Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Ther ; 25(2): 494-503, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28153096

ABSTRACT

To be effective against HIV type 1 (HIV-1), vaccine-induced T cells must selectively target epitopes, which are functionally conserved (present in the majority of currently circulating and reactivated HIV-1 strains) and, at the same time, beneficial (responses to which are associated with better clinical status and control of HIV-1 replication), and rapidly reach protective frequencies upon exposure to the virus. Heterologous prime-boost regimens using virally vectored vaccines are currently the most promising vaccine strategies; nevertheless, induction of robust long-term memory remains challenging. To this end, lentiviral vectors induce high frequencies of memory cells due to their low-inflammatory nature, while typically inducing only low anti-vector immune responses. Here, we describe construction of novel candidate vaccines ZVex.tHIVconsv1 and ZVex.tHIVconsv2, which are based on an integration-deficient lentiviral vector platform with preferential transduction of human dendritic cells and express a bivalent mosaic of conserved-region T cell immunogens with a high global HIV-1 match. Each of the two mosaic vaccines was individually immunogenic. When administered together in heterologous prime-boost regimens with chimpanzee adenovirus and/or poxvirus modified vaccinia virus Ankara (MVA) vaccines to BALB/c and outbred CD1-Swiss mice, they induced a median frequency of over 6,000 T cells/106 splenocytes, which were plurifunctional, broadly specific, and cross-reactive. These results support further development of this vaccine concept.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Vectors/genetics , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Lentivirus/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Conserved Sequence , Disease Models, Animal , Epitopes/genetics , Epitopes/immunology , Female , Gene Order , HIV Infections/virology , Humans , Immunity, Cellular , Mice , Peptides/genetics , Peptides/immunology
2.
Mol Ther ; 24(4): 832-42, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26743582

ABSTRACT

An effective human immunodeficiency virus type 1 (HIV-1) vaccine is the best solution for halting the acquired immune deficiency syndrome epidemic. Here, we describe the design and preclinical immunogenicity of T-cell vaccine expressing novel immunogens tHIVconsvX, vectored by DNA, simian (chimpanzee) adenovirus, and poxvirus modified vaccinia virus Ankara (MVA), a combination highly immunogenic in humans. The tHIVconsvX immunogens combine the three leading strategies for elicitation of effective CD8(+) T cells: use of regions of HIV-1 proteins functionally conserved across all M group viruses (to make HIV-1 escape costly on viral fitness), inclusion of bivalent complementary mosaic immunogens (to maximize global epitope matching and breadth of responses, and block common escape paths), and inclusion of epitopes known to be associated with low viral load in infected untreated people (to induce field-proven protective responses). tHIVconsvX was highly immunogenic in two strains of mice. Furthermore, the magnitude and breadth of CD8(+) T-cell responses to tHIVconsvX-derived peptides in treatment-naive HIV-1(+) patients significantly correlated with high CD4(+) T-cell count and low viral load. Overall, the tHIVconsvX design, combining the mosaic and conserved-region approaches, provides an indisputably better coverage of global HIV-1 variants than previous T-cell vaccines. These immunogens delivered in a highly immunogenic framework of adenovirus prime and MVA boost are ready for clinical development.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV Infections/therapy , Adenoviruses, Simian/immunology , Animals , CD4 Lymphocyte Count , HIV-1/physiology , HeLa Cells , Humans , Mice , Vaccinia virus/immunology , Viral Load
3.
Mol Ther ; 24(2): 375-384, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26581160

ABSTRACT

The biggest roadblock in development of effective vaccines against human immunodeficiency virus type 1 (HIV-1) is the virus genetic diversity. For T-cell vaccine, this can be tackled by focusing the vaccine-elicited T-cells on the highly functionally conserved regions of HIV-1 proteins, mutations in which typically cause a replicative fitness loss, and by computing multivalent mosaic proteins, which maximize the coverage of potential 9-mer T-cell epitopes of the input viral sequences. Our first conserved region vaccines HIVconsv employed clade alternating consensus sequences and showed promise in the initial clinical trials in terms of magnitude and breadth of elicited CD8(+) T-cells. Here, monitoring T-cells restricted by HLA-A*02:01 in transgenic mice, we assessed whether or not the tHIVconsv design (HIVconsv with a tissue plasminogen activator leader sequence) benefits from combining with a complementing conserved mosaic immunogen tHIVcmo, and compared the bivalent immunization to that with trivalent conserved mosaic vaccines. A hierarchy of tHIVconsv ≤ tHIVconsv+tHIVcmo < tCmo1+tCmo2+tCmo3 vaccinations for induction of CD8(+) T-cell responses was observed in terms of recognition of tested peptide variants. Thus, our HLA-A*02:01-restricted epitope data concur with previously published mouse and macaque observations and suggest that even conserved region vaccines benefit from oligovalent mosaic design.


