21 results on '"Angelo A. Cardoso"'
Search Results
2. Pre-clinical data supporting immunotherapy for HIV using CMV-HIV-specific CAR T cells with CMV vaccine
- Author
-
Min Guan, Laura Lim, Leo Holguin, Tianxu Han, Vibhuti Vyas, Ryan Urak, Aaron Miller, Diana L. Browning, Liliana Echavarria, Shasha Li, Shirley Li, Wen-Chung Chang, Tristan Scott, Paul Yazaki, Kevin V. Morris, Angelo A. Cardoso, M. Suzette Blanchard, Virginia Le Verche, Stephen J. Forman, John A. Zaia, John C. Burnett, and Xiuli Wang
- Subjects
chimeric antigen receptor T cell (CAR T) ,cytomegalovirus (CMV) vaccine ,broadly neutralizing antibody (bNAb) ,N6 ,HIV/AIDS ,immunotherapy ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
T cells engineered to express HIV-specific chimeric antigen receptors (CARs) represent a promising strategy to clear HIV-infected cells, but to date have not achieved clinical benefits. A likely hurdle is the limited T cell activation and persistence when HIV antigenemia is low, particularly during antiretroviral therapy (ART). To overcome this issue, we propose to use a cytomegalovirus (CMV) vaccine to stimulate CMV-specific T cells that express CARs directed against the HIV-1 envelope protein gp120. In this study, we use a GMP-compliant platform to engineer CMV-specific T cells to express a second-generation CAR derived from the N6 broadly neutralizing antibody, one of the broadest anti-gp120 neutralizing antibodies. These CMV-HIV CAR T cells exhibit dual effector functions upon in vitro stimulation through their endogenous CMV-specific T cell receptors or the introduced CARs. Using a humanized HIV mouse model, we show that CMV vaccination during ART accelerates CMV-HIV CAR T cell expansion in the peripheral blood and that higher numbers of CMV-HIV CAR T cells were associated with a better control of HIV viral load and fewer HIV antigen p24+ cells in the bone marrow upon ART interruption. Collectively, these data support the clinical development of CMV-HIV CAR T cells in combination with a CMV vaccine in HIV-infected individuals.
- Published
- 2022
- Full Text
- View/download PDF
3. Engineered extracellular vesicles directed to the spike protein inhibit SARS-CoV-2
- Author
-
Tristan A. Scott, Aroon Supramaniam, Adi Idris, Angelo A. Cardoso, Surya Shrivastava, Gabrielle Kelly, Nicole A. Grepo, Citradewi Soemardy, Roslyn M. Ray, Nigel A.J. McMillan, and Kevin V. Morris
- Subjects
extracellular vesicles ,nanobody ,SARS-CoV-2 ,COVID-19 ,therapeutic ,neutralization ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
SARS-CoV-2 (CoV-2) viral infection results in COVID-19 disease, which has caused significant morbidity and mortality worldwide. A vaccine is crucial to curtail the spread of SARS-CoV-2, while therapeutics will be required to treat ongoing and reemerging infections of SARS-CoV-2 and COVID-19 disease. There are currently no commercially available effective anti-viral therapies for COVID-19, urging the development of novel modalities. Here, we describe a molecular therapy specifically targeted to neutralize SARS-CoV-2, which consists of extracellular vesicles (EVs) containing a novel fusion tetraspanin protein, CD63, embedded within an anti-CoV-2 nanobody. These anti-CoV-2-enriched EVs bind SARS-CoV-2 spike protein at the receptor-binding domain (RBD) site and can functionally neutralize SARS-CoV-2. This work demonstrates an innovative EV-targeting platform that can be employed to target and inhibit the early stages of SARS-CoV-2 infection.
- Published
- 2022
- Full Text
- View/download PDF
4. COVID-19 vaccination elicits an evolving, cross-reactive antibody response to epitopes conserved with endemic coronavirus spike proteins
- Author
-
Evan A. Elko, Georgia A. Nelson, Heather L. Mead, Erin J. Kelley, Sophia T. Carvalho, Nathan G. Sarbo, Caroline E. Harms, Virginia Le Verche, Angelo A. Cardoso, Jennifer L. Ely, Annalee S. Boyle, Alejandra Piña, Sierra N. Henson, Fatima Rahee, Paul S. Keim, Kimberly R. Celona, Jinhee Yi, Erik W. Settles, Daniela A. Bota, George C. Yu, Sheldon R. Morris, John A. Zaia, Jason T. Ladner, and John A. Altin
- Subjects
CP: Immunology ,CP: Microbiology ,Biology (General) ,QH301-705.5 - Abstract
Summary: The COVID-19 pandemic has triggered the first widespread vaccination campaign against a coronavirus. Many vaccinated subjects are previously naive to SARS-CoV-2; however, almost all have previously encountered other coronaviruses (CoVs), and the role of this immunity in shaping the vaccine response remains uncharacterized. Here, we use longitudinal samples and highly multiplexed serology to identify mRNA-1273 vaccine-induced antibody responses against a range of CoV Spike epitopes, in both phylogenetically conserved and non-conserved regions. Whereas reactivity to SARS-CoV-2 epitopes shows a delayed but progressive increase following vaccination, we observe distinct kinetics for the endemic CoV homologs at conserved sites in Spike S2: these become detectable sooner and decay at later time points. Using homolog-specific antibody depletion and alanine-substitution experiments, we show that these distinct trajectories reflect an evolving cross-reactive response that can distinguish rare, polymorphic residues within these epitopes. Our results reveal mechanisms for the formation of antibodies with broad reactivity against CoVs.
- Published
- 2022
- Full Text
- View/download PDF
5. The Earliest T-Precursors in the Mouse Embryo Are Susceptible to Leukemic Transformation
- Author
-
Jixin Ding, Angelo A. Cardoso, Momoko Yoshimoto, and Michihiro Kobayashi
- Subjects
notch signaling ,notch intracellular domain ,yolk sac ,para-aortic splanchnopleura ,aorta-gonad-mesonephros region ,acute T cell leukemia ,Biology (General) ,QH301-705.5 - Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in pediatric patients. About 10–15% of pediatric ALL belong to T-cell ALL (T-ALL), which is characterized by aggressive expansion of immature T-lymphoblasts and is categorized as high-risk leukemia. Leukemia initiating cells represent a reservoir that is responsible for the initiation and propagation of leukemia. Its perinatal origin has been suggested in some childhood acute B-lymphoblastic and myeloblastic leukemias. Therefore, we hypothesized that child T-ALL initiating cells also exist during the perinatal period. In this study, T-ALL potential of the hematopoietic precursors was found in the para-aortic splanchnopleura (P-Sp) region, but not in the extraembryonic yolk sac (YS) of the mouse embryo at embryonic day 9.5. We overexpressed the Notch intracellular domain (NICD) in the P-Sp and YS cells and transplanted them into lethally irradiated mice. NICD-overexpressing P-Sp cells rapidly developed T-ALL while YS cells failed to display leukemia propagation despite successful NICD induction. These results suggest a possible role of fetal-derived T-cell precursors as leukemia-initiating cells.
- Published
- 2021
- Full Text
- View/download PDF
6. Sepsis Induces Hematopoietic Stem Cell Exhaustion and Myelosuppression through Distinct Contributions of TRIF and MYD88
- Author
-
Huajia Zhang, Sonia Rodriguez, Lin Wang, Soujuan Wang, Henrique Serezani, Reuben Kapur, Angelo A. Cardoso, and Nadia Carlesso
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Toll-like receptor 4 (TLR4) plays a central role in host responses to bacterial infection, but the precise mechanism(s) by which its downstream signaling components coordinate the bone marrow response to sepsis is poorly understood. Using mice deficient in TLR4 downstream adapters MYD88 or TRIF, we demonstrate that both cell-autonomous and non-cell-autonomous MYD88 activation are major causes of myelosuppression during sepsis, while having a modest impact on hematopoietic stem cell (HSC) functions. In contrast, cell-intrinsic TRIF activation severely compromises HSC self-renewal without directly affecting myeloid cells. Lipopolysaccharide-induced activation of MYD88 or TRIF contributes to cell-cycle activation of HSC and induces rapid and permanent changes in transcriptional programs, as indicated by persistent downregulation of Spi1 and CebpA expression after transplantation. Thus, distinct mechanisms downstream of TLR4 signaling mediate myelosuppression and HSC exhaustion during sepsis through unique effects of MyD88 and TRIF. : In this article, Carlesso and colleagues investigate the role of the TLR4 downstream adapters, MyD88 and TRIF, in the regulation of bone marrow response to sepsis. Their work shows that MyD88 activation is a major cause of myelosuppression during sepsis while having a modest impact on HSC, whereas cell-intrinsic TRIF activation compromises HSC self-renewal without directly affecting myeloid cells. Cell-intrinsic activation in HSC results in HSC exhaustion and transcriptional changes persisting over the long term even when the endotoxic environment is removed.
- Published
- 2016
- Full Text
- View/download PDF
7. Functional analysis of HOXA10 and HOXB4 in human medulloblastoma cell lines
- Author
-
Fábio Augusto Labre De Souza, Carolina Hassibe Thomé, Ricardo Santos de Oliveira, Aparecida Maria Fontes, Fernando Silva Ramalho, Fernanda Ursoli Ferreira Melo, Kuruvilla Joseph Abraham, Ricardo Bonfim-Silva, Hélio Rubens Machado, Angelo A. Cardoso, and Dimas Tadeu Covas
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Cell ,Mice, Nude ,Biology ,Real-Time Polymerase Chain Reaction ,Mice ,03 medical and health sciences ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,GENES HOMEOBOX ,Gene Silencing ,Cerebellar Neoplasms ,Hox gene ,Cell Proliferation ,Homeodomain Proteins ,Cell growth ,Wnt signaling pathway ,Cell migration ,Cell cycle ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Homeobox A10 Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,Cancer research ,Medulloblastoma ,Transcription Factors - Abstract
Medulloblastoma (MB) is a malignant childhood brain tumor which at molecular level is classified into at least four major subtypes: WNT, SHH, group C and group D differing in response to treatment. Previous studies have associated changes in expression levels and activation of certain HOX genes with MB development. In the present study, we investigate the role of HOX genes in two attributes acquired by tumor cells: migration and proliferation potential, as well as, in vivo tumorigenic potential. We analyzed UW402, UW473, DAOY and ONS-76 human pediatric MB cell lines and cerebellum primary cultures. Two-color microarray-based gene expression analysis was used to identify differentially expressed HOX genes. Among the various HOX genes significantly overexpressed in DAOY and ONS-76 cell lines compared to UW402 and UW473 cell lines, HOXA10 and HOXB4 were selected for further analysis. The expression levels of these HOX genes were validated by real-time PCR. A mouse model was used to study the effect of the HOXA10 and HOXB4 genes on the in vivo tumorigenic potential and the in vitro proliferative and migration potential of MB cell lines. Our results show that the inhibition of HOXA10 in DAOY cell line led to increased in vitro cell migration while in vitro cell proliferation or in vivo tumorigenic potential were unaffected. We also observed that induced expression of HOXB4 in the UW473 cell line significantly reduced in vitro cell proliferation and migration capability of UW473 cells with no effect on the in vivo tumorigenicity. This suggests that HOXA10 plays a role in migration events and the HOXB4 gene is involved in proliferation and migration processes of medulloblastoma cells, however, it appears that these genes are not essential for the tumorigenic process of these cells.
- Published
- 2017
- Full Text
- View/download PDF
8. Autonomous murine T-cell progenitor production in the extra-embryonic yolk sac before HSC emergence
- Author
-
Simon J. Conway, Prashanth Porayette, Momoko Yoshimoto, Mark H. Kaplan, Nicole L. Glosson, Angelo A. Cardoso, Nadia Carlesso, and Mervin C. Yoder
- Subjects
T-Lymphocytes ,T cell ,Cellular differentiation ,Immunology ,Thymus Gland ,Biology ,Biochemistry ,Mice ,T-Lymphocyte Subsets ,medicine ,Animals ,Yolk sac ,Progenitor cell ,Aorta ,Yolk Sac ,Progenitor ,Hematopoietic Stem Cell Transplantation ,Endothelial Cells ,Cell Differentiation ,Embryo ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Cell biology ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Animals, Newborn ,embryonic structures ,Spleen - Abstract
The extra-embryonic yolk sac (YS) is the first hematopoietic site in the mouse embryo and is thought to generate only primitive erythroid and myeloerythroid progenitor cells before definitive HSC emergence within the embryo on E10.5. Here, we have shown the existence of T cell–restricted progenitors in the E9.5 YS that directly engraft in recipient immunodeficient mice. T-cell progenitors were also produced in vitro from both YS and para-aortic splanchnopleura hemogenic endothelial cells, and these T-cell progenitors repopulated the thymus and differentiated into mature T-cell subsets in vivo on transplantation. Our data confirm that the YS produces T-lineage–restricted progenitors that are available to colonize the thymus and provide new insight into the YS as a definitive hematopoietic site in the mouse embryo.
- Published
- 2012
- Full Text
- View/download PDF
9. Loss of the E3 Ubiquitin Ligase SKP2 Limits De Oncogenic Potential of Notch in T-Cell Lymphoblastic Leukemia
- Author
-
Mark H. Kaplan, Lin Wang, Mark Y. Chiang, Mark Wunderlich, Joycelynne Palmer, Purvi Mehrotra, Amy Zollman, James C. Mulloy, Hujia Zhang, Angelo A. Cardoso, Mary A. Yui, George E. Sandusky, Sonia Rodriguez-Rodriguez, and Nadia Carlesso
- Subjects
Cancer Research ,biology ,business.industry ,T cell ,Lymphoblastic Leukemia ,Refractory Disease ,Cell Biology ,Hematology ,Ubiquitin ligase ,medicine.anatomical_structure ,Leukemia relapse ,Genetics ,Molecular targets ,SKP2 ,biology.protein ,Cancer research ,Medicine ,business ,Molecular Biology - Abstract
Despite marked clinical successes in the treatment of childhood T-ALL, leukemia relapse, refractory disease and induction failure (around 30% of patients) remain significant clinical problems. New insights in the molecular alterations of ALL have revealed new molecular targets, such as activating mutations of Notch1 (N1), identified in more than 50% of T-ALL patients. However, disruption of N1 signaling by gamma-secretase inhibitors failed to fulfill its clinical promise. Furthermore, little is known about N1 downstream mediators that can be potential therapeutic targets in T-ALL.
- Published
- 2018
- Full Text
- View/download PDF
10. Delta-Like 4 Induces Notch Signaling in Macrophages
- Author
-
James P. Canner, Jon C. Aster, Masanori Aikawa, Sai Man Timothy Tang, Nadia Carlesso, Erik Fung, Kunio Morishige, Joseph F. Arboleda-Velasquez, and Angelo A. Cardoso
- Subjects
Transcription, Genetic ,Adipose tissue macrophages ,Notch signaling pathway ,Inflammation ,Biology ,Proinflammatory cytokine ,Physiology (medical) ,medicine ,Humans ,Macrophage ,Gene silencing ,Receptor ,Receptor, Notch3 ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Receptors, Notch ,Macrophages ,Calcium-Binding Proteins ,Macrophage Activation ,Atherosclerosis ,Cell biology ,Gene Expression Regulation ,Immunology ,Intercellular Signaling Peptides and Proteins ,medicine.symptom ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Signal Transduction - Abstract
Background— Activated macrophages contribute to the pathogenesis of inflammatory diseases such as atherosclerosis. Although Notch signaling participates in various aspects of immunity, its role in macrophage activation remains undetermined. Methods and Results— To explore the role of Notch signaling in inflammation, we examined the expression and activity of Notch pathway components in human primary macrophages in vitro and in atherosclerotic plaques. Macrophages in culture express various Notch pathway components including all 4 receptors (Notch1 to Notch4). Notch3 selectively increased during macrophage differentiation; however, silencing by RNA interference demonstrated that all receptors are functional. The ligand Delta-like 4 (Dll4) increased in macrophages exposed to proinflammatory stimuli such as lipopolysaccharide, interleukin-1β, or minimally-modified low-density lipoprotein in a Toll-like receptor 4– and nuclear factor-κB–dependent fashion. Soluble Dll4 bound to human macrophages. Coincubation of macrophages with cells that expressed Dll4 triggered Notch proteolysis and activation; increased the transcription of proinflammatory genes such as inducible nitric oxide synthase, pentraxin 3 and Id1; resulted in activation of mitogen-activated protein kinase, Akt, and nuclear factor-κB pathways; and increased the expression of Dll4 in macrophages. Notch3 knockdown during macrophage differentiation decreased the transcription of genes that promote inflammation, such as inducible nitric oxide synthase, pentraxin 3, Id1, and scavenger receptor-A. These in vitro findings correlate with results of quantitative immunohistochemistry, which demonstrated the presence of Dll4 and other Notch components within macrophages in atherosclerotic plaques. Conclusion— Dll4-triggered Notch signaling may mediate inflammatory responses in macrophages and promote inflammation.
- Published
- 2007
- Full Text
- View/download PDF
11. Adaptive Design: A Review of the Technical, Statistical, and Regulatory Aspects of Implementation in a Clinical Trial.
- Author
-
Cerqueira, Franck Pires, Jesus, Angelo Miguel Cardoso, and Cotrim, Maria Dulce
- Subjects
PHYSIOLOGICAL adaptation ,BIOMARKERS ,CLINICAL trials ,COMMITTEES ,EXPERIMENTAL design ,MEDLINE ,ONLINE information services ,SYSTEMATIC reviews ,SAMPLE size (Statistics) - Abstract
Background: In an adaptive trial, the researcher may have the option of responding to interim safety and efficacy data in a number of ways, including narrowing the study focus or increasing the number of subjects, balancing treatment allocation or different forms of randomization based on responses of subjects prior to treatment. This research aims at compiling the technical, statistical, and regulatory implications of the employment of adaptive design in a clinical trial. Methods: Review of adaptive design clinical trials in Medline, PubMed, EU Clinical Trials Register, and ClinicalTrials.gov. Phase I and seamless phase I/II trials were excluded. We selected variables extracted from trials that included basic study characteristics, adaptive design features, size and use of independent data-monitoring committees (DMCs), and blinded interim analysis. Results: The research retrieved 336 results, from which 78 were selected for analysis. Sixty-seven were published articles, and 11 were guidelines, papers, and regulatory bills. The most prevalent type of adaptation was the seamless phase II/III design 23.1%, followed by adaptive dose progression 19.2%, pick the winner / drop the loser 16.7%, sample size re-estimation 10.3%, change in the study objective 9.0%, adaptive sequential design 9.0%, adaptive randomization 6.4%, biomarker adaptive design 3.8%, and endpoint adaptation 2.6%. Discussion Discussion: It is possible to infer that the use of Adaptive Design is an ethical and scientific advantage when properly planned and applied, since it increases the flexibility of the trial, shortens the overall clinical investigation time of a drug, and reduces the risk of patient exposure to adverse effects related to the experimental drug. Its greater methodologic and analytic complexity requires an adequate statistical methodology. Conclusions: The application of "adaptive clinical designs" for phase II/III studies appear to have been limited to trials with a small number of study centers, with smaller extensions of time and to experimental drugs with more immediate clinical effects that are amenable to risk/benefit decisions based on interim analyses. According to the reviewed studies, simple adaptive trial designs—such as early study terminations due to futility and sample size re-estimation—are becoming widely adopted throughout the pharmaceutical industry, especially in phase II and III studies. The pharmaceutical industry and contract research organizations (CROs) are implementing simple adaptations more frequently and the more complex adaptations—biomarker adaptive design, endpoint adaptation—are more sporadic. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
12. Leukemia-stimulated bone marrow endothelium promotes leukemia cell survival
- Author
-
Lara F. Costa, Stephen E. Sallan, Angelo A. Cardoso, J. Pedro Veiga, and Lee M. Nadler
- Subjects
Cancer Research ,Endothelium ,Cell Survival ,Angiogenesis ,Mice, SCID ,Biology ,Neovascularization ,Mice ,Microscopy, Electron, Transmission ,Bone Marrow ,Mice, Inbred NOD ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Proliferation ,Leukemia ,Neovascularization, Pathologic ,Cell growth ,Cell Biology ,Hematology ,medicine.disease ,Endothelial stem cell ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Immunology ,Microscopy, Electron, Scanning ,Cancer research ,Pseudopodia ,Bone marrow ,medicine.symptom - Abstract
Extensive endothelial cell proliferation and marked neovascularization are the most pronounced microenvironmental changes consistently observed in the bone marrow (BM) of patients with acute lymphoblastic leukemia (ALL). It is not known whether ALL cells induce this phenotype and whether they receive critical signals from the tumor-associated BM endothelium. Here, we show that leukemia cells actively stimulate BM endothelium, promote de novo angiogenesis, and induce neovascularization in the leukemic BM. Soluble factors, present in the leukemic BM microenvironment, promote the proliferation, migration, and morphogenesis of BM endothelial cells, which are critical processes in tumor angiogenesis. We also show in vitro that leukemia cells display directional motion towards assembled BM endothelium and following adherence exhibit cell polarization, pseudopodia, and ultrastructural features that suggest the existence of leukemia-endothelium cross-talk. Finally, we show that BM endothelium promotes leukemia cell survival through a mechanism mediated through the anti-apoptotic molecule bcl-2. These studies indicate that ALL cells actively recruit BM endothelium and mediate the leukemia-associated neovascularization observed in ALL. Therefore, disruption of interactions between leukemia cells and BM endothelium may constitute a valid therapeutic strategy.
- Published
- 2006
- Full Text
- View/download PDF
13. Interleukin-7 in T-cell acute lymphoblastic leukemia: An extrinsic factor supporting leukemogenesis?
- Author
-
Angelo A. Cardoso, João T. Barata, and Vassiliki A. Boussiotis
- Subjects
MAPK/ERK pathway ,Cancer Research ,medicine.medical_treatment ,T cell ,T-cell leukemia ,Protein Serine-Threonine Kinases ,Biology ,Phosphatidylinositol 3-Kinases ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Cell growth ,Interleukin-7 ,Hematology ,Cell biology ,Cytokine ,medicine.anatomical_structure ,Oncology ,Trans-Activators ,Mitogen-Activated Protein Kinases ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The malignant transformation and expansion of tumor cells involve both cell-autonomous mechanisms and microenvironment signals that regulate viability, nutrient utilization, metabolic activity and cell growth. In T-cell acute lymphoblastic leukemia (T-ALL), the co-culture of leukemic cells with stroma or the addition of particular cytokines prevents ex vivo spontaneous apoptosis. Interleukin-7 (IL-7), a cytokine produced by thymic and bone marrow stroma, increases the viability and proliferation of T-ALL cells. IL-7 induces the activation of Jak/STAT, MEK/Erk and PI3K/Akt signaling pathways in T-ALL cells. PI3K/Akt is the dominant pathway that mediates the effects of IL-7 on T-ALL. PI3K signaling is required for the induction of Bcl-2, the down-regulation of p27(kip1) and cell cycle progression. PI3K signaling is also required for the expression of the glucose transporter Glut1, uptake of glucose, activation of the metabolic machinery, increase in cell size, and maintenance of mitochondrial integrity. These observations suggest that substrates of molecular pathways activated by microenvironmental factors represent attractive molecular targets for the regulation of the viability and proliferation of T-ALL cells and provide the means for the development of novel treatment strategies.
- Published
- 2005
- Full Text
- View/download PDF
14. Failure to define window of time for autologous tumor vaccination in patients with newly diagnosed or relapsed acute lymphoblastic leukemia
- Author
-
Angelo A. Cardoso, Stephen E. Sallan, Lewis B. Silverman, Donna Neuberg, Lee M. Nadler, Heather L. Keczkemethy, Mark D. Fleming, W. Nicholas Haining, Eva C. Guinan, Richard Stone, Daniel J. DeAngelo, and Ilene Galinsky
- Subjects
Adult ,Male ,Cancer Research ,Time Factors ,Adolescent ,T-Lymphocytes ,medicine.medical_treatment ,Antigen-Presenting Cells ,Cancer Vaccines ,Immunotherapy, Adoptive ,Secondary Prevention ,Genetics ,medicine ,Humans ,CD40 Antigens ,Child ,Antigen-presenting cell ,Adverse effect ,Molecular Biology ,B cell ,CD40 ,biology ,business.industry ,Cell Biology ,Hematology ,Immunotherapy ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Vaccination ,medicine.anatomical_structure ,Child, Preschool ,Immunology ,biology.protein ,Female ,Myelopoiesis ,business ,Ex vivo - Abstract
Objectives We and others have shown that B cell precursor acute lymphoblastic leukemia cells (ALL) stimulated with CD40 ligand become efficient antigen-presenting cells (APC) capable of expanding autologous, tumor-specific T cells from patients. Translation of these preclinical findings to a novel treatment strategy required four separate issues to be determined: (1) if a CD40-ALL vaccine could be generated for clinical use; (2) whether clinical translation could be achieved; (3) whether the vaccination was safe; and (4) whether a window of time could be identified that would optimize the efficacy of vaccination. Patients and methods Nine patients with relapsed/refractory ALL were enrolled in a phase I trial of vaccination with autologous CD40-ALL. Immunologic reconstitution was measured in a separate cohort of 23 patients with newly diagnosed ALL. Results We successfully prepared autologous vaccines for all nine patients in the phase I trial. CD40-ALL were potent APC, capable of stimulating allogeneic and peptide-specific T cells in vitro. Two patients were vaccinated without adverse events. Five patients died or progressed before vaccination, suggesting that rapid disease progression limits vaccination in patients with relapse disease, thus limiting clinical translation. We therefore sought to identify a window of time for vaccination during which this approach might be feasible. To achieve this end, we evaluated immunological reconstitution in newly diagnosed patients with ALL patients. Despite recovery of myelopoiesis, most patients had profound defects in T, B, and natural killer (NK) cell numbers that failed to recover at any point during therapy. Conclusion Autologous tumor vaccination at a time of ALL relapse is not feasible. Alternative strategies for immunotherapy of ALL may require ex vivo generation of antigen specific T cells and adoptive therapy.
- Published
- 2005
- Full Text
- View/download PDF
15. CXCL13 (BCA-1) is produced by follicular lymphoma cells: role in the accumulation of malignant B cells
- Author
-
Jeffery L. Kutok, Rosalie de Beaumont, Giuliana Strola, Hervé Husson, Elizabeth G. Carideo, Angelo A. Cardoso, Olivier Munoz, Federico Caligaris-Cappio, Joachim L. Schultze, Arnold S. Freedman, and Paolo Ghia
- Subjects
CCL20 ,Follicular dendritic cells ,Cancer research ,CXCL10 ,Germinal center ,Hematology ,CXCL13 ,Biology ,CC chemokine receptors ,CXCL14 ,CXCL16 - Abstract
Follicular lymphomas (FLs) localize in lymphoid tissues and recapitulate the structure of normal secondary follicles. The chemokine/chemokine receptor pair CXCL13/CXCR5 is required for the architectural organization of B cells within lymphoid follicles. In this study, we showed that CXCL13 was secreted by FL cells. FL cells expressed CXCR5 and migrated in response to CXCL13. Furthermore, we observed a synergistic effect between CXCL13 and CXCL12 (SDF-1), a chemokine produced by stromal cells in lymphoid tissues. The production of CXCL13 by FL cells and CXCL12 by stromal cells probably directs and participates in the accumulation of FL cells within specific anatomic sites.
- Published
- 2002
- Full Text
- View/download PDF
16. MCP-1 modulates chemotaxis by follicular lymphoma cells
- Author
-
Serena M. Lugli, Laurence de Leval, Angelo A. Cardoso, Olivier Munoz, Joachim L. Schultze, Donna Neuberg, Hervé Husson, Elizabeth G. Carideo, and Arnold S. Freedman
- Subjects
Chemokine ,Lymphoma, B-Cell ,Receptors, CCR2 ,Translocation, Genetic ,Cell Line ,medicine ,Humans ,CCL17 ,CXCL14 ,Lymphoma, Follicular ,Chemokine CCL2 ,B cell ,Chromosomes, Human, Pair 14 ,Follicular dendritic cells ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Antibodies, Monoclonal ,Drug Synergism ,Chemotaxis ,Hematology ,Dendritic cell ,Flow Cytometry ,Chemokine CXCL12 ,Cell biology ,Chemotaxis, Leukocyte ,medicine.anatomical_structure ,Immunology ,biology.protein ,XCL2 ,Receptors, Chemokine ,Chromosomes, Human, Pair 18 ,Chemokines, CXC ,Dendritic Cells, Follicular - Abstract
The localization and establishment of follicular lymphoma (FL) cells in distinct anatomic sites probably involves chemokine and adhesion receptors on the neoplastic cells and appropriate chemokines and adhesion receptor ligands in the microenvironment. Several chemokines play an important role in normal B-cell trafficking and differentiation. Monocyte chemoattractant protein-1 (MCP-1) is a C-C chemokine that induces chemotaxis of a variety of lymphoid cells through its receptor CCR2. CCR2 is also expressed on B cells, and MCP-1 induces chemotaxis of normal B cells. In this report, we investigated expression and function of CCR2 on FL cells. We found FL cells as well as the t(14; 18)+ B-cell lymphoma line H2 expressed CCR2. MCP-1 potentiated SDF-1-induced chemotaxis of FL cells and H2 cells, but MCP-1 alone did not induce chemotaxis. The specificity of the effects of MCP-1 and SDF-1 was demonstrated by antibody blocking studies. Because FL cells are generally associated with follicular dendritic cells (FDCs), FDCs may be an important source of chemokines. We found that cultured FDCs produced MCP-1, and this production was enhanced by tumour necrosis factor. These data implicate MCP-1 in the migration and localization of FL cells.
- Published
- 2001
- Full Text
- View/download PDF
17. Identification of HER-2/neu immunogenic epitopes presented by renal cell carcinoma and other human epithelial tumors
- Author
-
David Alexandre Gross, Eric Angevin, Pedro Alves, Salem Chouaib, Lee M. Nadler, Sophie Tourdot, Kostas Kosmatopoulos, François A. Lemonnier, Antonio Scardino, Hüseyin Firat, Stéphanie Graff-Dubois, and Angelo A. Cardoso
- Subjects
medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,Biology ,medicine.disease ,Virology ,Epitope ,Virus ,Tumor antigen ,CTL ,Antigen ,medicine ,Cancer research ,Immunology and Allergy ,Cytotoxic T cell - Abstract
HER-2/neu is a tumor-associated antigen overexpressed in a large variety of human tumors. Eight HER-2/neu peptides displaying HLA-A*0201 anchoring motifs were selected and tested for their binding affinity to HLA-A*0201 and their capacity to elicit cytotoxic T lymphocyte (CTL) responses in both HLA-A*0201 transgenic mice and in HLA-A*0201(+) healthy donors. Two high-affinity (p5 and p48) and one intermediate-affinity (p1023) peptides triggered CTL responses in both transgenic mice and humans, comparable to those observed for the well-known HER2/neu dominant peptide p369. CTL induced in transgenic mice lysed HLA-A*0201(+) RMA cells infected with recombinant HER-2/neu but not cells infected with wild-type vaccinia virus. Human CTL lysed HLA-A*0201(+) HER-2/neu(+) tumor cells of different origins (breast, colon, lung and renal cancer) irrespective of the expression levels of HER-2/neu. Importantly, primed CTL specific for these epitopes were detected in freshly isolated tumor-infiltrating lymphocytes from three renal cell carcinoma patients. Therefore, the HER-2/neu peptides p5, p48 and p1023 may be good candidates for immunotherapy of a broad spectrum of tumors, including renal cell carcinoma.
- Published
- 2001
- Full Text
- View/download PDF
18. CD4-induced interaction of primary HIV-1 gp120 glycoproteins with the chemokine receptor CCR-5
- Author
-
Richard T. Wyatt, Craig Gerard, Joseph Sodroski, Nancy Ruffing, Cristina Parolin, Lijun Wu, Hyeryun Choe, Walter Newman, Norma P. Gerard, Alessândra Borsetti, Angelo A. Cardoso, and Elizabeth Desjardin
- Subjects
Receptors, CCR5 ,Chemokine receptor CCR5 ,viruses ,PATHOGENESIS ,C-C chemokine receptor type 7 ,C-C chemokine receptor type 6 ,HIV Antibodies ,HIV Envelope Protein gp120 ,V3 loop ,Binding, Competitive ,Membrane Fusion ,SDF-1 ,Mice ,Chemokine receptor ,Receptors, HIV ,Neutralization Tests ,Tumor Cells, Cultured ,Animals ,Humans ,Receptors, Cytokine ,Binding site ,Chemokine CCL4 ,Chemokine CCL3 ,HUMAN-IMMUNODEFICIENCY-VIRUS ,CXCR4 ,Binding Sites ,Multidisciplinary ,integumentary system ,biology ,fungi ,Antibodies, Monoclonal ,virus diseases ,Macrophage Inflammatory Proteins ,Chemokine receptor binding ,Molecular biology ,Peptide Fragments ,Recombinant Proteins ,CD4 Antigens ,HIV-1 ,biology.protein ,Drosophila ,CCL21 - Abstract
For efficient entry into target cells, primary macrophage-tropic and laboratory-adapted human immunodeficiency viruses type 1 (HIV-1) require particular chemokine receptors, CCR-5 and CXCR-4, respectively, as well as the primary receptor CD4 (refs 1-6). Here we show that a complex of gp120, the exterior envelope glycoprotein, of macrophage-tropic primary HIV-1 and soluble CD4 interacts specifically with CCR-5 and inhibits the binding of the natural CCR-5 ligands, macrophage inflammatory protein (MIP)-1alpha and MIP-1beta (refs 7, 8). The apparent affinity of the interaction between gp120 and CCR-5 was dramatically lower in the absence of soluble CD4. Additionally, in the absence of gp120, an interaction between a two-domain CD4 fragment and CCR-5 was observed. A gp120 fragment retaining the CD4-binding site and overlapping epitopes was able to interact with CCR-5 only if the V3 loop, which can specify HIV-1 tropism and chemokine receptor choice, was also present on the molecule. Neutralizing antibodies directed against either CD4-induced or V3 epitopes on gp120 blocked the interaction of gp12O-CD4 complexes with CCR-5. These results suggest that HIV-1 attachment to CD4 creates a high-affinity binding site for CCR-5, leading to membrane fusion and virus entry.
- Published
- 1996
- Full Text
- View/download PDF
19. Tumor necrosis factor-α and endothelial cells modulate Notch signaling in the bone marrow microenvironment during inflammation
- Author
-
Karen E. Pollok, Joanne E. Price, Angelo A. Cardoso, Jacquenilson Fernandes, Eugenia Cruz, Matthias Clauss, Nadia Carlesso, Sonia Rodríguez, Michael J. Ferkowicz, Christin Mumaw, Filipa Cristina, David T. Scadden, Angelo Chora, Hui Huang, and Luis Fernandez
- Subjects
Lipopolysaccharides ,Cancer Research ,Notch signaling pathway ,Mice, Inbred Strains ,Mice, Transgenic ,Inflammation ,Biology ,Ligands ,Article ,Mice ,Downregulation and upregulation ,Bone Marrow ,Genetics ,medicine ,Animals ,Humans ,Progenitor cell ,Molecular Biology ,Cells, Cultured ,Receptors, Notch ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Endothelial Cells ,Membrane Proteins ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Up-Regulation ,Cell biology ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,Intercellular Signaling Peptides and Proteins ,Tumor necrosis factor alpha ,Bone marrow ,Jagged-2 Protein ,medicine.symptom ,Signal Transduction - Abstract
Objective Homeostasis of the hematopoietic compartment is challenged and maintained during conditions of stress by mechanisms that are poorly defined. To understand how the bone marrow (BM) microenvironment influences hematopoiesis, we explored the role of Notch signaling and BM endothelial cells in providing microenvironmental cues to hematopoietic cells in the presence of inflammatory stimuli. Materials and Methods The human BM endothelial cell line (BMEC) and primary human BM endothelial cells were analyzed for expression of Notch ligands and the ability to expand hematopoietic progenitors in an in vitro coculture system. In vivo experiments were carried out to identify modulation of Notch signaling in BM endothelial and hematopoietic cells in mice challenged with tumor necrosis factor-α (TNF-α) or lipopolysaccharide (LPS), or in Tie2-tmTNF-α transgenic mice characterized by constitutive TNF-α activation. Results BM endothelial cells were found to express Jagged ligands and to greatly support progenitor's colony-forming ability. This effect was markedly decreased by Notch antagonists and augmented by increasing levels of Jagged2. Physiologic upregulation of Jagged2 expression on BMEC was observed upon TNF-α activation. Injection of TNF-α or LPS upregulated three- to fourfold Jagged2 expression on murine BM endothelial cells in vivo and resulted in increased Notch activation on murine hematopoietic stem/progenitor cells. Similarly, constitutive activation of endothelial cells in Tie2-tmTNF-α mice was characterized by increased expression of Jagged2 and by augmented Notch activation on hematopoietic stem/progenitor cells. Conclusions Our results provide the first evidence that BM endothelial cells promote expansion of hematopoietic progenitor cells by a Notch-dependent mechanism and that TNF-α and LPS can modulate the levels of Notch ligand expression and Notch activation in the BM microenvironment in vivo.
- Published
- 2008
- Full Text
- View/download PDF
20. APE1/Ref-1 regulates STAT3 transcriptional activity and APE1/Ref-1-STAT3 dual-targeting effectively inhibits pancreatic cancer cell survival.
- Author
-
Angelo A Cardoso, Yanlin Jiang, Meihua Luo, April M Reed, Safi Shahda, Ying He, Anirban Maitra, Mark R Kelley, and Melissa L Fishel
- Subjects
Medicine ,Science - Abstract
Pancreatic cancer is a largely incurable disease, and increasing evidence supports strategies targeting multiple molecular mediators of critical functions of pancreatic ductal adenocarcinoma cells. Intracellular redox state modulates the activity of various signal transduction pathways and biological processes, including cell survival, drug resistance and responsiveness to microenvironmental factors. Recently, it has been shown that the transcription factor STAT3 is under redox control, but the mechanisms involved in its regulation are unknown. Here, we demonstrate for the first time that STAT3 DNA binding and transcriptional activity is directly regulated by the redox function of the APE1/Ref-1 endonuclease, using overexpression and redox-specific mutational strategies, and gene knockdown. Also, pharmacological blockade of APE1/Ref-1 by the redox-selective inhibitor E3330 abrogates STAT3 DNA binding. Since APE1/Ref-1 also exerts redox control on other cancer-associated transcription factors, we assessed the impact of dual-targeting of STAT3 signaling and APE1/Ref-1 redox on pancreatic cancer cell functions. We observed that disruption of APE1/Ref-1 redox activity synergizes with STAT3 blockade to potently inhibit the proliferation and viability of human PDAC cells. Mechanistically, we show that STAT3-APE1/Ref-1 dual targeting promotes marked tumor cell apoptosis, with engagement of caspase-3 signaling, which are significantly increased in comparison to the effects triggered by single target blockade. Also, we show that STAT3-APE1/Ref-1 dual blockade results in significant inhibition of tumor cell migration. Overall, this work demonstrates that the transcriptional activity of STAT3 is directly regulated by the redox function of APE1/Ref-1, and that concurrent blockade of STAT3 and APE1/Ref-1 redox synergize effectively inhibit critical PDAC cell functions.
- Published
- 2012
- Full Text
- View/download PDF
21. Aberrant expression of functional BAFF-system receptors by malignant B-cell precursors impacts leukemia cell survival.
- Author
-
Sara Maia, Marc Pelletier, Jixin Ding, Yen-Ming Hsu, Stephen E Sallan, Sambasiva P Rao, Lee M Nadler, and Angelo A Cardoso
- Subjects
Medicine ,Science - Abstract
Despite exhibiting oncogenic events, patient's leukemia cells are responsive and dependent on signals from their malignant bone marrow (BM) microenvironment, which modulate their survival, cell cycle progression, trafficking and resistance to chemotherapy. Identification of the signaling pathways mediating this leukemia/microenvironment interplay is critical for the development of novel molecular targeted therapies.We observed that primary leukemia B-cell precursors aberrantly express receptors of the BAFF-system, BAFF-R, BCMA, and TACI. These receptors are functional as their ligation triggers activation of NF-κB, MAPK/JNK, and Akt signaling. Leukemia cells express surface BAFF and APRIL ligands, and soluble BAFF is significantly higher in leukemia patients in comparison to age-matched controls. Interestingly, leukemia cells also express surface APRIL, which seems to be encoded by APRIL-δ, a novel isoform that lacks the furin convertase domain. Importantly, we observed BM microenvironmental cells express the ligands BAFF and APRIL, including surface and secreted BAFF by BM endothelial cells. Functional studies showed that signals through BAFF-system receptors impact the survival and basal proliferation of leukemia B-cell precursors, and support the involvement of both homotypic and heterotypic mechanisms.This study shows an unforeseen role for the BAFF-system in the biology of precursor B-cell leukemia, and suggests that the target disruption of BAFF signals may constitute a valid strategy for the treatment of this cancer.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.