Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nutrients ; 14(22)2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36432471

ABSTRACT

Vitamin C is an important micronutrient for various immune cells. It increases phagocytic cell function and is necessary for T and natural killer (NK) cell development. Patients in need of an autologous hematopoietic stem cell transplantation (HSCT) are often vitamin C-depleted. We therefore hypothesized that vitamin C supplementation could improve immune recovery in autologous HSCT patients. This blinded, placebo-controlled trial included 44 patients randomized to receive vitamin C or a placebo. The following outcome measures used were clinical and immunological parameters, among others: time to neutrophil recovery, serum, and intracellular vitamin C values. Twenty-one patients received vitamin C, and 23 received a placebo. The time to neutrophil recovery did not differ between the two groups at 11.2 days (p = 0.96). There were no differences in hospitalization time (19.7 vs. 19.1 days, p = 0.80), the incidence of neutropenic fever (57% vs. 78%, p = 0.20), or 3-month overall survival (90.5% vs. 100%, p = 0.13). Bacteremia seemed to occur less in the vitamin C group (10% vs. 35%, p = 0.07). Our study shows no benefit from vitamin C supplementation on neutrophil recovery and hospitalization, despite possible lower rates of bacteremia in the vitamin C group. Therefore, we do not advise vitamin C supplementation in this treatment group.


Subject(s)
Bacteremia , Hematopoietic Stem Cell Transplantation , Lymphoma , Multiple Myeloma , Humans , Transplantation, Autologous , Multiple Myeloma/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Ascorbic Acid , Neutrophils , Lymphoma/therapy , Vitamins
2.
Int J Cancer ; 117(4): 596-604, 2005 Nov 20.
Article in English | MEDLINE | ID: mdl-15945101

ABSTRACT

The purpose of this phase 1/2 study was to evaluate toxicity, tumor evolution and immunologic response following administration of a fixed dose of a recombinant MAGE-3 protein by subcutaneous and intradermal routes in the absence of immunologic adjuvant. Thirty-two patients with detectable metastatic melanoma expressing gene MAGE-3 were included and 30 received at least one injection with a fixed dose of a ProtD-MAGE-3 fusion protein. The immunization schedule included 6 intradermal and subcutaneous injections at 3-week intervals. Afterward, patients without major tumor progression who required other treatments received additional vaccinations at increasing time intervals. The vaccine was generally well tolerated. Among the 26 patients who received at least 4 vaccinations, we observed 1 partial response and 4 mixed responses. For these 5 responding patients, time to progression varied from 3.5 to 51+ months. An anti-MAGE-3 CD4 T-lymphocyte response was detected in 1 out of the 5 responding patients. The majority of patients had no anti-MAGE-3 antibody response. The clinical and immunologic responses generated by the vaccine are rather limited. Nevertheless, given the potential antitumor efficacy and the very mild toxicity of vaccinations, further studies combining MAGE proteins and/or peptides with potent immunologic adjuvants are warranted, not only in metastatic melanoma, but also in the adjuvant setting.


Subject(s)
Antigens, Neoplasm/administration & dosage , Melanoma/drug therapy , Neoplasm Proteins/administration & dosage , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Disease Progression , Female , Humans , Injections, Subcutaneous , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Neoplasm Proteins/adverse effects , Neoplasm Proteins/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Survival Analysis
3.
J Immunol ; 173(7): 4699-707, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383606

ABSTRACT

Epidermal growth factor receptor (EGF-R) overexpression is common in a large number of solid tumors and represents a negative prognostic indicator. Overexpression of EGF-R is strongly tumor associated, and this tyrosine kinase type receptor is considered an attractive target for Ab therapy. In this study, we describe the evaluation of mAb 2F8, a high avidity human mAb (IgG1kappa) directed against EGF-R, developed using human Ig transgenic mice. mAb 2F8 effectively blocked binding of EGF and TGF-alpha to the EGF-R. At saturating concentrations, 2F8 completely blocked EGF-R signaling and inhibited the in vitro proliferation of EGF-R-overexpressing A431 cells. At much lower concentrations, associated with low receptor occupancy, 2F8 induced efficient Ab-dependent cell-mediated cytotoxicity (ADCC) in vitro. In vivo studies showed potent antitumor effects in models with A431 tumor xenografts in athymic mice. Ex vivo analysis of the EGF-R status in tumor xenografts in 2F8-treated mice revealed that there are two therapeutic mechanisms. First, blocking of EGF-R signaling, which is most effective at complete receptor saturation and therefore requires a relatively high Ab dose. Second, at very low 2F8 receptor occupancy, we observed potent antitumor effects in mice, which are likely based on the engagement of immune effector mechanisms, in particular ADCC. Taken together, our findings indicate that ADCC represents an important effector mechanism of this Ab, which is effective at relatively low dose.


