Register for free to join our community of investors and share your ideas. You will also get access to streaming quotes, interactive charts, trades, portfolio, live options flow and more tools.
Register for free to join our community of investors and share your ideas. You will also get access to streaming quotes, interactive charts, trades, portfolio, live options flow and more tools.
mojo,
thanks.
Haynes, the Director of CHAVI, now states, in work funded by CHAVI, that PS is responsible for blunting the immune system's fight against HIV.
(see bottom underlined paragraph of included snapshot)
Tony Fauci (Director of NIAID), says again, at the same Seattle event, that the goal is now to
"control acute viremia and lower viral setpoint".
(it's like his new mantra lately).
-----
j
CHAVI 012, April '07.
STUDY OBJECTIVES AND DESIGN
- snip -
Measuring T and B cell responses in mucosal samples of AHI, early HIV
infection, and control subjects, for study of residual immune cell activity after
massive apoptosis during AHI. This is to include cytokine production, antibody
production in vitro, and T cell functions in vitro. These samples will also
provide material for the CHAVI effort to develop mucosal assays to monitor
human vaccine clinical trials.
---------
BACKGROUND AND RATIONALE
-snip-
Innate immune responses can be activated very rapidly in response to pathogen
exposure or infection, and play important roles both in containment of early
pathogen replication and promotion of induction of the adaptive response. The
nature of the innate response in AHI and its interaction with the adaptive response
may be among the factors involved in determining the set point of plasma viremia
and the preservation of central memory CD4+ T cells, two factors that have been
shown to be critical metrics of disease prognosis. A key implication of this
hypothesis is the suggestion that innate responses may be harnessed to form an
important component of vaccine-elicited protective immunity, a setting in which
rapid triggering of effector functions following exposure to HIV or HIV-infected
cells is likely to be critical.
Despite the potential importance of innate responses in shaping events in primary
HIV infection, little is known about the innate response at this time. One study
reported transient detection of IFN-α in the serum in AHI, with levels peaking
prior to the peak in acute plasma viremia, indicative of a rapid type 1 IFN
response (von Sydow et al, 1991). Notably, there is a marked reduction in the
number of both plasmacytoid dendritic cells (pDCs, important IFN-producing
cells) and myeloid DCs (mDCs) in the blood during AHI (Pacanowski et al,
2001); whether this reflects recruitment to lymphoid tissues and/or loss is
unknown. The extent of infection of DC subsets during AHI is also unclear, as is
their functional capacity, although one study reported reduced costimulatory
molecule expression on DCs in lymphoid tissues in AHI (Lore et al, 2002). In
contrast to DCs, elevated numbers of natural killer (NK) cells are present in the
blood during AHI (Alter et al, 2005), although the functions exhibited by NK
cells at this time have not been characterized. Together, these studies show that
innate responses are activated early in AHI, but leave many important questions
about innate functions at this time unanswered. It is proposed to characterize the
innate effector mechanisms activated in primary HIV infection and explore their
role in control of early virus replication; and to address whether HIV impairs
aspects of innate immunity in AHI to promote its persistence. This will facilitate
the design of vaccination strategies to target key effector pathways and/or
circumvent infection-associated impairments in innate functions.
A broad array of novel assays will be pursued and developed in the CHAVI both
in support of this protocol, and through the use of the unique specimens made
available by this protocol. Flow cytometric studies will identify and characterize
the gut lymphocyte B cell, basophil, NK cell, T cell, dendritic, and monocyte/
macrophage populations. Immunglobulin levels will be assayed by sensitive
ELISA and surface plasmon resonance assays. B and T specificities will be
characterized by tetramer assays. Both microarray expression RNA analysis and
proteomic profiling will be done on B and T cell populations. Mucosal antibody
polyspecificity will be assessed by Luminex autoimmune antibody assays (Athena
platform). Latent infection of resting T cells will be quantified by limiting-
dilution assays of resting CD4 cells. Acutely expressed cytokines and apoptotic
microparticles hypothesized to play a key role in AHI pathogenesis will be
assayed. The CHAVI panel of cytokines including alpha IFN, TNF alpha, IL10,
TGF-beta, as well as a Luminex panel of T and B regulatory cytokines, and
plasma microparticles will be quantitated and as well phenotyped by flow
cytometry for source of cells of origin. Plasma TRAIL, FAS Ligand and TNFRII
will be determined to monitor apoptosis levels.
http://72.14.209.104/search?q=cache:BCKueWKqWTIJ:www.chavi.org/wysiwyg/downloads/CHAVI_012_protocol_v1.pdf+%22CHAVI+012%22&hl=en&ct=clnk&cd=1&gl=us&client=firefox-a
---------
j
free, CHAVI is Bavi,
Glad somebody noticed :)
Part of the apoptotic process is the blebbing off of microparticles from the membrane of the apoptotic cell.
It is now understood that apoptotic cells, AND the shed PS-exposing microparticles from those apoptotic cells, downregulate the immune response, via exposed PS.
This is true in viral infections, all cancers, etc...
It's what spreads the immunosuppressive environment.
In the info I posted this morning, Haynes and Pisetsky (remember all the microparticle papers I keep posting from Pisetsky?....) are saying that the exposed PS looks to be what's specifically responsible for allowing (via the downregulated immune respone) HIV to get a strong foot-hold. (Which should come as no surprise to those who read my posts of the work of Schlegel, Shilyansky, Henderson, Henson, and many others).
Obviously implying that PS is a darn attractive target in the early period of HIV infection, (which is exactly what Letvin's monkey work is about).
Lately Tony Fauci keeps saying that they hope to be able to get the immune system to fight harder/react quicker in the early period of HIV infection, thereby achieving a lower "set-point"/plateau of virus, which correlates directly with length of time to AIDS.
Duh, I wonder how they'll do that...
Remember the slide I often post, back in march, Haynes speaking of the planned monkey work, and I quote:
"With Norm Letvin, a protection trial is planned to determine if anti-PS antibodies can prevent infection, or early viral destruction of the immune system in acute SIV infection."
What work/paper would come next now after THIS presentation???
Haynes:
" PS+ apoptotic cells and microparticles have been reported to suppress antigen-specific immune responses
"suggests the hypothesis that immune cell apoptosis in the very earliest stages of acute HIV-1 infection may delay the onset of potentially protective anti-HIV immune responses".
SO WAHT DO YA DO ABOUT THAT??????......
The PS triggered immune-turn-off is what's responsible for the proliferation/pathogenesis of cancers, viruses, parasites, etc.
It carries implications for therapy against all viruses, cancers, tuberculosis, malaria, ...
and- CHAVI is Bavi....
j
highlighted -
MUST READ HAYNES -
http://www.hivvaccineenterprise.org/_dwn/Poster_Sessions.pdf
j
steve b,
Yes, they've mentioned finishing up the 1A trial around yr end.
They've also mentioned a few times that the FDA loosened up the inclusion/exclusion criteria which would allow for much easier patient enrollment.
Here is a slide from Brekken's recent Bavi presentation-
j
That's an interesting thought. Dr Ibrahim (MD Anderson) is doing breast cancer after all. I would think that once the MD Anderson (1A trial) is done, and bavi proves to be safe, then they could start adding cohorts in America (MD Anderson) to the current phase 2 trials, or announce additional trials at MDA. I'm really looking forward to the bavi/radiation trial that's been hinted at...
j
new - B cell battles...
Virol. 2007 Oct 17
HIV-1 gp41 Antibodies That Mask Membrane Proximal Region Epitopes: Antibody Binding Kinetics, Induction And Potential For Regulation in Acute Infection.
Alam SM, Scearce RM, Parks R, Plonk K, Plonk SG, Sutherland LL, Gorny MK, Zolla-Pazner S, Vanleeuwen S, Moody MA, Xia SM, Montefiori DC, Tomaras GD, Weinhold KJ, Karim SA, Hicks CB, Liao HX, Robinson J, Shaw GM, Haynes BF.
Duke Human Vaccine Institute, and Departments of Medicine and Immunology, Duke University School Medicine, Durham, NC 2 7710; New York University School of Medicine, New York, NY 15016; University of KwaZulu-Natal, Durban, South Africa, and Tulane University School of Medicine, New Orleans, LA; and University of Alabama at Birmingham; Birmingham, AL 35294-0024.
Two human monoclonal antibodies (mAbs) (2F5 and 4E10) against the HIV-1 envelope g41 cluster II membrane proximal external region (MPER) broadly neutralize HIV-1 primary isolates. However, these antibody specificities are rare, are not induced by Env immunization or HIV-1 infection, and are polyspecific and also react with lipids such as cardiolipin or phosphatidylserine. To probe MPER anti-gp41 antibodies that are produced in HIV-1 infection, we have made two novel murine mAbs, 5A9 and 13H11, against HIV-1 gp41 envelope that partially cross-blocked 2F5 mAb binding to Env, but did not neutralize HIV-1 primary isolates nor bind host lipids. Competitive inhibition assays using labeled 13H11 mAb and HIV-1+ patient plasma demonstrated cluster II 13H11-blocking plasma antibodies were made in 83% of chronically HIV-1 infected patients and were acquired between 5-10 weeks after acute HIV-1 infection. Both the mouse 13H11 mAb and the three prototypic cluster II human mAbs (98-6, 126-6, 167-D) blocked 2F5 binding to gp41 epitopes to variable degrees; the combination of 98-6 and 13H11 completely blocked 2F5 binding. These data provide support for the hypothesis that in some patients, B cells make non-neutralizing cluster II antibodies that may mask or otherwise down-modulate B cell responses to immunogenic regions of gp41 that could be recognized by B cells capable of producing antibodies like 2F5.
-------
j
free, one more -
1: Apoptosis. 2007 Feb
The role of membrane lipids in the induction of macrophage apoptosis by microparticles.
Huber LC, Jüngel A, Distler JH, Moritz F, Gay RE, Michel BA, Pisetsky DS, Gay S, Distler O.
Center of Experimental Rheumatology, University Hospital Zurich, Gloriastrasse 23, CH-8091, Zurich, Switzerland.
Microparticles are membrane-derived vesicles that are released from cells during activation or cell death. These particles can serve as mediators of intercellular cross-talk and induce a variety of cellular responses. Previous studies have shown that macrophages undergo apoptosis after phagocytosing microparticles. Here, we have addressed the hypothesis that microparticles trigger this process via lipid pathways. In these experiments, microparticles induced apoptosis in primary macrophage cells or cell lines (RAW 264.7 or U937) with up to a 5-fold increase. Preincubation of macrophages with phosphatidylinositol-3,5-bisphosphate (PtdIns(3,5)BP) reduced the microparticle-induced apoptosis in a dose-dependent manner. PtdIns(3,5)BP is a specific inhibitor of the acid sphingomyelinase and thus can block the generation of pro-apoptotic ceramides. Similarly, the pre-incubation of macrophages with PtdIns(3,5)BP prevented microparticle-induced upregulation of caspase 8, which is a major target molecule of ceramide action in the apoptosis pathway. PtdIns(3,5)BP, however, had no effect on the spontaneous rate of apoptosis. To evaluate further signaling pathways induced by microparticles, the extracellular signal regulated kinase (ERK-) 1 was investigated. This kinase plays a role in activating phospholipases A2 which cleaves membrane phospholipids into arachidonic acid; microparticles have been suggested to be a preferred substrate for phospholipases A2. As shown in our experiments, microparticles strongly increased the amount of phosphorylated ERK1/2 in RAW 264.7 macrophages in a time-dependent manner, peaking 15 min after co-incubation. Addition of PD98059, a specific inhibitor of ERK1, prevented the increase in apoptosis of RAW 264.7 macrophages. Together, these data suggest that microparticles perturb lipid homeostasis of macrophages and thereby induce apoptosis. These results emphasize the importance of biolipids in the cellular cross-talk of immune cells. Based on the fact that in clinical situations with excessive cell death such as malignancies, autoimmune diseases and following chemotherapies high levels of circulating microparticles might modulate phagocytosing cells, a suppression of the immune response might occur due to loss of macrophages.
http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=ShowDetailView&TermToSearch=17191114&...