Subject(s)
AIDS Vaccines/immunology , Epitopes, T-Lymphocyte/metabolism , HIV Infections/therapy , HLA-A2 Antigen/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Mice , Mice, Transgenic
4.
J Virol ; 89(11): 5760-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25810538

ABSTRACT

UNLABELLED: Cytotoxic T cells substantially contribute to the control of intracellular pathogens such as human immunodeficiency virus type 1 (HIV-1). Here, we evaluated the immunopeptidome of Jurkat cells infected with the vaccine candidate MVA.HIVconsv, which delivers HIV-1 conserved antigenic regions by using modified vaccinia virus Ankara (MVA). We employed liquid chromatography-tandem mass spectrometry (LC-MS/MS) to identify 6,358 unique peptides associated with the class I human leukocyte antigen (HLA), of which 98 peptides were derived from the MVA vector and 7 were derived from the HIVconsv immunogen. Human vaccine recipients responded to the peptide sequences identified by LC-MS/MS. Peptides derived from the conserved HIV-1 regions were readily detected as early as 1.5 h after MVA.HIVconsv infection. Four of the seven conserved peptides were monitored between 0 and 3.5 h of infection by using quantitative mass spectrometry (Q-MS), and their abundance in HLA class I associations reflected levels of the whole HIVconsv protein in the cell. While immunopeptides delivered by the incoming MVA vector proteins could be detected, all early HIVconsv-derived immunopeptides were likely synthesized de novo. MVA.HIVconsv infection generally altered the composition of HLA class I-associated human (self) peptides, but these changes corresponded only partially to changes in the whole cell host protein abundance. IMPORTANCE: The vast changes in cellular antigen presentation after infection of cells with a vectored vaccine, as shown here for MVA.HIVconsv, highlight the complexity of factors that need to be considered for efficient antigen delivery and presentation. Identification and quantitation of HLA class I-associated peptides by Q-MS will not only find broad application in T-cell epitope discovery but also inform vaccine design and allow evaluation of efficient epitope presentation using different delivery strategies.


Subject(s)
AIDS Vaccines/immunology , Antigens, Viral/analysis , HIV-1/immunology , Histocompatibility Antigens Class I/metabolism , Peptides/analysis , T-Lymphocytes, Cytotoxic/chemistry , Chromatography, Liquid , Genetic Vectors , Humans , Jurkat Cells , T-Lymphocytes, Cytotoxic/immunology , Tandem Mass Spectrometry , Time Factors , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
5.
Cancer Med ; 4(3): 457-71, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25641882

ABSTRACT

Latent membrane protein 2A (LMP2A) is expressed on almost all Epstein-Barr virus (EBV)-associated tumors and is a potential target for immunotherapeutic intervention and vaccination. However, LMP2A is not efficiently processed and presented on major histocompatibility antigens class I molecules to generate potent cytotoxic T-lymphocytes (CTL) responses capable of killing these tumors. The B subunit of Escherichia coli enterotoxin (EtxB), causes rapid internalization and processing of membrane-bound LMP2A on EBV-infected B cells, and facilitates loading of processed-LMP2A peptides onto MHC class I. This re-directed trafficking/delivery of LMP2A to the MHC class I machinery enhances recognition and killing by LMP2A-specific CTL in vitro. To test the potential of EtxB to enhance immune targeting of LMP2A expressed in solid tumors, we generated a murine tumor model (Renca-LMP2A), in which LMP2A is expressed as a transgenic neoantigen on a renal carcinoma (Renca) cell line and forms solid tumors when injected subcutaneously into BALB/c mice. The data show that in BALB/c mice which have only low levels of peripheral K(d)-LMP2A-specific CD8(+) T cells, merely a transient inhibition of tumor growth is achieved compared with naïve mice; suggesting that there is suboptimal LMP2A-specifc CTL recognition and poorly targeted tumor killing. However, importantly, treatment of these mice with EtxB led to a significant delay in the onset of tumor growth and significantly lower tumor volumes compared with similar mice that did not receive EtxB. Moreover, this remarkable effect of EtxB was achieved despite progressive reduction in tumor expression of LMP2A and MHC class I molecules. These data clearly demonstrate the potential efficacy of EtxB as a novel therapeutic agent that could render EBV-associated tumors susceptible to immune control.


Subject(s)
Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Enterotoxins/therapeutic use , Escherichia coli Proteins/therapeutic use , Neoplasms/drug therapy , Viral Matrix Proteins/metabolism , Animals , Cell Line, Tumor , Female , Immunization , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
6.
Immunology ; 145(1): 94-104, 2015 May.
Article in English | MEDLINE | ID: mdl-25495686

ABSTRACT

The frequency of CD4(+)  Foxp3(+) regulatory T (Treg) cells is often significantly increased in the blood of tumour-bearing mice and people with cancer. Moreover, Treg cell frequencies are often higher in tumours compared with blood and lymphoid organs. We wished to determine whether certain chemokines expressed within the tumour mass selectively recruit Treg cells, thereby contributing to their enrichment within the tumour-infiltrating lymphocyte pool. To achieve this goal, the chemokine profile of carcinogen-induced fibrosarcomas was determined, and the chemokine receptor expression profiles of both CD4(+)  Foxp3(-) and CD4(+)  Foxp3(+) T cells were compared. These analyses revealed that the tumours are characterized by expression of inflammatory chemokines (CCL2, CCL5, CCL7, CCL8, CCL12, CXCL9, CXCL10 and CX3CL1), reflected by an enrichment of activated Foxp3(-) and Foxp3(+) T cells expressing T helper type 1-associated chemokine receptors. Notably, we found that CXCR3(+) T cells were significantly enriched in the tumours although curiously we found no evidence that CXCR3 was required for their recruitment. Instead, CXCR3 marks a population of activated Foxp3(-) and Foxp3(+) T cells, which use multiple and overlapping ligand receptor pairs to guide their migration to tumours. Collectively, these data indicate that enrichment of Foxp3(+) cells in tumours characterized by expression of inflammatory chemokines, does not occur via a distinct chemokine axis, thus selective chemokine blockade is unlikely to represent a meaningful therapeutic strategy for preventing Treg cell accumulation in tumours.


Subject(s)
Carcinogens/toxicity , Chemokines/immunology , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Chemokines/genetics , Female , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Humans , Methylcholanthrene/toxicity , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology
7.
Clin Vaccine Immunol ; 21(11): 1565-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25230940

ABSTRACT

A likely requirement for a protective vaccine against human immunodeficiency virus type 1 (HIV-1)/AIDS is, in addition to eliciting antibody responses, induction of effective T cells. To tackle HIV-1 diversity by T-cell vaccines, we designed an immunogen, HIVconsv, derived from the most functionally conserved regions of the HIV-1 proteome and demonstrated its high immunogenicity in humans and rhesus macaques when delivered by regimens combining plasmid DNA, nonreplicating simian (chimpanzee) adenovirus ChAdV-63, and nonreplicating modified vaccinia virus Ankara (MVA) as vectors. Here, we aimed to increase the decision power for iterative improvements of this vaccine strategy in the BALB/c mouse model. First, we found that prolonging the period after the ChAdV63.HIVconsv prime up to 6 weeks increased the frequencies of HIV-1-specific, gamma interferon (IFN-γ)-producing T cells induced by the MVA.HIVconsv boost. Induction of strong responses allowed us to map comprehensively the H-2(d)-restricted T-cell responses to these regions and identified 8 HIVconsv peptides, of which three did not contain a previously described epitope and were therefore considered novel. Induced effector T cells were oligofunctional and lysed sensitized targets in vitro. Our study therefore provides additional tools for studying and optimizing vaccine regimens in this commonly used small animal model, which will in turn guide vaccine improvements in more expensive nonhuman primate and human clinical trials.


Subject(s)
AIDS Vaccines/immunology , Conserved Sequence/immunology , HIV-1/immunology , Interferon-gamma/metabolism , T-Lymphocytes/immunology , Viral Proteins/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/isolation & purification , Adenoviridae/genetics , Animals , Drug Carriers , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccinia virus/genetics
8.
Vaccine ; 32(44): 5801-8, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25173484

ABSTRACT

BACKGROUND: A safe, effective vaccine for breastfeeding infants born to HIV-1-positive mothers could complement antiretroviral therapy (ART) for prevention of mother-to-child transmission of HIV-1. To date, only a few HIV-1 vaccine candidates have been tested in infants. TRIAL DESIGN: A phase I/II randomized controlled trial PedVacc 002 was conducted to determine the safety and immunogenicity of a single, low dose of MVA.HIVA vaccine delivered intramuscularly to healthy 20-week-old infants born to HIV-1-positive mothers in Nairobi, Kenya. METHODS: Pregnant HIV-1-positive women in the 2nd/3rd trimester of gestation were enrolled, provided with ART and self-selected their infant-feeding modality. Infants received nevirapine and cotrimoxazole prophylaxis. At 20 weeks of age, eligible HIV-1-negative infants were randomized to vaccine versus no-treatment arms and followed to 48 weeks of age for assessments of vaccine safety, HIV-1-specific T-cell responses and antibodies to routine childhood vaccines. RESULTS: Between February and November 2010, 182 mothers were screened, 104 were eligible and followed on ART during pregnancy/postpartum, of whom 73 had eligible infants at 20 weeks postpartum. Thirty-six infants were randomized to vaccine and 37 to no treatment. Eighty-four percent of infants breastfed, and retention at 48 weeks was 99%. Adverse events were rare and similar between the two arms. HIV-1-specific T-cell frequencies in interferon-γ ELISPOT assay were transiently higher in the MVA.HIVA arm (p=0.002), but not above the threshold for a positive assay. Protective antibody levels were adequate and similar between arms for all routine childhood vaccines except HBV, where 71% of MVA.HIVA subjects compared to 92% of control subjects were protected (p=0.05). CONCLUSIONS: This trial tested for the first time an MVA-vectored candidate HIV-1 vaccine in HIV-1-exposed infants in Africa, demonstrating trial feasibility and vaccine safety, low immunogenicity, and compatibility with routine childhood vaccinations. These results are reassuring for use of the MVA vector in more potent prime-boost regimens.


Subject(s)
AIDS Vaccines/therapeutic use , HIV Infections/prevention & control , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy Complications, Infectious/immunology , Adult , Anti-HIV Agents/therapeutic use , Female , HIV Antibodies/blood , HIV Infections/drug therapy , HIV-1 , Humans , Infant , Interferon-gamma/immunology , Kenya , Male , Mothers , Pregnancy , T-Lymphocytes/immunology , Vaccines, DNA , Young Adult
9.
Front Microbiol ; 5: 439, 2014.
Article in English | MEDLINE | ID: mdl-25202303

ABSTRACT

Development of an effective HIV/AIDS vaccine remains a big challenge, largely due to the enormous HIV diversity which propels immune escape. Thus novel vaccine strategies are targeting multiple variants of conserved antibody and T cell epitopic regions which would incur a huge fitness cost to the virus in the event of mutational escape. Besides immunogen design, the delivery modality is critical for vaccine potency and efficacy, and should be carefully selected in order to not only maximize transgene expression, but to also enhance the immuno-stimulatory potential to activate innate and adaptive immune systems. To date, five HIV vaccine candidates have been evaluated for efficacy and protection from acquisition was only achieved in a small proportion of vaccinees in the RV144 study which used a canarypox vector for delivery. Conversely, in the STEP study (HVTN 502) where human adenovirus serotype 5 (Ad5) was used, strong immune responses were induced but vaccination was more associated with increased risk of HIV acquisition than protection in vaccinees with pre-existing Ad5 immunity. The possibility that pre-existing immunity to a highly promising delivery vector may alter the natural course of HIV to increase acquisition risk is quite worrisome and a huge setback for HIV vaccine development. Thus, HIV vaccine development efforts are now geared toward delivery platforms which attain superior immunogenicity while concurrently limiting potential catastrophic effects likely to arise from pre-existing immunity or vector-related immuno-modulation. However, it still remains unclear whether it is poor immunogenicity of HIV antigens or substandard immunological potency of the safer delivery vectors that has limited the success of HIV vaccines. This article discusses some of the promising delivery vectors to be harnessed for improved HIV vaccine efficacy.

