Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 15: 1385775, 2024.
Article in English | MEDLINE | ID: mdl-38572241

ABSTRACT

HIV-1 gp120 glycan binding to C-type lectin adhesion receptor L-selectin/CD62L on CD4 T cells facilitates viral attachment and entry. Paradoxically, the adhesion receptor impedes HIV-1 budding from infected T cells and the viral release requires the shedding of CD62L. To systematically investigate CD62L-shedding mediated viral release and its potential inhibition, we screened compounds specific for serine-, cysteine-, aspartyl-, and Zn-dependent proteases for CD62L shedding inhibition and found that a subclass of Zn-metalloproteinase inhibitors, including BB-94, TAPI, prinomastat, GM6001, and GI25423X, suppressed CD62L shedding. Their inhibition of HIV-1 infections correlated with enzymatic suppression of both ADAM10 and 17 activities and expressions of these ADAMs were transiently induced during the viral infection. These metalloproteinase inhibitors are distinct from the current antiretroviral drug compounds. Using immunogold labeling of CD62L, we observed association between budding HIV-1 virions and CD62L by transmission electron microscope, and the extent of CD62L-tethering of budding virions increased when the receptor shedding is inhibited. Finally, these CD62L shedding inhibitors suppressed the release of HIV-1 virions by CD4 T cells of infected individuals and their virion release inhibitions correlated with their CD62L shedding inhibitions. Our finding reveals a new therapeutic approach targeted at HIV-1 viral release.

2.
Nat Commun ; 14(1): 6380, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37821447

ABSTRACT

Severe COVID-associated lung injury is a major confounding factor of hospitalizations and death with no effective treatments. Here, we describe a non-classical fibrin clotting mechanism mediated by SARS-CoV-2 infected primary lung but not other susceptible epithelial cells. This infection-induced fibrin formation is observed in all variants of SARS-CoV-2 infections, and requires thrombin but is independent of tissue factor and other classical plasma coagulation factors. While prothrombin and fibrinogen levels are elevated in acute COVID BALF samples, fibrin clotting occurs only with the presence of viral infected but not uninfected lung epithelial cells. We suggest a viral-induced coagulation mechanism, in which prothrombin is activated by infection-induced transmembrane serine proteases, such as ST14 and TMPRSS11D, on NHBE cells. Our finding reveals the inefficiency of current plasma targeted anticoagulation therapy and suggests the need to develop a viral-induced ARDS animal model for treating respiratory airways with thrombin inhibitors.


Subject(s)
COVID-19 , Animals , Humans , SARS-CoV-2 , Thrombin , Prothrombin , Lung , Epithelial Cells , Fibrin
3.
PLoS One ; 18(2): e0281087, 2023.
Article in English | MEDLINE | ID: mdl-36780482

ABSTRACT

HIV infection remains incurable to date and there are no compounds targeted at the viral release. We show here HIV viral release is not spontaneous, rather requires caspases activation and shedding of its adhesion receptor, CD62L. Blocking the caspases activation caused virion tethering by CD62L and the release of deficient viruses. Not only productive experimental HIV infections require caspases activation for viral release, HIV release from both viremic and aviremic patient-derived CD4 T cells also require caspase activation, suggesting HIV release from cellular viral reservoirs depends on apoptotic shedding of the adhesion receptor. Further transcriptomic analysis of HIV infected CD4 T cells showed a direct contribution of HIV accessory gene Nef to apoptotic caspases activation. Current HIV cure focuses on the elimination of latent cellular HIV reservoirs that are resistant to infection-induced cell death. This has led to therapeutic strategies to stimulate T cell apoptosis in a "kick and kill" approach. Our current work has shifted the paradigm on HIV-induced apoptosis and suggests such approach would risk to induce HIV release and thus be counter-productive. Instead, our study supports targeting of viral reservoir release by inhibiting of caspases activation.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , nef Gene Products, Human Immunodeficiency Virus , Humans , Caspases/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Death , HIV Infections/drug therapy , HIV-1/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism
4.
Front Microbiol ; 12: 725741, 2021.
Article in English | MEDLINE | ID: mdl-34659153

ABSTRACT

HIV envelope glycoprotein is the most heavily glycosylated viral protein complex identified with over 20 glycans on its surface. This glycan canopy is thought to primarily shield the virus from host immune recognition as glycans are poor immunogens in general, however rare HIV neutralizing antibodies nevertheless potently recognize the glycan epitopes. While CD4 and chemokine receptors have been known as viral entry receptor and coreceptor, for many years the role of viral glycans in HIV entry was controversial. Recently, we showed that HIV envelope glycan binds to L-selectin in solution and on CD4 T lymphocytes. The viral glycan and L-selectin interaction functions to facilitate the viral adhesion and entry. Upon entry, infected CD4 T lymphocytes are stimulated to progressively shed L-selectin and suppressing this lectin receptor shedding greatly reduced HIV viral release and caused aggregation of diminutive virus-like particles within experimental infections and from infected primary T lymphocytes derived from both viremic and aviremic individuals. As shedding of L-selectin is mediated by ADAM metalloproteinases downstream of host-cell stimulation, these findings showed a novel mechanism for HIV viral release and offer a potential new class of anti-HIV compounds.

5.
Nat Commun ; 9(1): 2825, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30026537

ABSTRACT

CD4 and chemokine receptors mediate HIV-1 attachment and entry. They are, however, insufficient to explain the preferential viral infection of central memory T cells. Here, we identify L-selectin (CD62L) as a viral adhesion receptor on CD4+ T cells. The binding of viral envelope glycans to L-selectin facilitates HIV entry and infection, and L-selectin expression on central memory CD4+ T cells supports their preferential infection by HIV. Upon infection, the virus downregulates L-selectin expression through shedding, resulting in an apparent loss of central memory CD4+ T cells. Infected effector memory CD4+ T cells, however, remain competent in cytokine production. Surprisingly, inhibition of L-selectin shedding markedly reduces HIV-1 infection and suppresses viral release, suggesting that L-selectin shedding is required for HIV-1 release. These findings highlight a critical role for cell surface sheddase in HIV-1 pathogenesis and reveal new antiretroviral strategies based on small molecular inhibitors targeted at metalloproteinases for viral release.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV-1/immunology , Host-Pathogen Interactions , L-Selectin/genetics , Receptors, Virus/genetics , Virus Shedding/immunology , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/genetics , ADAM17 Protein/immunology , Animals , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Dipeptides/pharmacology , HEK293 Cells , HIV Core Protein p24/antagonists & inhibitors , HIV Core Protein p24/genetics , HIV Core Protein p24/immunology , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV-1/drug effects , HIV-1/growth & development , HeLa Cells , Humans , Hydroxamic Acids/pharmacology , Immunologic Memory/drug effects , L-Selectin/antagonists & inhibitors , L-Selectin/immunology , Lymphocyte Activation/drug effects , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Primary Cell Culture , Protease Inhibitors/pharmacology , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/immunology , Thiophenes/pharmacology , Virus Attachment/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects , Virus Replication/immunology , Virus Shedding/drug effects
6.
Protein Expr Purif ; 140: 65-73, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28842197

ABSTRACT

L-selectin (CD62L) is an extracellular protein with a lectin-like domain that mediates rolling adhesion of leukocytes to vascular endothelial cell surfaces. Currently, there are no solved structures for the ectodomain of CD62L, nor of CD62L in complex with its ligand. We have developed a rapid mammalian recombinant protein expression system using an amplifiable glutamine synthase based vector. Here, we further developed and applied this method to express and purify the entire extracellular region of CD62L. This resulted in excess of 20 mg/L yield of recombinant CD62L. In an attempt to understand the different expression levels among four similar CD62L constructs that differ primarily in signal sequences, we calculated the presence of potential RNA pseudoknots in their signal sequences. The results showed the presence of pseudoknots involving the start codon and between the signal sequence and gene in the mRNA of the non-expressing constructs, suggesting a potential inhibitory role of RNA pseudoknots in recombinant protein expression.


Subject(s)
L-Selectin/chemistry , RNA/chemistry , Recombinant Proteins/chemistry , Flow Cytometry , Humans , L-Selectin/biosynthesis , L-Selectin/genetics , Ligands , Nucleic Acid Conformation , Protein Domains/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
7.
Protein Expr Purif ; 105: 8-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25286402

ABSTRACT

We have previously developed a glutamine synthetase (GS)-based mammalian recombinant protein expression system that is capable of producing 5-30mg/L recombinant proteins. The over expression is based on multiple rounds of target gene amplification driven by methionine sulfoximine (MSX), an inhibitor of glutamine synthetase. However, like other stable mammalian over expression systems, a major shortcoming of the GS-based expression system is its lengthy turn-around time, typically taking 4-6months to produce. To shorten the construction time, we replaced the multi-round target gene amplifications with single-round in situ amplifications, thereby shortening the cell line construction to 2months. The single-round in situ amplification method resulted in highest recombinant CD62L expressing CHO cell lines producing ∼5mg/L soluble CD62L, similar to those derived from the multi-round amplification and selection method. In addition, we developed a MSX resistance assay as an alternative to utilizing ELISA for evaluating the expression level of stable recombinant CHO cell lines.


Subject(s)
Glutamate-Ammonia Ligase/chemistry , L-Selectin/isolation & purification , L-Selectin/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Glutamate-Ammonia Ligase/metabolism , HEK293 Cells , Humans , L-Selectin/genetics , Methionine Sulfoximine , Mutation/genetics , Recombinant Proteins/genetics
8.
Blood ; 120(18): 3635-46, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22968453

ABSTRACT

We conducted a gene therapy trial in 10 patients with adenosine deaminase (ADA)-deficient severe combined immunodeficiency using 2 slightly different retroviral vectors for the transduction of patients' bone marrow CD34(+) cells. Four subjects were treated without pretransplantation cytoreduction and remained on ADA enzyme-replacement therapy (ERT) throughout the procedure. Only transient (months), low-level (< 0.01%) gene marking was observed in PBMCs of 2 older subjects (15 and 20 years of age), whereas some gene marking of PBMC has persisted for the past 9 years in 2 younger subjects (4 and 6 years). Six additional subjects were treated using the same gene transfer protocol, but after withdrawal of ERT and administration of low-dose busulfan (65-90 mg/m(2)). Three of these remain well, off ERT (5, 4, and 3 years postprocedure), with gene marking in PBMC of 1%-10%, and ADA enzyme expression in PBMC near or in the normal range. Two subjects were restarted on ERT because of poor gene marking and immune recovery, and one had a subsequent allogeneic hematopoietic stem cell transplantation. These studies directly demonstrate the importance of providing nonmyeloablative pretransplantation conditioning to achieve therapeutic benefits with gene therapy for ADA-deficient severe combined immunodeficiency.


Subject(s)
Agammaglobulinemia/therapy , Bone Marrow Transplantation/methods , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation/methods , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Adolescent , Antigens, CD34/metabolism , Child , Child, Preschool , Female , Humans , Infant , Male , Retroviridae/genetics , Transduction, Genetic , Transplantation Conditioning , Young Adult
9.
Neuro Oncol ; 13(6): 591-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21636707

ABSTRACT

To assess the accumulation of myeloid-derived suppressor cells (MDSCs) in the peripheral blood of patients with glioma and to define their heterogeneity and their immunosuppressive function. Peripheral blood mononuclear cells (PBMCs) from healthy control subjects and from patients with newly diagnosed glioma were stimulated with anti-CD3/anti-CD28 and T cells assessed for intracellular expression of interferon (IFN)-γ. Antibody staining of PBMCs from glioma patients and healthy donors (CD33, HLADR, CD15, and CD14) followed by 4-color flow cytometry analysis-defined MDSC levels in the peripheral blood. To assess the role of MDSCs in suppressing T cell IFNγ production, PBMCs were depleted of MDSCs using anti-CD33 and anti-CD15 antibody-coated beads prior to T cell stimulation. Enzyme-linked immunosorbent assays were used to assess plasma arginase activity and the level of granulocyte colony-stimulating factor (G-CSF). Patients with glioblastoma have increased MDSC counts (CD33+HLADR-) in their blood that are composed of neutrophilic (CD15(+); >60%), lineage-negative (CD15(-)CD14(-); 31%), and monocytic (CD14(+); 6%) subsets. After stimulation, T cells from patients with glioblastoma had suppressed IFN-γ production when compared with healthy, age-matched donor T cells. Removal of MDSCs from the PBMCs with anti-CD33/CD15-coated beads significantly restored T cell function. Significant increases in arginase activity and G-CSF levels were observed in plasma specimens obtained from patients with glioblastoma. The accumulation of MDSCs in peripheral blood in patients with glioma likely promotes T cell immune suppression that is observed in this patient population. Increased plasma levels of arginase and G-CSF may relate to MDSC suppressor function and MDSC expansion, respectively, in patients with glioma.


Subject(s)
Glioblastoma/diagnosis , Glioblastoma/immunology , Leukocytes, Mononuclear/immunology , Myeloid Cells/immunology , Myeloid Cells/pathology , T-Lymphocytes/immunology , Arginase/metabolism , Brain Neoplasms/blood , Brain Neoplasms/diagnosis , Brain Neoplasms/immunology , Case-Control Studies , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioblastoma/blood , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Middle Aged
10.
J Clin Immunol ; 31(4): 690-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21487892

ABSTRACT

INTRODUCTION: Cycloxygenase-2 (COX-2) is an enzyme involved in prostaglandin E2 (PGE(2)) synthesis associated with higher renal cell carcinoma stage. COX-2 inhibition enhances interferon (IFN-α) anti-tumor immune effects in pre-clinical models. A phase II trial of celecoxib and IFN-α in a targeted population of metastatic renal cell carcinoma patients with maximal COX-2 expression was conducted. METHODS: Cytokine-naive metastatic renal cell carcinoma patients with tumors expressing ≥10% maximal COX-2 staining by immunohistochemistry received IFN-α 5 million units daily and celecoxib 400 mg orally twice daily in an open-label, single-arm phase II trial. RESULTS: There were 3 partial responses among 17 patients (objective response rate 18%; 95% confidence interval, 4-43%). Time to progression was 5.6 months. Increased tumor staining 3+ for COX-2 was associated with increased baseline peripheral blood PGE(2) levels, and these patients demonstrated less PGE(2) decrease with therapy. Patients with more 3+ COX-2 staining had significantly more CD3(+) (p = 0.004) and CD4(+) (p = 0.002) IFN-γ T cells at baseline and a significantly greater decrease in these cells with therapy. DISCUSSION: Celecoxib plus IFN-α in renal cell carcinoma (RCC) patients with maximally staining COX-2 tumors does not significantly enhance overall response rates over IFN monotherapy. CONCLUSION: COX-2-expressing RCC demonstrates inherent immunosuppression. COX-2 inhibition with IFN results in minimal immunomodulation and no augmented clinical activity in RCC.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Cyclooxygenase 2/biosynthesis , Interferon-alpha/therapeutic use , Kidney Neoplasms/drug therapy , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Adult , Aged , Carcinoma, Renal Cell/immunology , Celecoxib , Dendritic Cells/drug effects , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Drug Therapy, Combination/adverse effects , Female , Humans , Immunologic Factors/therapeutic use , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Kidney Neoplasms/pathology , Male , Middle Aged , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Th1 Cells/immunology , Th1-Th2 Balance , Th2 Cells/immunology , Treatment Outcome
11.
Int Immunopharmacol ; 11(7): 856-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21315783

ABSTRACT

Sunitinib is a receptor tyrosine kinase inhibitor (TKI) that is front-line therapy for metastatic renal cell carcinoma (mRCC). Its antitumor activity is related to its ability to block tumor cell and tumor vasculature cell signaling via several TKI receptors (i.e. vascular endothelial growth factor receptors VEGFRs, platelet-derived growth factors (PDGFs), and stem cell factors). Sunitinib also targets myeloid derived suppressor cells (MDSCs) significantly reducing their accumulation in the peripheral blood and reversing T cell (IFNγ) suppression in both mRCC patients and in murine tumor models. This reduction in immune suppression provides a rationale for combining sunitinib with immunotherapy for the treatment of certain tumor types. Despite these encouraging findings, however, we have observed that sunitinib has variable impact at reducing MDSCs and restoring T cell function within the tumor microenvironment. Given the immunosuppressive and proangiogenic activities of MDSC, it seems plausible that their persistence may contribute to the resistance that develops in sunitinib-treated patients. While sunitinib reduced tumor infiltrating MDSCs in Renca and CT26-bearing mice, coinciding with strong to modest decreases in tumor size respectively, it was ineffective at reducing MDSCs (<35% reduction in Gr1+CD11b+) or tumor burden in 4T1-bearing mice. Persistence of intratumor MDSCs was paralleled by depressed intratumor T cell IFNγ response and increased GM-CSF expression. Additionally, in vitro and in vivo experiments showed that GM-CSF prolongs survival of MDSCs, thus protecting them from the effects of sunitinib via a pSTAT5-dependent pathway. Although preliminary, there is evidence of intratumor MDSC resistance in some mRCC patients following sunitinib treatment. Intratumor MDSC persistence and T cell IFNγ response post nephrectomy in patients receiving sunitinib in a neoadjuvant setting are being compared to RCC patients undergoing nephrectomy without prior sunitinib treatment. Tumors from untreated patients showed suppressed T cell IFNγ response along with substantial expression of MDSCs (5% of total digested cells). Thus far, tumors from 5/8 neoadjuvant patients showed persistence of intratumor MDSCs and low T cell IFNγ production post sunitinib treatment, findings that parallel results from untreated tumors. In the remaining 3 neoadjuvant patients, intratumor MDSCs were detected at low levels which coincided with a T cell IFNγ response similar to that observed with normal donor peripheral T cells. GM-CSF's role in promoting MDSC survival in patient tumors is supported by the observation that GM-CSF is produced in short-term RCC cultures at levels capable of protecting MDSCs from sunitinib-induced cell death. Additionally, persistence of MDSC also may be associated with increased expression of proangiogenic proteins, such as MMP9, MMP8, and IL-8 produced by tumor stromal cells or infiltrating MDSCs. Indeed our findings suggest that the most dominate MDSC subset in RCC patients is the neutrophilic population that produces proangiogenic proteins. We propose that the development of sunitinib resistance is partly mediated by the survival of MDSCs intratumorally, thereby providing sustained immune suppression and angiogenesis.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Carcinoma, Renal Cell/immunology , Indoles/administration & dosage , Kidney Neoplasms/immunology , Myeloid Cells/metabolism , Pyrroles/administration & dosage , Animals , Antigens, Ly/biosynthesis , CD11b Antigen/biosynthesis , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/physiopathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Immunosuppression Therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/physiopathology , Kidney Neoplasms/therapy , Mice , Mice, Inbred BALB C , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoadjuvant Therapy , Neoplasm Metastasis , Sunitinib , Tumor Burden/drug effects , Tumor Escape
12.
Cancer Res ; 70(9): 3526-36, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20406969

ABSTRACT

The antiangiogenic drug sunitinib is a receptor tyrosine kinase inhibitor with significant, yet not curative, therapeutic effects in metastatic renal cell carcinoma (RCC). Sunitinib is also an immunomodulator, potently reversing myeloid-derived suppressor cell (MDSC) accumulation and T-cell inhibition in the blood even of nonresponder RCC patients. We observed that sunitinib similarly prevented MDSC accumulation and restored normal T-cell function to the spleens of tumor-bearing mice, independent of the capacity of sunitinib to inhibit tumor progression (RENCA>CT26>4T1). Both monocytic and neutrophilic splenic MDSC were highly repressible by sunitinib. In contrast, MDSC within the microenvironment of 4T1 tumors or human RCC tumors proved highly resistant to sunitinib and ambient T-cell function remained suppressed. Proteomic analyses comparing tumor to peripheral compartments showed that granulocyte macrophage colony-stimulating factor (GM-CSF) predicted sunitinib resistance and recombinant GM-CSF conferred sunitinib resistance to MDSC in vivo and in vitro. MDSC conditioning with GM-CSF uniquely inhibited signal transducers and activators of transcription (STAT3) and promoted STAT5 activation. STAT5ab(null/null) MDSC were rendered sensitive to sunitinib in the presence of GM-CSF in vitro. We conclude that compartment-dependent GM-CSF exposure in resistant tumors may account for the regionalized effect of sunitinib upon host MDSC modulation and hypothesize that ancillary strategies to decrease such regionalized escape will enhance the potency of sunitinib as an immunomodulator and a cancer therapy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Renal Cell/immunology , Immunologic Factors/pharmacology , Indoles/pharmacology , Kidney Neoplasms/immunology , Myeloid Cells/drug effects , Pyrroles/pharmacology , T-Lymphocyte Subsets/drug effects , Animals , CD11b Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/drug therapy , Female , Humans , Kidney Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Myeloid Cells/immunology , Sunitinib , Suppressor Factors, Immunologic/immunology , T-Lymphocyte Subsets/immunology
13.
Urology ; 75(2): 257-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19963243

ABSTRACT

OBJECTIVES: To validate enzyme-linked immunosorbent assay (ELISA) as an accurate and objective method to measure carbonic anhydrase IX (CAIX) expression in RCC tissue specimens and to also investigate the diagnostic and prognostic utility of serum CAIX levels. Recent studies found protein levels of CAIX, quantified using immunohistochemistry, correlated with clinical outcomes and therapeutic response to immunotherapy in advanced stage clear cell renal cell carcinoma (CCRCC). MATERIALS AND METHODS: A CAIX ELISA kit was used to measure the CAIX levels in the serum and tissue of 21 CCRCCs and 19 non-CCRCCs. CAIX immunohistochemical stains were performed on the corresponding formalin fixed, paraffin embedded tumor tissues. RESULTS: Tissue CAIX levels measured by ELISA highly correlated with the CAIX levels detected by immunohistochemistry (Pearson correlation coefficient = .802, P <.001). The CCRCC tumor tissue had significantly higher CAIX levels than non-CCRCC tissue (77 023 vs 938 pg/mL, P <.001) detected by ELISA. Serum CAIX levels in CCRCC patients were significantly higher than in non-CCRCC patients (126.1 vs 2.1 pg/mL, P = .013). The serum CAIX levels detected by ELISA correlated with the tumor size in CCRCC patients (Pearson correlation coefficient = .498, P = .036). CONCLUSIONS: ELISA provides a quantitative and objective method to measure CAIX levels in renal tumors. The tissue CAIX levels measured with ELISA highly correlate with the results obtained with the standard immunohistochemistry. Serum CAIX levels are significantly higher in CCRCC than in non-CCRCC and may aid the differential diagnosis of RCC. Serum CAIX levels also correlate with the tumor size in CCRCC patients.


