Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 11(1): 2115655, 2022.
Article in English | MEDLINE | ID: mdl-36052217

ABSTRACT

CCL22 is a macrophage-derived immunosuppressive chemokine that recruits regulatory T cells through the CCL22:CCR4 axis. CCL22 was shown to play a key role in suppressing anti-cancer immune responses in different cancer types. Recently, we showed that CCL22-specific T cells generated from cancer patients could kill CCL22-expressing tumor cells and directly influence the levels of CCL22 in vitro. The present study aimed to provide a rationale for developing a CCL22-targeting immunotherapy. Vaccination with CCL22-derived peptides induced CCL22-specific T-cell responses in both BALB/c and C57BL/6 mice, assessed by interferon-γ secretion ex vivo. Anti-tumor efficacy of the peptides was evaluated in mouse models engrafted with syngeneic tumor models showing a reduced tumor growth and prolonged survival of the treated mice. Vaccination induced changes in the cellular composition of immune cells that infiltrated the tumor microenvironment assessed with multicolor flow cytometry. In particular, the infiltration of CD8+ cells and M1 macrophages increased, which increased the CD8/Treg and the M1/M2 macrophage ratio. This study provided preclinical evidence that targeting CCL22 with CCL22 peptide vaccines modulated the immune milieu in the tumor microenvironment. This modulation led to an augmentation of anti-tumor responses. This study provided a rationale for developing a novel immunotherapeutic modality in cancer.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/pathology , T-Lymphocytes, Regulatory , Vaccines, Subunit
3.
Cancer Immunol Res ; 10(5): 571-580, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35290437

ABSTRACT

The immunosuppressive tumor microenvironment (TME) does not allow generation and expansion of antitumor effector cells. One of the potent immunosuppressive factors present in the TME is the indoleamine-pyrrole 2,3-dioxygenase (IDO) enzyme, produced mainly by cancer cells and suppressive immune cells of myeloid origin. In fact, IDO+ myeloid-derived suppressor cells (MDSC) and dendritic cells (DC) tend to be more suppressive than their IDO- counterparts. Hence, therapeutic approaches that would target the IDO+ cells in the TME, while sparing the antigen-presenting functions of IDO- myeloid populations, are needed. Using an IDO-specific peptide vaccine (IDO vaccine), we explored the possibility of generating effector cells against IDO and non-IDO tumor-derived antigens. For this, IDO-secreting (B16F10 melanoma) and non-IDO-secreting (TC-1) mouse tumor models were employed. We showed that the IDO vaccine significantly reduced tumor growth and enhanced survival of mice in both the tumor models, which associated with a robust induction of IDO-specific effector cells in the TME. The IDO vaccine significantly enhanced the antitumor efficacy of non-IDO tumor antigen-specific vaccines, leading to an increase in the number of total and antigen-specific activated CD8+ T cells (IFNγ+ and granzyme B+). Treatment with the IDO vaccine significantly reduced the numbers of IDO+ MDSCs and DCs, and immunosuppressive regulatory T cells in both tumor models, resulting in enhanced therapeutic ratios. Together, we showed that vaccination against IDO is a promising therapeutic option for both IDO-producing and non-IDO-producing tumors. The IDO vaccine selectively ablates the IDO+ compartment in the TME, leading to a significant enhancement of the immune responses against other tumor antigen-specific vaccines.


Subject(s)
Cancer Vaccines , Melanoma , Animals , Antigens, Neoplasm , Indoleamine-Pyrrole 2,3,-Dioxygenase , Melanoma/drug therapy , Mice , Tumor Microenvironment
4.
Nat Med ; 27(12): 2212-2223, 2021 12.
Article in English | MEDLINE | ID: mdl-34887574

