Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Oncogene ; 36(28): 4081-4086, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28263973

ABSTRACT

Combining immunotherapy with targeted therapy has increasingly become an appealing therapeutic paradigm for cancer treatment due to its great potential for generating durable and synergistic antitumor response. In this study, however, we unexpectedly found that two types of CpG-based tumor peptide vaccine treatments consistently negated the antitumor activity of a selective BRAF inhibitor in tumors with BRAF mutation rather than showing a synergistic antitumor effect. Our further studies demonstrated that CpG alone was sufficient to dampen BRAF inhibitor-induced antitumor responses, suggesting that the impaired antitumor activity of the BRAF inhibitor observed in mice receiving CpG-based peptide vaccine is mainly dependent upon the use of CpG. Mechanistically, CpG increased the number of circulating B cells, which produced elevated amounts of tumor necrosis factor-α (TNFα) that contributed to the increased tumor resistance to BRAF inhibitors. More importantly, B-cell depletion or TNFα neutralization can restore the antitumor effect of BRAF inhibition in mice receiving CpG treatment, indicating that TNFα-secreting B cells play an indispensable role in BRAF inhibitor resistance induced by CpG. Taken together, our results strongly suggest that precautions must be implemented when designing combinatorial approaches for cancer treatment, because distinct regimens, despite their respective therapeutic benefit as monotherapy, may together provide antagonistic clinical outcomes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes/physiology , Cancer Vaccines/adverse effects , Melanoma/drug therapy , Oligodeoxyribonucleotides/adverse effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , B-Lymphocytes/pathology , Cancer Vaccines/administration & dosage , Drug Antagonism , Female , Immunotherapy/adverse effects , Indoles/administration & dosage , Melanoma/pathology , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage , Sulfonamides/administration & dosage , Toll-Like Receptor 9/agonists , Tumor Cells, Cultured , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , gp100 Melanoma Antigen/administration & dosage , gp100 Melanoma Antigen/adverse effects
3.
Ann Oncol ; 26(5): 1012-1018, 2015 May.
Article in English | MEDLINE | ID: mdl-25669829

ABSTRACT

BACKGROUND: We carried out a phase I trial of the vascular endothelial growth factor inhibitor pazopanib and the histone deacetylase inhibitor vorinostat to determine the safety and efficacy. Because these agents are known to target factors activated by TP53 mutation and facilitate mutant p53 degradation, a subgroup analysis may be interesting in patients with TP53 mutant malignancies. PATIENTS AND METHODS: Patients with advanced solid tumors (n = 78) were enrolled following a 3 + 3 design, with dose expansion for those with responsive tumors. Hotspot TP53 mutations were tested when tumor specimens were available. RESULTS: Adverse events of ≥grade 3 included thrombocytopenia, neutropenia, fatigue, hypertension, diarrhea and vomiting. Overall, the treatment produced stable disease for at least 6 months or partial response (SD ≥6 months/PR) in 19% of the patients, median progression-free survival (PFS) of 2.2 months, and median overall survival (OS) of 8.9 months. In patients with detected hotspot TP53 mutant advanced solid tumors (n = 11), the treatment led to a 45% rate of SD ≥6 months/PR (1 PR and 3 SD ≥6 months), median PFS of 3.5 months, and median OS of 12.7 months, compared favorably with the results for patients with undetected hotspot TP53 mutations (n = 25): 16% (1 PR and 3 SD ≥6 months, P = 0.096), 2.0 months (P = 0.042), and 7.4 months (P = 0.1), respectively. CONCLUSION: The recommended phase II dosage was oral pazopanib at 600 mg daily plus oral vorinostat at 300 mg daily. The preliminary evidence supports further evaluation of the combination in cancer patients with mutated TP53, especially in those with metastatic sarcoma or metastatic colorectal cancer. CLINICAL TRIAL REGISTRATION: www.clinicaltrials.gov, NCT01339871.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Mutation , Neoplasms/drug therapy , Neovascularization, Pathologic , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Tumor Suppressor Protein p53/genetics , Administration, Oral , Adolescent , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease Progression , Disease-Free Survival , Drug Administration Schedule , Female , Histone Deacetylase Inhibitors/adverse effects , Humans , Hydroxamic Acids/adverse effects , Indazoles , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Proportional Hazards Models , Protein Stability , Proteolysis , Pyrimidines/adverse effects , Sulfonamides/adverse effects , Texas , Time Factors , Treatment Outcome , Tumor Suppressor Protein p53/metabolism , Vorinostat , Young Adult
4.
Ann Oncol ; 24(8): 2158-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23576709

ABSTRACT

BACKGROUND: The purpose of the study was to assess the outcome of patients with advanced melanoma treated with matched molecularly targeted therapy. PATIENTS AND METHODS: We reviewed 160 consecutive patients with metastatic melanoma treated in the phase I program (N = 35 protocols). Treatment was considered to be 'matched' (N = 84) if at least one drug in the regimen was known to inhibit the functional activity of at least one of the patient's mutations. RESULTS: Of 160 patients, 134 (83.7%) had adequate tissue for molecular analysis; 69% (110 of 160) had ≥1 mutation: 61.2% (82 of 134), BRAF; 20.7% (23 of 111), NRAS; 2.6% (2 of 77), KIT; 2.3% (1 of 44), KRAS; 20% (1 of 5), GNAQ; 11.1% (1 of 9), P53 and 2.6% (1 of 39), coexisting mutations in BRAF and PIK3CA. Eighty-four patients (52.4%) were treated with matched-targeted agents, most of whom had BRAF mutations (N = 74). Twenty-six percent of patients (41 of 160) achieved a complete or partial remission (CR/PR) [40% (34 of 84)) on a matched phase I protocol versus 9.2% (7 of 76) for those on a non-matched study (P ≤ 0.0001)]. The median progression-free survival (PFS) (95% CI) was longer for patients treated on a matched phase I trial than on their prior first standard treatment [5.27 (4.10, 6.44) versus 3.10 (1.92, 4.28) months, P = 0.023], but not on non-matched phase I treatment. Multivariable analysis showed that matched therapy was an independent predictor of higher CR/PR rates, prolonged PFS and survival. CONCLUSIONS: For melanoma patients, especially those with BRAF mutations, administering molecularly matched agents can be associated with better outcomes, including longer PFS compared with their first-line systemic therapy.


Subject(s)
Melanoma/drug therapy , Melanoma/genetics , Molecular Targeted Therapy , Precision Medicine , Adult , Aged , Aged, 80 and over , Class I Phosphatidylinositol 3-Kinases , Disease-Free Survival , Female , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/genetics , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Humans , Male , Melanoma/mortality , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins p21(ras) , Survival , Treatment Outcome , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Young Adult , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
5.
Vaccine ; 29(18): 3476-82, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21382485

ABSTRACT

Our laboratory previously described the strategy of fusing chemokine receptor ligands to antigens in order to generate immunogenic DNA vaccines. In the present study, we produced mouse ß-2 defensin (mBD2) fusion proteins using both ovalbumin (OVA) and gp100 as model antigens. Superior cross-presentation by dendritic cells (DC) was observed for mBD2 fused antigens over unfused antigens in vitro. In vivo, we observed significant increases in the expansion of adoptively transferred antigen-specific MHC class I, but not class II-restricted T cells after immunization with mBD2 fused antigen over antigen alone. This enhanced expansion of class I restricted T cells was Toll-like receptor 4 (TLR4) dependent, but CC chemokine receptor 6 (CCR6) independent. Superior tumor resistance was observed for mBD2-fusion protein vaccines, compared to unfused antigen, in both B16-OVA and B16 tumor models. These data suggest that production of mBD2 fusion proteins is feasible and that the vaccines facilitate in vivo expansion of adoptively transferred T cells through a TLR4-dependent mechanism.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Toll-Like Receptor 4/immunology , beta-Defensins/immunology , Animals , Antigen-Presenting Cells , Cross-Priming , Interferon-gamma/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Receptors, CCR6/immunology , Recombinant Fusion Proteins/immunology , gp100 Melanoma Antigen/immunology
6.
Int J Infect Dis ; 14(7): e548-52, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20005762

ABSTRACT

High-dose interleukin-2 (HDIL-2) has proven to be an effective treatment for metastatic renal cell carcinoma and melanoma. Previous studies have shown an increase in catheter-related bacteremia (CRB) in patients on HDIL-2. The primary objective of this study was to evaluate the effectiveness of minocycline and rifampin-coated catheters (M/R-C) in reducing CRB in cancer patients on HDIL-2. This was a retrospective study where non-coated catheters (NC-C) and M/R-C were used for the administration of HDIL-2 before and after December 2004, respectively. Data collected included demographics, cancer type, catheter type, antibiotic prophylaxis, and infection rates. A total of 107 episodes of catheter use for HDIL-2 were evaluated in 78 patients (30 episodes in patients with M/R-C vs. 77 with NC-C). A total of nine episodes of CRB were identified, all in patients with NC-C (M/R-C 0% vs. NC-C 12%; p=0.06). The median time to bacteremia was 11 days (range 1-315 days). A log-rank test showed a trend that the M/R-C group had lower probability of getting CRB than the NC-C group (p=0.06). The use of M/R-C in patients on HDIL-2 therapy for advanced melanoma and renal cell carcinoma may have reduced the risk of CRB to nil. CRB still occurred despite antibiotic prophylaxis in patients with NC-C.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacteremia/prevention & control , Catheters, Indwelling/adverse effects , Minocycline/administration & dosage , Neoplasms/complications , Rifampin/administration & dosage , Adult , Aged , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Bacteremia/etiology , Catheterization, Central Venous/adverse effects , Female , Humans , Interleukin-2/administration & dosage , Male , Middle Aged , Neoplasms/drug therapy , Treatment Outcome
7.
Ann Oncol ; 20(10): 1682-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19541791

ABSTRACT

BACKGROUND: To correlate serum cytokine and angiogenic factor (CAF) levels with overall survival (OS) in metastatic renal cell carcinoma (mRCC) treated with interferon-alpha (IFN-alpha). PATIENTS AND METHODS: Serum CAF levels were measured in 103 patients treated on a randomized trial with IFN-alpha 0.5 million units (MU) twice daily or 5 MU daily. Concentrations of 17 analytes were determined by multiplex bead immunoassays [vascular endothelial growth factor A (VEGF(A)) and several cytokines] or enzyme-linked immunosorbent assay (basic fibroblast growth factor). We used proportional hazards models to evaluate the effect of CAF levels and clinical factors on OS. RESULTS: Pretreatment serum interleukin (IL) 5, IL-12 p40, VEGF(A), and IL-6 levels and Memorial Sloan-Kettering Cancer Center risk grouping independently correlated with OS, with hazard ratios of 2.33, 2.00, 2.07, 1.82, and 0.39, respectively (concordance index = 0.69 for the combined model versus 0.60 for the CAF model versus 0.52 for the clinical model). Based on an index derived from these five risk factors (RFs), patients with 0-2 RF had a median OS time of 32 months versus 9 months for patients with 3-5 RF (P < 0.0001). CONCLUSIONS: Serum CAF profiling contributes to prognostic evaluation in mRCC and helps to identify a subset of patients with 20% 5-year OS.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/secondary , Cytokines/therapeutic use , Interferon-alpha/therapeutic use , Angiogenesis Inducing Agents/blood , Carcinoma, Renal Cell/pathology , Chi-Square Distribution , Cytokines/blood , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor 2/blood , Follow-Up Studies , Humans , Immunoassay , Interleukin-12 Subunit p40/blood , Interleukin-12 Subunit p40/therapeutic use , Interleukin-5/blood , Interleukin-5/therapeutic use , Interleukin-6/blood , Interleukin-6/therapeutic use , Kaplan-Meier Estimate , Male , Proportional Hazards Models , Randomized Controlled Trials as Topic , Risk Factors , Survival Analysis , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/therapeutic use
8.
J Inherit Metab Dis ; 31(3): 319-36, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18509745

ABSTRACT

Enzyme replacement was introduced as treatment for non-neuronopathic Gaucher disease more than 15 years ago. To ensure the best use of this costly ultra-orphan agent, a systematic disease management approach has been proposed by an international panel; this includes the development, by consensus, of achievable treatment goals. Here we critically review these goals and monitoring guidelines and incorporate emerging experience of the disease in the therapeutic era, as well as contemporary clinical research. This review makes recommendations related specifically to the management of pregnancy; the appropriate use of splenectomy and bisphosphonate treatment; the relevance of biochemical markers to disease monitoring; and the use of semi-quantitative methods for assessing bone marrow infiltration. In addition, we identify key areas for development, including the requirement for a validated index of disease severity; the need to correlate widely used biomarkers with long-term disease outcomes, and the desirability of establishing agreed standards for monitoring of bone disease particularly in infants and children with Gaucher disease.