Subject(s)
Antigens, Neoplasm/analysis , Antigens, Neoplasm/biosynthesis , Carbonic Anhydrases/analysis , Carbonic Anhydrases/biosynthesis , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/enzymology , Enzyme-Linked Immunosorbent Assay , Kidney Neoplasms/diagnosis , Kidney Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/blood , Carbonic Anhydrase IX , Carbonic Anhydrases/blood , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/chemistry , Diagnosis, Differential , Female , Humans , Kidney Neoplasms/blood , Kidney Neoplasms/chemistry , Male , Middle Aged , Prognosis , Young Adult
14.
Clin Cancer Res ; 15(6): 2148-57, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276286

ABSTRACT

PURPOSE: Immune dysfunction reported in renal cell carcinoma (RCC) patients may contribute to tumor progression. Myeloid-derived suppressor cells (MDSC) represent one mechanism by which tumors induce T-cell suppression. Several factors pivotal to the accumulation of MDSC are targeted by the tyrosine kinase inhibitor, sunitinib. The effect of sunitinib on MDSC-mediated immunosuppression in RCC patients has been investigated. EXPERIMENTAL DESIGN: Patient peripheral blood levels of MDSC and regulatory T-cell (Treg) and T-cell production of IFN-gamma were evaluated before and after sunitinib treatment. Correlations between MDSC and Treg normalization as well as T-cell production of IFN-gamma were examined. The in vitro effect of sunitinib on patient MDSC was evaluated. RESULTS: Metastatic RCC patients had elevated levels of CD33(+)HLA-DR(-) and CD15(+)CD14(-) MDSC, and these were partially overlapping populations. Treatment with sunitinib resulted in significant reduction in MDSC measured by several criteria. Sunitinib-mediated reduction in MDSC was correlated with reversal of type 1 T-cell suppression, an effect that could be reproduced by the depletion of MDSC in vitro. MDSC reduction in response to sunitinib correlated with a reversal of CD3(+)CD4(+)CD25(hi)Foxp3(+) Treg cell elevation. No correlation existed between a change in tumor burden and a change in MDSC, Treg, or T-cell production of IFN-gamma. In vitro addition of sunitinib reduced MDSC viability and suppressive effect when used at >/=1.0 microg/mL. Sunitinib did not induce MDSC maturation in vitro. CONCLUSIONS: Sunitinib-based therapy has the potential to modulate antitumor immunity by reversing MDSC-mediated tumor-induced immunosuppression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Myeloid Cells/immunology , Pyrroles/pharmacology , Suppressor Factors, Immunologic/drug effects , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/immunology , Female , Humans , Indoles/therapeutic use , Interferon-gamma/biosynthesis , Kidney Neoplasms/immunology , Male , Middle Aged , Pyrroles/therapeutic use , Sunitinib , Suppressor Factors, Immunologic/physiology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology
15.
Clin Cancer Res ; 14(20): 6674-82, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18927310

ABSTRACT

PURPOSE: Immune dysfunction is well documented in renal cell carcinoma (RCC) patients and likely contributes to tumor evasion. This dysfunction includes a shift from a type-1 to a type-2 T-cell cytokine response and enhanced T-regulatory (Treg) cell expression. Given the antitumor activity of select tyrosine kinase inhibitors such as sunitinib in metastatic RCC (mRCC) patients, it is relevant to assess their effect on the immune system. EXPERIMENTAL DESIGN: Type-1 (IFNgamma) and type-2 (interleukin-4) responses were assessed in T cells at baseline and day 28 of treatment with sunitinib (50 mg/d) by measuring intracellular cytokines after in vitro stimulation with anti-CD3/anti-CD28 antibodies. RESULTS: After one cycle of treatment, there was a significant increase in the percentage of IFNgamma-producing T cells (CD3(+), P < 0.001; CD3(+)CD4(+), P = 0.001), a reduction in interleukin-4 production (CD3(+) cells, P = 0.05), and a diminished type-2 bias (P = 0.005). The increase in type-1 response may be partly related to modulation of Treg cells. The increased percentage of Treg cells noted in mRCC patients over healthy donors (P = 0.001) was reduced after treatment, although not reaching statistical significance. There was, however, an inverse correlation between the increase in type-1 response after two cycles of treatment and a decrease in the percentage of Treg cells (r = -0.64, P = 0.01). In vitro studies suggest that the effects of sunitinib on Treg cells are indirect. CONCLUSIONS: The demonstration that sunitinib improved type-1 T-cell cytokine response in mRCC patients while reducing Treg function provides a basis for the rational combination of sunitinib and immunotherapy in mRCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Immunosuppression Therapy , Indoles/therapeutic use , Pyrroles/therapeutic use , T-Lymphocytes, Regulatory/drug effects , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Renal Cell/metabolism , Case-Control Studies , Female , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Kidney/drug effects , Kidney/immunology , Kidney/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Male , Middle Aged , Recombinant Proteins , Sunitinib , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
16.
J Immunother ; 30(6): 634-43, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17667527

ABSTRACT

The administration of dendritic cell vaccines is a promising approach for cancer immunotherapy. Langerhans cells (LCs) that are genetically modified to express viral or tumor antigens are a dendritic cell subset of particular interest because they elicit potent antigen-specific immune responses. For clinical investigation, transduced, functional LCs must be generated in sufficient numbers using a scalable closed system that conforms to current good manufacturing practices. We therefore developed a process to expand CD34+ hematopoietic progenitor cell-derived LCs in serum-free medium using hydrophobic culture bags and compared their biologic function to that of LCs grown in plates or flasks. We obtained significantly higher yields of mature LCs in bags compared with plates or flasks. LCs grown in bags displayed comparable maturation phenotypes and were transduced by GaLV-pseudotyped retroviral vectors with the same efficiency as LCs grown in plates or flasks. Bag-expanded LCs effectively stimulated the proliferation of allogeneic T lymphocytes and the production of interferon-gamma by autologous CD8 T cells against the viral influenza matrix peptide or human tyrosinase. We have thus developed a scalable closed process to expand genetically modified, biologically functional CD34+ hematopoietic progenitor cell-derived LCs for phase I clinical trials.


Subject(s)
Cell Culture Techniques , Cytokines/immunology , Cytokines/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Antigens, CD34/immunology , Cell Proliferation , Cryopreservation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Monophenol Monooxygenase/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic , Viral Matrix Proteins/immunology
17.
Blood ; 109(2): 503-6, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16973956

ABSTRACT

A patient with adenosine deaminase-deficient severe combined immune deficiency (ADA-SCID) was enrolled in a study of retroviral-mediated ADA gene transfer to bone marrow hematopoietic stem cells. After the discontinuation of ADA enzyme replacement, busulfan (75 mg/m2) was administered for bone marrow cytoreduction, followed by infusion of autologous, gene-modified CD34+ cells. The expected myelosuppression developed after busulfan but then persisted, necessitating the administration of untransduced autologous bone marrow back-up at day 40. Because of sustained pancytopenia and negligible gene marking, diagnostic bone marrow biopsy and aspirate were performed at day 88. Analyses revealed hypocellular marrow and, unexpectedly, evidence of trisomy 8 in 21.6% of cells. Trisomy 8 mosaicism (T8M) was subsequently diagnosed by retrospective analysis of a pretreatment marrow sample that might have caused the lack of hematopoietic reconstitution. The confounding effects of this preexisting marrow cytogenetic abnormality on the response to gene transfer highlights another challenge of gene therapy with the use of autologous hematopoietic stem cells.


Subject(s)
Adenosine Deaminase/deficiency , Chromosomes, Human, Pair 8/genetics , Genetic Therapy , Mosaicism , Pancytopenia/therapy , Severe Combined Immunodeficiency/therapy , Trisomy , Adenosine Deaminase/genetics , Child, Preschool , Cytogenetic Analysis , Female , Humans , Pancytopenia/etiology , Retrospective Studies , Severe Combined Immunodeficiency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...