--------
j
free,
The abstract describes that one pretty well.
Just keep these mind -
AND - read the links in this post -
http://investorshub.advfn.com/boards/read_msg.asp?message_id=23454574
-------
j
free- very pertinent post.....
"Hence, their ['microparticle/'microvesicle'/'exosome'...] secretion by neoplastic cells may in the future become a novel pathway to target for therapeutic intervention in cancer patients."
Yes.
Absolutely correct.
---------
j
Kim / Emi / Tanabe: tumor immune evasion paper-
now available in full, for free -
a MUST READ
Tumor-Driven Evolution of Immunosuppressive Networks during Malignant Progression
http://cancerres.aacrjournals.org/cgi/content/full/66/11/5527
excerpts:
ABSTRACT:
Tumors evolve mechanisms to escape immune control by a process called immune editing, which provides a selective pressure in the tumor microenvironment that could lead to malignant progression. A variety of tumor-derived factors contribute to the emergence of complex local and regional immunosuppressive networks, including vascular endothelial growth factor, interleukin-10, transforming growth factor-ß, prostaglandin E2, and soluble phosphatidylserine, soluble Fas, soluble Fas ligand, and soluble MHC class I–related chain A proteins. Although deposited at the primary tumor site, these secreted factors could extend immunosuppressive effects into the local lymph nodes and the spleen, promoting invasion and metastasis. Vascular endothelial growth factors play a key role in recruiting immature myeloid cells from the bone marrow to enrich the microenvironment as tumor-associated immature dendritic cells and tumor-associated macrophages. The understanding of the immunosuppressive networks that evolve is incomplete, but several features are emerging. Accumulation of tumor-associated immature dendritic cells may cause roving dendritic cells and T cells to become suppressed by the activation of indoleamine 2,3-dioxygenase and arginase I by tumor-derived growth factors. Soluble phosphatidylserines support tumor-associated macrophages by stimulating the release of anti-inflammatory mediators that block antitumor immune responses. Soluble Fas, soluble FasL, and soluble MHC class I–related chain A proteins may help tumor cells escape cytolysis by cytotoxic T cells and natural killer cells, possibly by counterattacking immune cells and causing their death. In summary, tumor-derived factors drive the evolution of an immunosuppressive network which ultimately extends immune evasion from the primary tumor site to peripheral sites in patients with cancer.
.....
soluble phosphatidylserine (sPS), acts as an inducer of an anti-inflammatory response to TAMs, resulting in the release of anti-inflammatory mediators, such as IL-10, TGF-ß, and PGE2, that inhibit an immune response to DCs and T cells
.....
Phosphatidylserine, an ATP-dependent aminophospholipid, triggers the phagocytosis of apoptotic cells by phagocytes such as macrophages and DCs. The clearance of apoptotic cells is mainly mediated by macrophages influenced by the interaction between phosphatidylserine and phosphatidylserine receptor. Phagocytosis of apoptotic cells by macrophages occurs without the induction of inflammatory reactions (25, 26). The presence of apoptotic cells during macrophage activation has been shown to increase the secretion of anti-inflammatory mediators, such as IL-10, TGF-ß, and PGE2, and to decrease the secretion of proinflammatory cytokines such as IL-12 (27). Although phagocytosis of both apoptotic and necrotic cells is mediated by the phosphatidylserine/phosphatidylserine receptor interaction, the reduced cytotoxicity of TAMs after ingestion of apoptotic cells may be explained by the anti-inflammatory response to cancer cells that was caused by several negative mediators. The sequential immune responses by TAMs and DCs can be abrogated by sPS derived from cancer cells. In the sequential interactions, the tumor-derived sPS reacts with phosphatidylserine receptor in TAMs and DCs to inhibit the innate immune response. The enforced anti-inflammatory response by sPS—not only by interaction with the phosphatidylserine receptor on TAMs, but also with the phosphatidylserine receptor on iDCs to inhibit their maturation—may cause immune escape and tumor proliferation. Furthermore, the tumor-derived sPS bind to their own phosphatidylserine receptor, resulting in the production of IL-10, TGF-ß, and PGE2 to facilitate tumor proliferation.
.....
Concluding Remarks
Cancer immune evasion initiated from the primary tumor site, which is mediated by TDSFs and altered tumor surface antigens, is able to extend to secondary lymphoid organs and peripheral vessels, resulting in the formation of an immunosuppressive network that inhibits an antitumor immune response. The increased number of iMCs recruited from the bone marrow, triggered by VEGF and other TDSFs, as well as their TiDCs and TAM, play a critical role in suppressing the immune response at not only the primary site but also at secondary lymphoid organs and peripheral vessels. Given that tumor cells are regulated by a proinflammatory mechanism that stimulates tumor growth, whereas immune cells are regulated by an anti-inflammatory mechanism that causes immune suppression, proinflammatory or anti-inflammatory treatments have been controversial and considered as a double-edged sword in cancer therapy. However, because a proinflammatory response is mediated by massive tumor cell death, and other immunostimulatory agents are crucial for provoking an antitumor immune response, a proinflammatory response does not necessarily promote tumor growth. In contrast, anti-inflammatory treatment with, for example, NF-{kappa}B inhibitor may affect the activation of immune cells. Selective targeted therapy for effects downstream of NF-{kappa}B may be more effective than nonspecific inhibition of NF-{kappa}B. Thus, primary cancer therapy needs to be focused on inducing a proinflammatory response, which can be connected to the activation of an immune response to tumor antigens and tumor-associated antigens in cancer cells.
Many previous reports indicated that tumors infiltrated with lymphocytes or mature DCs had a good prognosis. Furthermore, recent reports on the use of DNA microarrays for gene expression analysis also suggested that an increase in expression of immune response–related genes is associated with a good prognosis (105, 106). Despite the fact that treatment with anticancer drugs and molecular targeting agents induces infiltration of T cells into the tumor, the molecular mechanism by which anticancer treatment activates an immune response against the tumor-induced immunosuppressive network in individual cancer patients still remains to be elucidated. Further studies will be required for the analysis of individual immune responses to tumor cells following anticancer treatment to disrupt and modulate the immunosuppressive network. Such studies may provide new insights and help to overcome a critical obstacle in cancer therapy.
-------
j
Charlie..........
"131I-TNT was immunogenic in only a small minority"
is a good thing.
j
goat- and this is a must read-
a new offer....
http://investorshub.advfn.com/boards/read_msg.asp?message_id=21706236
------
j
goat, HSPD 18...
"Is the DTRA grant the only one they are working on?
Doubtful by looking at AVII and others..When you get one, you get another...."
Take a look at HSPD 18...
http://investorshub.advfn.com/boards/read_msg.asp?message_id=21791012
-------
j
cjg, re: Donald McDonald at today's symposium,
nice to see Donald McDonald there. He's a giant in antiangiogenesis work. I hope he becomes interested, (or more interested), in PS as the fundamental impetus of vascular sprouting. (He was the author of the paper last year that demonstrated how rapidly vessels regrow after cessation of VEGF inhibition therapy).
to quote from his paper-
(co-authored by Bruce Freimark who is now working for Peregrine...)
Abstract:
Inhibitors of VEGF signaling can block angiogenesis and reduce tumor vascularity, but little is known about the reversibility of these changes after treatment ends. In the present study, regrowth of blood vessels in spontaneous RIP-Tag2 tumors and implanted Lewis lung carcinomas in mice was assessed after inhibition of VEGF receptor signaling by AG-013736 or AG-028262 for 7 days. Both agents caused loss of 50%–60% of tumor vasculature. Empty sleeves of basement membrane were left behind. Pericytes also survived but had less –SMA immunoreactivity. One day after drug withdrawal, endothelial sprouts grew into empty sleeves of basement membrane. Vessel patency and connection to the bloodstream followed close behind. By 7 days, tumors were fully revascularized, and the pericyte phenotype returned to baseline. Importantly, the regrown vasculature regressed as much during a second treatment as it did in the first. Inhibition of MMPs or targeting of type IV collagen cryptic sites by antibody HUIV26 did not eliminate the sleeves or slow revascularization. These results suggest that empty sleeves of basement membrane and accompanying pericytes provide a scaffold for rapid revascularization of tumors after removal of anti-VEGF therapy and highlight their importance as potential targets in cancer therapy.
http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1578604
------------
Is it becoming clear that there's a more "upstream" target that needs to be dealt with?!!...
------------
* According to Schroit & Fidler, Anionic phospholipids (namely PS) are responsible for the endothelial cell sprouts which eventually form new blood vessels. PS becomes the target which the sprouts on endothelial cells reach out for.
http://cancerres.aacrjournals.org/cgi/content/full/65/24/11529
According to Peter Henson, Stephen Dow etc., exposed PS is responsible for stimulating VEGF production by macrophages.
Author List: L. U’Ren1,2, P. Henson3, Shyra Gardhi3, and S. Dow1,2.
Associated Departments/Locations for Authors: 1Department of Microbiology, Immunology, and Pathology, Colorado State University, Ft Collins, CO. 2Animal Cancer Center, Colorado State University. 3Integrated Dept of Immunology, National Jewish Medical and Research Center, Denver CO.
Abstract:
Background. Phosphatidylserine (PS) is normally expressed on the inner leaflet of living cells and is translocated to the outer leaflet as cells becomes apoptotic. Surface expression of PS stimulates engulfment of cells by macrophages. Previous studies have found that engulfment of PS+ apoptotic cells by macrophages triggers release of anti-inflammatory cytokines. It is also known that tumor-associated macrophages (TAM) are associated with increased tumor growth and angiogenesis. Therefore, we investigated the effects of PS+ tumor cells on production of the pro-angiogenic factor VEGF by macrophages. Methods. Tumor cell lines were screened by flow cytometry for expression of PS, using Annexin V staining. Normal macrophages were obtained from the peritoneal cavity of mice, while tumor associated macrophages (TAM) were sorted from enzymatically digested tumor tissues of mice. VEGF production was measured by elisa. Results. Incubation of macrophages with PS+ tumor cells (live cells or apoptotic cells) triggered release of significant quantities of VEGF. This response could be partially inhibited when the apoptotic tumor cells were pre-incubated with Annexin V. We also observed that exosomes derived from a PS expressing tumor cell line (MCA2.1-1) could significantly increase VEGF production by peritoneal macrophages, and that this response could be completely blocked by the addition of Annexin V. The addition of exosomes derived from a non-PS expressing tumor cell line (B16) did not induce macrophage VEGF production. Conclusions. PS appears to play an important role in regulating the production of VEGF by tumor associated macrophages and may therefore promote tumor angiogenesis. Our results indicate that PS expressed on tumor cells or on tumor exosomes can both stimulate production of macrophage VEGF. Thus, tumor cells or their secreted membranes may promote tumor angiogenesis via their interaction with tumor associated macrophages.
----------
j
2 Shilyansky papers due out this winter -
Phosphatidylserine-dependent suppression of dendritic cell function: a novel mechanism for tumor immune evasion. In preparation.
Chen X, Doffek K, Gavin C, Orentas R, Johnson B, Matsui K, Sugg SL, Shilyansky J.
Phosphatidylserine on the surface of apoptotic cells inhibits NFkB and p38 MAPK activation and induces tolerogenic dendritic cells. In preparation.
Doffek K, Gavin C, Chen X, Sugg SL, Shilyansky J.
http://www.healthcare.uiowa.edu/surgery/joel-shilyansky.html
-----
j
stealthy L,
Annexin V.
Here's Shilyansky's recent AACR abstract -
#213 (Apr15)
Neuroblastoma Express Phosphatidlyserine: Mechanism for Immune Evasion
Kara Doffek, Xiaocia Yan, Sonia L. Sugg, Bryon Johnson, Joel Shilyansky. Medical College of Wisconsin, Milwaukee, WI, Univ. of Iowa
ABSTRACT:
Introduction: We examined the effect of phosphatidylserine (PS) on tumor immunity. PS is a membrane phospholipid that is restricted to the inner surface of plasma membrane in living cells, but flips to the cell surface during apoptosis. PS may be also expressed on the surface of live tumor cells. PS was reported to promote tolerance, possibly by inhibiting antigen presentation and inflammation. The effect of tumor PS on the immune response against neuroblastoma (NB), a childhood malignancy, was examined in a murine model.
Methods: PS on NB cell surface was determined using Annexin-V (AnV) binding and flow cytometry. To block PS in vivo, NB cells were engineered to secrete AnV protein covalently fused to FLAG, which specifically binds and blocks PS. Western blot was used to determine AnV expression. A/J mice were injected with 104 NB cells subcutaneously and tumor growth was determined.
Results: NB cells were transfected with pre-pro-trypsin AnV-FLAG construct. Western blot analysis was used to quantify AnV production in supernatants and identify NB clones that secreted AnV (AnV-NB). Clones that proliferated in vitro at the same rate as wild type (wt) NB cells were selected. Supernatants from AnV-NB cells blocked PS on the surface of wt NB cells preventing staining with FITC-conjugated recombinant AnV.
The findings suggest that secreted AnV blocked PS. The wt and AnV-NB cells were then injected subcutaneously into A/J mice. Tumor homogenates showed continuous AnV expression in vivo. In immunocompetent mice, wt tumors grew faster than AnV-NB cells. In mice depleted of T cells with anti-Thy1.2, no difference in growth rate between wt and AnV-NB tumors was noted.
Mice were then immunized two times, 7 days apart, with AnV-NB or control cells. 7 days later mice were injected subcutaneously with 105 wt NB cells. Immunization with An-NB, but not control cells, protected mice from wt NB challenge.
Conclusions: The study demonstrated that mouse NB cells express PS on the cell surface. Blocking PS in vivo by engineering NB cells to secrete AnV slowed tumor growth in immunocompetent but not T cell depleted mice. Importantly, immunization with AnV-NB cells was protective. The findings suggest that PS inhibited anti-tumor T cell immunity in mice. The mechanism of PS action may be indirect inhibition of anti-tumor CTL responses, or activation of regulatory T cells. The studies support the hypothesis that PS expression is a potential mechanism for tumor immune evasion.
-------
j
PS on microparticles inhibits DC's -
Shilyansky / Haynes
Shilyansky
PHOSPHATIDYLSERINE ON ENDOTHELIUM DERIVED
MICROPARTICLES INHIBITS DENDRITIC CELL FUNCTION
PHOSPHATIDYLSERINE ON ENDOTHELIUM DERIVED
MICROPARTICLES INHIBITS DENDRITIC CELL FUNCTION
John C. Densmore, M.D.1, Xiao Chen, M.D.1, Kara
Doffek1, Sonia L. Sugg, M.D.2, Keith T. Oldham, M.D.1,
Joel Shilyansky, M.D.1, 1Children’s Hospital of Wisconsin,
Milwaukee, WI, U.S.A., 2Medical College of Wisconsin,
Milwaukee, WI, U.S.A.
http://www.eapsa.org/surgeons/APSA_Prelim_05.pdf
--------------------------
Haynes (CHAVI / Gates)
"OT" new Shilyansky grant-
http://cmg.medicine.uiowa.edu/blogs/facstaff/
Drs. Joel Shilyansky, UI associate professor and director of pediatric surgery and the Robert and Hélène Soper Chair in Pediatric Surgery and Rajeev Vibhakar, UI assistant professor of pediatrics, have been awarded a total of $100,000 in 2007 Aiming for a Cure Foundation grants, which were made through the Holden Comprehensive Cancer Center at the UI. The two-year grants were effective Oct. 1. 100507vibhakar1.jpg
The Aiming for a Cure Foundation raises funds to benefit pediatric oncology patients and families at UI Children’s Hospital at UI Hospitals and Clinics. The goal is to improve the quality of care and life for these children through the funding of research and patient care. The award recipients are both members of the Holden Comprehensive Cancer Center.
Shilyansky received $50,000 for a study on neuroblastoma, a childhood cancer that forms in nerve tissue and is often incurable. Shilyansky’s team will focus on a phospholipid called phosphatidylserine that helps neuroblastoma subvert patients’ immune system. The team will investigate whether a certain protein that can block phosphatidylserine would increase immunity against the tumor. This approach could eventually be used clinically to train patients’ immune system to recognize and destroy cancer cells. The approach also could possibly lead to more generally applicable and effective treatment for neuroblastoma and other cancers...
-----
j
the most important pic I've seen so far is this one -
j
re: mabs -
mabs are antibodies. One of the important differentiators is the region of the ab that sticks to it's target, called the 'variable' region. Think of typical antibodies as having two feet that stick. Peregrine is the only company working with therapeutic abs that deliberately target phospholpids. PS and other phospholipids have traditionally been considered "off limits" targets, but recently the folks at MD Anderson, (Alan Schroit etc.), have made discoveries into important differences into what look to be safe abs to phospholipids, (and why). If Peregrine's anti-PS works, ( actually I suppose I should say 'if it keeps working'), PS will be a very important target, as PS is the reason that so many diseases and critters get the better of us. (That wasn't really known at the time Thorpe was designing his anti-PS. His work came about for a different reason - because of his discovery of PS becoming exposed on tumor vasculature, but the understanding that PS itself is immunosuppressive - that it alters our immune response in a very fundamental way, was/is new, and it carries implications for treating "disease" itself, in the broadest sense imaginable). Our immune system recognizes PS as a universal symptom of dying host cells, and reacts accordingly - by clearing it up, but with a 'kinder/gentler' pro-growth response, and diseases like cancer, viruses, malaria, tuberculosis, etc, etc,..., seem to exploit this aspect of our immune response.
---------
j
S. King: 1:13:25
re: the two planned phase two bavi/chemo trials, (one of which is breast in which they're batting two-for-two for objective responses, and that was in very very advanced patients)....
SK: "These are two-staged design trials, in which an initial group of patients is treated, we'll evaluate the patients for tumor response as well as safety parameters, and then depending on the results we'll either go on to treat another set of patients, bringing the total for lung cancer to 49, and the total for breast cancer to 46. It's also an evaluation point, - if we see some spectacular things happening it gives us the ability to potentially move toward randomized clinical trials, or if we feel like we want more data- the ability to treat the additional patients to get a better read on anti-tumor activity."
-------
comments???......
j
------------
S. King:
"These are two of those patients in the carboplatin/paclitaxel arm, both with metastatic breast cancer. You can see in the left-hand panel here, highlighted in red is the tumor masses before treatment, and then eight weeks post-treatment you can see that essentially these target lesions have completely resolved. In fact we believe these probably would have been considered complete responses except for the fact that there were probably metastatic lesions which did not completely resolve. But again - very encouraging results in early clinical studies."
---------
S King: re: NEW PHASE II BAVI+TAXOLCARBO VS. BREAST CANCER IN INDIA
"This trial is designed to assess the safety & efficacy of Bavi in combo with paclitaxel & carboplatin [Bristol-Myer’s “TaxolCarbo”] in patients with Metastatic Breast Cancer. We chose BREAST CANCER as a 2nd Ph.2 target based on compelling signs of activity in the pilot combo-therapy study completed earlier this year. In that study, both women with metastatic breast cancer, receiving the same combination, showed promising signs of activity, with each having partial tumor responses [‘partial response’ = greater than a 30% reduction in tumor size]. Although this was a small study, these are very encouraging results in an indication in which these is a high un-met need for better treatments."
------------
Two for two.
j
S. King:
"These are two of those patients in the carboplatin/paclitaxel arm, both with metastatic breast cancer. You can see in the left-hand panel here, highlighted in red is the tumor masses before treatment, and then eight weeks post-treatment you can see that essentially these target lesions have completely resolved. In fact we believe these probably would have been considered complete responses except for the fact that there were probably metastatic lesions which did not completely resolve. But again - very encouraging results in early clinical studies."
---------
S King: re: NEW PHASE II BAVI+TAXOLCARBO VS. BREAST CANCER IN INDIA
"This trial is designed to assess the safety & efficacy of Bavi in combo with paclitaxel & carboplatin [Bristol-Myer’s “TaxolCarbo”] in patients with Metastatic Breast Cancer. We chose BREAST CANCER as a 2nd Ph.2 target based on compelling signs of activity in the pilot combo-therapy study completed earlier this year. In that study, both women with metastatic breast cancer, receiving the same combination, showed promising signs of activity, with each having partial tumor responses [‘partial response’ = greater than a 30% reduction in tumor size]. Although this was a small study, these are very encouraging results in an indication in which these is a high un-met need for better treatments."
------------
Two for two.
j
mojo,
the current Peregrine used to be named Techniclone. In '97 Techniclone acquired Vascular Targeting Technologies, which was formerly named Peregrine :) ............
Here's a Steve King bio from Reuters that may help also -
http://stocks.us.reuters.com/stocks/OfficersDirectorsDetails.asp?rpc=66&symbol=PPHM.O&office....
King, Steven W.
Brief Biography
Mr. King has served as the President and Chief Executive Officer of Peregrine Pharmaceuticals, Inc., since March 19, 2003. From August 2002 to such date, Mr. King served as Chief Operating Officer of Peregrine. From February 2000 to August 2002, Mr. King served as our Vice President of Technology and Product Development. Mr. King joined Peregrine in 1997 in the capacity of Director of Research and Development. Mr. King was responsible for planning and launching our wholly owned contract manufacturing subsidiary, Avid Bioservices, Inc., in 2002. Mr. King has served as the President of Avid since its inception. Mr. King was previously employed at Vascular Targeting Technologies, Inc., (formerly known as Peregrine Pharmaceuticals, Inc.) a company we acquired in 1997, which held the rights to the Vascular Targeting Agent technology. Mr. King previously worked with Dr. Philip Thorpe, inventor of our Anti-Phosphatidylserine (ôAnti-PSö) Immunotherapeutic and VTA technology platforms, at the University of Texas Southwestern Medical Center at Dallas and is a co-inventor on over 40 U.S. and foreign patents and patent applications in the Vascular Targeting Agent field. Mr. King received his Bachelors and Masters degrees from Texas Tech. University in Cell and Molecular Biology.
------
j
mojo,
Peregrine was in Princeton NJ at the time.
here's some info for you from the PPHM page -
------
http://www.peregrineinc.com/content.php?mi=MjU=#2
When was Peregrine founded?
Peregrine was founded in June 3, 1981 under its former name Techniclone International Corporation, a California corporation. On March 24, 1997, Techniclone International Corporation was merged with and into Techniclone Corporation, a corporation incorporated in the State of Delaware on September 25, 1996. This merger was effected for the purpose of effecting a change in the Company's state of incorporation from California to Delaware. The Company changed its name from Techniclone Corporation to Peregrine Pharmaceuticals, Inc. in October 2000.
When did Peregrine go public?
Peregrine's initial public offering was on December 16, 1982.
When did Peregrine acquire the rights to the VTA technology?
On April 24, 1997, the Company acquired all of the outstanding stock of Vascular Targeting Technologies, Inc. (formerly known as Peregrine Pharmaceuticals, Inc.) in exchange for 5,080,000 shares of the Company's common stock and the assumption of net liabilities of approximately $484,000. Vascular Targeting Technologies, Inc. was a development stage company involved in the research and development of vascular targeting agents. The acquisition was accounted for as a purchase. The excess of the purchase price over net tangible assets acquired (cash and notes receivable) and liabilities assumed (accounts payable and accrued liabilities) represents the difference between the fair value of the Company's common stock exchanged and the fair value of net assets purchased. The excess purchase price of $27,154,402 over the net tangible assets acquired represents the amount paid for acquired technologies and related intangible assets. The excess purchase price for the acquisition had been charged to operations as of the effective date of the acquisition as the related technologies have not reached technological feasibility and the technology had no known future alternative uses other than the possibility for treating cancer patients.
When did Peregrine acquire the rights to the TNT technology?
On January 18, 1994, Peregrine and Cancer Biologics, Inc. ("CBI") entered into an Agreement and Plan of Merger (the "Agreement and Plan of Merger") which contemplated the merger of CBI with and into Peregrine. On June 10, 1994, the shareholders of the Company approved the merger pursuant to the Agreement and Plan of Merger. The merger between CBI and the Company was completed on July 26, 1994. The assets of CBI acquired by the Company consist primarily of research and development of the TNT antibody technology. As a result of the merger, the Company incurred an immediate charge to operations for purchased in-process research and development of approximately $4,850,000. This amount represents the excess of the fair market value of the Company's common stock issued over the net assets acquired of CBI, plus an additional non-recurring charge relating to CBI stock options assumed by the Company.
--------
j
DECEMBER 1, 1994
PATENT LICENSE AGREEMENT
BETWEEN UNIVERSITY OF TEXAS SYSTEM
AND
PEREGRINE PHARMACEUTICALS. INC.
THIS AGREEMENT is made by and between the BOARD OF REGENTS (BOARD)
OF THE UNIVERSITY OF TEXAS SYSTEM (SYSTEM), an agency of the State of Texas,
whose address is 201 West 7th Street, Austin, Texas 78701 and PEREGRINE
PHARMACEUTICALS, INC....
...snip...
page 110:
7.1 The Term of this Agreement shall extend from the Effective Date set forth
hereinabove to the full end of the term or terms for which PATENT RIGHTS have not
expired and if only TECHNOLOGY RIGHTS are licensed and no PATENT RIGHTS are
applicable, then, on a per-product basis, (i) with respect to LICENSED PRODUCTS which
have an FDA-approved therapeutic indication for humans, for a term of seven (7) years from
the date of COMMERCIAL INTRODUCTION of any such product and (ii) with respect to
LICENSED PRODUCTS which have an FDA-approved diagnostic indication for humans, for
a term of seven (7) years from the date of COMMERCIAL INTRODUCTION of such
product.
-------
j
not much change in overall shorts as of end of Sept
September End Month 2007 4,429,317 (2.06)
from Esparza's report -
http://www.hivvaccineenterprise.org/_dwn/news/ghve_report_2007.pdf
snip -
page 42:
Recent studies have demonstrated FcR-dependent antiviral effects of HIV-1-positive serum samples and MAbs in macrophages and immature DCs in cases where the antibodies had little or no detectable activity in a conventional neutralization assay. Other recent studies have shown an antiviral effect of non-neutralizing antibodies in an assay termed “antibody-dependent cell-mediated virus inhibition” (ADCVI) that measures cytolytic and noncytolytic killing of infected cells by FcR-bearing effector cells. Two independent groups also reported antibody-dependent complement-mediated virus lysis and inactivation activity in serum from early HIV-1 seroconverters. These combined observations provide strong rationale to screen sera for these activities in greater detail even if neutralizing activity is absent.
As new immunogens are designed and tested, it will be important to compare them to each other and to earlier prototypes with respect to the magnitude and breadth of the neutralizing antibody responses each generates. In order to adequately monitor neutralization breadth and potency and to compare and prioritize immunogens, assays are needed that are sensitive, quantitative, high throughput and have correlative value. Substantial improvements were made in the past several years in assay technology and in available reference reagents. Thus, cumbersome and expensive assays using PBMC and uncloned viruses are being replaced with a new technology that utilizes molecularly cloned Env-pseudotyped viruses and Tat-induced luciferase reporter gene expression in genetically engineered cells lines. This new technology affords greater sensitivity, reproducibility, high throughput, cost-effectiveness and scientific value compared to PBMC assays and, as a result, it has been responsible for an explosion of new data that was not possible before. Steps are being taken by the CAVD to transfer this new technology to multiple laboratories around the world and to implement a validated proficiency testing program to assure inter-laboratory equivalency in assay performance.
