ali; Peter D. Katsikis; Hao Shen; David S. Roos; W. Nicholas Haining; Georg M. Lauer; E. John Wherry

Epidemiological evidence suggests that chronic infections impair immune responses to unrelated pathogens and vaccines. The underlying mechanisms, however, are unclear and distinguishing effects on priming versus development of immunological memory has been challenging. We investigated whether bystander chronic infections impact differentiation of memory CD8(+) T cells, the hallmark of protective immunity against intracellular pathogens. Chronic bystander infections impaired development of memory CD8(+) T cells in several mouse models and humans. These effects were independent of initial priming and were associated with chronic inflammatory signatures. Chronic inflammation negatively impacted the number of bystander CD8(+) T cells and their memory development. Distinct underlying mechanisms of altered survival and differentiation were revealed with the latter regulated by the transcription factors T-bet and Blimp-1. Thus, exposure to prolonged bystander inflammation impairs the effector to memory transition. These data have relevance for immunity and vaccination during persisting infections and chronic inflammation.


Journal of Immunology | 2014

Circulating CXCR5+PD-1+ Response Predicts Influenza Vaccine Antibody Responses in Young Adults but not Elderly Adults

Ramin S. Herati; Morgan A. Reuter; Douglas V. Dolfi; Kathleen D. Mansfield; Htin Aung; Osama Z. Badwan; Raj Kurupati; Senthil Kannan; Hildegund C.J. Ertl; Kenneth E. Schmader; Michael R. Betts; David H. Canaday; E. John Wherry

Although influenza vaccination is recommended for all adults annually, the incidence of vaccine failure, defined as weak or absent increase in neutralizing Ab titers, is increased in the elderly compared with young adults. The T follicular helper cell (Tfh) subset of CD4 T cells provides B cell help in germinal centers and is necessary for class-switched Ab responses. Previous studies suggested a role for circulating Tfh cells (cTfh) following influenza vaccination in adults, but cTfh have not been studied in elderly adults in whom weak vaccine responses are often observed. In this study, we studied cTfh expressing CXCR5 and programmed death-1 (PD-1). cTfh from elderly adults were present at reduced frequency, had decreased in vitro B cell help ability, and had greater expression of ICOS compared with young adults. At 7 d after inactivated influenza vaccination, cTfh correlated with influenza vaccine–specific IgM and IgG responses in young adults but not in elderly adults. In sum, we have identified aging-related changes in cTfh that correlated with reduced influenza vaccine responses. Future rational vaccine design efforts should incorporate Tfh measurement as an immune correlate of protection, particularly in the setting of aging.


Journal of Virology | 2013

Persistent Enteric Murine Norovirus Infection Is Associated with Functionally Suboptimal Virus-Specific CD8 T Cell Responses

Vesselin Tomov; Lisa C. Osborne; Douglas V. Dolfi; Gregory F. Sonnenberg; Laurel A. Monticelli; Kathleen D. Mansfield; Herbert W. Virgin; David Artis; E. J. Wherry

ABSTRACT Norovirus (NV) gastroenteritis is a major contributor to global morbidity and mortality, yet little is known about immune mechanisms leading to NV control. Previous studies using the murine norovirus (MNV) model have established a key role for T cells in MNV clearance. Despite these advances, important questions remain regarding the magnitude, location, and dynamics of the MNV-specific T cell response. To address these questions, we identified MNV-specific major histocompatibility complex (MHC) class I immunodominant epitopes using an overlapping peptide screen. One of these epitopes (amino acids 519 to 527 of open reading frame 2 [ORF2519-527]) was highly conserved among all NV genogroups. Using MHC class I peptide tetramers, we tracked MNV-specific CD8 T cells in lymphoid and mucosal sites during infection with two MNV strains with distinct biological behaviors, the acutely cleared strain CW3 and the persistent strain CR6. Here, we show that enteric MNV infection elicited robust T cell responses primarily in the intestinal mucosa and that MNV-specific CD8 T cells dynamically regulated the expression of surface molecules associated with activation, differentiation, and homing. Furthermore, compared to MNV-CW3 infection, chronic infection with MNV-CR6 resulted in fewer and less-functional CD8 T cells, and this difference was evident as early as day 8 postinfection. Finally, MNV-specific CD8 T cells were capable of reducing the viral load in persistently infected Rag1 −/− mice, suggesting that these cells are a crucial component of NV immunity. Collectively, these data provide fundamental new insights into the adaptive immune response to two closely related NV strains with distinct biological behaviors and bring us closer to understanding the correlates of protective antiviral immunity in the intestine.


Journal of Leukocyte Biology | 2013

Increased T-bet is associated with senescence of influenza virus-specific CD8 T cells in aged humans

Douglas V. Dolfi; Kathleen D. Mansfield; Antonio Polley; Susan A. Doyle; Gordon J. Freeman; Hanspeter Pircher; Kenneth E. Schmader; E. John Wherry

Aged individuals have increased morbidity and mortality following influenza and other viral infections, despite previous exposure or vaccination. Mouse and human studies suggest increased senescence and/or exhaustion of influenza virus‐specific CD8 T cells with advanced age. However, neither the relationship between senescence and exhaustion nor the underlying transcriptional pathways leading to decreased function of influenza virus‐specific cellular immunity in elderly humans are well‐defined. Here, we demonstrate that increased percentages of CD8 T cells from aged individuals express CD57 and KLRG1, along with PD‐1 and other inhibitory receptors, markers of senescence, or exhaustion, respectively. Expression of T‐box transcription factors, T‐bet and Eomes, were also increased in CD8 T cells from aged subjects and correlated closely with expression of CD57 and KLRG1. Influenza virus‐specific CD8 T cells from aged individuals exhibited decreased functionality with corresponding increases in CD57, KLRG1, and T‐bet, a molecular regulator of terminal differentiation. However, in contrast to total CD8 T cells, influenza virus‐specific CD8 T cells had altered expression of inhibitory receptors, including lower PD‐1, in aged compared with young subjects. Thus, our data suggest a prominent role for senescence and/or terminal differentiation for influenza virus‐specific CD8 T cells in elderly subjects.


Advances in Experimental Medicine and Biology | 2007

CD28 and Cd27 Costimulation of Cd8+ T Cells: A Story of Survival

Douglas V. Dolfi; Peter D. Katsikis

Although the requirement of CD28 and CD27 costimulation has been clearly demonstrated during primary CD8+ T cell responses and this costimulation acts by providing proliferation and survival cues to naive CD8+ T cells, a number of questions also arise from these studies. Is the requirement for CD28 and CD27 costimulation restricted to the initiation of the immune response in the lymph nodes, where presumably the initial contact between naive CD8+ T cell and DC occurs? What is the purpose of the dramatic influx of DC to sites of inflammation such as the lung during influenza virus infection and the formation of inflammatory BALT (iBALT)?(104) Are such DC at the site of inflammation and at later stages of the immune response providing cytokines or costimulation to effector CD8+ T cells? If DC are required for optimal secondary responses (100), is CD28 costimulation the missing signal or is it other members of the B7:CD28 family or TNF family? Given that a number of investigators are actively addressing these questions, the answers we expect will be soon to come and open exciting new opportunities for immune enhancement or dampening strategies and vaccine adjuvants.

Collaboration


Dive into the Douglas V. Dolfi's collaboration.

Top Co-Authors

Avatar

E. John Wherry

University of Pennsylvania

View shared research outputs
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

Carly G.K. Ziegler

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel P. Beiting

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David S. Roos

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge