1
|
Atay C, Medina-Echeverz J, Hochrein H, Suter M, Hinterberger M. Armored modified vaccinia Ankara in cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 379:87-142. [PMID: 37541728 DOI: 10.1016/bs.ircmb.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
Cancer immunotherapy relies on unleashing the patient´s immune system against tumor cells. Cancer vaccines aim to stimulate both the innate and adaptive arms of immunity to achieve durable clinical responses. Some roadblocks for a successful cancer vaccine in the clinic include the tumor antigen of choice, the adjuvants employed to strengthen antitumor-specific immune responses, and the risks associated with enhancing immune-related adverse effects in patients. Modified vaccinia Ankara (MVA) belongs to the family of poxviruses and is a versatile vaccine platform that combines several attributes crucial for cancer therapy. First, MVA is an excellent inducer of innate immune responses leading to type I interferon secretion and induction of T helper cell type 1 (Th1) immune responses. Second, it elicits robust and durable humoral and cellular immunity against vector-encoded heterologous antigens. Third, MVA has enormous genomic flexibility, which allows for the expression of multiple antigenic and costimulatory entities. And fourth, its replication deficit in human cells ensures a excellent safety profile. In this review, we summarize the current understanding of how MVA induces innate and adaptive immune responses. Furthermore, we will give an overview of the tumor-associated antigens and immunomodulatory molecules that have been used to armor MVA and describe their clinical use. Finally, the route of MVA immunization and its impact on therapeutic efficacy depending on the immunomodulatory molecules expressed will be discussed.
Collapse
Affiliation(s)
- Cigdem Atay
- Bavarian Nordic GmbH, Fraunhoferstr.13, Planegg, Germany
| | | | | | - Mark Suter
- Prof. em. University of Zurich, Switzerland
| | | |
Collapse
|
2
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
3
|
White D, Cote-Martin A, Bleck M, Garaffa N, Shaaban A, Wu H, Liu D, Young D, Scheer J, Lorenz IC, Nixon A, Fine JS, Byrne FR, Mbow ML, Moreno-Garcia ME. Programmed Cell Death-1 (PD-1) anchoring to the GPI-linked co-receptor CD48 reveals a novel mechanism to modulate PD-1-dependent inhibition of human T cells. Mol Immunol 2023; 156:31-38. [PMID: 36889184 DOI: 10.1016/j.molimm.2023.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023]
Abstract
Activation of PD-1 by anchoring it to Antigen Receptor (AR) components or associated co-receptors represents an attractive approach to treat autoimmune conditions. In this study, we provide evidence that CD48, a common lipid raft and Src kinase-associated coreceptor, induces significant Src kinase-dependent activation of PD-1 upon crosslinking, while CD71, a receptor excluded from these compartments, does not. Functionally, using bead-conjugated antibodies we demonstrate that CD48-dependent activation of PD-1 inhibits proliferation of AR-induced primary human T cells, and similarly, PD-1 activation using PD-1/CD48 bispecific antibodies inhibits IL-2, enhances IL-10 secretion, and reduces NFAT activation in primary human and Jurkat T cells, respectively. As a whole, CD48-dependent activation of PD-1 represents a novel mechanism to fine tune T cell activation, and by functionally anchoring PD-1 with receptors other than AR, this study provides a conceptual framework for rational development of novel therapies that activate inhibitory checkpoint receptors for treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Della White
- Departments of Immunology and Respiratory Research, USA.
| | | | - Marina Bleck
- Departments of Immunology and Respiratory Research, USA
| | | | | | - Helen Wu
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Dongmei Liu
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - David Young
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Justin Scheer
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Ivo C Lorenz
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Andrew Nixon
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Jay S Fine
- Departments of Immunology and Respiratory Research, USA
| | | | - M Lamine Mbow
- Departments of Immunology and Respiratory Research, USA; Cancer Immunology and Immune Modulation, USA
| | | |
Collapse
|
4
|
Consalvo KM, Kirolos SA, Sestak CE, Gomer RH. Sex-Based Differences in Human Neutrophil Chemorepulsion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:354-367. [PMID: 35793910 PMCID: PMC9283293 DOI: 10.4049/jimmunol.2101103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/02/2022] [Indexed: 05/25/2023]
Abstract
A considerable amount is known about how eukaryotic cells move toward an attractant, and the mechanisms are conserved from Dictyostelium discoideum to human neutrophils. Relatively little is known about chemorepulsion, where cells move away from a repellent signal. We previously identified pathways mediating chemorepulsion in Dictyostelium, and here we show that these pathways, including Ras, Rac, protein kinase C, PTEN, and ERK1 and 2, are required for human neutrophil chemorepulsion, and, as with Dictyostelium chemorepulsion, PI3K and phospholipase C are not necessary, suggesting that eukaryotic chemorepulsion mechanisms are conserved. Surprisingly, there were differences between male and female neutrophils. Inhibition of Rho-associated kinases or Cdc42 caused male neutrophils to be more repelled by a chemorepellent and female neutrophils to be attracted to the chemorepellent. In the presence of a chemorepellent, compared with male neutrophils, female neutrophils showed a reduced percentage of repelled neutrophils, greater persistence of movement, more adhesion, less accumulation of PI(3,4,5)P3, and less polymerization of actin. Five proteins associated with chemorepulsion pathways are differentially abundant, with three of the five showing sex dimorphism in protein localization in unstimulated male and female neutrophils. Together, this indicates a fundamental difference in a motility mechanism in the innate immune system in men and women.
Collapse
Affiliation(s)
| | - Sara A Kirolos
- Department of Biology, Texas A&M University, College Station, TX
| | - Chelsea E Sestak
- Department of Biology, Texas A&M University, College Station, TX
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX
| |
Collapse
|
5
|
Dahal A, Parajuli P, Singh SS, Shrestha L, Sonju JJ, Shrestha P, Chatzistamou I, Jois S. Targeting protein–protein interaction for immunomodulation: A sunflower trypsin inhibitor analog peptidomimetic suppresses RA progression in CIA model. J Pharmacol Sci 2022; 149:124-138. [PMID: 35641025 PMCID: PMC9208026 DOI: 10.1016/j.jphs.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/10/2022] [Accepted: 04/18/2022] [Indexed: 11/29/2022] Open
Abstract
Protein–protein interactions (PPI) of co-stimulatory molecules CD2-CD58 are important in the early stage of an immune response, and increased expression of these co-stimulatory molecules is observed in the synovial region of joints in rheumatoid arthritis (RA) patients. A CD2 epitope region that binds to CD58 was grafted on to sunflower trypsin inhibitor (SFTI) template structure to inhibit CD2-CD58 PPI. The peptide was incorporated with an organic moiety dibenzofuran (DBF) in its structure. The designed peptidomimetic was studied for its ability to inhibit CD2-CD58 interactions in vitro, and its thermal and enzymatic stability was evaluated. Stability studies indicated that the grafted peptidomimetic was stable against trypsin cleavage. In vivo studies using the collagen-induced arthritis (CIA) model in mice indicated that the peptidomimetic was able to slow down the progress of arthritis, an autoimmune disease in the mice model. These studies suggest that with the grafting of organic functional groups in the stable peptide template SFTI stabilizes the peptide structure, and these peptides can be used as a template to design stable peptides for therapeutic purposes.
Collapse
Affiliation(s)
- Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA
| | - Pravin Parajuli
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA
| | - Leeza Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA
| | - Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA
| | - Prajesh Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & Immunology (PMI), School of Medicine, USC, SC 6439 Garners Ferry Rd, Columbia, SC, 29208, USA
| | - Seetharama Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe LA, 71201, USA.
| |
Collapse
|
6
|
Al-Aghbar MA, Jainarayanan AK, Dustin ML, Roffler SR. The interplay between membrane topology and mechanical forces in regulating T cell receptor activity. Commun Biol 2022; 5:40. [PMID: 35017678 PMCID: PMC8752658 DOI: 10.1038/s42003-021-02995-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
T cells are critically important for host defense against infections. T cell activation is specific because signal initiation requires T cell receptor (TCR) recognition of foreign antigen peptides presented by major histocompatibility complexes (pMHC) on antigen presenting cells (APCs). Recent advances reveal that the TCR acts as a mechanoreceptor, but it remains unclear how pMHC/TCR engagement generates mechanical forces that are converted to intracellular signals. Here we propose a TCR Bending Mechanosignal (TBM) model, in which local bending of the T cell membrane on the nanometer scale allows sustained contact of relatively small pMHC/TCR complexes interspersed among large surface receptors and adhesion molecules on the opposing surfaces of T cells and APCs. Localized T cell membrane bending is suggested to increase accessibility of TCR signaling domains to phosphorylation, facilitate selective recognition of agonists that form catch bonds, and reduce noise signals associated with slip bonds.
Collapse
Affiliation(s)
- Mohammad Ameen Al-Aghbar
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Ashwin K Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
7
|
Khongthong S, Theapparat Y, Roekngam N, Tantisuwanno C, Otto M, Piewngam P. Characterization and immunomodulatory activity of sulfated galactan from the red seaweed Gracilaria fisheri. Int J Biol Macromol 2021; 189:705-714. [PMID: 34474051 PMCID: PMC10999990 DOI: 10.1016/j.ijbiomac.2021.08.182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Polysaccharides from the red seaweed Gracilaria fisheri possess many functions, which include antioxidant, antiviral, and antibacterial activities. However, detailed data on their immunomodulatory activities are scarce. Here, we isolated sulfated galactans (SG) from G. fisheri. We found that the predominant SG from G. fisheri, termed SG-1, had an estimated molecular mass of 100 kDa and activated murine J774A.1 macrophages via the dectin-1 signaling pathway. Furthermore, we observed enhancement of nitric oxide (NO) secretion, increased expression of inducible nitric oxide synthase (iNOS) mRNA, and increased mRNA levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukins IL-1β and IL-6 by SG-1 in macrophages. Moreover, there was higher expression of intercellular adhesion molecule 1 (ICAM-1) and co-stimulatory molecules (B7-1 and B7-2) mRNA. Treatment with G. fisheri SG-1 at 50 μg/mL generally achieved or exceeded the pro-inflammatory activities of 100 ng/mL lipopolysaccharide. Our study demonstrates immune-stimulatory activities of G. fisheri SG that may be of value for immune-potentiating treatment in humans or livestock.
Collapse
Affiliation(s)
- Sunisa Khongthong
- Faculty of Veterinary Science, Rajamangala University of Technology Srivijaya, Nakhon Si Thammarat, Thailand
| | - Yongyuth Theapparat
- Center of Excellence in Functional Foods and Gastronomy, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Natthrit Roekngam
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Science, Prince of Songkla University, HatYai, Songkhla, Thailand
| | | | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA.
| | - Pipat Piewngam
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
CRISPR-based screens uncover determinants of immunotherapy response in multiple myeloma. Blood Adv 2021; 4:2899-2911. [PMID: 32589729 DOI: 10.1182/bloodadvances.2019001346] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/11/2020] [Indexed: 02/01/2023] Open
Abstract
Cancer cells commonly develop resistance to immunotherapy by loss of antigen expression. Combinatorial treatments that increase levels of the target antigen on the surface of cancer cells have the potential to restore efficacy to immunotherapy. Here, we use our CRISPR interference- and CRISPR activation-based functional genomics platform to systematically identify pathways controlling cell surface expression of the multiple myeloma immunotherapy antigen B-cell maturation antigen (BCMA). We discovered that pharmacologic inhibition of HDAC7 and the Sec61 complex increased cell surface BCMA, including in primary patient cells. Pharmacologic Sec61 inhibition enhanced the antimyeloma efficacy of a BCMA-targeted antibody-drug conjugate. A CRISPR interference chimeric antigen receptor T cells (CAR-T cells) coculture screen enabled us to identify both antigen-dependent and antigen-independent mechanisms controlling response of myeloma cells to BCMA-targeted CAR-T cells. Thus, our study shows the potential of CRISPR screens to uncover mechanisms controlling response of cancer cells to immunotherapy and to suggest potential combination therapies.
Collapse
|
9
|
Orlik C, Deibel D, Küblbeck J, Balta E, Ganskih S, Habicht J, Niesler B, Schröder-Braunstein J, Schäkel K, Wabnitz G, Samstag Y. Keratinocytes costimulate naive human T cells via CD2: a potential target to prevent the development of proinflammatory Th1 cells in the skin. Cell Mol Immunol 2020; 17:380-394. [PMID: 31324882 PMCID: PMC7109061 DOI: 10.1038/s41423-019-0261-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/27/2019] [Indexed: 12/01/2022] Open
Abstract
The interplay between keratinocytes and immune cells, especially T cells, plays an important role in the pathogenesis of chronic inflammatory skin diseases. During psoriasis, keratinocytes attract T cells by releasing chemokines, while skin-infiltrating self-reactive T cells secrete proinflammatory cytokines, e.g., IFNγ and IL-17A, that cause epidermal hyperplasia. Similarly, in chronic graft-versus-host disease, allogenic IFNγ-producing Th1/Tc1 and IL-17-producing Th17/Tc17 cells are recruited by keratinocyte-derived chemokines and accumulate in the skin. However, whether keratinocytes act as nonprofessional antigen-presenting cells to directly activate naive human T cells in the epidermis remains unknown. Here, we demonstrate that under proinflammatory conditions, primary human keratinocytes indeed activate naive human T cells. This activation required cell contact and costimulatory signaling via CD58/CD2 and CD54/LFA-1. Naive T cells costimulated by keratinocytes selectively differentiated into Th1 and Th17 cells. In particular, keratinocyte-initiated Th1 differentiation was dependent on costimulation through CD58/CD2. The latter molecule initiated STAT1 signaling and IFNγ production in T cells. Costimulation of T cells by keratinocytes resulting in Th1 and Th17 differentiation represents a new explanation for the local enrichment of Th1 and Th17 cells in the skin of patients with a chronic inflammatory skin disease. Consequently, local interference with T cell-keratinocyte interactions may represent a novel strategy for the treatment of Th1 and Th17 cell-driven skin diseases.
Collapse
Affiliation(s)
- Christian Orlik
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Daniel Deibel
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Johanna Küblbeck
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Emre Balta
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Sabina Ganskih
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Jüri Habicht
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Beate Niesler
- Institute of Human Genetics, Department of Human Molecular Genetics, and nCounter Core Facility, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Jutta Schröder-Braunstein
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Guido Wabnitz
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany.
| |
Collapse
|
10
|
Agrawal B. New therapeutic targets for cancer: the interplay between immune and metabolic checkpoints and gut microbiota. Clin Transl Med 2019; 8:23. [PMID: 31468283 PMCID: PMC6715761 DOI: 10.1186/s40169-019-0241-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023] Open
Abstract
Transformation and growth of tumor cells are associated with profound alterations in neighbouring cells and their environment, together forming the tumor microenvironment (TME). The TME provides a conducive but complex milieu for the tumors to thrive while incapacitating the immune cells that home there as part of our natural immunosurveillance mechanism. The orchestration of this successful survival strategy by tumor cells is associated with exploitation of numerous metabolic and immune checkpoints, as well as metabolic reprogramming in the tumor cells. Together these form an intricate network of feedback mechanisms that favor the growing tumor. In addition, an ecosystem of microbiota, proximal or distal to tumors, influences the successful survival or elimination of tumor cells mediated by immune cells. Discovery and clinical application of immune checkpoint inhibitors (ICIs) i.e., monoclonal antibodies (mAbs) blocking specific immune checkpoints CTLA-4 and PD-1/PD-L1, have revolutionized therapy of various cancers. However, they are still associated with limited response rates, severe immune-related adverse events, development of resistance, and more serious exacerbation of cancer progression termed hyper-progressive disease. Checkpoint inhibitors only represent a milestone and not the finish-line in the quest for treating and curing cancer. Efforts are underway to investigate and develop inhibitors of other immune as well as metabolic checkpoint molecules. Future therapy for various cancers is projected to target immune and metabolic checkpoints and the microbiota together.
Collapse
Affiliation(s)
- Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
11
|
The Role of Co-Stimulatory Molecules in Chagas Disease. Cells 2018; 7:cells7110200. [PMID: 30405039 PMCID: PMC6262639 DOI: 10.3390/cells7110200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/29/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, is a potentially life-threatening tropical disease endemic to Latin American countries that affects approximately 8 million people. In the chronic phase of the disease, individuals are classified as belonging to the indeterminate clinical form or to the cardiac and/or digestive forms when clinical symptoms are apparent. The relationship between monocytes and lymphocytes may be an important point to help clarify the complexity that surrounds the clinical symptoms of the chronic phase of Chagas disease. The co-stimulatory signals are essential to determining the magnitude of T cell response to the antigen. The signals are known to determine the regulation of subsequent adaptive immune response. However, little is known about the expression and function of these molecules in Chagas disease. Therefore, this review aims to discuss the possible role of main pathways of co-stimulatory molecule-receptor interactions in this pathology that could be crucial to understand the disease dynamics.
Collapse
|
12
|
Agrawal B, Gupta N, Konowalchuk JD. MUC1 Mucin: A Putative Regulatory (Checkpoint) Molecule of T Cells. Front Immunol 2018; 9:2391. [PMID: 30405607 PMCID: PMC6204366 DOI: 10.3389/fimmu.2018.02391] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/26/2018] [Indexed: 01/30/2023] Open
Abstract
T lymphocytes are at the center of inducing an effective adaptive immune response and maintaining homeostasis. T cell responses are initiated through interactions between antigen presenting cells (APCs) and T cells. The type and strength of signals delivered through the T cell receptor (TCR) may modulate how the cells respond. The TCR-MHC (T cell receptor-major histocompatibility complex molecules) complex dictates the specificity, whereas co-stimulatory signals induced by interaction of various accessory cell surface molecules strengthen and optimize T cell responses. Multiple immune regulatory mechanisms brought about by co-inhibitory molecules expressed on T cells play a key role in orchestrating successful and non-damaging immunity. These co-inhibitory molecules are also referred to as initiators of immune check-points or co-inhibitory pathways. Knowledge of co-inhibitory pathways associated with activated T lymphocytes has allowed a better understanding of (a) the inflammatory and anti-inflammatory processes associated with infectious diseases and autoimmune diseases, and (b) mechanisms by which tumors evade immune attack. Many of these regulatory pathways are non-redundant and function in a highly concerted manner. Targeting them has provided effective approaches in treating cancer and autoimmune diseases. For this reason, it is valuable to identify any co-inhibitory molecules that affect these pathways. MUC1 mucin (CD227) has long been known to be expressed by epithelial cells and overexpressed by a multitude of adenocarcinomas. As long ago as 1998 we made a surprising discovery that MUC1 is also expressed by activated human T cells and we provided the first evidence of the role of MUC1 as a novel T cell regulator. Subsequent studies from different laboratories, as well as ours, supported an immuno-regulatory role of MUC1 in infections, inflammation, and autoimmunity that corroborated our original findings establishing MUC1 as a novel T cell regulatory molecule. In this article, we will discuss the experimental evidence supporting MUC1 as a putative regulatory molecule or a “checkpoint molecule” of T cells with implications as a novel biomarker and therapeutic target in chronic diseases such as autoimmunity, inflammation and cancer, and possibly infections.
Collapse
Affiliation(s)
- Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nancy Gupta
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jeffrey D Konowalchuk
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Proteomic analysis of lipopolysaccharide activated human monocytes. Mol Immunol 2018; 103:257-269. [PMID: 30326359 DOI: 10.1016/j.molimm.2018.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/20/2018] [Accepted: 09/25/2018] [Indexed: 12/21/2022]
Abstract
Monocytes are key mediators of innate immunity and comprise an important cellular defence against invading pathogens. However, exaggerated or dysregulated monocyte activation can lead to severe immune-mediated pathology such as sepsis or chronic inflammatory diseases. Thus, detailed insight into the molecular mechanisms of monocyte activation is essential to understand monocyte-driven inflammatory pathologies. We therefore investigated the global protein changes in human monocytes during lipopolysaccharide (LPS) activation to mimic bacterial activation. Purified human monocytes were stimulated with LPS for 17 h and analyzed by state-of-the-art liquid chromatography tandem mass spectrometry (LC-MS/MS). The label-free quantitative proteome analysis identified 2746 quantifiable proteins of which 101 had a statistically significantly different abundance between LPS-stimulated cells and unstimulated controls. Additionally, 143 proteins were exclusively identified in either LPS stimulated cells or unstimulated controls. Functional annotation clustering demonstrated that LPS, most significantly, regulates proteasomal- and lysosomal proteins but in opposite directions. Thus, seven proteasome subunits were upregulated by LPS while 11 lysosomal proteins were downregulated. Both systems are critically involved in processing of proteins for antigen-presentation and together with LPS-induced regulation of CD74 and tapasin, our data suggest that LPS can skew monocytic antigen-presentation towards MHC class I rather than MHC class II. In summary, this study provides a sensitive high throughput protein analysis of LPS-induced monocyte activation and identifies several LPS-regulated proteins not previously described in the literature which can be used as a source for future studies.
Collapse
|
14
|
Li Y, Zhu M, Huo Y, Zhang X, Liao M. Anti-fibrosis activity of combination therapy with epigallocatechin gallate, taurine and genistein by regulating glycolysis, gluconeogenesis, and ribosomal and lysosomal signaling pathways in HSC-T6 cells. Exp Ther Med 2018; 16:4329-4338. [PMID: 30542382 PMCID: PMC6257822 DOI: 10.3892/etm.2018.6743] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/21/2017] [Indexed: 12/24/2022] Open
Abstract
A previous study by our group indicated that combined treatment with taurine, epigallocatechin gallate (EGCG) and genistein protects against liver fibrosis. The aim of the present study was to elucidate the antifibrotic mechanism of this combination treatment using isobaric tag for relative and absolute quantification (iTRAQ)-based proteomics in an activated rat hepatic stellate cell (HSC) line. In the present study, HSC-T6 cells were incubated with taurine, EGCG and genistein, and cellular proteins were extracted and processed for iTRAQ labeling. Quantification and identification of proteins was performed using two-dimensional liquid chromatography coupled with tandem mass spectrometry. Proteomic analysis indicated that the expression of 166 proteins were significantly altered in response to combination treatment with taurine, EGCG and genistein. A total 76 of these proteins were upregulated and 90 were downregulated. Differentially expressed proteins were grouped according to their association with specific Kyoto Encyclopedia of Genes and Genomes pathways. The results indicated that the differentially expressed proteins hexokinase-2 and lysosome-associated membrane glycoprotein 1 were associated with glycolysis, gluconeogenesis and lysosome signaling pathways. The expression of these proteins was validated using western blot analysis; the expression of hexokinase-2 was significantly decreased and the expression of lysosome-associated membrane glycoprotein 1 was significantly increased in HSC-T6 cells treated with taurine, EGCG and genistein compared with the control, respectively (P<0.05). These results were in accordance with the changes in protein expression identified using the iTRAQ approach. Therefore, the antifibrotic effect of combined therapy with taurine, EGCG and genistein may be associated with the activation of several pathways in HSCs, including glycolysis, gluconeogenesis, and the ribosome and lysosome signaling pathways. The differentially expressed proteins identified in the current study may be useful for treatment of liver fibrosis in the future.
Collapse
Affiliation(s)
- Yan Li
- Guangxi University Library, Guangxi University, Nanning, Guangxi 530004, P.R. China
| | - Min Zhu
- Guangxi University Library, Guangxi University, Nanning, Guangxi 530004, P.R. China
| | - Yani Huo
- Medical Scientific Research Centre, Key Laboratory of High-Incidence-Tumor Prevention and Treatment, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Xuerong Zhang
- Medical Scientific Research Centre, Key Laboratory of High-Incidence-Tumor Prevention and Treatment, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Ming Liao
- Medical Scientific Research Centre, Key Laboratory of High-Incidence-Tumor Prevention and Treatment, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
15
|
El Menshawy N, Eissa M, Abdeen HM, Elkhamisy EM, Joseph N. CD58; leucocyte function adhesion-3 (LFA-3) could be used as a differentiating marker between immune and non-immune thyroid disorders. ACTA ACUST UNITED AC 2018; 27:721-727. [PMID: 29706856 PMCID: PMC5910489 DOI: 10.1007/s00580-018-2657-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 02/05/2018] [Indexed: 12/02/2022]
Abstract
The link between Graves’ disease (GD) and Hashimoto’s thyroiditis (HT) has been debated for decades due to the shared pathological and immunological components. Immune intolerance and inappropriate immune reaction against self-thyroid cells are distinctive features of both diseases, but definitive data for the clinical presentation of autoimmune thyroid disease remains unclear. To analyse the expression of T-regulatory cells, CD58, the CD4/CD8 ratio and the neutrophil/lymphocyte ratio and to determine if these parameters could be used as differentiating markers between auto- and non-immune thyroid diseases, 75 patients were enrolled in this study—40 with autoimmune thyroid disease (HT and GD ), 15 with non-immune thyroid disease, and 20 healthy controls. Multicolour flow cytometry was used to analyse CD58, T-regulatory cells (Treg) expressing CD4, CD25, HLA-DR and CD8 using different stained fluorescent labelled monoclonal antibodies. The neutrophils and lymphocyte ratio was also measured. Lower expression of Treg with higher expression of CD58 (LFA-3) was found in the autoimmune diseases when compared with the non-immune and control groups. ROC analysis showed that CD58 with sensitivity 88% and specificity 100% with cut-off value more than or equal to 29.9 indicates Hashimoto’s disease, while lower value indicates colloid goitre, and higher or equal to 29.84 indicates Graves’ disease and lower indicates colloid goitre with 100% sensitivity and specificity. CD58 could be used as differentiating marker between immune and non-immune thyroid disorders.
Collapse
Affiliation(s)
- Nadia El Menshawy
- 1Clinical Pathology Department, Hematology Unit, Mansoura Medical School, Mansoura University, Mansoura, Egypt
| | - Mohammed Eissa
- 2Clinical Pathology Department, Faculty of Medicine, Zagazig University and King Khalid University, Zagazig, Egypt
| | - Hanaa M Abdeen
- 3Biochemistry Department, Mansoura Medical School, Mansoura University, Mansoura, Egypt
| | - Enas M Elkhamisy
- 4Internal Medicine Department, Specialized Medicine Hospital, Mansoura Medical School, Mansoura University, Mansoura, Egypt
| | - Nabil Joseph
- 5Community Medicine Department, Mansoura Medical School, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Molecular mechanisms and biomedical applications of glucosamine as a potential multifunctional therapeutic agent. Life Sci 2016; 152:21-9. [PMID: 27012765 DOI: 10.1016/j.lfs.2016.03.028] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/30/2022]
Abstract
Glucosamine and its acetylated derivative, N-acetyl glucosamine, are naturally occurring amino sugars found in human body. They are important components of glycoproteins, proteoglycans and glycosaminoglycans. Scientific studies have supported that glucosamine has the beneficial pharmacological effects to relieve osteoarthritis symptoms. Glucosamine can also be as a promising candidate for the prevention and/or treatment of some other diseases due to its anti-oxidant and anti-inflammatory activities. Most of its function is exerted by modulation of inflammatory responses especially through Nuclear Factor-κB (NF-κB) that can control inflammatory cytokine production and cell survival. In this review, we present a concise update on additional new therapeutic applications of glucosamine including treatment of cardiovascular disease, neurological deficits, skin disorders, cancer and the molecular mechanistic rationale for these uses. This article will also examine safety profile and adverse effects of glucosamine in human.
Collapse
|
17
|
Rocha-Perugini V, Sánchez-Madrid F, Martínez Del Hoyo G. Function and Dynamics of Tetraspanins during Antigen Recognition and Immunological Synapse Formation. Front Immunol 2016; 6:653. [PMID: 26793193 PMCID: PMC4707441 DOI: 10.3389/fimmu.2015.00653] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/18/2015] [Indexed: 12/31/2022] Open
Abstract
Tetraspanin-enriched microdomains (TEMs) are specialized membrane platforms driven by protein–protein interactions that integrate membrane receptors and adhesion molecules. Tetraspanins participate in antigen recognition and presentation by antigen-presenting cells (APCs) through the organization of pattern-recognition receptors (PRRs) and their downstream-induced signaling, as well as the regulation of MHC-II–peptide trafficking. T lymphocyte activation is triggered upon specific recognition of antigens present on the APC surface during immunological synapse (IS) formation. This dynamic process is characterized by a defined spatial organization involving the compartmentalization of receptors and adhesion molecules in specialized membrane domains that are connected to the underlying cytoskeleton and signaling molecules. Tetraspanins contribute to the spatial organization and maturation of the IS by controlling receptor clustering and local accumulation of adhesion receptors and integrins, their downstream signaling, and linkage to the actin cytoskeleton. This review offers a perspective on the important role of TEMs in the regulation of antigen recognition and presentation and in the dynamics of IS architectural organization.
Collapse
Affiliation(s)
- Vera Rocha-Perugini
- Servicio de Inmunología, Instituto de Investigación Sanitaria La Princesa, Hospital de la Princesa, Madrid, Spain; Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Instituto de Investigación Sanitaria La Princesa, Hospital de la Princesa, Madrid, Spain; Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Gloria Martínez Del Hoyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , Madrid , Spain
| |
Collapse
|
18
|
Bazzi S, Modjtahedi H, Mudan S, Akle C, Bahr GM. Analysis of the immunomodulatory properties of two heat-killed mycobacterial preparations in a human whole blood model. Immunobiology 2015; 220:1293-304. [PMID: 26253276 DOI: 10.1016/j.imbio.2015.07.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/15/2015] [Accepted: 07/23/2015] [Indexed: 12/20/2022]
Abstract
The significant role played by mycobacteria in modulating immune responses through enhancing the crosstalk between innate and adaptive immunity has been highlighted in several studies. Owing to their unique antigenic profile, heat killed (HK) preparations of rapid-growing mycobacteria, currently undergoing clinical development, have been assessed as adjuvant therapy in various diseases. The purpose of this study is to investigate the regulation of leukocyte surface receptors, in whole blood from healthy donors, following in vitro stimulation with HK Mycobacterium vaccae (M. vaccae) or M. obuense. We have demonstrated the ability of both mycobacterial preparations to target monocytes and neutrophils and to regulate the surface expression of selected adhesion receptors, antigen-presenting and costimulatory receptors, pattern recognition receptors, complement and Fc receptors, as well as cytokine/chemokine receptors. Toll-like receptors (TLRs) 1 and 2 were also shown to be involved in mediating the M. obuense-induced upregulation of selected surface receptors on monocytes. Whole blood stimulation with M. vaccae or M. obuense resulted in a significant increase in the secretion of a specific set of cytokines and chemokines. Both mycobacterial preparations induced strong antigen-specific proliferative responses in peripheral blood mononuclear cells. Collectively, our data shows that M. vaccae and M. obuense have the potential to act as potent immunomodulators. Future research based on these findings may reveal novel immune pathways induced by these preparations with potential implication for their use in diverse immunotherapeutic approaches.
Collapse
Affiliation(s)
- Samer Bazzi
- School of Life Sciences, Faculty of Science, Engineering and Computing, Kingston University, Kingston upon Thames, Surrey KT1 2EE, United Kingdom; Faculty of Medicine and Medical Sciences, University of Balamand, 33 Amioun, Al Kurah, Lebanon.
| | - Helmout Modjtahedi
- School of Life Sciences, Faculty of Science, Engineering and Computing, Kingston University, Kingston upon Thames, Surrey KT1 2EE, United Kingdom.
| | - Satvinder Mudan
- Division of Clinical Sciences, St George's, University of London, London SW170RE, United Kingdom; Department of Academic Surgery, Royal Marsden Hospital, London SW3 6JJ, United Kingdom.
| | - Charles Akle
- The London Clinic, London W1G 6JA, United Kingdom.
| | - Georges M Bahr
- Faculty of Medicine and Medical Sciences, University of Balamand, 33 Amioun, Al Kurah, Lebanon.
| |
Collapse
|
19
|
McMichael AJ. Excimer laser: a module of the alopecia areata common protocol. J Investig Dermatol Symp Proc 2013; 16:S77-S79. [PMID: 24326569 DOI: 10.1038/jidsymp.2013.31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Alopecia areata (AA) is an autoimmune condition characterized by T cell-mediated attack of the hair follicle. The inciting antigenic stimulus is unknown. A dense perbulbar lymphocytic infiltrate and reproducible immunologic abnormalities are hallmark features of the condition. The cellular infiltrate primarily consists of activated T lymphocytes and antigen-presenting Langerhans cells. The xenon chloride excimer laser emits its total energy at the wavelength of 308 nm and therefore is regarded as a "super-narrowband" UVB light source. Excimer laser treatment is highly effective in psoriasis, another T cell-mediated disorder that shares many immunologic features with AA. The excimer laser is superior in inducing T cell apoptosis in vitro compared with narrowband UVB, with paralleled improved clinical efficacy. The excimer laser has been used successfully in patients with AA. In this context, evaluation of the potential benefit of 308-nm excimer laser therapy in the treatment of AA is clinically warranted. Herein, the use of a common treatment protocol with a specifically designed module to study the outcome of excimer laser treatment on moderate-to-severe scalp AA in adults is described.
Collapse
Affiliation(s)
- Amy J McMichael
- Department of Dermatology, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| |
Collapse
|
20
|
Kim HR, Jeon BH, Lee HS, Im SH, Araki M, Araki K, Yamamura KI, Choi SC, Park DS, Jun CD. IGSF4 is a novel TCR ζ-chain-interacting protein that enhances TCR-mediated signaling. ACTA ACUST UNITED AC 2011; 208:2545-60. [PMID: 22084409 PMCID: PMC3256964 DOI: 10.1084/jem.20110853] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Immunoglobulin superfamily member 4 (IGSF4) is a known ligand of CRTAM, a receptor expressed in activated NKT and CD8(+) T cells, but its function in T cell immunity has not been elucidated. In this study, we show that IGSF4 directly interacts with the T cell receptor (TCR) ζ-chain and enhances TCR signaling by enhancing ζ-chain phosphorylation. Ectopic overexpression of IGSF4 enhances TCR-mediated T cell activation. In contrast, IGSF4 knockdown shows a dramatic decrease in markers associated with T cell activation compared with those in control small interfering RNA. The transmembrane domain is essential for TCR ζ-chain association and clustering to the immunological synapse, and the ectodomain is associated with T cell interaction with antigen-presenting cells (APCs). IGSF4-deficient mice have impaired TCR-mediated thymocyte selection and maturation. Furthermore, these mice reveal attenuated effector T cell functions accompanied by defective TCR signaling. Collectively, the results indicate that IGSF4 plays a central role in T cell functioning by dual independent mechanisms, control of TCR signaling and control of T cell-APC interaction.
Collapse
Affiliation(s)
- Hye-Ran Kim
- Immune Synapse Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Teschner D, Wenzel G, Distler E, Schnürer E, Theobald M, Neurauter AA, Schjetne K, Herr W. In vitro stimulation and expansion of human tumour-reactive CD8+ cytotoxic T lymphocytes by anti-CD3/CD28/CD137 magnetic beads. Scand J Immunol 2011; 74:155-64. [PMID: 21517928 DOI: 10.1111/j.1365-3083.2011.02564.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adoptive immunotherapy with tumour-reactive CD8(+) cytotoxic T lymphocytes (CTLs) requires efficient in vitro approaches allowing the expansion of CTLs to large numbers prior infusion. Here, we investigated the antigen-independent activation and the expansion of human T cells in peripheral blood mononuclear cells (PBMCs) and in tumour-reactive CTLs using Dynabeads coated with monoclonal antibodies to CD3 and to the costimulatory molecules CD28 and CD137 (4-1BB). T cells in PBMCs showed an increased expansion rate of 15- to 17-fold during a 2-week culture period using antibody-conjugated beads with interleukin-2 (IL-2) added versus IL-2 alone. No significant difference between CD3/CD28 beads and CD3/CD28/CD137 beads was observed (P = 0.4). In contrast, expansion of tumour-reactive CD8(+) CTLs over 2 weeks was more efficient using CD3/CD28/CD137 beads (14.4-fold ± 1.2) compared with CD3/CD28 beads (10.6-fold ± 0.7) (P = 0.03) and matched well to the control arm using weekly stimulation with tumour cells. Although all modes of in vitro stimulation decreased the expression of central memory markers CD62L and CCR7 on CTLs, bead-activated cultures expressed consistently higher levels than tumour-stimulated cultures. CTLs analysed after bead-induced expansion versus weekly tumour stimulation showed equal IFN-γ production in ELISPOT assay. Furthermore, cytotoxicity assays demonstrated an either unchanged or slightly reduced capability of tumour cell lysis for antigen-independent stimulated CTLs versus those that maintained on weekly tumour stimulation, regardless of which type of beads was used. Our data suggest that the conjugation of anti-CD137 antibodies to conventional CD3/CD28 beads results in a minor but significant increase in the expansion capacity for tumour-reactive CD8(+) CTLs.
Collapse
Affiliation(s)
- D Teschner
- Department of Medicine III, Hematology and Oncology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Gokhale A, Weldeghiorghis TK, Taneja V, Satyanarayanajois SD. Conformationally constrained peptides from CD2 to modulate protein-protein interactions between CD2 and CD58. J Med Chem 2011; 54:5307-19. [PMID: 21755948 DOI: 10.1021/jm200004e] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cell adhesion molecule CD2 and its ligand CD58 provide good examples of protein-protein interactions in cells that participate in the immune response. To modulate the cell adhesion interaction, peptides were designed from the discontinuous epitopes of the β-strand region of CD2 protein. The two strands were linked by a peptide bond. β-Strands in the peptides were nucleated by inserting a β-sheet-inducing (D)-Pro-Pro sequence or a dibenzofuran (DBF) turn mimetic with key amino acid sequences from CD2 protein that binds to CD58. The solution structures of the peptides (5-10) were studied by NMR and molecular dynamics simulations. The ability of these peptides to inhibit cell adhesion interaction was studied by E-rosetting and lymphocyte epithelial assays. Peptides 6 and 7 inhibit the cell adhesion activity with an IC(50) of 7 and 11 nM, respectively, in lymphocyte epithelial adhesion assay. NMR and molecular modeling results indicated that peptides 6 and 7 exhibited β-hairpin structure in solution.
Collapse
Affiliation(s)
- Ameya Gokhale
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana 71201, United States
| | | | | | | |
Collapse
|
23
|
Findlay L, Sharp G, Fox B, Ball C, Robinson CJ, Bird C, Stebbings R, Eastwood D, Wadhwa M, Poole S, Thorpe R, Thorpe SJ. Endothelial cells co-stimulate peripheral blood mononuclear cell responses to monoclonal antibody TGN1412 in culture. Cytokine 2011; 55:141-51. [DOI: 10.1016/j.cyto.2011.03.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 03/18/2011] [Indexed: 12/01/2022]
|
24
|
Arlen PM, Gulley JL. Current perspectives in prostate cancer vaccines. Anticancer Agents Med Chem 2010; 9:1052-7. [PMID: 19719454 DOI: 10.2174/187152009789735062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 08/17/2009] [Indexed: 11/22/2022]
Abstract
The use of vaccines as a potential therapeutic modality for the treatment of cancer has been extensively studied. Recent advances include identification and characterization of tumor-associated antigens, novel vaccine delivery systems, and the combination of vaccines with immune stimulants and other therapeutic modalities. Immunotherapy as a modality for treatment of prostate cancer has received significant attention. There are several characteristics of prostate cancer that make it an ideal target for immunotherapy. Prostate cancer's relative indolence allows sufficient time to generate immune responses, which may take weeks or months to mount. In addition, prostate cancer-associated antigens direct the immune response to prostate cancer cells, thus sparing normal vital tissue. This review focuses on promising new vaccines and novel perspectives in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Philip M Arlen
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1750, USA
| | | |
Collapse
|
25
|
Lombardi V, Singh AK, Akbari O. The role of costimulatory molecules in allergic disease and asthma. Int Arch Allergy Immunol 2009; 151:179-89. [PMID: 19786798 DOI: 10.1159/000242355] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The prevalence of allergic diseases has increased rapidly in recent years. It is well established that the deleterious allergic response is initiated by T-cell recognition of major histocompatibility class II-peptide complexes at the surface of antigen-presenting cells. While this first signal gives antigen specificity to the adaptive immune response, a second nonspecific costimulatory signal is required by T cells to become fully activated. This signal is provided by interactions between antigen-presenting cells and T cells through molecules borne at the surfaces of the two cell types. Depending on the type of molecules involved, this secondary signal can promote the development of an inflammatory allergic reaction or may favor immune regulation. Several molecules of the B7 family (CD80, CD86, PD-1, ICOS, CTLA-4) and tumor necrosis factor receptor family (OX40, CD30, 4-1BB, Fas, CD27, CD40) play an important role in delivering costimulatory signals in early and late phases of allergic response. Therefore, costimulatory molecules involved in promotion or prevention of allergic immune responses are potential targets for the development of novel therapeutic approaches. This review aims to recapitulate our current understanding of the relationship between allergic diseases and costimulatory molecules.
Collapse
Affiliation(s)
- Vincent Lombardi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033-9605, USA
| | | | | |
Collapse
|
26
|
Abstract
Tumor immunotherapy harnesses the potential of the host immune system to recognize and eradicate neoplastic tissue. The efficiency of the immune system in mediating tumor regression depends on the induction of antigen-specific T-cell responses through physiologic immune surveillance, priming by vaccination, or following adoptive transfer of T-cells. Although a variety of tumor-associated antigens have been identified and many immunotherapeutic strategies have been tested, objective clinical responses are rare. The reasons for this include the inability of current immunotherapy approaches to generate efficient T-cell responses, the presence of regulatory cells that inhibit T-cell responses, and other tumor escape mechanisms. The activation of effector T-cells depends on interactions between the T-cell receptor (TCR) and cognate antigen presented as peptides within the major histocompatibility complex (MHC) and costimulatory signals delivered by CD28, which binds to B7.1 and B7.2. More recently, several new molecular receptors and ligands have been identified that integrate into stimulatory or inhibitory activity for T-cells. These signals have been loosely associated with the costimulatory molecules but actually represent a diverse group of molecular pathways that have unique and overlapping functions. This review will focus on these pathways and emphasize their role in mediating T-cell activation for the purpose of enhancing tumor immunotherapy. As we gain a better understanding of the molecular and cellular consequences of T-cell signaling through the costimulatory pathways, a more rational approach to the activation or inhibition of T-cell responses can be developed for the treatment of cancer and other immune-mediated diseases.
Collapse
Affiliation(s)
- Robert C Ward
- The Tumor Immunology Laboratory, Division of Surgical Oncology, Columbia University, New York, New York 10032, USA
| | | |
Collapse
|
27
|
Toor AA, Stiff PJ, Nickoloff BJ, Rodriguez T, Klein JL, Gordon KB. Alefacept in corticosteroid refractory graft versus host disease: Early results indicate promising activity. J DERMATOL TREAT 2009; 18:13-8. [PMID: 17365261 DOI: 10.1080/09546630601121045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Steroid refractory graft versus host disease (GVHD) presents a significant therapeutic challenge due to the limited efficacy and safety of second-line treatments. Three patients with extensively pretreated, refractory GVHD were treated with a targeted anti-T-cell agent, alefacept, and demonstrated rapid and clinically significant improvement in their GVHD, facilitating tapering of corticosteroids. The pathological and immunohistochemical findings of GVHD also improved, validating our clinical impression. These preliminary findings indicate that alefacept may have beneficial activity in GVHD warranting further study.
Collapse
Affiliation(s)
- Amir A Toor
- Bone Marrow Transplantation Program, Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | | | | | | | | | | |
Collapse
|
28
|
Madan RA, Arlen PM, Mohebtash M, Hodge JW, Gulley JL. Prostvac-VF: a vector-based vaccine targeting PSA in prostate cancer. Expert Opin Investig Drugs 2009; 18:1001-11. [PMID: 19548854 PMCID: PMC3449276 DOI: 10.1517/13543780902997928] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prostvac is a prostate cancer vaccine regimen consisting of a recombinant vaccinia vector as a primary vaccination, followed by multiple booster vaccinations employing a recombinant fowlpox vector. Both vectors contain the transgenes for prostate-specific antigen (PSA) and multiple T-cell co-stimulatory molecules (TRICOM). The PSA-TRICOM vaccines infect antigen-presenting cells (APCs) and generate proteins that are expressed on the surface of the APCs in an immune context. The interaction of these APCs with T cells initiates a targeted immune response and T cell-mediated tumor cell destruction. Preliminary clinical trials have indicated negligible toxicity, and Phase II trials have suggested a survival benefit after treatment with Prostvac, especially in patients with indolent disease characteristics. Preclinical and clinical data indicate that radiation, hormonal therapy, and chemotherapy may be combined with Prostvac to enhance the vaccine's efficacy. Additional strategies are in development to further enhance the clinical benefits of Prostvac, and a Phase III trial is being planned in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Ravi A. Madan
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip M. Arlen
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mahsa Mohebtash
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James L. Gulley
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
29
|
Arlen PM, Mohebtash M, Madan RA, Gulley JL. Promising novel immunotherapies and combinations for prostate cancer. Future Oncol 2009; 5:187-96. [PMID: 19284377 PMCID: PMC2705770 DOI: 10.2217/14796694.5.2.187] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The field of therapeutic cancer vaccines is currently in a state of active preclinical and clinical investigation, and certain novel therapies involving tumor immunotherapy have recently come to the forefront of prostate cancer research. While no therapeutic cancer vaccine has yet been approved by the US FDA, recent findings have demonstrated that new paradigms of combination therapies involving vaccines, employed in clinical trials with appropriate design and end points, may ultimately lead to cancer vaccines being used to treat various malignancies. Several characteristics of prostate cancer make it an ideal target for immunotherapy. Its relative indolence allows sufficient time to generate immune responses, which usually take weeks or months to mount. In addition, prostate cancer-associated antigens direct the immune response to prostate cancer cells, thus sparing normal tissue. This review focuses on the future of promising new vaccines and novel perspectives in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Philip M Arlen
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Mahsa Mohebtash
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Ravi A Madan
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - James L Gulley
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, 10 Center, Drive, Building 10, Room 8B09, MSC, 1750, Bethesda, MD 20892-1750, USA, Tel.: +1 301 435 2956, Fax: +1 301 480 5094,
| |
Collapse
|
30
|
Abstract
Gene transfer into mature T cells with gammaretroviral vectors requires prestimulation, as only mitotic cells are susceptible to integration of the gammaretroviral proviral genome. Costimulation via the CD3/ TCR complex and a second costimulatory molecule, such as CD28 was found to better preserve functionality of the T lymphocytes during ex vivo expansion than stimulation with anti-CD3 alone. The protocols described here for prestimulation and transduction of human and murine T cells with gammaretroviral vectors were optimized for high-level gene transfer and maximum yield of functional T lymphocytes.
Collapse
Affiliation(s)
- Sebastian Newrzela
- Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | | | | |
Collapse
|
31
|
Requirement of L-selectin for gammadelta T lymphocyte activation and migration during allergic pleurisy: co-relation with eosinophil accumulation. Int Immunopharmacol 2009; 9:303-12. [PMID: 19135179 DOI: 10.1016/j.intimp.2008.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 12/01/2008] [Accepted: 12/02/2008] [Indexed: 11/20/2022]
Abstract
Intra-thoracic antigenic challenge (ovalbumin, 12.5 microg/cavity) led to increased numbers of gammadelta T lymphocytes in pleural cavities, blood and thoracic lymph nodes in sensitized mice within 48 h. Part of these cells expressed CD62L, which increased on gammadelta T cell surfaces obtained from lymph nodes after ovalbumin (OVA) challenge. Selectin blockade by fucoidan pre-treatment (10 mg/kg, i.v.) impaired in vivo increase in CD25(+) and c-fos(+) gammadelta T cell numbers in lymph nodes, indicating a role for selectins on gammadelta T lymphocyte activation and proliferation. In vivo selectin blockade by fucoidan or alpha-CD62L mAb (200 microg/mice, i.p.) also inhibited OVA-induced gammadelta T cell accumulation in pleural cavities. Confirming the direct effect of CD62L on gammadelta T cell transmigration, the migration of i.v. adoptively-transferred CFSE-labeled gammadelta T lymphocytes into pleural cavities of challenged recipient mice was impaired by fucoidan ex vivo treatment. It is noteworthy that eosinophil influx was also impaired in those mice, indicating that reduced eosinophil migration by CD62L in vivo blockade depended on gammadelta T cell migration via CD62L molecules. Accordingly, pleural gammadelta T lymphocytes from fucoidan-treated mice presented reduced OVA-induced IL-5 and CCL11 production. Supporting these data, the depletion of Vgamma4 T lymphocytes, which are pulmonary gammadelta T cells, decreased OVA-induced eosinophil influx into allergic site. Such results demonstrate that CD62L is crucial for the activation of gammadelta T cells in lymph nodes, for their migration into inflamed tissue and for the modulation of eosinophil influx during allergic response.
Collapse
|
32
|
Taylor A, Verhagen J, Akkoç T, Wenig R, Flory E, Blaser K, Akdis M, Akdis CA. IL-10 suppresses CD2-mediated T cell activation via SHP-1. Mol Immunol 2008; 46:622-9. [PMID: 18952289 DOI: 10.1016/j.molimm.2008.07.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 07/11/2008] [Accepted: 07/27/2008] [Indexed: 12/31/2022]
Abstract
Interleukin (IL)-10 is an essential suppressive cytokine and plays a key role in peripheral T cell tolerance to allergens, autoantigens, transplantation antigens and tumor antigens. However, the molecular mechanisms of direct T cell suppression by IL-10 are not fully understood. Here, we demonstrate that IL-10 directly inhibits CD2 signaling in T cells. T cell stimulation via CD2 alone induces activation and proliferation, when endogenous IL-10 sources are eliminated from cultures. IL-10 utilizes the src-homology-2 domain containing tyrosine phosphatase (SHP-1) to directly suppress T cell activation. The role of SHP-1 in IL-10-mediated suppression of CD2 co-stimulation on T cells is demonstrated by using dominant-negative SHP-1 over-expressing T cells and silencing endogenous SHP-1 by small inhibitory RNA. Findings are confirmed using both SHP-1-deficient mice and IL-10-deficient mice. CD2-induced proliferation is suppressed by exogenous IL-10 in IL-10-deficient, but not SHP-1-deficient murine T cells. In conclusion, SHP-1-mediated inhibition of CD2 signaling represents a novel mechanism for direct T cell suppression by IL-10.
Collapse
Affiliation(s)
- Alison Taylor
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ghosh N, Singh PN, Kumar V. Novel immunobiologics for psoriasis. Indian J Pharmacol 2008; 40:95-102. [PMID: 20040934 PMCID: PMC2792605 DOI: 10.4103/0253-7613.42300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 05/28/2008] [Accepted: 06/21/2008] [Indexed: 11/04/2022] Open
Abstract
Psoriasis is one of the most common human skin diseases and is considered to have key genetic contributions. It is characterized by excessive growth and aberrant differentiation of keratinocytes, but is reversible with appropriate therapy with the possibilities of recurrence. The trigger of the keratinocyte response is thought to be the activation of the cellular immune system with T cells, dendritic cells and various immune related cytokines and chemokines being implicated in pathogenesis. Immunosuppressants like cyclosporine and methotrexate were used earlier in the treatment of psoriasis, however their use was associated with severe adverse effects due to down regulation of immune system. The most recent advances in therapies for psoriasis target specific immune components of psoriasis and promise to have high therapeutic efficacy with low adverse effects. This review focuses on the novel therapies aimed to specifically modulate the dysregulated immune system with minimal adverse effects.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Pharmacology Research Laboratory, Department of Pharmaceutics, Institute of Technology, Banaras Hindu University, Varanasi-221 005, India
| | | | | |
Collapse
|
34
|
Berndt U, Philipsen L, Bartsch S, Wiedenmann B, Baumgart DC, Hämmerle M, Sturm A. Systematic high-content proteomic analysis reveals substantial immunologic changes in colorectal cancer. Cancer Res 2008; 68:880-8. [PMID: 18245490 DOI: 10.1158/0008-5472.can-07-2923] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The immune system is a significant determinant of epithelial tumorigenesis, but its role in colorectal cancer pathogenesis is not well understood. The function of the immune system depends upon the integrity of the protein network environment, and thus, we performed MELC immunofluorescence microscopy focusing on the lamina propria. By analyzing structurally intact tissues from colorectal cancer, ulcerative colitis, and healthy colonic mucosa, we used this unique and novel highly multiplexed robotic-imaging technology, which allows visualizing dozens of proteins simultaneously, and explored the toponome in colorectal cancer mucosa for the first time. We identified 1,930 motifs that distinguish control from colorectal cancer tissue. In colorectal cancer, the number of activated T cells is increased, explained by a lack of bax, caspase-3, and caspase-8. Whereas CD4(+)CD25(+) T cells are decreased and are, other than in ulcerative colitis, not activated, cytotoxic T cells are significantly increased in colorectal cancer. Furthermore, the number of activated human lymphocyte antigen (HLA)-DR(+) T-cells is increased in colorectal cancer, pointing to an altered antigen presentation. In colorectal cancer, CD3(+)CD29(+) expression and assembly of the LFA-1 and LFA-3 receptor are differentially changed, indicating a distinct regulation of T-cell adhesion in colorectal cancer. We also identified increased numbers of natural killer and CD44(+) cells in the colorectal cancer mucosa and nuclear factor-kappaB as regulator of apoptosis in these cell populations. High-content proteomic analysis showed that colorectal cancer induces a tremendous modification of protein expression profiles in the lamina propria. Thus, topological proteomic analysis may help to unravel the role of the adaptive immune system in colorectal cancer and aid the development of new antitumor immunotherapy approaches.
Collapse
Affiliation(s)
- Uta Berndt
- Department of Medicine, Division of Gastroenterology and Hepatology, Charité-Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Hui G, Hashimoto C. The requirement of CD80, CD86, and ICAM-1 on the ability of adjuvant formulations to potentiate antibody responses to a Plasmodium falciparum blood-stage vaccine. Vaccine 2007; 25:8549-56. [PMID: 18006124 DOI: 10.1016/j.vaccine.2007.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Revised: 09/18/2007] [Accepted: 10/01/2007] [Indexed: 01/04/2023]
Abstract
Many adjuvants are known to enhance expression of co-stimulatory and adhesion molecules secondarily to the activation of immune cells. Whether interactions via these molecules are obligatory in adjuvants' ability to potentiation vaccine immunogenicity is less clear. We investigated the ability of eight adjuvant formulations to potentiate the immunogenicity of a malaria vaccine in mice deficient in the prominent co-stimulatory molecules, CD80 and CD86; and the adhesion ligand, ICAM-1. While no adjuvants could bypass co-stimulatory requirements, more formulations exhibited dependency for CD86 than for CD80. In CD80 or CD86 KO mice, formulations with the saponin derivative, QS21 could efficiently default to the other B7 molecule. This effect was dominant over other adjuvant constituents. The requirement for ICAM-1 could be readily bypassed using adjuvant formulations containing immunomodulators; whereas this was not the case with emulsion-type adjuvants in which reduction in adjuvanticity was associated with decreases in antigen-specific IFN-gamma responses. These studies may help to guide the formulation of vaccine adjuvants to maintain effectiveness in hosts with altered immunological environment that often result from infections.
Collapse
Affiliation(s)
- George Hui
- Department of Tropical Medicine and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, HI 96813, United States.
| | | |
Collapse
|
36
|
Zhou H, Zou P, Chen ZC, You Y. A novel vicious cycle cascade in tumor chemotherapy. Med Hypotheses 2007; 69:1230-3. [PMID: 17555885 DOI: 10.1016/j.mehy.2007.03.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 03/28/2007] [Indexed: 01/17/2023]
Abstract
Dendritic cells (DCs) play a critical role in both the cellular and humoral immune response to tumor cells. The participation of co-stimulator molecules on DCs is of vital importance for full activation of T cells. Conventional tumor chemotherapy cannot only induce apoptosis of malignant cells, but also do harm to benign DCs. Expression levels of co-stimulatory molecules (especially CD80 and CD86) are downregulated during anticancer drug treatment. These DCs are defective in their antigen-presenting function being lack of sufficient co-stimulatory molecules. Immune defective DCs provide negative immune signals to CD4+ and CD8+ T cells. Thereby these T cells are induced to become tolerant to the tumor cells, and function as T regulatory/suppressor cells to induce tolerogenic DCs by cell-cell contact. These DCs tolerized to tumor antigens may further render tolerogenicity to many more T cells via intercellular contact, and thus lead to a vicious cycle cascade. This model allowed us to envisage modified strategies of antitumor chemotherapy. We may use anticancer drugs with higher specificity to tumor cells, or in combination with immunopotentiator to disrupt the vicious cycle cascade, and expect better clinical result.
Collapse
Affiliation(s)
- Hao Zhou
- Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science & Technology, 1277 Jiefang Road, Wuhan, Hubei 430022, China
| | | | | | | |
Collapse
|
37
|
Madan RA, Arlen PM, Gulley JL. PANVAC-VF: poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert Opin Biol Ther 2007; 7:543-54. [PMID: 17373905 DOI: 10.1517/14712598.7.4.543] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PANVAC is a cancer vaccine therapy delivered through two viral vectors--recombinant vaccinia and recombinant fowlpox--which are given sequentially. Both vectors contain transgenes for the tumor-associated antigens epithelial mucin 1 and carcinoembryonic antigen, which are altered or overexpressed in most carcinomas. The vectors also contain transgenes for three human T cell costimulatory molecules required to enhance immune response: B7.1, intracellular adhesion molecule-1 and leukocyte function-associated antigen-3. PANVAC is injected subcutaneously and processed by the body's antigen-presenting cells. Preclinical studies have demonstrated the efficacy of PANVAC in inducing both carcinoembryonic antigen- and mucin 1-specific cytotoxic T lymphocyte responses in vitro and in murine models. Other strategies that enhance the immune response include the use of granulocyte-macrophage colony-stimulating factor and a prime-boost administration sequence. Clinical trials have demonstrated PANVAC's safety and its ability to induce antigen-specific T cell responses. Early clinical trials are evaluating PANVAC alone and in combination with conventional chemotherapy and/or radiation. Studies to date hold promise for the use of PANVAC as a means to stimulate the immune system against malignancies and to provide clinical benefit.
Collapse
Affiliation(s)
- Ravi A Madan
- Clinical Immunotherapy Group, National Cancer Institute (NCI), Laboratory of Tumor Immunology and Biology, National Institutes of Health (NIH), 10 Center Drive, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
38
|
Abstract
The development of cancer vaccines, aimed to enhance the immune response against a tumor, is a promising area of research. A better understanding of both the molecular mechanisms that govern the generation of an effective immune response and the biology of a tumor has contributed to substantial progress in the field. Areas of intense investigation in cancer immunotherapy will be discussed here, including: (1) the discovery and characterization of novel tumor antigens to be used as targets for vaccination; (2) the investigation of different vaccine-delivery modalities such as cellular-based vaccines, protein- and peptide-based vaccines, and vector-based vaccines; (3) the characterization of biological adjuvants to further improve the immunogenicity of a vaccine; and (4) the investigation of multimodal therapies where vaccines are being combined with other oncological treatments such as radiation and chemotherapy. A compilation of data from preclinical studies conducted in vitro as well as in animal models is presented here. The results from these studies would certainly support the development of new vaccination strategies toward cancer vaccines with enhanced clinical efficacy.
Collapse
Affiliation(s)
- Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
39
|
Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology 2006; 117:433-42. [PMID: 16556256 PMCID: PMC1782242 DOI: 10.1111/j.1365-2567.2006.02321.x] [Citation(s) in RCA: 510] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Specific immune suppression and induction of tolerance are essential processes in the regulation and circumvention of immune defence. The balance between allergen-specific type 1 regulatory (Tr1) cells and T helper (Th) 2 cells appears to be decisive in the development of allergy. Tr1 cells consistently represent the dominant subset specific for common environmental allergens in healthy individuals. In contrast, there is a high frequency of allergen-specific interleukin-4 (IL-4)-secreting T cells in allergic individuals. Allergen-specific immunotherapy can induce specific Tr1 cells that abolish allergen-induced proliferation of Th1 and Th2 cells, as well as their cytokine production. Tr1 cells utilize multiple suppressor mechanisms, such as IL-10 and transforming growth factor-beta (TGF-beta) as secreted cytokines and various surface molecules, such as cytotoxic T-lymphocyte antigen 4 and programmed death-1. IL-10 only inhibits T cells stimulated by low numbers of triggered T-cell receptors, which depend on CD28 costimulation. IL-10 inhibits CD28 tyrosine phosphorylation, preventing the binding of phosphatidylinositol 3-kinase p85 and consequently inhibiting the CD28 signalling pathway. In addition, IL-10 and TGF-beta secreted by Tr1 cells skew the antibody production from immunoglobulin E (IgE) towards the non-inflammatory isotypes IgG4 and IgA, respectively. Induction of antigen-specific Tr1 cells can thus re-direct an inappropriate immune response against allergens or auto-antigens using a broad range of suppressor mechanisms.
Collapse
Affiliation(s)
- Alison Taylor
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland
| | | | | | | | | |
Collapse
|
40
|
Tzu J, Mamelak AJ, Sauder DN. Current advancements in the treatment of psoriasis: Immunobiologic agents. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.cair.2006.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Linton PJ, Lustgarten J, Thoman M. T cell function in the aged: Lessons learned from animal models. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.cair.2006.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
42
|
Wanschitz F, Nell A, Patruta S, Wagner A, Ewers R. Influence of three currently used bone replacing materials on the in vitro proliferation of human peripheral blood mononuclear cells. Clin Oral Implants Res 2005; 16:570-4. [PMID: 16164463 DOI: 10.1111/j.1600-0501.2005.01150.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES A cell culture system for biocompatibility testing of bone grafting materials is described. We investigated the in vitro viability and proliferative response of peripheral blood mononuclear cells (PBMC) from 10 healthy donors in the presence of three materials currently used for bone grafting: Algipore, Bio-Oss and Bone Source, for immunologic biocompatibility testing. MATERIAL AND METHODS PBMC isolated from venous blood from 10 healthy donors were incubated for 4 days with each bone replacing material, in the presence and absence of interleukin-2 (IL-2). After 4 days, H3-thymidine was added for 18 h and the incorporated radioactivity was measured with a beta-plate counter. RESULTS Basal PBMC counts were 152.9+/-66.2 counts per minute (c.p.m.) (mean+/-SD), in the presence of 0.4 U IL-2/well 206.5+/-83 c.p.m. were measured. With Algipore and Bio-Oss, which are deproteinized bone replacing materials, the proliferation rate of PBMC with IL-2 was not significantly modified: for Algipore 151+/-51 c.p.m./+IL-2 188.8+/-62 c.p.m., for Bio-Oss 144.5+/-64.9 c.p.m./+IL-2 176.3+/-71.23 c.p.m. For Bone Source 164.2+/-80.4/+IL-2 188.3+/-81 c.p.m. were measured. CONCLUSION This in vitro experiment indicates, that the investigated bone replacing materials are not acting as specific antigens/haptens and are not generating increased proliferative responses of human PBMC from healthy donors. Even with IL-2, that induces proliferation of T lymphocytes, which encountered their specific antigen, the proliferation rate of PBMC from healthy donors was not increased after incubation with this bone grafting materials.
Collapse
Affiliation(s)
- Felix Wanschitz
- Department of Oral and Maxillofacial Surgery, University of Vienna, Medical School, General Hospital, Vienna, Austria.
| | | | | | | | | |
Collapse
|
43
|
Vancheri C, Mastruzzo C, Trovato-Salinaro E, Gili E, Lo Furno D, Pistorio MP, Caruso M, La Rosa C, Crimi C, Failla M, Crimi N. Interaction between human lung fibroblasts and T-lymphocytes prevents activation of CD4+ cells. Respir Res 2005; 6:103. [PMID: 16159396 PMCID: PMC1243241 DOI: 10.1186/1465-9921-6-103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Accepted: 09/13/2005] [Indexed: 12/31/2022] Open
Abstract
Background T lymphocytes are demonstrated to play an important role in several chronic pulmonary inflammatory diseases. In this study we provide evidence that human lung fibroblasts are capable of mutually interacting with T-lymphocytes leading to functionally significant responses by T-cells and fibroblasts. Methods Human lung fibroblast were co-cultured with PMA-ionomycin activated T-CD4 lymphocytes for 36 hours. Surface as well as intracellular proteins expression, relevant to fibroblasts and lymphocytes activation, were evaluated by means of flow cytometry and RT-PCR. Proliferative responses of T lymphocytes to concanavalin A were evaluated by the MTT assay. Results In lung fibroblasts, activated lymphocytes promote an increase of expression of cyclooxygenase-2 and ICAM-1, expressed as mean fluorescence intensity (MFI), from 5.4 ± 0.9 and 0.7 ± 0.15 to 9.1 ± 1.5 and 38.6 ± 7.8, respectively. Fibroblasts, in turn, induce a significant reduction of transcription and protein expression of CD69, LFA-1 and CD28 in activated lymphocytes and CD3 in resting lymphocytes. In activated T lymphocytes, LFA-1, CD28 and CD69 expression was 16.6 ± 0.7, 18.9 ± 1.9 and 6.6 ± 1.3, respectively, and was significantly reduced by fibroblasts to 9.4 ± 0.7, 9.4 ± 1.4 and 3.5 ± 1.0. CD3 expression in resting lymphocytes was 11.9 ± 1.4 and was significantly reduced by fibroblasts to 6.4 ± 1.1. Intracellular cytokines, TNF-alpha and IL-10, were evaluated in T lymphocytes. Co-incubation with fibroblasts reduced the number of TNF-alpha positive lymphocytes from 54,4% ± 6.12 to 30.8 ± 2.8, while IL-10 positive cells were unaffected. Finally, co-culture with fibroblasts significantly reduced Con A proliferative response of T lymphocytes, measured as MTT absorbance, from 0.24 ± 0.02 nm to 0.16 ± 0.02 nm. Interestingly, while the activation of fibroblasts is mediated by a soluble factor, a cognate interaction ICAM-1 mediated was demonstrated to be responsible for the modulation of LFA-1, CD28 and CD69. Conclusion Findings from this study suggest that fibroblasts play a role in the local regulation of the immune response, being able to modulate effector functions of cells recruited into sites of inflammation.
Collapse
Affiliation(s)
- Carlo Vancheri
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Claudio Mastruzzo
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Elisa Trovato-Salinaro
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Elisa Gili
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Debora Lo Furno
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Maria P Pistorio
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Massimo Caruso
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Cristina La Rosa
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Claudia Crimi
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Marco Failla
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| | - Nunzio Crimi
- Department of Internal and Specialistic Medicine, Section of Respiratory Medicine, University of Catania, Catania, 95125, Italy
| |
Collapse
|
44
|
Salucci V, Lena AM, Ciliberto G, Scarselli E, La Monica N. Adenovirus Transduction and Culture Conditions Affect the Immunogenicity of Murine Dendritic Cells. Scand J Immunol 2005; 62:206-17. [PMID: 16179007 DOI: 10.1111/j.1365-3083.2005.01658.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adenovirus vectors encoding carcinoembryonic antigen (Ad-CEA) or costimulatory molecules CD80, intercellular adhesion molecule-1 (ICAM-1) and leucocyte function-associated antigen-3 (LFA-3) (Ad-STIM) were used to transduce murine bone marrow-derived dendritic cells (BMDC). BMDC were characterized for expression of activation markers and for their ability to elicit protective immunity against MC38-CEA tumours in wildtype and CEA-transgenic (CEA-tg) mice. To determine optimal culture conditions, studies were conducted using BMDC cultured in heterologous bovine serum or autologous mouse serum. Transduction of cells grown in presence of heterologous serum increased the expression of costimulatory molecules, major histocompatibility complex class II, of IL-6 and IL-12. Upon vaccination, tumour protection was not specific and was observed also with untransduced cells. Transduced BMDC cultured in the presence of autologous serum showed low expression of the activation markers, did not express IL-6 and had reduced ability to stimulate T-cell proliferation. Nonetheless, CEA-specific CD8+ T-cell response was enhanced upon coinfection of Ad-STIM and Ad-CEA in both mouse strains, although this immune response was not sufficient to protect CEA-tg mice from tumour challenge. These studies support the use of BMDC transduced with Ad vectors encoding tumour antigens for cancer immunotherapy and demonstrate that culture conditions greatly affect the immunological properties of these cells.
Collapse
Affiliation(s)
- V Salucci
- Istituto di Ricerche di Biologia Molecolare (IRBM), Pomezia, Italy
| | | | | | | | | |
Collapse
|
45
|
Palena C, Foon KA, Panicali D, Yafal AG, Chinsangaram J, Hodge JW, Schlom J, Tsang KY. Potential approach to immunotherapy of chronic lymphocytic leukemia (CLL): enhanced immunogenicity of CLL cells via infection with vectors encoding for multiple costimulatory molecules. Blood 2005; 106:3515-23. [PMID: 16081691 PMCID: PMC1895050 DOI: 10.1182/blood-2005-03-1214] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a disease of CD5(+) B lymphocytes (designated as CLL cells) that are inefficient antigen-presenting cells. Their poor ability to present antigens to the T cells, largely due to an inadequate costimulatory capacity, is manifested as a failure to stimulate proliferation of both allogeneic and autologous T cells. We have investigated the ability of in vitro manipulated CLL cells, via hyperexpression of a triad of costimulatory molecules (B7-1, intercellular adhesion molecule 1 [ICAM-1], and leukocyte-function-associated antigen 3 [LFA-3], designated TRICOM), to stimulate effective antitumor T-cell responses. A recombinant modified vaccinia virus strain Ankara (MVA), which is a highly attenuated, replication-impaired virus variant, was successfully used to infect and deliver the simultaneous expression of the 3 human costimulatory molecules in TRICOM on the surface of the CLL cells. Proliferation of allogeneic and autologous T cells was observed when MVA-TRICOM-infected CLL cells were used as stimulators in proliferation assays. Cytotoxic T lymphocytes, generated in vitro by stimulation of autologous T cells with MVA-TRICOM-infected CLL cells, showed cytotoxicity against unmodified/uninfected CLL cells. Therefore, our findings suggest that the use of CLL cells infected ex vivo with MVA-TRICOM or direct injection of MVA-TRICOM in patients with CLL has potential for the immunotherapy of CLL.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, CD/genetics
- Antigens, CD/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Proliferation
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/immunology
- Female
- Genetic Vectors
- Humans
- Immunotherapy/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Male
- Middle Aged
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Vaccinia virus
Collapse
Affiliation(s)
- Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Schlom J, Tsang KY, Kantor JA, Abrams SI, Zaremba S, Greiner J, Hodge JW. Cancer vaccine development. Expert Opin Investig Drugs 2005; 7:1439-52. [PMID: 15992042 DOI: 10.1517/13543784.7.9.1439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A new era involving the evaluation of recombinant cancer vaccines has begun with the concurrent emergence of insights and technologies in the fields of molecular biology and immunology. These advances include: The identification and cloning of an array of genes associated with the neoplastic process, such as oncogenes, suppressor genes, genes encoding oncofoetal antigens and tissue-lineage determinants. The development of a variety of viral and bacterial vectors to deliver and present gene products. The identification of numerous T-cell costimulatory molecules and an understanding of their mode of action. The cloning and analysis of the modes of action of an array of cytokines and other immunomodulatory molecules. More sophisticated knowledge of the mode(s) of antigen presentation and T-cell activation. One current challenge in cancer therapy is the delineation of strategies toward the rational design and implementation of recombinant vaccines that will be of therapeutic benefit to cancer patients and/or members of groups at high risk for specific neoplasias. Numerous concepts are emerging in this regard. The study of immunologic intervention using laboratory animal models demonstrates that no one approach will prevail for all cancer types or, perhaps, for the various stages of the neoplastic process of a given tumour type. The immunological role(s) of CD8+, CD4+, natural killer and other cell types, as well as the roles of antibodies, must all be taken into consideration. This article reviews some of the strategies currently undergoing evaluation toward the development of recombinant vaccines for several carcinoma types.
Collapse
Affiliation(s)
- J Schlom
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 8B07, Bethesda, MD 20892-1750, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Morse MA, Clay TM, Hobeika AC, Osada T, Khan S, Chui S, Niedzwiecki D, Panicali D, Schlom J, Lyerly HK. Phase I study of immunization with dendritic cells modified with fowlpox encoding carcinoembryonic antigen and costimulatory molecules. Clin Cancer Res 2005; 11:3017-24. [PMID: 15837756 DOI: 10.1158/1078-0432.ccr-04-2172] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To determine the safety and immunologic and clinical efficacy of a dendritic cell vaccine modified to hyperexpress costimulatory molecules and tumor antigen. EXPERIMENTAL DESIGN In this phase I study, we administered one or two cycles of four triweekly s.c./intradermal injections of ex vivo generated dendritic cells modified with a recombinant fowlpox vector encoding carcinoembryonic antigen (CEA) and a triad of costimulatory molecules [rF-CEA(6D)-TRICOM]. Controls consisted of immature dendritic cells loaded with tetanus toxoid and a HLA A2-restricted peptide derived from cytomegalovirus pp65 protein. RESULTS Fourteen patients (11 with colorectal cancer and 3 with non-small cell lung cancer) were enrolled and 12 completed at least one cycle of immunization. There were no grade 3/4 toxicities directly referable to the immunizations. One patient had a decrease in the CEA level from 46 to 6.8 and a minor regression in adenopathy that occurred several months after completion of the immunizations. Five other patients were stable through at least one cycle of immunization (3 months). Direct analysis of peripheral blood mononuclear cells using the ELISpot assay showed an increase in the frequency of CEA-specific T cells in 10 patients (range, 10-541 CEA-specific cells/10(5) peripheral blood mononuclear cells). There was a trend for a greater peak frequency of CEA-specific T cells among those with either a minor response or a stable disease following at least one cycle of therapy. A second cycle was not associated with higher T-cell frequencies. Cytokine flow cytometry showed CEA-specific immune response among both CD4(+) and CD8(+) T cells in all immune responders. CONCLUSION This immunization strategy is safe and activates potent CEA-specific immune responses.
Collapse
Affiliation(s)
- Michael A Morse
- Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Slavin-Chiorini DC, Catalfamo M, Kudo-Saito C, Hodge JW, Schlom J, Sabzevari H. Amplification of the lytic potential of effector/memory CD8+ cells by vector-based enhancement of ICAM-1 (CD54) in target cells: implications for intratumoral vaccine therapy. Cancer Gene Ther 2004; 11:665-80. [PMID: 15354200 DOI: 10.1038/sj.cgt.7700741] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We demonstrated that enhanced expression of the costimulatory molecules CD80, CD54 and CD48 (designated rF-TRICOM) on target cells, as delivered via a recombinant fowlpox vector, results in an increased state of stimulation of CD8+ T cells, and consequent increased lysis of target cells. CTL studies in conjunction with antibody-blocking studies demonstrated that the enhanced effector activity of these CD8+ T cells is mediated mainly through CD54. Intracellular staining of CD8+ cells that interact with target cells infected with rF-TRICOM showed that they contain higher amounts of perforin and have a higher level of perforin message. Enhanced expression of costimulatory molecules (specifically CD54) on target cells using rF-TRICOM vectors also leads to the formation of stable conjugates/synapses between targets and T cells. The interaction of T cells with target cells that overexpress costimulatory molecules upon infection with rF-TRICOM leads to enhanced signaling through Lck, ZAP70, and STAT-1 in CD8+ T cells and heightened lytic activity of CD8+ cells through the formation of a greater number of immunological synapses. This, in turn, leads to enhanced signaling in T cells. Finally, studies were conducted in mice in which CEA is a self-antigen in an attempt to understand the potential clinical relevancy of intratumoral vaccine therapy. Mice were transplanted subcutaneously with CEA expressing tumors. Intratumoral (i.t.) vaccination was administered 8 days post tumor transplant. Mice vaccinated i.t. with rF-TRICOM demonstrated significantly reduced tumor growth and 40% of the mice had complete tumor regression. The antitumor effects were further improved by the addition of tumor antigen (CEA) in the vaccination by utilizing rF-CEA/TRICOM, with 80% of the mice experiencing complete tumor regression. These studies thus support the concept of intratumoral vaccination employing vectors expressing costimulatory molecules.
Collapse
Affiliation(s)
- Dale C Slavin-Chiorini
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | |
Collapse
|
49
|
Kalamasz D, Long SA, Taniguchi R, Buckner JH, Berenson RJ, Bonyhadi M. Optimization of Human T-Cell Expansion Ex Vivo Using Magnetic Beads Conjugated with Anti-CD3 and Anti-CD28 Antibodies. J Immunother 2004; 27:405-18. [PMID: 15314550 DOI: 10.1097/00002371-200409000-00010] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
T-cell receptor engagement and accompanying costimulatory signals control the level of activation and functional potential of individual T cells. The authors previously developed a novel technology in which human T cells are activated and expanded in culture ex vivo using anti-CD3 and anti-CD28 monoclonal antibodies covalently linked to superparamagnetic beads (Xcyte Dynabeads). In this study the addition of N-acetyl L-cysteine (NAC) to the cultures markedly increased the expansion of T cells from human peripheral blood mononuclear cells without diminishing cell function. NAC increased the rate of T-cell division, reduced apoptosis, and increased the percentage of antigen-specific memory T cells in the cultures. The effect of varying the ratio of beads to T cells (1:10-10:1) at culture initiation was also evaluated. Polyclonal T cells were expanded at all bead-to-T cell ratios tested (range 1:10-10:1). While high bead-to-T cell ratios (5:1 and 10:1) deleted, low ratios (1:10 and 1:5) preserved memory T cells directed against cytomegalovirus, Epstein-Barr virus, and influenza virus antigens. Adding more anti-CD3/anti-CD28 beads during the culture led to further expansion of T cells. Experiments also revealed that reducing the amount of anti-CD3 antibodies relative to the amount of anti-CD28 antibodies on the beads favored the proliferation of antigen-specific T cells. In summary, these data indicate that T cell-stimulating effects of anti-CD3/anti-CD28 beads can be further manipulated to control the expansion of antigen-specific memory T cells and can be used to rapidly expand antigen-specific T cells ex vivo for potential clinical applications.
Collapse
Affiliation(s)
- Dale Kalamasz
- Xcyte Therapies, Inc., Seattle, Washington 98104, USA
| | | | | | | | | | | |
Collapse
|
50
|
Kohm AP, Miller SD. Role of ICAM-1 and P-selectin expression in the development and effector function of CD4+CD25+regulatory T cells. J Autoimmun 2003; 21:261-71. [PMID: 14599851 DOI: 10.1016/s0896-8411(03)00117-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Dynamic regulatory mechanisms prevent autoreactive T cell activation. Upon T cell receptor crosslinking, CD4+CD25+ T regulatory (T(R)) cells block both the proliferation and cytokine production of CD4+CD25- effector cells in an apparent antigen non-specific manner. Within the T(R)population, L-selectin (CD62L)(hi)T(R)cells have been described as more efficient suppressors of T cell proliferation than CD62L(low)T(R)cells. We have previously reported that CD4+CD25+CD62L(hi)T(R)cells express elevated levels of two additional adhesion molecules, ICAM-1 (CD54) and P-selectin (CD62P) in comparison to non-T(R)cells. In the current study, we investigated the functional contribution of CD54 and CD62P expression to the suppressive phenotype of T(R)cells both in vitro and in vivo. While the CD4+CD25+ T(R)cell population was demonstrated to be significantly larger in CD62P-/- mice than in wild-type C57BL/6 mice, CD62P-/- T(R)cell function was deficient in vitro, but not in vivo. Interestingly, we detected no deficiencies in T(R)cell numbers or effector function in CD54-/- mice suggesting that T(R)cells may differ from effector CD4+ T cells in the requirement for CD54 expression within the immunological synapse. Collectively, these findings indicate that CD62P may influence T(R)cell differentiation/development and that T(R)cell activation occurs independently of CD54 expression.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigen-Presenting Cells/immunology
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CD3 Complex/immunology
- CD4 Antigens/analysis
- CD4 Antigens/immunology
- Cell Count
- Coculture Techniques
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Glycoproteins/immunology
- Glycoproteins/pharmacology
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Interferon-gamma/metabolism
- Interleukins/metabolism
- L-Selectin/immunology
- L-Selectin/metabolism
- Lectins, C-Type
- Lymph Nodes/cytology
- Lymphocyte Activation/immunology
- Lymphocyte Transfusion
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein
- P-Selectin/genetics
- P-Selectin/immunology
- Peptide Fragments/immunology
- Peptide Fragments/pharmacology
- Receptors, Interleukin-2/analysis
- Receptors, Interleukin-2/immunology
- Spleen/cytology
- T-Lymphocytes/chemistry
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Th1 Cells/immunology
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Adam P Kohm
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|