ABSTRACT

Anti-programmed death (PD)-1 (aPD1) therapy is an effective treatment for metastatic melanoma (MM); however, over 50% of patients progress due to resistance. We tested a first-in-class immune-modulatory vaccine (IO102/IO103) against indoleamine 2,3-dioxygenase (IDO) and PD ligand 1 (PD-L1), targeting immunosuppressive cells and tumor cells expressing IDO and/or PD-L1 (IDO/PD-L1), combined with nivolumab. Thirty aPD1 therapy-naive patients with MM were treated in a phase 1/2 study ( https://clinicaltrials.gov/ , NCT03047928). The primary endpoint was feasibility and safety; the systemic toxicity profile was comparable to that of nivolumab monotherapy. Secondary endpoints were efficacy and immunogenicity; an objective response rate (ORR) of 80% (confidence interval (CI), 62.7-90.5%) was reached, with 43% (CI, 27.4-60.8%) complete responses. After a median follow-up of 22.9 months, the median progression-free survival (PFS) was 26 months (CI, 15.4-69 months). Median overall survival (OS) was not reached. Vaccine-specific responses assessed in vitro were detected in the blood of >93% of patients during vaccination. Vaccine-reactive T cells comprised CD4+ and CD8+ T cells with activity against IDO- and PD-L1-expressing cancer and immune cells. T cell influx of peripherally expanded T cells into tumor sites was observed in responding patients, and general enrichment of IDO- and PD-L1-specific clones after treatment was documented. These clinical efficacy and favorable safety data support further validation in a larger randomized trial to confirm the clinical potential of this immunomodulating approach.


Subject(s)
Adjuvants, Immunologic/administration & dosage , B7-H1 Antigen/immunology , Cancer Vaccines/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Melanoma/therapy , Nivolumab/therapeutic use , Skin Neoplasms/therapy , Humans , Melanoma/pathology , Skin Neoplasms/pathology
5.
Cancer Immunol Res ; 9(11): 1316-1326, 2021 11.
Article in English | MEDLINE | ID: mdl-34518197

ABSTRACT

Expression of the L-arginine catabolizing enzyme arginase 1 (ARG1) is a central immunosuppressive mechanism mediated by tumor-educated myeloid cells. Increased activity of ARG1 promotes the formation of an immunosuppressive microenvironment and leads to a more aggressive phenotype in many cancers. Intrinsic T-cell immunity against ARG1-derived epitopes in the peripheral blood of cancer patients and healthy subjects has previously been demonstrated. To evaluate the antitumor efficacy of ARG1-derived peptide vaccines as a monotherapy and as a combinational therapy with checkpoint blockade, different in vivo syngeneic mouse tumor models were utilized. To evaluate the antitumor effects, flow cytometry analysis and IHC were performed on tumors, and ELISPOT assays were performed to characterize immune responses. We show that ARG1-targeting therapeutic vaccines were able to activate endogenous antitumor immunity in several in vivo syngeneic mouse tumor models and to modulate the cell composition of the tumor microenvironment without causing any associated side effects or systemic toxicity. ARG1-targeting vaccines in combination with anti-PD-1 also resulted in increased T-cell infiltration, decreased ARG1 expression, reduced suppressive function of tumor-educated myeloid cells, and a shift in the M1/M2 ratio of tumor-infiltrating macrophages. These results indicated that the induced shift toward a more proinflammatory microenvironment by ARG1-targeting immunotherapy favors effective tumor control when combined with anti-PD-1 checkpoint blockade. Our data illustrate the ability of ARG1-based immune modulatory vaccination to elicit antigen-specific immunosurveillance and imply the feasibility of this novel immunotherapeutic approach for clinical translation.


Subject(s)
Arginase/metabolism , Myeloid Cells/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Vaccines/therapeutic use , Animals , Cell Line, Tumor , Female , Humans , Mice , Tumor Microenvironment , Vaccines/pharmacology , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 27(3): 689-703, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33122346

ABSTRACT

Therapeutic cancer vaccines, an exciting development in cancer immunotherapy, share the goal of creating and amplifying tumor-specific T-cell responses, but significant obstacles still remain to their success. Here, we briefly outline the principles underlying cancer vaccine therapy with a focus on novel vaccine platforms and antigens, underscoring the renewed optimism. Numerous strategies have been investigated to overcome immunosuppressive mechanisms of the tumor microenvironment (TME) and counteract tumor escape, including improving antigen selection, refining delivery platforms, and use of combination therapies. Several new cancer vaccine platforms and antigen targets are under development. In an effort to amplify tumor-specific T-cell responses, a heterologous prime-boost antigen delivery strategy is increasingly used for virus-based vaccines. Viruses have also been engineered to express targeted antigens and immunomodulatory molecules simultaneously, to favorably modify the TME. Nanoparticle systems have shown promise as delivery vectors for cancer vaccines in preclinical research. T-win is another platform targeting both tumor cells and the TME, using peptide-based vaccines that engage and activate T cells to target immunoregulatory molecules expressed on immunosuppressive and malignant cells. With the availability of next-generation sequencing, algorithms for neoantigen selection are emerging, and several bioinformatic platforms are available to select therapeutically relevant neoantigen targets for developing personalized therapies. However, more research is needed before the use of neoepitope prediction and personalized immunotherapy becomes commonplace. Taken together, the field of therapeutic cancer vaccines is fast evolving, with the promise of potential synergy with existing immunotherapies for long-term cancer treatment.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Nanoparticle Drug Delivery System , Neoplasms/therapy , Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Humans , Immunogenicity, Vaccine , Neoplasms/genetics , Neoplasms/immunology , Precision Medicine/methods , Precision Medicine/trends , T-Lymphocytes/immunology , Tumor Escape , Tumor Microenvironment/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
7.
J Immunother Cancer ; 8(2)2020 07.
Article in English | MEDLINE | ID: mdl-32690770

ABSTRACT

BACKGROUND: The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase 1 (IDO1), which subverts T-cell immunity at multiple levels, is itself subject to inherent T-cell reactivity. This intriguing deviation from central tolerance has been interpreted as counterbalancing IDO1-mediated immunosuppression. Based on this hypothesis, clinical studies employing an IDO1 peptide-based vaccine approach for cancer treatment have been initiated, but there remains a pressing need to further investigate the immunological ramifications of stimulating the anti-IDO1 T-cell response in this manner. METHODS: CT26 colon carcinoma tumors were evaluated for expression of IDO1 protein by western blot analysis, immunofluorescence microscopy and flow cytometry. Mouse IDO1-derived peptides, predicted to bind either major histocompatibility complex (MHC) class I or II of the H2d BALB/c strain, were emulsified in 50% Montanide for prophylactic or therapeutic vaccine treatment of CT26 tumor-bearing mice initiated either 7 days prior to or following tumor cell injection, respectively. In some therapeutic treatment experiments, administration of programmed cell death protein 1-binding antibody (anti-PD1 antibody) or epacadostat was concurrently initiated. Tumor size was determined by caliper measurements and comparative tumor growth suppression was assessed by longitudinal analyses of tumor growth data. For adoptive transfer, T cells from complete responder animals were isolated using paramagnetic beads and fluorescence-activated cell sorting. RESULTS: This study identifies mouse MHC class I-directed and II-directed, IDO1-derived peptides capable of eliciting antitumor responses, despite finding IDO1 expressed exclusively in tumor-infiltrating immune cells. Treatment of established tumors with anti-PD1 antibody and class I-directed but not class II-directed IDO1 peptide vaccines produced an enhanced antitumor response. Likewise, class I-directed and II-directed IDO1 peptides elicited an enhanced combinatorial response, suggesting distinct mechanisms of action. Consistent with this interpretation, adoptive transfer of isolated CD8+ T cells from class I and CD4+ T cells from class II peptide-vaccinated responder mice delayed tumor growth. The class II-directed response was completely IDO1-dependent while the class I-directed response included an IDO1-independent component consistent with antigen spread. CONCLUSIONS: The in vivo antitumor effects demonstrated with IDO1-based vaccines via targeting of the tumor microenvironment highlight the utility of mouse models for further exploration and refinement of this novel vaccine-based approach to IDO1-directed cancer therapy and its potential to improve patient response rates to anti-PD1 therapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vaccines, Subunit/therapeutic use , Animals , Cancer Vaccines/pharmacology , Cell Line, Tumor , Female , Humans , Mice , Mice, Transgenic , Vaccines, Subunit/pharmacology
8.
Chin Clin Oncol ; 7(2): 19, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29764164