Subject(s)
Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites, Antibody , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Immunologic , ErbB Receptors/physiology , Female , Growth Inhibitors/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology , Neoplasms/prevention & control , Protein Binding , Signal Transduction/immunology , Xenograft Model Antitumor Assays/methods
4.
Int Immunol ; 16(8): 1091-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15192052

ABSTRACT

Dendritic cells (DC) can trigger naive CD8(+) T cell responses by their capacity to cross-present exogenous antigens via the major histocompatibility complex class I pathway. The myeloid class I IgG receptor, FcgammaRI (CD64), is expressed on DC, and in vivo targeting of antigens to FcgammaRI induces strong humoral and cellular immune responses. We studied the capacity of human FcgammaRI (hFcgammaRI) to facilitate DC-mediated cross presentation and T cell activation, and assessed the effect of CpG oligodeoxynucleotides on this process. We generated hFcgammaRI expressing immature DC from hFcgammaRI transgenic and immature DC from non-transgenic mice. Antigens were targeted to Fcgamma receptors as ovalbumin immune complexes, or selectively to hFcgammaRI via ovalbumin-CD64 mAb fusion proteins. Co-incubation of immature DC with CpG ODN led to markedly increased MHC class I presentation of FcgammaR-targeted antigens. When OVA was selectively targeted to hFcgammaRI, few differences were observed between Tg and NTg DC. However, upon co-incubation with CpG ODN, hFcgammaRI-triggered cross presentation was enhanced. These results document the capacity of hFcgammaRI on DC to trigger cross presentation via MHC class I upon co-culture with CpG ODN.


Subject(s)
Antigen Presentation/drug effects , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Genes, MHC Class I/immunology , Oligodeoxyribonucleotides/pharmacology , Receptors, IgG/immunology , Animals , Antigen Presentation/immunology , Humans , Mice , Mice, Transgenic , Ovalbumin/immunology , Receptors, IgG/genetics
5.
Anticancer Drugs ; 15(6): 587-91, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15205601

ABSTRACT

Our objective was to study the influence of pegylated interferon-alpha2b (PEG-IFN-alpha) on the metabolism of amino acids and pteridines. We used an exploratory study into plasma concentrations of large neutral amino acids, 5-hydroxyindolacetic acid (5-HIAA), total biopterin (BIOP) and neopterin (NEOP) in 40 high-risk melanoma patients. Patients were randomized to treatment with PEG-IFN-alpha once a week in a dose of 6 microg/kg/week s.c. during 8 weeks, followed by a maintenance treatment of 3 microg/kg/week s.c. or to observation only. We found that treatment with PEG-IFN-alpha decreases tryptophan (TRP) concentrations in the first 3 months of treatment to a maximum of 25.3% compared to controls [95% confidence interval (CI): 14.9 to 34.4]. The TRP:LNAA ratio, an index for the availability of TRP to the central nervous system (CNS), decreases during 6 months with 18.8% (95% CI: 11.9 to 25.2). Concentrations of NEOP rose; however, concentrations of BIOP, the sum of tetrahydrobiopterin [BH4] and its oxidative products, did not decrease. The ratio of phenylalanine to tyrosine was increased with 11.7% (95% CI: 1.0 to 23.5) during 6 months. We conclude that, like conventional IFN-alpha, PEG-IFN-alpha lowers TRP concentrations and decreases the availability of TRP to the CNS. PEG-IFN-alpha has a similar influence on pteridine metabolism as standard IFN-alpha. If a lowered availability of TRP and a consequent decrease of serotonergic neurotransmission are indeed a mechanism underlying neuropsychiatric side-effects of IFN-alpha, patients on PEG-IFN-alpha are not at a lower risk of developing neuropsychiatric side-effects as patients on conventional IFN-alpha.


Subject(s)
Amino Acids/metabolism , Hydroxyindoleacetic Acid/metabolism , Interferon-alpha , Interferon-alpha/therapeutic use , Melanoma/drug therapy , Polyethylene Glycols , Pteridines/metabolism , Adult , Aged , Amino Acids/drug effects , Biopterins/metabolism , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interferon-alpha/pharmacokinetics , Male , Middle Aged , Neopterin/metabolism , Phenylalanine/drug effects , Phenylalanine/metabolism , Pilot Projects , Randomized Controlled Trials as Topic , Recombinant Proteins , Time Factors , Tryptophan/drug effects , Tryptophan/metabolism , Tyrosine/drug effects , Tyrosine/metabolism
6.
Cancer Res ; 63(17): 5595-600, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500400

ABSTRACT

Immunostimulatory CpG oligodeoxynucleotides (ODNs) can enhance the therapeutic effect of monoclonal antibodies (mAbs) by enhancing antibody-dependent cell-mediated cytotoxicity (ADCC). Distinct classes of CpG ODNs have been found recently to stimulate different effector cell populations. We used murine cancer models to explore the role of various effector cell populations in the antitumor activity seen with mAbs combined with CpG ODNs of the A and B classes. In the 38C13 syngeneic murine lymphoma model, both CpG A and CpG B enhanced the efficacy of murine antilymphoma mAb. Depletion of natural killer (NK) cells alone markedly decreased the efficacy of therapy with mAbs plus CpG A. In contrast, depletion of both NK cells and granulocytes was required to decrease the efficacy of mAb plus CpG B. A human (h) Fc gamma receptor I (FcgammaRI)-expressing transgenic (Tg) mouse model was used to explore the role of FcgammaRI in therapy with mAb and CpG ODN. CpG B induced up-regulation of FcgammaRI in hFcgammaRI Tg mice, whereas CpG A did not. In vitro CpG B also enhanced ADCC of HER-2/neu-expressing tumor cells by the FcgammaRI-directed bispecific antibody MDX-H210 using hFcgammaRI-positive effector cells. In a solid tumor model, tumor growth was inhibited in Tg mice treated with a combination of MDX-H210 and CpG B. These data suggest that CpG A enhance ADCC largely by activating NK cells. In contrast, other effector cell populations, including granulocytes, contribute to the antitumor activity of CpG B and mAbs. FcgammaRI plays an important role in this activity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Monoclonal/pharmacology , CpG Islands/immunology , Granulocytes/immunology , Immunization, Passive/methods , Killer Cells, Natural/immunology , Oligonucleotides/pharmacology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Division/drug effects , Cell Division/immunology , Drug Synergism , Female , Granulocytes/drug effects , Granulocytes/metabolism , Humans , Immunoglobulin G/immunology , Killer Cells, Natural/drug effects , Lymphoma/immunology , Lymphoma/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Oligonucleotides/immunology , Receptor, ErbB-2/immunology , Receptors, IgG/biosynthesis , Receptors, IgG/immunology , Up-Regulation
7.
Cancer Immunol Immunother ; 52(3): 133-44, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12649742

ABSTRACT

The clinical development of interleukin 12 (IL-12) as a single agent for systemic cancer therapy has been hindered by its significant toxicity and disappointing anti-tumor effects. The lack of efficacy was accompanied by, and probably related to, the declining biological effects of IL-12 in the course of repeated administrations at doses approaching the maximum tolerated dose (MTD). Nevertheless, IL-12 remains a very promising immunotherapeutic agent because recent cancer vaccination studies in animal models and humans have demonstrated its powerful adjuvant properties. Therefore, IL-12 may re-enter the arena of cancer therapy. Here, we review the immune modulating characteristics of IL-12 considered responsible for the adjuvant effects, as well as the results of animal and human cancer vaccination studies with IL-12 applied as an adjuvant. In addition, we discuss how studies with systemic IL-12 in cancer patients, and several other lines of evidence, indicate that IL-12 may exert optimal adjuvant effects only at low dose levels. Therefore, the MTD may not constitute the maximum effective dose of IL-12 for adjuvant application.


Subject(s)
Cancer Vaccines , Chemotherapy, Adjuvant , Interleukin-12/therapeutic use , Animals , Dendritic Cells , Dose-Response Relationship, Drug , Down-Regulation , Humans , Inflammation , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Maximum Tolerated Dose , Neoplasms/immunology , Neoplasms/metabolism , Recombinant Proteins/metabolism , T-Lymphocytes/immunology , Time Factors
8.
Blood ; 101(1): 253-8, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12485936

ABSTRACT

Antibody-reliant destruction of tumor cells by immune effector cells is mediated by antibody-dependent cellular cytotoxicity, in which Fc receptor (FcR) engagement is crucial. This study documents an important role for the beta(2) integrin Mac-1 (CD11b/CD18) in FcR-mediated protection against melanoma. CD11b-deficient mice, those that lack Mac-1, were less protected by melanoma-specific monoclonal antibody TA99 than wild-type (WT) mice. Significantly more lung metastases and higher tumor loads were observed in Mac-1(-/-) mice. Histologic analyses revealed no differences in neutrophil infiltration of lung tumors between Mac-1(-/-) and WT mice. Importantly, Mac-1(-/-) phagocytes retained the capacity to bind tumor cells, implying that Mac-1 is essential during actual FcR-mediated cytotoxicity. In summary, this study documents Mac-1 to be required for FcR-mediated antimelanoma immunity in vivo and, furthermore, supports a role for neutrophils in melanoma rejection.


Subject(s)
Antibody Formation , Macrophage-1 Antigen/immunology , Melanoma/immunology , Receptors, Fc/immunology , Animals , Antibodies, Neoplasm , CD11b Antigen/genetics , CD11b Antigen/immunology , Chemotaxis, Leukocyte , Granulocyte Colony-Stimulating Factor/pharmacology , Lung Neoplasms/secondary , Macrophage-1 Antigen/genetics , Melanoma/pathology , Mice , Mice, Knockout , Neutrophils/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...