10.
Front Immunol ; 5: 202, 2014.
Article in English | MEDLINE | ID: mdl-24847327

ABSTRACT

The field of HIV prevention has indeed progressed in leaps and bounds, but with major limitations of the current prevention and treatment options, the world remains desperate for an HIV vaccine. Sadly, this continues to be elusive, because more than 30 years since its discovery there is no licensed HIV vaccine. Research aiming to define immunological biomarkers to accurately predict vaccine efficacy have focused mainly on systemic immune responses, and as such, studies defining correlates of protection in the genitorectal mucosa, the primary target site for HIV entry and seeding are sparse. Clearly, difficulties in sampling and analysis of mucosal specimens, as well as their limited size have been a major deterrent in characterizing the type (mucosal antibodies, cytokines, chemokines, or CTL), threshold (magnitude, depth, and breadth) and viral inhibitory capacity of HIV-1-specific immune responses in the genitorectal mucosa, where they are needed to immediately block HIV acquisition and arrest subsequent virus dissemination. Nevertheless, a few studies document the existence of HIV-specific immune responses in the genitorectal mucosa of HIV-infected aviremic and viremic controllers, as well as in highly exposed persistently seronegative (HEPS) individuals with natural resistance to HIV-1. Some of these responses strongly correlate with protection from HIV acquisition and/or disease progression, thus providing significant clues of the ideal components of an efficacious HIV vaccine. In this study, we provide an overview of the key features of protective immune responses found in HEPS, elite and viremic controllers, and discuss how these can be achieved through mucosal immunization. Inevitably, HIV vaccine development research will have to consider strategies that elicit potent antibody and cellular immune responses within the genitorectal mucosa or induction of systemic immune cells with an inherent potential to home and persist at mucosal sites of HIV entry.

11.
Front Immunol ; 5: 90, 2014.
Article in English | MEDLINE | ID: mdl-24639678

ABSTRACT

Excessive immune responses directed against foreign pathogens, self-antigens, or commensal microflora can cause cancer establishment and progression if the execution of tight immuno-regulatory mechanisms fails. On the other hand, induction of potent tumor antigen-specific immune responses together with stimulation of the innate immune system is a pre-requisite for effective anti-tumor immunity, and if suppressed by the strong immuno-regulatory mechanisms can lead to cancer progression. Therefore, it is crucial that the inevitable co-existence of these fundamental, yet conflicting roles of immune-regulatory cells is carefully streamlined as imbalances can be detrimental to the host. Infection with chronic persistent viruses is characterized by severe immune dysfunction resulting in T cell exhaustion and sometimes deletion of antigen-specific T cells. More often, this is due to increased immuno-regulatory processes, which are triggered to down-regulate immune responses and limit immunopathology. However, such heightened levels of immune disruption cause a concomitant loss of tumor immune-surveillance and create a permissive microenvironment for cancer establishment and progression, as demonstrated by increased incidences of cancer in immunosuppressed hosts. Paradoxically, while some cancers arise as a consequence of increased immuno-regulatory mechanisms that inhibit protective immune responses and impinge on tumor surveillance, other cancers arise due to impaired immuno-regulatory mechanisms and failure to limit pathogenic inflammatory responses. This intricate complexity, where immuno-regulatory cells can be beneficial in certain immune settings but detrimental in other settings underscores the need for carefully formulated interventions to equilibrate the balance between immuno-stimulatory and immuno-regulatory processes.

12.
Mol Ther ; 22(2): 464-475, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24166483

ABSTRACT

Virus diversity and escape from immune responses are the biggest challenges to the development of an effective vaccine against HIV-1. We hypothesized that T-cell vaccines targeting the most conserved regions of the HIV-1 proteome, which are common to most variants and bear fitness costs when mutated, will generate effectors that efficiently recognize and kill virus-infected cells early enough after transmission to potentially impact on HIV-1 replication and will do so more efficiently than whole protein-based T-cell vaccines. Here, we describe the first-ever administration of conserved immunogen vaccines vectored using prime-boost regimens of DNA, simian adenovirus and modified vaccinia virus Ankara to uninfected UK volunteers. The vaccine induced high levels of effector T cells that recognized virus-infected autologous CD4(+) cells and inhibited HIV-1 replication by up to 5.79 log10. The virus inhibition was mediated by both Gag- and Pol- specific effector CD8(+) T cells targeting epitopes that are typically subdominant in natural infection. These results provide proof of concept for using a vaccine to target T cells at conserved epitopes, showing that these T cells can control HIV-1 replication in vitro.


Subject(s)
AIDS Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , HIV Infections/immunology , HIV-1/immunology , T-Lymphocytes/immunology , AIDS Vaccines/genetics , Adolescent , Adult , Amino Acid Sequence , Cells, Cultured , Conserved Sequence/immunology , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Female , HIV Infections/prevention & control , HIV-1/genetics , Humans , Male , Middle Aged , Molecular Sequence Data , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Virus Replication/immunology , Young Adult , gag Gene Products, Human Immunodeficiency Virus/immunology , pol Gene Products, Human Immunodeficiency Virus/immunology
13.
Int J Cancer ; 134(9): 2156-67, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24142504

ABSTRACT

The tumor microenvironment comprises newly formed blood and lymphatic vessels which shape the influx, retention and departure of lymphocytes within the tumor mass. Thus, by influencing the intratumoral composition of lymphocytes, these vessels affect the manner in which the adaptive immune system responds to the tumor, either promoting or impairing effective antitumor immunity. In our study, we utilized a mouse model of carcinogen-induced fibrosarcoma to examine the composition of tumor-infiltrating lymphocytes during tumor progression. In particular, we sought to determine whether CD4(+) Foxp3(+) regulatory T cells (Tregs) became enriched during tumor progression thereby contributing to tumor-driven immunosuppression. This was not the case as the proportion of Tregs and effector CD4(+) T cells actually declined within the tumor owing to the unexpected accumulation of naïve T cells. However, we found no evidence for antigen-driven migration of these T cells or for their participation in an antitumor immune response. Our data support the notion that lymphocytes can enter tumors via aberrantly formed blood and lymphatic vessels. Such findings suggest that targeting both the tumor vasculature and lymphatics will alter the balance of lymphocyte subpopulations that enter the tumor mass. A consideration of this aspect of tumor immunology may be critical to the success of solid cancer immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fibrosarcoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neovascularization, Pathologic/immunology , Tumor Microenvironment/immunology , Animals , Carcinogens/toxicity , Disease Models, Animal , Disease Progression , Female , Fibrosarcoma/pathology , Flow Cytometry , Immunohistochemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology
14.
Vaccine ; 31(47): 5594-601, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-23831324

ABSTRACT

BACKGROUND: The systemic toxicity of three candidate HIV-1 vaccines plasmid pSG2.HIVconsv DNA (D), ChAdV63.HIVconsv (C) and MVA.HIVconsv (M) expressing chimeric immunogen derived from the most conserved regions of the HIV-1 proteome was evaluated in two repeat-dose studies in the male and female BALB/c mice. METHODS: In study UNO011, mice received three doses of 2×10(7) plaque-forming units of MVA.HIVconsv vaccine (MMM). In study UNO012, mice received 3 doses of 50µg of pSG2.HIVconsv DNA followed by a single dose of 5.95×10(9) virus particles of ChAdV63.HIVconsv vaccine (DDDC). Similarly constituted control groups received the vehicle alone (phosphate buffered saline) at the same volume-dose. All vaccines were administered by intramuscular needle injection into the right hind limb at 14-day intervals and animals were sacrificed 7 days after the last dose. Assessment of local and systemic toxicity was made. Induction of HIV-1-specific responses was confirmed. Parameters assessed included clinical condition, body weight, food consumption, ophthalmoscopy, haematology, blood chemistry, organ weight and macroscopic and microscopic pathology. RESULTS: In both studies, treatment with the candidate vaccines elicited strong HIV-1-specific T-cell responses. The vaccine treatment was well-tolerated without any adverse systemic toxicological changes. The local toxicity findings observed in these studies were consistent with the predicted response to a vaccine/substance administration by intramuscular injection. CONCLUSIONS: The three novel anti-HIV-1 vaccines were well tolerated when administered by intramuscular injection to BALB/c mice. These results supported an application for authorisation by the Medicines and Healthcare Products Regulatory Agency of the UK to test these vaccines for the first time in phase I clinical trials in healthy both uninfected subjects and HIV-1-infected patients stable on antiretroviral treatment.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/adverse effects , HIV-1/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Adenoviridae/genetics , Animals , Drug Carriers , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Injections, Intramuscular , Male , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccinia virus/genetics
15.
Eur J Immunol ; 43(10): 2613-25, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23857287

ABSTRACT

Balancing the generation of immune responses capable of controlling virus replication with those causing immunopathology is critical for the survival of the host and resolution of influenza-induced inflammation. Based on the capacity of interleukin-6 (IL-6) to govern both optimal T-cell responses and inflammatory resolution, we hypothesised that IL-6 plays an important role in maintaining this balance. Comparison of innate and adaptive immune responses in influenza-infected wild-type control and IL-6-deficient mice revealed striking differences in virus clearance, lung immunopathology and generation of heterosubtypic immunity. Mice lacking IL-6 displayed a profound defect in their ability to mount an anti-viral T-cell response. Failure to adequately control virus was further associated with an enhanced infiltration of inflammatory monocytes into the lung and an elevated production of the pro-inflammatory cytokines, IFN-α and TNF-α. These events were associated with severe lung damage, characterised by profound vascular leakage and death. Our data highlight an essential role for IL-6 in orchestrating anti-viral immunity through an ability to limit inflammation, promote protective adaptive immune responses and prevent fatal immunopathology.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Influenza A virus/physiology , Interleukin-6/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/immunology , Animals , CD4-Positive T-Lymphocytes/virology , Cell Movement/genetics , Cells, Cultured , Cytokines/metabolism , Female , Inflammation Mediators/metabolism , Interleukin-6/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/pathology , Viral Load/genetics , Virus Replication/genetics
16.
Front Immunol ; 4: 197, 2013.
Article in English | MEDLINE | ID: mdl-23874342

ABSTRACT

CD4(+)Foxp3(+) regulatory T cells (Tregs) have a fundamental role in maintaining immune balance by preventing autoreactivity and immune-mediated pathology. However this role of Tregs extends to suppression of anti-tumor immune responses and remains a major obstacle in the development of anti-cancer vaccines and immunotherapies. This feature of Treg activity is exacerbated by the discovery that Treg frequencies are not only elevated in the blood of cancer patients, but are also significantly enriched within tumors in comparison to other sites. These observations have sparked off the quest to understand the processes through which Tregs become elevated in cancer-bearing hosts and to identify the specific mechanisms leading to their accumulation within the tumor microenvironment. This manuscript reviews the evidence for specific mechanisms of intra-tumoral Treg enrichment and will discuss how this information may be utilized for the purpose of manipulating the balance of tumor-infiltrating T cells in favor of anti-tumor effector cells.

17.
Cancer Res ; 72(21): 5473-82, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22962270

ABSTRACT

The evolution of immune blockades in tumors limits successful antitumor immunity, but the mechanisms underlying this process are not fully understood. Depletion of regulatory T cells (Treg), a T-cell subset that dampens excessive inflammatory and autoreactive responses, can allow activation of tumor-specific T cells. However, cancer immunotherapy studies have shown that a persistent failure of activated lymphocytes to infiltrate tumors remains a fundamental problem. In evaluating this issue, we found that despite an increase in T-cell activation and proliferation following Treg depletion, there was no significant association with tumor growth rate. In contrast, there was a highly significant association between low tumor growth rate and the extent of T-cell infiltration. Further analyses revealed a total concordance between low tumor growth rate, high T-cell infiltration, and the presence of high endothelial venules (HEV). HEV are blood vessels normally found in secondary lymphoid tissue where they are specialized for lymphocyte recruitment. Thus, our findings suggest that Treg depletion may promote HEV neogenesis, facilitating increased lymphocyte infiltration and destruction of the tumor tissue. These findings are important as they point to a hitherto unidentified role of Tregs, the manipulation of which may refine strategies for more effective cancer immunotherapy.


Subject(s)
Chemotaxis, Leukocyte/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Venules/immunology , Animals , Cell Proliferation , Endothelium, Vascular/immunology , Flow Cytometry , Immunohistochemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/pathology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
18.
J Infect Dis ; 199(12): 1771-9, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19419333

ABSTRACT

BACKGROUND: Cellular immune responses characterized by interferon gamma (IFN-gamma) production enhance clearance and confer protective immunity against Chlamydia trachomatis infection but have not been simultaneously investigated in systemic and mucosal compartments. METHODS: With use of the IFN-gamma enzyme-linked immunosorbent spot assay, we investigated immune responses to Chlamydia elementary body (EB) and 3 genotypically variant heat shock protein 60 (CHSP60) antigens using peripheral blood mononuclear cells and endometrial mononuclear cells obtained from a female sex worker cohort with high levels of exposure to C. trachomatis. RESULTS: Although we observed a marginally higher frequency of IFN-gamma responses to EB in peripheral blood mononuclear cells, compared with the frequency in endometrial mononuclear cells, the magnitudes of systemic and mucosal responses were similar except for preferential targeting of CHSP60 type 2 by endometrial mononuclear cells. Systemic and mucosal responses were highly correlated for EB and CHSP60 types 1 and 2 but not type 3. The frequency and magnitude of systemic responses specific for EB and CHSP60 type 1 were greater for CD4+ T cells than they were for CD8+ T cells, whereas preferential targeting by CHSP60 types 2 and 3 was undetectable. IFN-gamma response to CHSP60 type 1 by peripheral blood mononuclear cells was inversely correlated with systemic antibody titers to CHSP60 type 1. CONCLUSION: Systemic and mucosal IFN-gamma responses are correlated, with preferential systemic targeting of CD4+ T cells. Furthermore, CHSP60 type 1 response is largely CD4+ T cell mediated and follows discrete T helper 1 and T helper 2 pathways.


Subject(s)
Chaperonin 60/pharmacology , Chlamydia trachomatis/immunology , Interferon-gamma/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/microbiology , Adolescent , Adult , Chaperonin 60/classification , Chaperonin 60/immunology , Chlamydia Infections/epidemiology , Chlamydia Infections/immunology , Cohort Studies , Endometrium/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interferon-gamma/genetics , Leukocytes, Mononuclear/immunology , Sex Work , Young Adult
19.
Eur J Immunol ; 38(12): 3549-60, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19016530

ABSTRACT

Majority of HIV-2-infected individuals meet the criteria of long-term non-progressors. This has been linked to superior qualitative HIV-2-specific cellular immune responses that correlate with viral control. However, it is unknown whether this is due to frequent targeting of immunodominant Gag epitopes in HIV-2 than HIV-1 infection. We describe a comprehensive comparison of the magnitude, breadth and frequency of Gag responses and the degree of cross-recognition of frequently targeted, immunodominant Gag peptides in a cross-sectional study of asymptomatic HIV-1- and HIV-2-infected individuals. Fresh PBMC from 20 HIV-1- and 20 HIV-2-infected patients with similar CD4(+) T-cell counts (p=0.36) were stimulated with pools of HIV-1 and/or HIV-2 Gag peptides in an IFN-gamma ELISPOT assay. We found no difference in the cumulative magnitude of IFN-gamma responses (p=0.75) despite significantly lower plasma viral loads in HIV-2-infected people (p<0.0001). However, Gag211-290 was targeted with significantly higher magnitude in HIV-2-infected subjects (p=0.03) although this did not correlate with viral control. There was no difference in frequently targeted Gag peptides, the breadth, immunodominance or cross-recognition of Gag peptide pools between the two infections. This suggests that other factors may control viral replication in HIV-2 infection.


Subject(s)
Gene Products, gag/immunology , HIV Infections/epidemiology , HIV Infections/immunology , HIV-1/immunology , HIV-2/immunology , Interferon-gamma/immunology , Adult , Aged , Amino Acid Sequence , Cells, Cultured , Cohort Studies , Cross Reactions/immunology , Female , Gambia/epidemiology , Gene Products, gag/chemistry , HIV-1/chemistry , HIV-2/chemistry , Humans , Male , Middle Aged , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology
20.
J Immunol ; 179(1): 597-606, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579081

ABSTRACT

During chronic HIV-1 infection, continuing viral replication is associated with impaired proliferative capacity of virus-specific CD8+ T cells and with the expansion and persistence of oligoclonal T cell populations. TCR usage may significantly influence CD8+ T cell-mediated control of AIDS viruses; however, the potential to modulate the repertoire of functional virus-specific T cells by immunotherapy has not been explored. To investigate this, we analyzed the TCR Vbeta usage of CD8+ T cells populations which were expanded following vaccination with modified vaccinia virus Ankara expressing a HIV-1 gag/multiepitope immunogen (MVA.HIVA) in HIV-1-infected patients receiving highly active antiretroviral therapy. Vaccinations induced the re-expansion of HIV-1-specific CD8+ T cells and these showed broad TCR Vbeta usage which was maintained for at least 1 year in some individuals. By contrast, virus-specific CD8+ T cell populations in the same donors which failed to expand after vaccination and in unvaccinated controls were oligoclonal. Simultaneously, we observed that CD8+ T cells recognizing vaccine-derived HIV-1 epitopes displayed enhanced capacity to proliferate and to inhibit HIV-1 replication in vitro, following MVA.HIVA immunizations. Taken together, these data indicate that an attenuated viral-vectored vaccine can modulate adaptive CD8+ T cell responses to HIV-1 and improve their antiviral functional capacity. The potential therapeutic benefit of this vaccination approach warrants further investigation.


Subject(s)
AIDS Vaccines/immunology , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Epitopes, T-Lymphocyte/administration & dosage , HIV-1/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , CD8-Positive T-Lymphocytes/pathology , Chronic Disease , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, pol/genetics , Gene Products, pol/immunology , Genes, T-Cell Receptor beta , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/genetics , HIV-1/growth & development , Humans , Receptors, Antigen, T-Cell, alpha-beta/genetics , nef Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...