Subject(s)
Bone Diseases/diagnosis , Diphosphonates/therapeutic use , Gaucher Disease/therapy , Pregnancy Complications/therapy , Splenectomy , Absorptiometry, Photon , Biomarkers , Female , Gaucher Disease/complications , Humans , Magnetic Resonance Imaging , Pregnancy
9.
Cancer Gene Ther ; 14(5): 499-508, 2007 May.
Article in English | MEDLINE | ID: mdl-17332777

ABSTRACT

Activation and expansion of T cells are important in disease resolution, but tumors do not usually satisfy these immune requirements. Therefore, we employed a novel strategy whereby dual-specific T cells were generated that could respond to both tumor and influenza virus, reasoning that immunization with influenza virus would activate and expand tumor-specific cells, and inhibit tumor growth. Dual-specific T cells were generated by gene modification of influenza virus-specific mouse T cells with a chimeric gene-encoding reactivity against the erbB2 tumor-associated antigen. Dual-specific T cells were demonstrated to respond against both tumor and influenza in vitro, and expanded in vitro in response to influenza to a much greater degree than in response to tumor cells. Following adoptive transfer and immunization of tumor-bearing mice with influenza virus, dual-specific T cells expanded greatly in numbers in the peritoneal cavity and spleen. This resulted in a significant increase in time of survival of mice. However, tumors were not eradicated, which may have been due to the observed poor penetration of tumor by T cells. This is the first demonstration that the potent immunogenic nature of an infectious agent can be utilized to directly impact on T-cell expansion and activity against tumor in vivo.


Subject(s)
Immunotherapy, Adoptive/methods , Mammary Neoplasms, Animal/therapy , Orthomyxoviridae/immunology , Receptor, ErbB-2/antagonists & inhibitors , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/immunology , Mice , Mice, Inbred Strains , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology
10.
Gene Ther ; 14(4): 316-23, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17024104

ABSTRACT

Melanoma patients with metastases have a very low survival rate and limited treatment options. Therefore, the targeting of melanoma cells when they begin to invade and metastasize would be beneficial. An adhesion molecule that is upregulated at the vertical growth phase is the melanoma cell adhesion molecule (MCAM/MUC18). MUC18 is expressed in late primary and metastatic melanoma with little or no expression on normal melanocytes. We utilized the alphavirus-based DNA plasmid, SINCp, encoding murine MUC18 (SINCp c-muMUC18) for vaccination against B16F10 murine melanoma cells expressing murine MUC18. This vaccine effectively protected mice from lethal challenges with melanoma-expressing murine MUC18 in both primary and metastatic tumor models. Vaccination against MUC18 elicited effective humoral and CD8+ T-cell immune responses against melanoma. We propose that targeting molecules important in tumor invasion may be useful in the design of future strategies for the prevention and treatment of melanoma.


Subject(s)
Genetic Therapy/methods , Melanoma/therapy , Skin Neoplasms/therapy , Vaccines, DNA/administration & dosage , Alphavirus/genetics , Animals , CD146 Antigen/genetics , Cell Line, Tumor , DNA Fragmentation , Flow Cytometry , Immunotherapy/methods , Lymphocyte Activation , Melanoma/immunology , Melanoma/secondary , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/immunology , Neoplasms, Experimental , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology
11.
Gene Ther ; 9(16): 1085-92, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12140736

ABSTRACT

Genetic modification of human lymphocytes is being employed in strategies to correct enzyme deficiencies, encode cytokines and to redirect lymphocytes to antigenic targets other than those encoded by their endogenous T cell receptor. However, expression of transgenes in primary lymphocytes is generally low. Reasoning that vector modification may lead to increased transgene expression and subsequent increases in function, we have performed two retroviral vector modifications and report their effect on the functional expression in primary lymphocytes. A chimeric receptor specific for the colon carcinoma-associated antigen, EGP40, was initially incorporated into the retroviral vector LXSN. In this vector, receptor expression is driven by the Moloney murine leukemia virus LTR, and neomycin phosphotransferase expression driven by the SV40 promoter. Replacement of SV40 with an internal ribosomal entry site (IRES) increased the transgene activity of a mouse T cell line and human PBL as judged by increased cytokine release in response to antigen positive target cells. A further increase in transgene function was generated by the additional incorporation of a splice acceptor motif into the construct. Human PBL transduced with vector incorporating both IRES and intron were consistently more effective at lysing antigen positive colorectal carcinoma cells.


Subject(s)
Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Colonic Neoplasms/pathology , Genetic Therapy/methods , Genetic Vectors/genetics , Immunotherapy/methods , Adult , Animals , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Epithelial Cell Adhesion Molecule , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Mice , Moloney murine leukemia virus/genetics , RNA Splice Sites , Transduction, Genetic , Transgenes/genetics , Tumor Cells, Cultured
12.
Cancer Res ; 61(21): 7920-4, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691813

ABSTRACT

Endogenous retroviral gene products have been found in some human tumors, and therefore, may serve as antigens for immunotherapy approaches. The murine colorectal carcinoma CT26 and melanoma B16 have recently been found to express the endogenous retroviral gene products gp70 and p15E, respectively, that can serve as antigens recognized by T cells. To date, though, there has been no demonstration of tumor treatment using an endogenous retroviral protein. In this study, we demonstrate that mice immunized with recombinant vaccinia encoding the gp70 H2-L(d)-restricted minimal determinant were protected from CT26 tumor challenge. Splenocytes from mice immunized with vaccinia gp70 specifically secreted IFN-gamma in response to gp70 peptide-pulsed stimulators. Although this strategy could protect against subsequent tumor challenge, it was ineffective against established tumors. Therefore, to investigate the treatment of established CT26 or B16 lung metastases, mice were treated with cultured dendritic cells (DCs) pulsed with gp70 or p15E peptide. Significant inhibition of established lung metastases required immunization with peptide-pulsed DCs pretreated with CD40 ligand that has been demonstrated to increase the T-cell stimulatory activity of DCs. The ability to immunize against endogenous retroviral tumor antigens may have relevance in the induction of antitumor immunity for some human cancers.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Cancer Vaccines/immunology , Retroviridae Proteins, Oncogenic/immunology , Viral Envelope Proteins/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , CD40 Ligand/immunology , Cancer Vaccines/genetics , Colonic Neoplasms/therapy , Dendritic Cells/immunology , Immunotherapy, Adoptive , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Retroviridae Proteins, Oncogenic/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Envelope Proteins/genetics
13.
J Immunol ; 167(8): 4758-64, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591807

ABSTRACT

Involvement of tumor-Ag specific CD4(+) and CD8(+) T cells could be critical in the generation of an effective immunotherapy for cancer. In an attempt to optimize the T cell response against defined tumor Ags, we previously developed a method allowing transgene expression in human dendritic cells (DCs) using retroviral vectors. One advantage of using gene-modified DCs is the potential ability to generate CD8(+) T cells against multiple class I-restricted epitopes within the Ag, thereby eliciting a broad antitumor immune response. To test this, we generated tumor-reactive CD8(+) T cells with DCs transduced with the melanoma Ag gp100, for which a number of HLA-A2-restricted epitopes have been described. Using gp100-transduced DCs, we were indeed able to raise T cells recognizing three distinct HLA-A2 epitopes within the Ag, gp100(154-162), gp100(209-217), and gp100(280-288). We next tested the ability of transduced DCs to raise class II-restricted CD4(+) T cells. Interestingly, stimulation with gp100-transduced DCs resulted in the generation of CD4(+) T cells specific for a novel HLA-DRbeta1*0701-restricted epitope of gp100. The minimal determinant of this epitope was defined as gp100(174-190) (TGRAMLGTHTMEVTVYH). These observations suggest that retrovirally transduced DCs have the capacity to present multiple MHC class I- and class II-restricted peptides derived from a tumor Ag, thereby eliciting a robust immune response against that Ag.


Subject(s)
Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/immunology , Melanoma/immunology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Antigen Presentation , Antigens, CD34/isolation & purification , Epitopes , Genetic Vectors , HLA-A2 Antigen/immunology , HLA-DR Antigens/immunology , HLA-DRB1 Chains , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Peptide Fragments/immunology , Peptides , Retroviridae/genetics , Transformation, Genetic , gp100 Melanoma Antigen
14.
J Immunother ; 24(4): 363-73, 2001.
Article in English | MEDLINE | ID: mdl-11565838

ABSTRACT

This report describes a phase I study of the adoptive transfer of cloned melanoma antigen-specific T lymphocytes for therapy of patients with advanced melanoma. Clones were derived from peripheral blood lymphocytes or tumor-infiltrating lymphocytes of patients who had received prior immunization with the melanoma-associated antigen, gpl00. In response to its cognate antigen, each clone used for treatment secreted large amounts of interferon-gamma and granulocyte-macrophage colony-stimulating factor, lesser amounts of interleukin (IL)-2 and tumor necrosis factor-alpha, and little or no IL-4 and IL-10. Clones also demonstrated recognition of human leukocyte antigen-matched melanomas using cytokine secretion and lysis assays. Twelve patients received 2 cycles of cells alone; 11 patients received additional cycles of cells and were randomized between two schedules of IL-2 (125,000 IU/kg subcutaneously daily for 12 days versus 720,000 IU/kg intravenously every 8 h for 4 days). A total of 51 cycles of cells were administered, with an average of 1 x 10(10) cells per cycle. Peripheral blood samples were analyzed for persistence of transferred cells by T-cell receptor-specific polymerase chain reaction. Transferred cells reached a maximum level at 1 h after transfer but rapidly declined to undetectable levels by 2 weeks. One minor response and one mixed response were observed (both in the high-dose IL-2 arm). This report demonstrates the safety and feasibility of cloned T-cell transfer as a therapy for patients with cancer. The lack of clinical effectiveness of this protocol suggests that transfer of different or additional cell types or that modulation of the recipient host environment is required for successful therapy.


Subject(s)
Immunotherapy, Adoptive , Interleukin-2/pharmacology , Melanoma/therapy , Membrane Glycoproteins/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , Adult , Aged , Clone Cells , Female , Humans , Interferon-gamma/metabolism , Male , Melanoma/immunology , Melanoma/secondary , Middle Aged , gp100 Melanoma Antigen
15.
Proc Natl Acad Sci U S A ; 98(18): 10362-7, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504927

ABSTRACT

A number of perturbations of B cells has been described in the setting of HIV infection; however, most remain poorly understood. To directly address the effect of HIV replication on B cell function, we investigated the capacity of B cells isolated from HIV-infected patients to respond to a variety of stimuli before and after reduction of viremia by effective antiretroviral therapy. B cells taken from patients with high levels of plasma viremia were defective in their proliferative responses to various stimuli. Viremia was also associated with the appearance of a subpopulation of B cells that expressed reduced levels of CD21. After fractionation into CD21(high)- and CD21(low)-expressing B cells, the CD21(low) fraction showed dramatically reduced proliferation in response to B cell stimuli and enhanced secretion of immunoglobulins when compared with the CD21(high) fraction. Electron microscopic analysis of each fraction revealed cells with plasmacytoid features in the CD21(low) B cell population but not in the CD21(high) fraction. These results indicate that HIV viremia induces the appearance of a subset of B cells whose function is impaired and which may be responsible for the hypergammaglobulinemia associated with HIV disease.


Subject(s)
B-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/pathogenicity , Antigens, CD19/genetics , Antigens, CD19/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/ultrastructure , B-Lymphocytes/ultrastructure , Base Sequence , DNA Primers/genetics , Gene Expression , HIV Infections/genetics , Humans , Hypergammaglobulinemia/etiology , Immunophenotyping , Lymphocyte Activation , Microscopy, Electron , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , Viremia/genetics , Viremia/immunology
16.
Hum Gene Ther ; 11(18): 2445-52, 2000 Dec 10.
Article in English | MEDLINE | ID: mdl-11119416

ABSTRACT

The destruction of newly forming tumor vasculature is a promising approach to inhibit tumor growth. The goal of the present study was to investigate whether human lymphocytes gene modified to express a chimeric receptor specific for the angiogenic endothelial cell receptor, KDR, could react against KDR(+) cells. Gene-modified lymphocytes specifically lysed KDR(+) cells and secreted cytokines in response to KDR(+) target cells including human umbilical vein endothelial cells (HUVECs). Anti-KDR lymphocytes induced HUVECs to secrete the chemokine interleukin 8 and upregulate the adhesion molecules VCAM and E-selectin, which may be important in the recruitment of further immune effector cells to tumor. These KDR-specific lymphocytes may be useful in the adoptive immunotherapy of a broad range of cancers by inducing immune-mediated destruction of tumor neovasculature.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , T-Lymphocytes/metabolism , CD28 Antigens/genetics , Cell Adhesion , Cell Line , Cells, Cultured , Cytokines/biosynthesis , E-Selectin/metabolism , Endothelium, Vascular/cytology , Flow Cytometry , Gene Transfer Techniques , Humans , Interleukin-8/biosynthesis , Protein Structure, Tertiary , Receptors, Fc/metabolism , Receptors, Vascular Endothelial Growth Factor , Transduction, Genetic , Umbilical Veins/cytology , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
17.
Hum Gene Ther ; 11(17): 2377-87, 2000 Nov 20.
Article in English | MEDLINE | ID: mdl-11096442

ABSTRACT

Adoptive immunotherapy with genetically modified T lymphocytes is being utilized in clinical trials for the treatment of a broad range of diseases including cancer and HIV infection. To improve on these treatments, and to better understand their mechanisms of action, it is necessary to develop techniques to generate large numbers of cells and characterize the functional heterogeneity of the cells produced. In this study, patient peripheral blood lymphocytes were transduced with a chimeric antigen receptor (MOv-gamma) derived from a mouse monoclonal antibody against folate-binding protein, which is overexpressed on many ovarian cancers. Thus, irrespective of their original specificity, normal human T lymphocytes were redirected to react against ovarian cancer cells. Lymphocytes from five patients were transduced and grown to large numbers, with a median expansion of more than 7000-fold. When proliferation was inadequate, the cells were expanded by stimulation utilizing anti-CD3, IL-2, and irradiated allogeneic PBMCs. The cells maintained their functional ability to recognize ovarian cancer over several months. Cloning of transduced cells was undertaken to determine the level of gene expression and function of individual cells making up the bulk population. Transduced CD4(+) and CD8(+) cell clones were isolated from the bulk and demonstrated antitumor activity. These clones had a diverse repertoire with respect to secretion of cytokines, and individual clones maintained their cytokine profile on subsequent expansion. These studies establish the feasibility of consistently generating large numbers of gene-modified tumor-reactive lymphocytes, with a stable and diverse cytokine repertoire, that could be utilized for patient treatment.


Subject(s)
Antibodies, Monoclonal/genetics , Ovarian Neoplasms/genetics , Recombinant Proteins/genetics , T-Lymphocytes/physiology , Transduction, Genetic/methods , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , CD3 Complex/immunology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cells, Cultured , Culture Media , Cytokines/metabolism , Female , Genetic Therapy/methods , Humans , Interleukin-2/metabolism , Melanoma/pathology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Phenotype , Recombinant Fusion Proteins/genetics , Recombinant Proteins/metabolism , Reference Values , T-Lymphocytes/radiation effects , Tumor Cells, Cultured
18.
Eur J Immunol ; 30(11): 3291-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11093145

ABSTRACT

Dendritic cells (DC) are specialized cells of the immune system responsible for the initiation and regulation of both cellular and humoral responses. DC function is highly dependent on their level of maturation. In this study, we postulated that full DC maturation would require a combination of activating signals. When cultured monocyte-derived DC received stimulation with CD40 ligand (CD40L) and lipopolysaccharide (LPS) together, the IL-12 secretion increased 5-60-fold and the IL-10 secretion increased 5-15-fold when compared with either stimulation alone. In addition, poly I.C, a double-stranded RNA analog that mimics viral infection, also synergized with CD40L to stimulate DC to secrete high levels of IL-12 and IL-10. Flow cytometry revealed an up-regulation in the expression of CD80, CD86 and CD83 following activation with a soluble trimeric form of CD40L (CD40Ls) or LPS. However, no further up-regulation was observed when both CD40Ls and LPS were used together compared with a single stimulatory signal, suggesting that there was no correlation between the expression of these markers and the level of IL-12/IL-10 secretion. Finally, specific cytotoxic T lymphocytes (CTL) were generated using DC pulsed with a modified HLA-A2-restricted peptide epitope derived from the melanoma antigen MART-1. DC activated with a combination of CD40Ls and LPS were more efficient in eliciting MART-specific reactivity compared to DC activated with CD40Ls or LPS alone. These results demonstrate that multiple maturational signals have a positive impact on the ability of DC to secrete IL-12 and IL-10 and more importantly, to generate antigen-specific T lymphocytes.