ABSTRACT

Historically, the development of cancer vaccines has focused on the central role of tumor antigens in eliciting tumor-specific immune responses, with limited success. Recent advances with checkpoint blockade approaches have brought about a renewed appreciation of the importance of targeting immune suppression in cancer patients. Here we discuss a novel approach to cancer immunotherapy, namely to target recently described T cells that uniquely control cells with immune suppressive functions. Accumulating evidence support the existence of self-reactive T cells that are specific to antigens derived from immunoregulatory proteins ("immunoregulatory antigens"), such as indoleamine 2,3-dioxygenase (IDO) and PD-L1. Vaccination approaches to potentiate these T cells have proven safe with minimal toxicity in the clinical phase I trials conducted thus far. Given that immunoregulatory antigens can be new targets for cancer immunotherapy, we propose here that they could be considered as a new class of tumor antigens. Targeting such antigens has advantages over targeting classical tumor antigens, as there is no requirement for identification of relevant antigens that are specific for the cancer type, and the targets are genetically stable. Furthermore, targeting immunoregulatory antigen-specific T cells potentially has dual mode of actions (I) targeting immune suppression and thereby potentiating anti-tumor effector T cell responses and (II) direct killing of immunoregulatory antigen-expressing tumor cells.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy/methods , Humans
9.
Basic Clin Pharmacol Toxicol ; 117(5): 306-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25903922

ABSTRACT

HIV protease inhibitors (PIs) are antiretroviral agents, which have been found to also affect several cellular processes, such as inflammation and cell progression. In studies on non-steroidal, anti-inflammatory drugs, the addition of a nitric oxide (NO) moiety has been shown to both reduce their toxicity and enhance their pharmacological efficacy. Along this line of research, several derivatives of PIs have been synthesized by covalent attachment of NO moiety to the parental molecules. Previous work has indicated that NO-hybridization of the prototypical PI, Saquinavir leads to a derivative named Saquinavir-NO that while retaining the antiretroviral effect, acquires antitumoural and immunomodulatory properties along with reduced toxicity in vitro and in vivo. These data prompted us to evaluate the effects of NO-hybridization on two other PIs, Lopinavir and Ritonavir. The two NO-derivatives were compared head to head with their parental compounds on human primary peripheral blood mononuclear cells as well as on human primary macrophages. Lopinavir-NO and Lopinavir were also screened in an in vivo model of autoimmune hepatitis. Our results prove that Lopinavir-NO exerts markedly superior effects as compared to the parental compound both in vitro and in vivo. On the contrary, Ritonavir-NO effects overlapped those of Ritonavir. These data demonstrate that NO-hybridization of Lopinavir generates a derivative with significantly stronger immunomodulatory effects that are apparently related to an action of the compound on T-cell secretory capacity. Lopinavir-NO deserves additional studies for its possible use in T-cell-mediated autoimmune diseases including, but not limited to autoimmune hepatitis.


Subject(s)
HIV Protease Inhibitors/pharmacology , Hepatitis, Autoimmune/drug therapy , Immunologic Factors/pharmacology , Leukocytes, Mononuclear/drug effects , Lopinavir/pharmacology , Macrophages/drug effects , Nitric Oxide/pharmacology , Ritonavir/pharmacology , Animals , Concanavalin A , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Drug Design , Female , Hepatitis, Autoimmune/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lopinavir/analogs & derivatives , Macrophages/immunology , Macrophages/metabolism , Mice , Nitric Oxide/analogs & derivatives , Ritonavir/analogs & derivatives , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
J Neuroimmunol ; 259(1-2): 55-65, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23602714

ABSTRACT

NO-hybridization of the HIV protease inhibitor Saquinavir generates a new chemical entity named Saq-NO, that retains the anti-viral activity and exerts lower toxicity. We show that Saq-NO inhibited the generation of various cytokines in ConA-stimulated unfractionated murine spleen cells and rat lymph nodes stimulated with ConA as well as in purified CD4(+) T cells in vitro and reduced the circulating levels of cytokines in mice challenged with anti-CD3 antibody. Furthermore, Saq-NO reduced IL-17 and IFN-γ production in myelin basic protein (MBP)-specific cells isolated from rats immunized with MBP. These findings translated well into the in vivo setting as Saq-NO ameliorated the course of the disease in two preclinical models of multiple sclerosis. Our results demonstrate that Saq-NO exerts immunomodulatory effects that warrant studies on its application in autoimmune diseases.


Subject(s)
Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Saquinavir/analogs & derivatives , Animals , Benzofurans , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinolines , Rats , Rats, Inbred Strains , Ribosomal Protein S6 Kinases/metabolism , Saquinavir/pharmacology , Spleen/cytology
11.
Mol Pharmacol ; 82(4): 700-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22798453

ABSTRACT

We have examined the influence of the nitric oxide (NO)-modified anti-inflammatory drug (S,R)-3-phenyl-4,5-dihydro-5-isoxasole acetic acid (VGX-1027) named GIT-27NO or the NO-modified antiviral drug saquinavir (Saq) named Saq-NO on two colon cancer cell lines, mouse CT26CL25 and human HCT116. The effects of the drugs on cell viability, apoptosis, proliferation, and metastatic potential were analyzed. The release of NO and oxygen and nitrogen species was also determined. The efficacy of the drugs was evaluated in vivo in BALB/c mice injected with CT26CL25 cells. Both agents suppressed the growth of colon cancer cells in vitro and reduced tumor volume in syngeneic BALB/c mice. However, their mechanisms of action were different because GIT-27NO released larger amounts of nitrite than Saq-NO in cell cultures and its antitumor action depended on the intracellular NO release inside the cells. On the contrary, Saq-NO released barely detectable amounts of NO and its antitumor action was NO-independent. In fact, cotreatment with an NO-peroxynitrite scavenger revealed that GIT-27NO but not Saq-NO acts through peroxynitrite-mediated cell destruction. At the cellular level, GIT-27NO prevalently induced proapoptotic signals followed by caspase-dependent apoptosis. In contrast, Saq-NO blocked cell proliferation, changed the adhesive, migratory, and invasive properties of the cells, and decreased metastatic potential in vivo. In conclusion, differences in NO release and oxidative stress generation between GIT-27NO and Saq-NO resulted in different mechanisms that caused cell death.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Nitric Oxide/chemistry , Oxazoles/pharmacology , Saquinavir/pharmacology , Acetates/chemistry , Acetates/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Caspases/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Transplantation , Nitric Oxide/physiology , Oxazoles/chemistry , Oxazoles/therapeutic use , Saquinavir/chemistry , Saquinavir/therapeutic use
12.
Oncol Rep ; 28(2): 682-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22665020

ABSTRACT

We have recently shown that covalent attachment of the nitric oxide (NO) moiety to the HIV protease inhibitor Saquinavir (Saq) produced a qualitatively new chemical entity, named Saquinavir-NO (Saq-NO), with enhanced anticancer properties and reduced toxicity both in vitro and in vivo. The aim of this study was to address several unanswered questions both on the pharmacological profile of Saq-NO as well as on the in vivo role of NO in the oncogenesis of A375 human melanoma cells. To this end, we have evaluated here the impact of single and combined effects of Saq-NO, Saq, the NO-donor DETA NONOate and the iNOS inhibitor L-NAME on the in vitro as well as in vivo growth of the iNOS positive A375 cells. Our data confirm clear-cut evidence for a strong and powerful anti-melanoma action of Saq-NO that is not duplicable by the combined use of Saq and DETA NONOate. Surprisingly, but also in agreement with the complex and multifaceted role of endogenous NO in A375 cells, both DETA NONOate and L-NAME significantly suppressed the in vivo growth of xenotransplants.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Saquinavir/analogs & derivatives , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Nitric Oxide Donors/pharmacology , Saquinavir/pharmacology , Xenograft Model Antitumor Assays
13.
Cell Cycle ; 11(6): 1174-82, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22370480

ABSTRACT

We previously reported that the NO-modified form of HIV protease inhibitor Saquinavir (Saq) is a potent antitumoral agent efficient against numerous tumor cell lines in vitro and in vivo. In acute toxicity studies, doses of Saq-NO equivalent to DL100 of the parental drug were completely nontoxic. Beside direct effect on malignant cell growth, Saq-NO sensitizes certain type of cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated cell death. In this study, we evaluated the effects of Saq-NO on androgen-dependent prostate cancer LNCaP. Saq-NO inhibited both the growth of LNCaP cells in vitro and in xenograft models. Suppression of tumor growth was accompanied with cell cycle arrest in G 0/G 1 phase and established a persistent inhibition of proliferation. Furthermore, Saq-NO reverted sensitivity of LNCaP cells to TRAIL but not to TNF. Treatment of cells with Saq-NO induced transient upregulation of Akt and ERK1/2. This, however, did not represent the primary mode of action of Saq-NO, as elimination with specific inhibitors did not compromise the chemotherapic efficacy of the drug. However, permanent abrogation of phosphorylation of the S6 protein, which is the downstream target of both signaling pathways, was observed. Diminished S6 phosphorylation was associated with re-established sensitivity to TRAIL and reduction of X-linked inhibitor of apoptosis protein (XIAP). In summary, NO modification of Saq led to a new chemical entity with stronger and more pleiotropic antitumor activity than the parental drug.


Subject(s)
Prostatic Neoplasms/pathology , Ribosomal Protein S6 Kinases/metabolism , Saquinavir/analogs & derivatives , TNF-Related Apoptosis-Inducing Ligand/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Cell Death , Cell Line, Tumor , Cell Proliferation , Cell Survival , G1 Phase Cell Cycle Checkpoints , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Prostatic Neoplasms/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ribosomal Protein S6 Kinases/genetics , Saquinavir/administration & dosage , Saquinavir/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
14.
Vitam Horm ; 86: 63-82, 2011.
Article in English | MEDLINE | ID: mdl-21419267

ABSTRACT

Dendritic cells (DCs), the most potent antigen-presenting cells of the immune system, express nuclear receptors for 1,25-dihydroxyvitamin D(3) (VD3) and they are one of its main targets. In the presence of VD3, DCs differentiate into a phenotype that resembles semimature DCs, with reduced T cell costimulatory molecules and hampered IL-12 production. These VD3-modulated DCs induce T cell tolerance in vitro using multiple mechanisms such as rendering T cells anergic, dampening of Th1 responses, and recruiting and differentiating regulatory T cells. Due to their ability to specifically target pathological T cells, VD3-modulated DCs are safe and potentially more effective alternatives to currently available immunoregulatory therapies. While a number of considerations remain, including the establishment of a reliable quality control measure to ensure the safety and efficacy of VD3-DCs in vivo and the optimal frequency, dose, and route of DC administration to achieve therapeutic effects in humans, adoptive VD3-DC transfer represents one of the most promising approaches to future treatment of autoimmune diseases.


Subject(s)
Autoimmune Diseases/therapy , Dendritic Cells/drug effects , Immunotherapy , Vitamin D/pharmacology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Calcitriol/metabolism , Calcitriol/pharmacology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Humans , Vitamin D/metabolism
15.
Hum Immunol ; 71(1): 67-73, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19819280

ABSTRACT

Dendritic cells (DCs) exposed to tumor antigens followed by treatment with T(h)1-polarizing differentiation signals have paved the way for the development of DC-based cancer vaccines. Critical parameters for assessment of the optimal functional state of DCs and prediction of the vaccine potency of activated DCs have in the past been based on measurements of differentiation surface markers like HLA-DR, CD80, CD83, CD86, and CCR7 and the level of secreted cytokines like interleukin-12p70. However, the level of these markers does not provide a complete picture of the DC phenotype and may be insufficient for prediction of clinical outcome for DC-based therapy. We therefore looked for additional biomarkers by investigating the differential expression of microRNAs (miRNAs) in mature DCs relative to immature DCs. A microarray-based screening revealed that 12 miRNAs were differentially expressed in the two DC phenotypes. Of these, four miRNAs, hsa-miR-155, hsa-miR-146a, hsa-miR-125a-5p, and hsa-miR-29a, were validated by real-time polymerase chain reaction and northern blotting. The matured DCs from 12 individual donors were divided into two groups of highly and less differentiated DCs, respectively. A pronounced difference at the level of miRNA induction between these two groups was observed, suggesting that quantitative evaluation of selected miRNAs potentially can predict the immunogenicity of DC vaccines.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Dendritic Cells/immunology , MicroRNAs/analysis , Neoplasms/immunology , Neoplasms/therapy , Antigens, CD/analysis , Antigens, CD/genetics , Cell Differentiation , Cells, Cultured , Dendritic Cells/chemistry , Dendritic Cells/cytology , Flow Cytometry , Gene Expression Regulation , Genetic Markers , Humans , Immunoglobulins/analysis , Immunoglobulins/genetics , Immunotherapy , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , MicroRNAs/genetics , Neoplasms/genetics , Receptors, CCR7/analysis , Receptors, CCR7/genetics , CD83 Antigen
16.
Clin Cancer Res ; 15(24): 7726-7736, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19996212

ABSTRACT

PURPOSE: We evaluated the clinical benefit of an allogeneic melanoma cell lysate (MCL)-pulsed autologous dendritic cell (DC) vaccine in advanced colorectal cancer patients expressing at least one of six MAGE-A antigens overexpressed by the cell line source of the lysate. EXPERIMENTAL DESIGN: DCs were cultured from peripheral blood mononuclear cells (PBMC), pulsed with the allogeneic MCL, and matured using cytokines that achieved high CD83- and CCR7-expressing DCs. Each patient received up to 10 intradermal vaccinations (3-5 x 10(6) cells per dose) at biweekly intervals. RESULTS: Twenty patients received a total of 161 vaccinations. Treatment was well tolerated and quality of life measurements did not vary much across time. One patient experienced partial response [5%; 95% confidence interval (CI), 1-24%] and seven achieved stable disease (35%; 95% CI, 18-57%), one of whom also achieved late tumor regression, yielding a clinical benefit response rate of 40% (95% CI, 22-61%). Although overall median progression-free survival was 2.4 months (95% CI, 1.9-4.1 months), five patients (25%) experienced prolonged progression-free survival (>6 months), two of whom (10%) remain progression-free for >27 and >37 months, respectively. This result is particularly meaningful as all patients had progressive disease before treatment. Overall, DC vaccination was associated with a serial decline in regulatory T cells. Using an antibody array, we characterized plasma protein profiles in responding patients that may correlate with vaccine activity and report a prevaccination protein signature distinguishing responders from nonresponders. CONCLUSION: This phase II vaccine study using mature, MCL-pulsed DCs has shown promising results and warrants further evaluation in a prospective randomized setting. (Clin Cancer Res 2009;15(24):7726-36).

17.
Cancer Immun ; 9: 9, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19813699

ABSTRACT

Tumor antigens are the primary target of therapeutic cancer vaccines. We set out to define and compare the expression pattern of tumor antigen genes in esophagus carcinoma biopsies and in an allogeneic tumor lysate-based cancer vaccine, MelCancerVac. Cells used for vaccine production were treated with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-aza-CdR) to determine whether this treatment could improve the profile of tumor antigen genes expressed in these cells. In addition, the presence of MAGE-A tumor antigen protein was evaluated in the purified tumor cell lysate used in the production of the vaccine. Quantitative PCR was used to assay 74 tumor antigen genes in patients with squamous cell carcinoma of the esophagus. 81% (13/16) of tumors expressed more than five cancer/testis (CT) antigens. A total of 96 genes were assayed in the tumor cell clone (DDM1.7) used to make tumor cell lysate for vaccine preparation. Gene expression in DDM1.7 cells was compared with three normal tissues; 16 tumor antigen genes were induced more than ten-fold relative to normal tissues. Treatment with 5-aza-CdR induced expression of an additional 15 tumor antigens to a total of 31. MAGE-A protein was detected in cell lysate by Western blot at an estimated concentration of 0.2 micrograms/ml or 0.01% of the total protein.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Gene Expression Profiling , Neoplasm Proteins/genetics , Aged , Antigens, Neoplasm/immunology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Clone Cells , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Epithelial Cells/immunology , Epithelial Cells/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Female , Humans , Male , Middle Aged , Neoplasm Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction
18.
J Transl Med ; 6: 81, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19105846

ABSTRACT

The International Society for the Biological Therapy of Cancer (iSBTc) has initiated in collaboration with the United States Food and Drug Administration (FDA) a programmatic look at innovative avenues for the identification of relevant parameters to assist clinical and basic scientists who study the natural course of host/tumor interactions or their response to immune manipulation. The task force has two primary goals: 1) identify best practices of standardized and validated immune monitoring procedures and assays to promote inter-trial comparisons and 2) develop strategies for the identification of novel biomarkers that may enhance our understating of principles governing human cancer immune biology and, consequently, implement their clinical application. Two working groups were created that will report the developed best practices at an NCI/FDA/iSBTc sponsored workshop tied to the annual meeting of the iSBTc to be held in Washington DC in the Fall of 2009. This foreword provides an overview of the task force and invites feedback from readers that might be incorporated in the discussions and in the final document.


Subject(s)
Biomarkers , Immunotherapy , Research , Clinical Trials as Topic , Education , Humans , Neoplasms/diagnosis , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/physiopathology , Reproducibility of Results , Research/economics , Research Design , United States , United States Food and Drug Administration
19.
Oncol Rep ; 20(6): 1305-11, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020707

ABSTRACT

Patients with disseminated colorectal cancer have a poor prognosis. Preliminary studies have shown encouraging results from vaccines based on dendritic cells. The aim of this phase II study was to evaluate the effect of treating patients with advanced colorectal cancer with a cancer vaccine based on dendritic cells pulsed with an allogenic tumor cell lysate. Twenty patients with advanced colorectal cancer were consecutively enrolled. Dendritic cells (DC) were generated from autologous peripheral blood mononuclear cells and pulsed with allogenic tumor cell lysate containing high levels of cancer-testis antigens. Vaccines were biweekly administered intradermally with a total of 10 vaccines per patient. CT scans were performed and responses were graded according to the RECIST criteria. Quality of life was monitored with the SF-36 questionnaire. Toxicity and adverse events were graded according to the National Cancer Institute's common Toxicity Criteria. Four patients were graded with stable disease. Two remained stable throughout the entire study period. Analysis of changes in the patients' quality of life revealed stability in the subgroups: 'physical function' (p=0.872), 'physical role limitation' (p=0.965), 'bodily pain' (p=0.079), 'social function' (p=0.649), 'emotional role limitation' (p=0.252) and 'mental health' (p=0.626). The median survival from inclusion was 5.3 months (range 0.2-29.2 months) with one patient still being alive almost 30 months after inclusion in the trial. Treatment with this DC-based cancer vaccine was safe and non-toxic. Stable disease was found in 24% (4/17) of the patients. The quality of life remained for most categories high and stable throughout the study period.


Subject(s)
Cancer Vaccines , Colorectal Neoplasms/therapy , Dendritic Cells/metabolism , Adult , Aged , Antigens, Neoplasm/metabolism , Biopsy , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Quality of Life , Surveys and Questionnaires , Time Factors
20.
Vaccine ; 26(36): 4716-24, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18616968

ABSTRACT

We have previously established a cancer vaccine using autologous DCs, generated by in vitro stimulation with IL-4 and GM-CSF, and pulsed with six HLA-A*0201 binding wild-type p53 derived peptides. This vaccine was used in combination with low-dose interleukin-2 in a recently published clinical Phase II trial where 26 HLA-A2+ patients with progressive late-stage metastatic breast cancer (BC) were included. Almost 1/3rd of the patients obtained stable disease or minor regression during treatment with a positive correlation to tumour over-expression of p53. In the present study, we performed a comprehensive analysis of the effector stage of the p53-specific CD8+ T cells by the use of Dextramer Technology and multicolour FACS. Pre- and post-treatment blood samples from eight BC patients were analysed. Independent of clinical outcome p53-specific T cells were phenotypic distinctly antigen experienced (CD44high, CCR-7low and CD62Llow). Furthermore, fresh blood from 18 cancer patients included in the vaccination trial were prospectively examined for more general treatment associated quantitative and qualitative changes in T cell subpopulations. We found that the frequency of CD4+ CD25high regulatory T cells was almost doubled after only 4 weeks of weekly vaccination and low-dose IL-2. In addition, a decrease in the percentage of CD27highCCR-7high CD4/CD8 naïve T cells was measured particularly in patients with progressive disease during vaccination. Finally, prior to immunotherapy a higher percentage of both CD28 and CD27 positive CD8naïve/early effector memory T cells were present in chemotherapy-treated patients.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/immunology , Dendritic Cells/immunology , Interleukin-2/administration & dosage , Lymphocyte Subsets/immunology , Tumor Suppressor Protein p53/immunology , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...