Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
World J Surg Oncol ; 18(1): 150, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32620130

ABSTRACT

BACKGROUND: Currently, nivolumab and ipilimumab are the most widely used immune checkpoint inhibitors. We performed a meta-analysis to evaluate the efficacy and treatment-related adverse events (TRAEs) of nivolumab plus ipilimumab therapy in cancer treatment. METHODS: We examined data from PubMed, Web of Science, EBSCO, and Cochrane Library. Eleven articles fulfilled our criteria, which we divided into 3 groups: nivolumab plus ipilimumab versus nivolumab (the dose used for monotherapy is 3 mg/kg), nivolumab plus ipilimumab versus ipilimumab (the dose used for monotherapy is 3 mg/kg), and nivolumab 1 mg/kg plus ipilimumab 3 mg/kg (N1I3) versus nivolumab 3 mg/kg plus ipilimumab 1 mg/kg (N3I1). We measured the complete response (CR), partial response (PR), objective response rate (ORR), and TRAEs in any grade and grade 3 or higher. RESULTS: The overall effect estimate favored the combined immunotherapy group in terms of the ORR (RR: 1.40, p < 0.001) and PR (RR: 1.50, p < 0.001) than nivolumab alone. Compared with ipilimumab alone, the combined immunotherapy group had better CR (RR: 4.89, p < 0.001), PR (RR: 2.75, p < 0.001), and ORR (RR: 3.31, p < 0.001). Finally, N1I3 showed better PR (RR: 1.35, p = 0.006) and ORR (RR: 1.21, p = 0.03) than N3I1. The incidence of any TRAEs was similar between both groups (RR: 1.05, p = 0.06). However, the incidence of serious adverse events (grade 3 or higher) was lower in group N3I1 than group N1I3 (RR: 1.51, p < 0.001). CONCLUSION: This meta-analysis showed that the curative effect of nivolumab plus ipilimumab was better than that of nivolumab or ipilimumab monotherapy. In the combined immunotherapy group, N1I3 was more effective than N3I1. Although the side effects were slightly increased in N1I3 group, overall safety was acceptable.


Subject(s)
Immune Checkpoint Inhibitors , Nivolumab , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Humans , Ipilimumab , Prognosis
2.
Cancer Res ; 75(6): 996-1008, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25623234

ABSTRACT

The mobilization of bone marrow-derived cells (BMDC) to distant tissues before the arrival of disseminated tumor cells has been shown preclinically to facilitate metastasis through the establishment of metastatic niches. Primary tumor hypoxia has been demonstrated to play a pivotal role in the production of chemokines and cytokines responsible for the mobilization of these BMDCs, especially in breast cancer. Carbonic anhydrase IX (CAIX, CA9) expression is highly upregulated in hypoxic breast cancer cells through the action of hypoxia-inducible factor-1 (HIF1). Preclinical evidence has demonstrated that CAIX is required for breast tumor growth and metastasis; however, the mechanism by which CAIX exerts its prometastatic function is not well understood. Here, we show that CAIX is indispensable for the production of granulocyte colony-stimulating factor (G-CSF) by hypoxic breast cancer cells and tumors in an orthotopic model. Furthermore, we demonstrate that tumor-expressed CAIX is required for the G-CSF-driven mobilization of granulocytic myeloid-derived suppressor cells (MDSC) to the breast cancer lung metastatic niche. We also determined that CAIX expression is required for the activation of NF-κB in hypoxic breast cancer cells and constitutive activation of the NF-κB pathway in CAIX-depleted cells restored G-CSF secretion. Together, these findings identify a novel hypoxia-induced CAIX-NF-κB-G-CSF cellular signaling axis culminating in the mobilization of granulocytic MDSCs to the breast cancer lung metastatic niche.


Subject(s)
Carbonic Anhydrases/physiology , Cell Movement , Granulocyte Colony-Stimulating Factor/biosynthesis , Myeloid Cells/physiology , Neoplasms, Experimental/pathology , Animals , CD11b Antigen/analysis , Carbonic Anhydrase IX , Cell Hypoxia , Chemokine CXCL10/physiology , Female , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Neoplasm Metastasis
3.
J Med Chem ; 54(24): 8271-7, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22077347

ABSTRACT

A series of 7-substituted coumarins incorporating various glycosyl moieties were synthesized and investigated for the inhibition of the zinc enzyme carbonic anhydrase (CA, EC 4.2.1.1). These coumarins were very weak or ineffective as inhibitors of the housekeeping, off target isoforms CA I and II, but some of them inhibited tumor-associated CA IX and XII in the low nanomolar range. They also significantly inhibited the growth of primary tumors by the highly aggressive 4T1 syngeneic mouse mammary tumor cells at 30 mg/kg, constituting interesting candidates for the development of conceptually novel anticancer drugs. Because CA IX is overexpressed in hypoxic tumors and exhibits very limited expression in normal tissues, such compounds may be useful for treating cancers not responsive to classic chemo- and radiotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/metabolism , Coumarins/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbonic Anhydrase IX , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Cell Hypoxia , Coumarins/chemistry , Coumarins/pharmacology , Female , Humans , Mice , Mice, Inbred BALB C , Structure-Activity Relationship , Xenograft Model Antitumor Assays
4.
Cancer Res ; 71(9): 3364-76, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21415165

ABSTRACT

Carbonic anhydrase IX (CAIX) is a hypoxia and HIF-1-inducible protein that regulates intra- and extracellular pH under hypoxic conditions and promotes tumor cell survival and invasion in hypoxic microenvironments. Interrogation of 3,630 human breast cancers provided definitive evidence of CAIX as an independent poor prognostic biomarker for distant metastases and survival. shRNA-mediated depletion of CAIX expression in 4T1 mouse metastatic breast cancer cells capable of inducing CAIX in hypoxia resulted in regression of orthotopic mammary tumors and inhibition of spontaneous lung metastasis formation. Stable depletion of CAIX in MDA-MB-231 human breast cancer xenografts also resulted in attenuation of primary tumor growth. CAIX depletion in the 4T1 cells led to caspase-independent cell death and reversal of extracellular acidosis under hypoxic conditions in vitro. Treatment of mice harboring CAIX-positive 4T1 mammary tumors with novel CAIX-specific small molecule inhibitors that mimicked the effects of CAIX depletion in vitro resulted in significant inhibition of tumor growth and metastasis formation in both spontaneous and experimental models of metastasis, without inhibitory effects on CAIX-negative tumors. Similar inhibitory effects on primary tumor growth were observed in mice harboring orthotopic tumors comprised of lung metatstatic MDA-MB-231 LM2-4(Luc+) cells. Our findings show that CAIX is vital for growth and metastasis of hypoxic breast tumors and is a specific, targetable biomarker for breast cancer metastasis.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carbonic Anhydrase Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbonic Anhydrase IX , Carbonic Anhydrases/biosynthesis , Carbonic Anhydrases/deficiency , Carbonic Anhydrases/metabolism , Cell Growth Processes/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Xenograft Model Antitumor Assays
5.
J Med Chem ; 54(6): 1896-902, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21361354

ABSTRACT

A series of ureido-substituted benzenesulfonamides was prepared that showed a very interesting profile for the inhibition of several human carbonic anhydrases (hCAs, EC 4.2.1.1), such as hCAs I and II (cytosolic isoforms) and hCAs IX and XII (transmembrane, tumor-associated enzymes). Excellent inhibition of all these isoforms has been observed with various members of the series, depending on the substitution pattern of the urea moiety. Several low nanomolar CA IX/XII inhibitors also showing good selectivity for the transmembrane over the cytosolic isoforms have been discovered. One of them, 4-{[(3'-nitrophenyl)carbamoyl]amino}benzenesulfonamide, significantly inhibited the formation of metastases by the highly aggressive 4T1 mammary tumor cells at pharmacologic concentrations of 45 mg/kg, constituting an interesting candidate for the development of conceptually novel antimetastatic drugs.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Benzene Derivatives/chemical synthesis , Carbonic Anhydrases/metabolism , Mammary Neoplasms, Experimental/pathology , Sulfonamides/chemical synthesis , Urea/analogs & derivatives , Urea/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Benzene Derivatives/pharmacology , Carbonic Anhydrase IX , Catalytic Domain , Female , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Models, Molecular , Neoplasm Metastasis , Nitrobenzenes/chemical synthesis , Nitrobenzenes/pharmacology , Protein Binding , Structure-Activity Relationship , Sulfonamides/pharmacology , Urea/pharmacology
6.
Cancer Res ; 70(19): 7562-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20729277

ABSTRACT

Matrix metalloproteinases (MMP), strongly associated pathogenic markers of cancer, have undergone extensive drug development programs. Marimastat, a noncovalent MMP inhibitor, was conjugated with FITC to label cellular metalloproteinase cancer targets in MDA-MB-231 cells in vitro. Punctate localization of active transmembrane MMP14 was observed. For molecular-targeted positron emission tomography imaging of syngeneic 67NR murine mammary carcinoma in vivo, marimastat was (18)F-labeled using a shelf-stable arylboronic ester conjugate as a captor for aqueous [(18)F]fluoride in a novel, rapid one-step reaction at ambient temperature. [(18)F]Marimastat-aryltrifluoroborate localized to the tumors, with labeling being blocked in control animals first loaded with >10-fold excess unlabeled marimastat. The labeled drug cleared primarily via the hepatobiliary and gastrointestinal tract, with multiple animals imaged in independent experiments, confirming the ease of this new labeling strategy.


Subject(s)
Breast Neoplasms/diagnostic imaging , Enzyme Inhibitors , Fluorine Radioisotopes , Hydroxamic Acids , Mammary Neoplasms, Experimental/diagnostic imaging , Matrix Metalloproteinase Inhibitors , Radiopharmaceuticals , Animals , Borates/chemistry , Breast Neoplasms/enzymology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Female , Fluorine Radioisotopes/chemistry , Humans , Hydroxamic Acids/chemistry , Isoenzymes , Isotope Labeling/methods , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Inbred BALB C , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis
7.
Dev Dyn ; 237(10): 2755-68, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18773493

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been linked to metastatic propensity. The 4T1 tumor is a clinically relevant model of spontaneous breast cancer metastasis. Here we characterize 4T1-derived cell lines for EMT, in vitro invasiveness and in vivo metastatic ability. Contrary to expectations, 67NR cells, which form primary tumors but fail to metastasize, express vimentin and N-cadherin, but not E-cadherin. 4T1 cells express E-cadherin and ZO-1, but are migratory, invasive, and metastasize to multiple sites. 66cl4 cells form lung metastases and display a mixed phenotype, but are not as migratory or invasive as 67NR cells. These findings demonstrate that the metastatic ability of breast cancer cells does not strictly correlate with genotypic and phenotypic properties of EMT per se, and suggest that other processes may govern metastatic capability. Gene expression analysis of primary tumors did not identify differences in EMT markers, but did reveal candidate genes that may influence metastatic ability.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation , Mesoderm/cytology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Cell Transplantation , Chemotaxis , Disease Models, Animal , Female , Gene Expression Profiling , Mesoderm/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/pathology
8.
Clin Cancer Res ; 14(5): 1494-501, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316574

ABSTRACT

PURPOSE: Tpn is a member of the MHC class I loading complex and functions to bridge the TAP peptide transporter to MHC class I molecules. Metastatic human carcinomas often express low levels of the antigen-processing components Tapasin and TAP and display few functional surface MHC class I molecules. As a result, carcinomas are unrecognizable by effector CTLs. The aim of this study is to examine if Tapasin (Tpn) plays a critical role in the escape of tumors from immunologic recognition. EXPERIMENTAL DESIGN: To test our hypothesis, a nonreplicating adenovirus vector encoding human Tpn (AdhTpn) was constructed to restore Tpn expression in vitro and in vivo in a murine lung carcinoma cell line (CMT.64) that is characterized by down-regulation of surface MHC class I due to deficiency in antigen-processing components. RESULTS: Ex vivo, Tpn expression increased surface MHC class I and restored susceptibility of tumor cells to antigen-specific CTL killing, and AdhTpn infection of dendritic cells also significantly increased cross-presentation and cross-priming. Furthermore, tumor-bearing animals inoculated with AdhTpn demonstrated a significant increase in CD8(+) and CD4(+) T cells and CD11c(+) dendritic cells infiltrating the tumors. Provocatively, whereas syngeneic mice bearing tumors that were inoculated with AdhTpn a significant reduction in tumor growth and increased survival compared with vector controls, combining AdhTpn inoculation with AdhTAP1 resulted in a significant augmentation of protection from tumor-induced death than either component alone. CONCLUSIONS: This is the first demonstration that Tpn alone can enhance survival and immunity against tumors but additionally suggests that Tpn and TAP should be used together as components of immunotherapeutic vaccine protocols to eradicate tumors.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Antigen Presentation , Lung Neoplasms/therapy , Membrane Transport Proteins/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cross-Priming , Dendritic Cells/immunology , Disease-Free Survival , Histocompatibility Antigens Class I/immunology , Humans , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Survival Rate , T-Lymphocytes, Cytotoxic
9.
Vaccine ; 25(12): 2331-9, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17229499

ABSTRACT

Despite continued progress in understanding the pathophysiology of tumours, curative therapeutic options are still lacking for the metastatic form of the disease. One approach that has gathered considerable interest is the creation of therapeutic vaccines using genetically engineered non-replicating viruses as vehicles to revive immunosurveillance mechanisms that may eradicate residual tumour cells. A perceived problem with this approach is that the number of non-replicating viruses used as a vaccine inoculum does not remotely approximate the total number of cells in the body, nor even the number of tumour cells in the case of large tumour burden or metastasis. Here, we addressed the hypothesis that a limited amount of inoculum (1x10(8) PFU) of recombinant non-replicating adenovirus encoding human TAP1 (AdhTAP1) can induce protective immunity against 1.5x10(5) TAP-deficient, metastatic melanoma cells transplanted into a normal mouse (total of approximately 1x10(11) body cells). We show that efficacious anti-tumour cytolytic T cell responses are indeed induced by injecting melanoma-bearing animals with small numbers of recombinant viruses, resulting in increases in tumour-infiltrating dendritic cells, enhanced memory T cell subpopulations and, most importantly, in increased animal survival. This novel approach uses a limited input inoculum relative to the tumour cell mass, and thus achieves an efficacious outcome that has so far eluded other vaccine, immunotherapeutic or gene therapeutic strategies where there is a requisite for the majority of tumour cells to be transduced for beneficial outcome to be achieved.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Adenoviridae/genetics , Cancer Vaccines/immunology , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Dendritic Cells/immunology , Female , H-2 Antigens/immunology , H-2 Antigens/metabolism , Humans , Immunoblotting , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL
10.
Cancer Res ; 65(17): 7926-33, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140964

ABSTRACT

A wide variety of human carcinomas have low expression of tumor-associated antigen presentation in the context of MHC class I antigens due to defects in the antigen presentation pathway. This immune evasion mechanism renders many tumors unrecognizable by host immune surveillance mechanisms. The present study examines the expression of HLA, tapasin, transporter associated with antigen processing 1 (TAP1), and beta2 microglobulin in human small cell lung carcinoma and non-small cell lung carcinoma. Immunohistochemical staining showed severe impairment of the antigen presentation pathway in all patients. In order to recover tumor immunogenicity, a nonreplicating adenovirus expressing human TAP1 (AdhTAP1) was used to restore the expression of TAP1 in the antigen presentation pathway-deficient mouse lung carcinoma cell line, CMT.64. Infection of CMT.64 cells with AdhTAP1 increased MHC class I antigen surface expression, antigen presentation, and susceptibility to antigen-specific CTLs. Fluorescence-activated cell sorting and ELISPOT analysis showed that AdhTAP1 treatment significantly increased dendritic cell cross-presentation and cross-priming of tumor antigens. Furthermore, ex vivo and in vivo AdhTAP1 treatment significantly retarded tumor growth and increased survival of mice bearing CMT.64 tumors. Fluorescence-activated cell sorting analysis and immunohistochemical staining showed a significant increase in CD8+ and CD4+ T cells and CD11c+ dendritic cells infiltrating the tumors. The results show that TAP should be considered as a part of the immunotherapies for various cancers because it is likely to provide a general method for increasing immune responses against tumors regardless of the antigenic composition of the tumor or the MHC haplotypes of the host.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Antigen Presentation/immunology , Antiporters/biosynthesis , HLA Antigens/biosynthesis , Immunoglobulins/biosynthesis , Lung Neoplasms/immunology , beta 2-Microglobulin/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Adenoviridae/genetics , Aged , Antiporters/genetics , Antiporters/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/immunology , Carcinoma, Small Cell/metabolism , Cell Line, Tumor , Dendritic Cells/immunology , Female , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Membrane Transport Proteins , Middle Aged , Spleen/cytology , Spleen/immunology , Spleen/metabolism , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
11.
Vaccine ; 21(25-26): 4022-35, 2003 Sep 08.
Article in English | MEDLINE | ID: mdl-12922139

ABSTRACT

Previous non-human primate studies have shown replication competent adenovirus (Ad) HIVenv/rev and SIVenv/rev recombinants to be promising vaccine candidates. To broaden induced immunity in rhesus macaques, an Ad type 5 host range (Ad5hr) mutant vector with an inserted SIV gag gene was added to the vaccine regimen. Immunity to the encoded SIV Env, Rev, and Gag gene products was evaluated following two immunizations with the same recombinants. The vaccines were administered intranasally plus orally via stomach tube at weeks 0 and 12. The recombinants replicated well in the upper respiratory tract but poorly in the gut, suggesting enteric-coated capsules might improve oral delivery to the intestine. SIV-specific cellular immunity was induced in all 16 immunized macaques. Fourteen exhibited positive interferon-gamma (IFN-gamma) ELISPOT responses, and nine, including two lacking IFN-gamma responses, exhibited SIV-specific T-cell proliferative activity. IFN-gamma secreting peripheral blood mononuclear cells (PBMCs) in response to SIV Gag, Env, and Rev peptides were induced in 73, 53, and 27% of macaques, respectively, and were boosted two- to four-fold by the second immunization. A persistent response to Gag was evident at least 10 weeks thereafter. p11C tetramer staining confirmed elicitation of SIV Gag-specific CD8+ T-cells in Mamu-A*01 macaques. Proliferative responses were more frequent after the second immunization, and binding antibody titers to SIV gp120 were significantly boosted by the immunization regimen. We conclude that a second administration of recombinants based in the same Ad5hr vector can effectively boost immunity to inserted gene products, obviating development of several recombinants in different Ad serotypes for multiple immunizations.


Subject(s)
Gene Products, gag/immunology , Gene Products, rev/immunology , Simian Immunodeficiency Virus/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , Blotting, Western , Cell Division , Cryopreservation , Enzyme-Linked Immunosorbent Assay , Female , Immunization, Secondary , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Macaca mulatta , Neutralization Tests , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology
12.
J Virol ; 77(15): 8354-65, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12857905

ABSTRACT

In this study we investigated the ability of a replication-competent Ad5hr-SIVenv/rev and Ad5hr-SIVgag recombinant priming/gp120 boosting regimen to induce protective immunity in rhesus macaques against pathogenic simian immunodeficiency virus(mac251). Immunization of macaques by two sequential administrations of the same recombinants by the same route resulted in boosting and persistence of SIV-specific cellular immune responses for 42 weeks past the initial immunization. Anti-SIV gp120 immunoglobulin G (IgG) and IgA antibodies were induced in secretory fluids, and all macaques exhibited serum neutralizing antibody activity. After intrarectal SIV(mac251) challenge, all of the macaques became infected. However, relative protection, as assessed by statistically significant lower SIV viral loads in plasma at both acute infection and set point, was observed in 8 out of 12 immunized non-Mamu-A(*)01 animals. Elevated mean cellular immune responses to Gag and Env, neutralizing antibody activity, and IgG and IgA binding antibody levels were observed in the eight protected macaques. Statistically significant correlations with protective outcome were observed for cellular immune responses to SIV Env and Gag and for SIV gp120-specific IgG antibodies in nasal and vaginal fluids. Two macaques that exhibited the greatest and most persistent viremia control also exhibited strong CD8(+) T-cell antiviral activity. The results suggest that a spectrum of immune responses may be necessary for adequate control of viral replication and disease progression and highlight a potential role for nonneutralizing antibodies at mucosal sites.


Subject(s)
Adenoviridae/genetics , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Administration, Rectal , Animals , Antibodies, Viral/blood , Female , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, rev/genetics , Gene Products, rev/immunology , Immunity, Mucosal , Immunization , Immunization, Secondary , Macaca mulatta , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , RNA, Viral/blood , Recombination, Genetic , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Vaccines, Synthetic , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication
13.
J Virol ; 77(16): 8607-20, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12885879

ABSTRACT

Immunity elicited by multicomponent vaccines delivered by replication-competent Ad5hr-simian immunodeficiency virus (SIV) recombinants was systematically investigated. Rhesus macaques were immunized mucosally at weeks 0 and 12 with Ad5hr-SIV(smH4) env/rev, with or without Ad5hr-SIV(mac239) gag or Ad5hr-SIV(mac239) nef, or with all three recombinants. The total Ad5hr dosage was comparably adjusted among all animals with empty Ad5hr-DeltaE3 vector. The macaques were boosted with SIV gp120 in monophosphoryl A-stable emulsion adjuvant at 24 and 36 weeks. Controls received Ad5hr-DeltaE3 vector or adjuvant only. By ELISPOT analysis, all four SIV gene products elicited potent cellular immune responses that persisted 42 weeks post-initial immunization. Unexpectedly, modulation of this cellular immune response was observed among macaques receiving one, two, or three Ad5hr-SIV recombinants. Env responses were significantly enhanced throughout the immunization period in macaques immunized with Ad5hr-SIV env/rev plus Ad5hr-SIV gag and tended to be higher in macaques that also received Ad5hr-SIV nef. Macaques primed with all three recombinants displayed significant down-modulation in numbers of gamma interferon (IFN-gamma)-secreting cells specific for SIV Nef, and the Env- and Gag-specific responses were also diminished. Modulation of antibody responses was not observed. Down-modulation was seen only during the period of Ad5hr-recombinant priming, not during subunit boosting, although SIV-specific IFN-gamma-secreting cells persisted. The effect was not attributable to Ad5hr replication differences among immunization groups. Vaccine delivery via replication-competent live vectors, which can persistently infect new cells and continuously present low-level antigen, may be advantageous in overcoming competition among complex immunogens for immune recognition. Effects of current multicomponent vaccines on individual immune responses should be evaluated with regard to future vaccine design.


Subject(s)
Adenoviridae/genetics , Genes, Viral , Immunity, Cellular , Membrane Glycoproteins/immunology , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Adenoviridae/physiology , Animals , Antibodies, Viral/biosynthesis , Cell Division , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , Macaca mulatta , Male , T-Lymphocytes/cytology , Viral Vaccines/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...