Subject(s)
Antigens, Neoplasm/immunology , Cell Communication/immunology , Dendritic Cells/immunology , T-Lymphocytes/immunology , Antigen Presentation , CD40 Antigens/immunology , CD40 Ligand/immunology , Humans , Lymphocyte Activation/immunology , Signal Transduction/immunology
19.
Hum Gene Ther ; 11(13): 1901-9, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10986562

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells and are capable of activating naive T cells. Gene transfer of tumor antigen and cytokine genes into DCs could be an important strategy for immunotherapeutic applications. Dendritic cells derived from peripheral blood monocytes do not divide and are therefore poor candidates for gene transfer by Moloney murine leukemia virus (Mo-MuLV)-based retroviral vectors. Lentiviral vectors are emerging as a powerful tool for gene delivery into dividing and nondividing cells. A three-plasmid expression system pseudotyped with the envelope from vesicular stomatitis virus (VSV-G) was used to generate lentiviral vector particles expressing enhanced green fluorescent protein (EGFP). Peripheral blood monocyte-derived DCs were cultured in the presence of GM-CSF and IL-4 and transduced with lentiviral or Mo-MuLV-based vectors expressing EGFP. FACS analysis of lentiviral vector-transduced DCs derived either from normal healthy volunteers or from melanoma patients demonstrated transduction efficiency ranging from 70 to 90% compared with 2-8% using Mo-MuLV-based vectors pseudotyped with VSV-G. Comparison of lentiviral vectors expressing EGFP driven by CMV or human PGK promoters showed similar levels of transgene expression. Lentiviral vector preparations produced in the absence of HIV accessory proteins transduced DCs at efficiencies equal to vectors produced with accessory proteins. Alu-HIV-1 LTR PCR demonstrated the genomic integration of the lentiviral vector in the transduced DCs. Transduced cells showed characteristic dendritic cell phenotype and strong allostimulatory capacity and maintained the ability to respond to activation signals such as CD40 ligand and lipopolysaccharide. These results provide evidence that lentiviral vectors are efficient tools for gene transfer and expression in monocyte-derived DCs that could be useful for immunotherapeutic applications.


Subject(s)
Dendritic Cells/physiology , Gene Transfer Techniques , Lentivirus/genetics , Monocytes/cytology , Alu Elements/genetics , Antigens, CD , Cytomegalovirus/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Green Fluorescent Proteins , HIV-1/genetics , Humans , Immunoglobulins/metabolism , Interleukin-12/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/metabolism , Moloney murine leukemia virus/genetics , Monocytes/virology , Promoter Regions, Genetic , T-Lymphocytes/immunology , Vesicular stomatitis Indiana virus/genetics , Viral Proteins/genetics , CD83 Antigen
20.
Cancer Gene Ther ; 7(2): 284-91, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10770638

ABSTRACT

Transduction with chimeric T-cell receptor genes can be used to redirect primary T lymphocytes to recognize specific antigens (Ags), including ovarian and breast cancer Ags. To extend this approach to colon cancer we report here redirection of T cells using a chimeric receptor recognizing the colon cancer-associated Ag EGP40. Chimeric T cell receptors were constructed by ligating single-chain genes of either of two EGP40-specific monoclonal antibodies (CO17.1 A, GA733) to the Fc receptor gamma-signaling chain. Retroviral vectors incorporating these constructs were used to transduce a murine T-cell line and human peripheral blood lymphocytes. These modified T cells were analyzed for specific recognition of colon cancer lines by measuring cytokine release and lytic activity against tumor targets. Murine lymphocytes transduced with the chimeric receptor based on GA733, but not CO17.1A, released cytokine specifically in response to EGP40-expressing colon cancer cell lines. Recognition of colon cancer targets by murine lymphocytes was blocked by the addition of GA733 antibody or soluble EGP40 Ag, confirming that colon cancer recognition is based on specific chimeric receptor-Ag interaction. Human lymphocytes transduced with chimeric GA733 specifically recognized colon carcinoma cells in cytokine release assays and lysed EGP40-expressing tumor cells. Genetic modification of T cells can be used to redirect T cells against EGP40-expressing tumor cells. The expression of chimeric GA733 in the autologous lymphocytes of patients may provide a source of tumor-reactive cells with therapeutic application against colon cancer.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transfection/immunology , 3T3 Cells , Adult , Animals , Antibodies, Monoclonal/metabolism , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/metabolism , Cell Line , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Epithelial Cell Adhesion Molecule , Humans , K562 Cells , Mice , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/metabolism , Receptors, IgG/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/virology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL