Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
J Hematol Oncol ; 15(1): 42, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35410313

ABSTRACT

Inevitable relapses remain as the major therapeutic challenge in patients with mantle cell lymphoma (MCL) despite FDA approval of multiple targeted therapies and immunotherapies. Fc gamma receptors (FcγRs) play important roles in regulating antibody-mediated immunity. FcγRIIB, the unique immune-checkpoint inhibitory member of the FcγR family, has been implicated in immune cell desensitization and tumor cell resistance to the anti-CD20 antibody rituximab and other antibody-mediated immunotherapies; however, little is known about its expression and its immune-modulatory function in patients with aggressive MCL, especially those with multi-resistance. In this study, we found that FcγRIIB was ubiquitously expressed in both MCL cell lines and primary patient samples. FcγRIIB expression is significantly higher in CAR T-relapsed patient samples (p < 0.0001) compared to ibrutinib/rituximab-naïve, sensitive or resistant samples. Rituximab-induced CD20 internalization in JeKo-1 cells was completely blocked by concurrent treatment with BI-1206, a recombinant human monoclonal antibody targeting FcγRIIB. Combinational therapies with rituximab-ibrutinib, rituximab-venetoclax and rituximab-CHOP also induced CD20 internalization which was again effectively blocked by BI-1206. BI-1206 significantly enhanced the in vivo anti-MCL efficacy of rituximab-ibrutinib (p = 0.05) and rituximab-venetoclax (p = 0.02), but not the rituximab-CHOP combination in JeKo-1 cell line-derived xenograft models. In patient-derived xenograft (PDX) models, BI-1206, as a single agent, showed high potency (p < 0.0001, compared to vehicle control) in one aggressive PDX model that is resistant to both ibrutinib and venetoclax but sensitive to the combination of rituximab and lenalidomide (the preclinical mimetic of R2 therapy). BI-1206 sensitized the efficacy of rituximab monotherapy in a PDX model with triple resistance to rituximab, ibrutinib and CAR T-therapies (p = 0.030). Moreover, BI-1206 significantly enhanced the efficacy of the rituximab-venetoclax combination (p < 0.05), which led to long-term tumor remission in 25% of mice. Altogether, these data support that targeting this new immune-checkpoint blockade enhances the therapeutic activity of rituximab-based regimens in aggressive MCL models with multi-resistance.


Subject(s)
Antineoplastic Agents , Lymphoma, Mantle-Cell , Receptors, Chimeric Antigen , Adult , Animals , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20 , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Lymphoma, Mantle-Cell/drug therapy , Mice , Neoplasm Recurrence, Local/drug therapy , Receptors, Chimeric Antigen/therapeutic use , Rituximab/pharmacology , Rituximab/therapeutic use
2.
J Infect Dis ; 219(11): 1749-1754, 2019 05 05.
Article in English | MEDLINE | ID: mdl-30715363

ABSTRACT

Disease progression of human immunodeficiency virus type 1 (HIV-1) is delayed by HIV type 2 (HIV-2) in individuals with dual HIV-1/HIV-2 infection. The protective mechanisms, however, are still to be revealed. In the current study we examined type-specific and cross-reactive antibody-dependent cellular cytotoxicity (ADCC) in HIV-1 and HIV-2 monoinfection or dual infection. Of note, intertype cross-reactive antibodies that mediated HIV-1 envelope glycoprotein (Env)-targeted ADCC were frequently identified in HIV-2-infected individuals. Furthermore, the magnitude of HIV-1 cross-reactive ADCC activity during HIV-2 infections depended on the HIV-1 Env origin and was associated with the duration of infection. These results suggest that preexisting antibodies against HIV-2, which mediate intertype ADCC, might contribute to control of HIV-1 during dual infection.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Cross Reactions/immunology , Glycoproteins/immunology , HIV Infections/immunology , HIV-1/immunology , HIV-2/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , HIV Antibodies/immunology , HIV Infections/virology , Humans
3.
Br J Community Nurs ; 24(2): 80-86, 2019 Feb 02.
Article in English | MEDLINE | ID: mdl-30698480

ABSTRACT

Older patients with multimorbidity and extensive healthcare needs are at risk of frequent readmission to hospital after discharge. With a Swedish report entitled 'Follow-up 48-72' as its basis, the present study aimed to describe nurses' experiences of follow-up visits to older patients with multimorbidity 48 to 72 hours after discharge from hospital. Semi-structured interviews were conducted with 10 nurses experienced with such home visits to older patients, and the material was analysed by qualitative content analysis. The results indicate that such visits by nurses can relieve patient anxiety, as patients are often unsure of the next steps, in terms of medication and care. According to the nurses, these visits created trust in the nurse-patient relationship and ensured patient safety. Follow-up visits soon after discharge from hospital should become a part of routine nursing, especially for older people with multimorbidity.


Subject(s)
Attitude of Health Personnel , Continuity of Patient Care , House Calls , Practice Patterns, Nurses' , Adult , Aged , Community Health Nursing , Female , Health Services for the Aged , Humans , Interviews as Topic , Male , Middle Aged , State Medicine , United Kingdom
4.
AIDS Res Hum Retroviruses ; 34(2): 206-217, 2018 02.
Article in English | MEDLINE | ID: mdl-28982260

ABSTRACT

The induction of both neutralizing antibodies and non-neutralizing antibodies with effector functions, for example, antibody-dependent cellular cytotoxicity (ADCC), is desired in the search for effective vaccines against HIV-1. In the pursuit of novel immunogens capable of inducing an efficient antibody response, rabbits were immunized with selected antigens using different prime-boost strategies. We immunized 35 different groups of rabbits with Env antigens from clinical HIV-1 subtypes A and B, including immunization with DNA alone, protein alone, and DNA prime with protein boost. The rabbit sera were screened for ADCC activity using a GranToxiLux-based assay with human peripheral blood mononuclear cells as effector cells and CEM.NKRCCR5 cells coated with HIV-1 envelope as target cells. The groups with the highest ADCC activity were further characterized for cross-reactivity between HIV-1 subtypes. The immunogen inducing the most potent and broadest ADCC response was a trimeric gp140. The ADCC activity was highest against the HIV-1 subtype corresponding to the immunogen. The ADCC activity did not necessarily reflect neutralizing activity in the pseudovirus-TZMbl assay, but there was an overall correlation between the two antiviral activities. We present a rabbit vaccination model and an assay suitable for screening HIV-1 vaccine candidates for the induction of ADCC-mediating antibodies in addition to neutralizing antibodies. The antigens and/or immunization strategies capable of inducing antibodies with ADCC activity did not necessarily induce neutralizing activity and vice versa. Nevertheless, we identified vaccine candidates that were able to concurrently induce both types of responses and that had ADCC activity that was cross-reactive between different subtypes. When searching for an effective vaccine candidate, it is important to evaluate the antibody response using a model and an assay measuring the desired function.


Subject(s)
AIDS Vaccines/immunology , Antibody-Dependent Cell Cytotoxicity , HIV Antibodies/blood , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/isolation & purification , Animals , Antibodies, Neutralizing/blood , Cross Reactions , Female , HIV Infections/virology , HIV-1/isolation & purification , Humans , Immunization Schedule , Leukocytes, Mononuclear/immunology , Male , Rabbits , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, DNA/isolation & purification , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , env Gene Products, Human Immunodeficiency Virus/isolation & purification
5.
Vet Immunol Immunopathol ; 195: 25-32, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29249314

ABSTRACT

BACKGROUND: Influenza A virus in swine herds represents a major problem for the swine industry and poses a constant threat for the emergence of novel pandemic viruses and the development of more effective influenza vaccines for pigs is desired. By optimizing the vector backbone and using a needle-free delivery method, we have recently demonstrated a polyvalent influenza DNA vaccine that induces a broad immune response, including both humoral and cellular immunity. OBJECTIVES: To investigate the protection of our polyvalent influenza DNA vaccine approach in a pig challenge study. METHODS: By intradermal needle-free delivery to the skin, we immunized pigs with two different doses (500µg and 800µg) of an influenza DNA vaccine based on six genes of pandemic origin, including internally expressed matrix and nucleoprotein and externally expressed hemagglutinin and neuraminidase as previously demonstrated. Two weeks following immunization, the pigs were challenged with the 2009 pandemic H1N1 virus. RESULTS: When challenged with 2009 pandemic H1N1, 0/5 vaccinated pigs (800µg DNA) became infected whereas 5/5 unvaccinated control pigs were infected. The pigs vaccinated with the low dose (500µg DNA) were only partially protected. The DNA vaccine elicited binding-, hemagglutination inhibitory (HI) - as well as cross-reactive neutralizing antibody activity and neuraminidase inhibiting antibodies in the immunized pigs, in a dose-dependent manner. CONCLUSION: The present data, together with the previously demonstrated immunogenicity of our influenza DNA vaccine, indicate that naked DNA vaccine technology provides a strong approach for the development of improved pig vaccines, applying realistic low doses of DNA and a convenient delivery method for mass vaccination.


Subject(s)
Influenza Vaccines/therapeutic use , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , Vaccines, DNA/therapeutic use , Animals , Influenza A Virus, H1N1 Subtype/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Swine , Swine Diseases/immunology , Vaccines, DNA/immunology
6.
Hum Vaccin Immunother ; 13(8): 1823-1830, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28613978

ABSTRACT

DNA vaccines induce broad immunity, which involves both humoral and strong cellular immunity, and can be rapidly designed for novel or evolving pathogens such as influenza. However, the humoral immunogenicity in humans and higher animals has been suboptimal compared with that of traditional vaccine approaches. We tested whether the emulsion-based and α-tocopherol containing adjuvant Diluvac Forte® has the ability to enhance the immunogenicity of a naked DNA vaccine (i.e., plasmid DNA). As a model vaccine, we used plasmids encoding both a surface-exposed viral glycoprotein (hemagglutinin) and an internal non-glycosylated nucleoprotein in the Th1/Th2 balanced CB6F1 mouse model. The naked DNA (50 µg) was premixed at a 1:1 volume/volume ratio with Diluvac Forte®, an emulsion containing different concentrations of α-tocopherol, the emulsion alone or endotoxin-free phosphate-buffered saline (PBS). The animals received 2 intracutaneous immunizations spaced 3 weeks apart. When combined with Diluvac Forte® or the emulsion containing α-tocopherol, the DNA vaccine induced a more potent and balanced immunoglobulin G (IgG)1 and IgG2c response, and both IgG subclass responses were significantly enhanced by the adjuvant. The DNA vaccine also induced CD4+ and CD8+ vaccine-specific T cells; however, the adjuvant did not exert a significant impact. We concluded that the emulsion-based adjuvant Diluvac Forte® enhanced the immunogenicity of a naked DNA vaccine encoding influenza proteins and that the adjuvant constituent α-tocopherol plays an important role in this immunogenicity. This induction of a potent and balanced humoral response without impairment of cellular immunity constitutes an important advancement toward effective DNA vaccines.


Subject(s)
Adjuvants, Immunologic , Immunity, Humoral , Immunogenicity, Vaccine , Influenza Vaccines/immunology , Vaccines, DNA/immunology , alpha-Tocopherol/immunology , Animals , Antibodies, Viral , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immunity, Cellular , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Vaccination
7.
Vaccine ; 34(32): 3634-40, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27211039

ABSTRACT

BACKGROUND: Pigs are natural hosts for influenza A viruses, and the infection is widely prevalent in swine herds throughout the world. Current commercial influenza vaccines for pigs induce a narrow immune response and are not very effective against antigenically diverse viruses. To control influenza in pigs, the development of more effective swine influenza vaccines inducing broader cross-protective immune responses is needed. Previously, we have shown that a polyvalent influenza DNA vaccine using vectors containing antibiotic resistance genes induced a broadly protective immune response in pigs and ferrets using intradermal injection followed by electroporation. However, this vaccination approach is not practical in large swine herds, and DNA vaccine vectors containing antibiotic resistance genes are undesirable. OBJECTIVES: To investigate the immunogenicity of an optimized version of our preceding polyvalent DNA vaccine, characterized by a next-generation expression vector without antibiotic resistance markers and delivered by a convenient needle-free intradermal application approach. METHODS: The humoral and cellular immune responses induced by three different doses of the optimized DNA vaccine were evaluated in groups of five to six pigs. The DNA vaccine consisted of six selected influenza genes of pandemic origin, including internally expressed matrix and nucleoprotein and externally expressed hemagglutinin and neuraminidase. RESULTS: Needle-free vaccination of growing pigs with the optimized DNA vaccine resulted in specific, dose-dependent immunity down to the lowest dose (200µg DNA/vaccination). Both the antibody-mediated and the recall lymphocyte immune responses demonstrated high reactivity against vaccine-specific strains and cross-reactivity to vaccine-heterologous strains. CONCLUSION: The results suggest that polyvalent DNA influenza vaccination may provide a strong tool for broad protection against swine influenza strains threatening animal as well as public health. In addition, the needle-free administration technique used for this DNA vaccine will provide an easy and practical approach for the large-scale vaccination of swine.


Subject(s)
Cross Reactions , Immunity, Cellular , Immunity, Humoral , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Hemagglutination Inhibition Tests , Immunogenicity, Vaccine , Needles , Neutralization Tests , Swine , T-Lymphocytes/immunology , Vaccination/methods
8.
J Acquir Immune Defic Syndr ; 71(1): 8-16, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26761268

ABSTRACT

BACKGROUND: Virus-specific CD8(+) T-cell responses are believed to play an important role in the control of HIV-1 infection; however, what constitutes an effective HIV-1 CD8(+) T-cell response remains a topic of debate. The ex vivo viral suppressive capacity was measured of CD8(+) T cells from 44 HIV-1-positive individuals. The phenotypic and cytokine profiles, and also the specificity of the CD8(+) T cells, were correlated with the suppression of HIV-1 replication. We also aimed to determine whether antiretroviral therapy (ART) had any positive effect on the HIV-1 suppressive CD8(+) T cells. METHOD: Ex vivo suppression assay was used to evaluate the ability of CD8(+) T cells to suppress HIV-1 replication in autologous CD4(+) T cells. The CD107a, interferon-γ, interleukin-2, tumor necrosis factor-α (TNF-α), and macrophage inflammatory protein-1ß (MIP-1ß) responses to HIV-1 were evaluated by intracellular staining. The phenotypic profile of CD8(+) T cells was determined by whole blood staining. RESULTS: The expression of CD57 on effector CD8(+) T cells correlated with the suppression of HIV-1 replication and to the duration of ART. CD107a and tumor necrosis factor-α expression levels were significantly higher in individuals with ex vivo suppressive activity compared with individuals without suppressive activity. CONCLUSIONS: Standard in vitro assays measuring one or several cytokines do not correlate with the functional viral suppressive capacity of CD8(+) T cells from HIV-1-positive individuals. The best correlation of viral suppression was found to be CD57 expression. CD57 expression correlated with the duration of ART, suggesting that ART restores the cytotoxic capacity of CD8(+) T lymphocytes.


Subject(s)
CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Virus Replication , Anti-Retroviral Agents/pharmacology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemokine CCL4/metabolism , HIV-1/immunology , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Viral Load/immunology , Virus Replication/immunology
9.
AIDS Res Hum Retroviruses ; 32(5): 434-42, 2016 May.
Article in English | MEDLINE | ID: mdl-26621287

ABSTRACT

The development of therapeutic and prophylactic HIV vaccines for African countries is urgently needed, but the question of what immunogens to use needs to be answered. One approach is to include HIV envelope immunogens derived from HIV-positive individuals from a geographically concentrated epidemic with more limited viral genetic diversity for a region-based vaccine. To address if there is a basis for a regional selected antibody vaccine, we have screened two regionally separate cohorts from Guinea-Bissau and Denmark for neutralizing antibody activity and antibody-dependent cellular cytotoxicity (ADCC) against local and nonlocal circulating HIV-1 strains. The neutralizing activity did not demonstrate higher potential against local circulating strains according to geography and subtype determination, but the plasma from Danish individuals demonstrated significantly higher inhibitory activity than that from Guinea-Bissau individuals against both local and nonlocal virus strains. Interestingly, an opposite pattern was observed with ADCC activity, where Guinea-Bissau individual plasma demonstrated higher activity than Danish plasma and was specifically against the local circulating subtype. Thus, on basis of samples from these two cohorts, no local-specific neutralizing activity was detected, but a local ADCC response was identified in the Guinea-Bissau samples, suggesting potential use of regional immunogens for an ADCC-inducing vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , HIV Antibodies/immunology , HIV Antigens/immunology , HIV-1/immunology , AIDS Vaccines/immunology , Antibodies, Neutralizing/blood , Base Sequence/genetics , Denmark , Female , Guinea-Bissau , HIV Antibodies/blood , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV Infections/virology , Humans , Male , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
10.
PLoS One ; 10(12): e0145249, 2015.
Article in English | MEDLINE | ID: mdl-26696395

ABSTRACT

Understanding alterations in HIV-specific immune responses during antiretroviral therapy (ART), such as antibody-dependent cellular cytotoxicity (ADCC), is important in the development of novel strategies to control HIV-1 infection. This study included 53 HIV-1 positive individuals. We evaluated the ability of effector cells and antibodies to mediate ADCC separately and in combination using the ADCC-PanToxiLux assay. The ability of the peripheral blood mononuclear cells (PBMCs) to mediate ADCC was significantly higher in individuals who had been treated with ART before seroconversion, compared to the individuals initiating ART at a low CD4+ T cell count (<350 cells/µl blood) and the ART-naïve individuals. The frequency of CD16 expressing natural killer (NK) cells correlated with both the duration of ART and Granzyme B (GzB) activity. In contrast, the plasma titer of antibodies mediating ADCC declined during ART. These findings suggest improved cytotoxic function of the NK cells if initiating ART early during infection, while the levels of ADCC mediating antibodies declined during ART.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , HIV Antibodies/immunology , HIV Infections , HIV-1/immunology , Killer Cells, Natural/immunology , Adult , Aged , CD4 Lymphocyte Count , Female , GPI-Linked Proteins/immunology , Granzymes/immunology , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Killer Cells, Natural/pathology , Male , Middle Aged , Receptors, IgG/immunology
11.
PLoS One ; 10(10): e0139573, 2015.
Article in English | MEDLINE | ID: mdl-26426913

ABSTRACT

CD8+ T cell-restricted immunity is important in the control of HIV-1 infection, but continued immune activation results in CD8+ T cell dysfunction. Early initiation of antiretroviral treatment (ART) and the duration of ART have been associated with immune reconstitution. Here, we evaluated whether restoration of CD8+ T cell function in HIV-1-infected individuals was dependent on early initiation of ART. HIV-specific CD107a, IFNγ, IL-2, TNFα and MIP-1ß expression by CD8+ T cells and the frequency of CD8+ T cells expressing PD-1, 2B4 and CD160 were measured by flow cytometry. The frequency of CD8+ T cells expressing the inhibitory markers PD-1, 2B4 and CD160 was lower in ART-treated individuals compared with ART-naïve individuals and similar to the frequency in HIV-uninfected controls. The expression of the three markers was similarly independent of when therapy was initiated. Individuals treated before seroconversion displayed an HIV-specific CD8+ T cell response that included all five functional markers; this was not observed in individuals treated after seroconversion or in ART-naïve individuals. In summary, ART appears to restore the total CD8+ T cell population to a less exhausted phenotype, independent of the time point of initiation. However, to preserve multifunctional, HIV-1-specific CD8+ T cells, ART might have to be initiated before seroconversion.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Viral Load/immunology , Adult , Aged , Case-Control Studies , Female , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged
12.
Hum Vaccin Immunother ; 11(8): 1983-90, 2015.
Article in English | MEDLINE | ID: mdl-25746201

ABSTRACT

The threat posed by the 2009 pandemic H1N1 virus emphasized the need for new influenza A virus vaccines inducing a broad cross-protective immune response for use in both humans and pigs. An effective and broad influenza vaccine for pigs would greatly benefit the pork industry and contribute to public health by diminishing the risk of emerging highly pathogenic reassortants. Current inactivated protein vaccines against swine influenza produce only short-lived immunity and have no efficacy against heterologous strains. DNA vaccines are a potential alternative with advantages such as the induction of cellular and humoral immunity, inherent safety and rapid production time. We have previously developed a DNA vaccine encoding selected influenza proteins of pandemic origin and demonstrated broad protective immune responses in ferrets and pigs. In this study, we evaluated our DNA vaccine expressed by next-generation vectors. These new vectors can improve gene expression, but they are also efficiently produced on large scales and comply with regulatory guidelines by avoiding antibiotic resistance genes. In addition, a new needle-free delivery of the vaccine, convenient for mass vaccinations, was compared with intradermal needle injection followed by electroporation. We report that when our DNA vaccine is expressed by the new vectors and delivered to the skin with the needle-free device in the rabbit model, it can elicit an antibody response with the same titers as a conventional vector with intradermal electroporation. The needle-free delivery is already in use for traditional protein vaccines in pigs but should be considered as a practical alternative for the mass administration of broadly protective influenza DNA vaccines.


Subject(s)
Influenza A virus/immunology , Influenza, Human/prevention & control , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , Vaccines, DNA/immunology , Administration, Cutaneous , Animals , Antibodies, Viral/blood , Humans , Influenza A virus/genetics , Orthomyxoviridae Infections/prevention & control , Rabbits , Swine , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
13.
J Virol ; 89(7): 3542-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25589645

ABSTRACT

UNLABELLED: The spontaneous control of human and simian immunodeficiency viruses (HIV/SIV) is typically associated with specific major histocompatibility complex (MHC) class I alleles and efficient CD8(+) T-cell responses, but many controllers maintain viral control despite a nonprotective MHC background and weak CD8(+) T-cell responses. Therefore, the contribution of this response to maintaining long-term viral control remains unclear. To address this question, we transiently depleted CD8(+) T cells from five SIV-infected cynomolgus macaques with long-term viral control and weak CD8(+) T-cell responses. Among them, only one carried the protective MHC allele H6. After depletion, four of five controllers experienced a transient rebound of viremia. The return to undetectable viremia was accompanied by only modest expansion of SIV-specific CD8(+) T cells that lacked efficient SIV suppression capacity ex vivo. In contrast, the depletion was associated with homeostatic activation/expansion of CD4(+) T cells that correlated with viral rebound. In one macaque, viremia remained undetectable despite efficient CD8(+) cell depletion and inducible SIV replication from its CD4(+) T cells in vitro. Altogether, our results suggest that CD8(+) T cells are not unique contributors to the long-term maintenance of low viremia in this SIV controller model and that other mechanisms, such as weak viral reservoirs or control of activation, may be important players in control. IMPORTANCE: Spontaneous control of HIV-1 to undetectable levels is associated with efficient anti-HIV CD8(+) T-cell responses. However, in some cases, this response fades over time, although viral control is maintained, and many HIV controllers (weak responders) have very low frequencies of HIV-specific CD8(+) T cells. In these cases, the importance of CD8 T cells in the maintenance of HIV-1 control is questionable. We developed a nonhuman primate model of durable SIV control with an immune profile resembling that of weak responders. Transient depletion of CD8(+) cells induced a rise in the viral load. However, viremia was correlated with CD4(+) T-cell activation subsequent to CD8(+) cell depletion. Regain of viral control to predepletion levels was not associated with restoration of the anti-SIV capacities of CD8(+) T cells. Our results suggest that CD8(+) T cells may not be involved in maintenance of viral control in weak responders and highlight the fact that additional mechanisms should not be underestimated.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Animals , Leukocyte Reduction Procedures , Macaca fascicularis , Survivors , Viral Load
14.
J Acquir Immune Defic Syndr ; 68(2): 103-11, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25394194

ABSTRACT

BACKGROUND: Natural killer (NK) cell phenotype and function have recently gained much attention as playing crucial roles in antibody-dependent cellular cytotoxicity (ADCC). We investigated NK cell function, as measured by ADCC, in HIV-1-positive individuals before and 6 months after highly active antiretroviral therapy (HAART) initiation. METHOD: The ability of antibodies and NK cells to mediate ADCC was investigated separately and in combination in an autologous model. The NK cell subset distribution and NK cell phenotype (ie, expression of maturation and activation markers within NK cell subsets) were analyzed. RESULTS: The ability of NK cells to mediate ADCC was significantly increased after only 6 months of HAART and was not explained by a normalization of NK cell subsets (CD56 CD16 and CD56 CD16 NK cells) but rather by normalization in the frequency of NK cells expressing CCR7 and CD27. For individuals with no increase in ADCC after 6 months of HAART, the frequency of NK cells expressing NKp46 was downregulated. The ability of antibodies to mediate ADCC alone and in combination in an autologous model was not improved. CONCLUSIONS: HAART improves the ability of NK cells to mediate ADCC after 6 months. This improvement does not correlate with general immune restoration, as measured by CD4 T-cell counts, but rather to a decrease in the frequency of NK cells expressing CCR7 and CD27.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Antibody-Dependent Cell Cytotoxicity , HIV Antibodies/immunology , HIV Infections/drug therapy , HIV Infections/immunology , Killer Cells, Natural/immunology , Adult , Aged , Female , Humans , Immunophenotyping , Killer Cells, Natural/chemistry , Longitudinal Studies , Male , Middle Aged , Receptors, CCR7/analysis , Tumor Necrosis Factor Receptor Superfamily, Member 7/analysis , Young Adult
16.
AIDS Res Hum Retroviruses ; 29(11): 1504-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23634822

ABSTRACT

We have designed a therapeutic HIV-1 vaccine concept based on peptides together with the adjuvant CAF01. Peptides represented 15 HLA-supertype-restricted subdominant and conserved CD8 T cell epitopes and three CD4 T-helper cell epitopes. In this phase I clinical trial, safety and immunogenicity were assessed in untreated HIV-1-infected individuals in Guinea-Bissau, West Africa. Twenty-three HIV-1-infected individuals were randomized to receive placebo (n=5) or vaccine (n=18). Safety was appraised by clinical follow-up combined with monitoring of biochemistry, hematology, CD4 T cell counts, and HIV-1 viral loads. T cell immunogenicity was monitored longitudinally by interferon (IFN)-γ ELISpot. New vaccine-specific T cell responses were induced in 6/14 vaccinees for whom ELISpot data were valid. CD4 T cell counts and viral loads were stable. The study shows that therapeutic immunization is feasible and safe in Guinea-Bissau and that it is possible to redirect T cell immunity with CAF01-adjuvanted HIV-1 peptide vaccine during untreated HIV-1 infection in some patients. However, relatively few preexisting and vaccine-induced HIV-1 T cell responses to CD8 T cell epitopes were detected against HIV-1 using IFN-γ ELISpot in this chronically infected African population.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/therapeutic use , Antigens, Viral/therapeutic use , HIV Infections/therapy , HIV-1/immunology , Immunotherapy/adverse effects , Immunotherapy/methods , AIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Antigens, Viral/immunology , CD4 Lymphocyte Count , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/therapeutic use , Female , Guinea-Bissau , Humans , Interferon-gamma/metabolism , Liposomes/administration & dosage , Male , Middle Aged , Phosphoproteins/immunology , Viral Load , Young Adult
17.
PLoS One ; 8(5): e63575, 2013.
Article in English | MEDLINE | ID: mdl-23691069

ABSTRACT

Induction of broad T-cell immune responses is regarded as critical for vaccines against the human immunodeficiency virus type 1 (HIV-1) which exhibit high diversity and, therefore, focus has been on inducing cytotoxic CD8 T-cell responses against the more conserved parts of the virus, such as the Gag protein. Herein, we have used the p24 protein which contains a range of conserved T-cell epitopes. We demonstrate that a vaccine of HIV-1 subtype B consensus group-specific antigen (Gag) p24 protein with the CD8-inducing liposomal cationic adjuvant formulation (CAF) 05, induces both CD4 and CD8 T-cell responses in CB6F1 mice. The adjuvanted vaccine also induced functional antigen-specific cytotoxicity in vivo. Furthermore, we found that when fragmenting the Gag p24 protein into overlapping Gag p24 peptides, a broader T-cell epitope specificity was induced in the humanized human leukocyte antigen (HLA)-A2/DR-transgenic mouse model. Thus, combining overlapping Gag p24 peptides with CAF05 appears to be a promising and simple strategy for inducing broader T-cell responses to multiple conserved epitopes which will be relevant for both prophylactic and therapeutic HIV-1 vaccines.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Core Protein p24/immunology , HIV-1/immunology , Immunity, Cellular/immunology , Amino Acid Sequence , Analysis of Variance , Animals , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/immunology , HIV Core Protein p24/genetics , HLA-A2 Antigen/genetics , Mice , Mice, Transgenic , Molecular Sequence Data
18.
J Acquir Immune Defic Syndr ; 63(4): 418-27, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23507659

ABSTRACT

BACKGROUND: Still no effective HIV-1 prophylactic or therapeutic vaccines are available. However, as the proportion of HIV-1-infected individuals on antiretroviral treatment is increasing, knowledge about the residual immune response is important for the possible development of an HIV-1 vaccine. METHODS: In this study, the magnitude, breadth, and quality of the HIV-1-specific T-cell response in HIV-1-infected viremic individuals (n = 19) and individuals on highly active antiretroviral treatment (HAART) (n = 14) using multicolor flow cytometry were determined. RESULTS: We found that magnitude and breadth of the CD8 T-cell response were significantly higher in viremic individuals than individuals on HAART (P < 0.0001 and P < 0.0001, respectively) and that the functionality of the overall HIV-1-specific response was significantly different in individuals on HAART and viremic individuals (P = 0.0020). In individuals on HAART, the remaining responses were primarily detected upon stimulation with overlapping peptides from Gag p24, integrase, and Nef. The Gag p24 response was more polyfunctional than corresponding responses observed in viremic individuals. CONCLUSIONS: Identification of highly immunogenic regions also recognized by individuals on HAART may be important for HIV-1 vaccine development. Irrespective of HLA haplotype, specific regions within the HIV-1 genome that is targeted more frequently in individuals on HAART have been identified. However, further studies are required to establish if these particular regions could be interesting for a future vaccine that might limit the time and opportunity for escape mutations.


Subject(s)
Antiviral Agents/pharmacology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/immunology , Viral Load , Antiretroviral Therapy, Highly Active , Antiviral Agents/therapeutic use , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/metabolism , Female , HIV Core Protein p24/immunology , HIV Infections/genetics , HIV Integrase/immunology , HLA Antigens/genetics , Humans , Male , RNA, Viral/blood , nef Gene Products, Human Immunodeficiency Virus/immunology
19.
Clin Immunol ; 146(2): 120-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23314272

ABSTRACT

We investigated the potential of inducing additional T-cell immunity during chronic HIV-1 infection directed to subdominant HIV-1 epitopes from common HLA-supertypes. Ten treatment-naïve HIV-1-infected individuals were immunized with peptides in the adjuvant CAF01. One individual received placebo. T-cell immunogenicity was examined longitudinally by a flow cytometry (CD107a, IFNγ, TNFα, IL-2 and/or MIP1ß expression) as well as IFNγ ELISPOT. Safety was evaluated by clinical follow up combined with monitoring of biochemistry, hematology, CD4 T-cell counts and viral load. New CD4 and CD8 T-cell responses specific for one or more vaccine epitopes were induced in 10/10 vaccinees. The responses were dominated by CD107a and MIP1ß expression. There were no significant changes in HIV-1 viral load or CD4 T-cell counts. Our study demonstrates that the peptide/CAF01 vaccine is safe and that it is possible to generate new HIV-1 T-cell responses to defined epitopes in treatment-naïve HIV-1-infected individuals.


Subject(s)
Adjuvants, Immunologic/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/therapeutic use , HIV Infections/immunology , HIV Infections/therapy , Immunodominant Epitopes/therapeutic use , Peptides/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , HIV Infections/prevention & control , HLA-A Antigens/administration & dosage , HLA-A Antigens/genetics , HLA-B Antigens/administration & dosage , HLA-B Antigens/genetics , HLA-C Antigens/administration & dosage , HLA-C Antigens/genetics , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Male , Middle Aged , Peptides/administration & dosage , Peptides/immunology , Single-Blind Method , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...