43 Report of Activities | 2005 – 2007
The new assay technology gained momentum after early validation studies showed an acceptable level of agreement with the results obtained in PBMC assays. These early validation studies, which employed a relatively small number of human MAbs and HIV-1-positive serum samples for comparisons between assays, found no cases where the new assay technology was considerably less sensitive than PBMC-based assays. In fact, in general the new technology was more sensitive. However, as the number and types of different antibodies tested in both assays grew over time, several cases were identified where neutralization was considerably more potent or only detected in the PBMC assay, including some cases involving anti-lipid antibodies derived from immunization of mice with lipids, anti-lipid antibodies derived from a patient with autoimmune disease, as well as some cases involving gp120- and gp41-specific MAbs.
Thus, the recent recognition that new assay technologies lack sensitivity for certain neutralizing antibodies raises important questions about current plans to globalize a single assay for routine use and points to the need for a better understanding of the mechanisms of neutralization. It may be necessary to use more than one assay to assure that all neutralizing antibodies are detected. Efforts are needed to:
Study the biologic basis of differential neutralization in the two assay systems and to determine how the ++new assay technology might be modified to detect all neutralizing antibodies,
Run parallel assays in PBMC with large numbers of serum samples from HIV-1-infected individuals and ++multiple preclinical and clinical trials to determine how often and why neutralization is missed in the new assay, and
Develop and explore additional assays for neutralizing antibodies. Progress in this area will greatly de++pend on an effort to develop a more standardized approach to the PBMC assay.
Meeting participants also expressed interest in assays that measure neutralizing antibodies that block cell-cell spread of the virus. HIV-1 can spread by cell-cell contact either though direct synaptic transfer or by transfer to susceptible cells of exogenous virus that is captured by adhesion molecules (e.g., DC-SIGN, complement receptors) on the surface of either DCs or B lymphocytes. Neutralization assays based on these distinct modes of cell-cell spread have been described but have not been properly standardized or validated. It is recommended that these and similar types of assays be explored in greater detail for their biologic relevance, and that they be standardized and validated for possible use as endpoint assays in preclinical and clinical vaccine trials.
Important decisions need to be made about the type(s) of antibodies and assays that have greatest relevance to HIV-1 vaccines. Simply using highly standardized assays that reliably measure antiviral activities in vitro may not be sufficient if they do not predict a corresponding outcome in vivo. The preferred way to make informed decisions would be to employ a variety of different assays to study the antibody response in a clinical trial in which the vaccine was at least partially protective. Because no such vaccine is currently available for HIV-1, studies in animal models are the next best choice. In this regard, two animal models are widely used for HIV vaccine development: SIV and chimeric simian-human immunodeficiency virus (SHIV) infection in monkeys. Highly quantitative passive transfer experiments in either model with antibodies that exhibit different functions could be used to address the biologic relevance of in vitro assays. Because most MAbs and polyclonal antisera that need to be characterized would be derived from HIV-1-infected subjects, chimeric SHIVs containing HIV-1 envelope glycoproteins would offer greatest value. Unfortunately,
Section II: 2. Humoral responses to HIV
44 Global HIV Vaccine Enterprise
very few SHIVs are currently available and, among these, most are derived from a single genetic subtype (clade B) and exhibit properties that may not be well-suited to assay validation. The creation of new and better SHIVs from non-clade B viruses would facilitate assay standardization as well as vaccine challenge models.
This workshop identified several critical gaps in the current understanding of B cell regulatory pathways that impede a more rational development of an effective antibody-based HIV-1 vaccine. Closing these gaps may lead to a better understanding of the poor immunogenicity of Env and ways to elicit desirable neutralizing antibody responses. For example, broadly neutralizing antibodies in patient serum bind epitopes that are present on monomeric gp120, yet this is a poor immunogen for neutralizing antibody induction in vaccine recipients. Moreover, as mentioned above, epitopes for the known broadly neutralizing MAbs are poorly immunogenic in infected individuals and as vaccine candidates. Thus, Env as an immunogen appears to either bypass one or more key steps in the B cell inductive pathway for which little is known, or may actively induce negative or down-regulation of production of some broadly neutralizing specificities. Similar negative regulatory pathways do not explain the poor immunogenicity of other Env epitopes, as many show no clear evidence of autoreactivity.
In general, antibody responses are initiated when mature naive B lymphocytes encounter antigen via their B cell receptor (BCR) and subsequently engage antigen-specific activated CD4+ T helper cells. Through cognate interactions between multiple co-stimulatory molecules on specific B and T cell subsets, and in the presence of a suitable cytokine environment, a germinal center reaction ensues in which the naive B cells proliferate and undergo clonal expansion, Ig class switch recombination and somatic hypermutation to differentiate into memory B cells. Subsequent co-stimulatory signals delivered by CD4+ T helper cells induce terminal differentiation of memory B cells into plasma cells that then migrate to the bone marrow to complete their differentiation process and secrete antibody. Other mature B lymphocytes that normally reside in the marginal zone (MZ) undergo rapid differentiation into antibody secreting plasma cells, including the production of both antigen-specific and polyreactive antibodies in the absence of T cell help, but these responses tend to be short-lived.
Receptor-ligand interactions and intracellular signaling pathways that govern the production of antibody-producing plasma cells and the persistence of plasma and memory B cells are poorly understood. Additional information on the mechanisms responsible for B cell migration, selection and differentiation within and between specialized anatomical sites, particularly within lymphoid follicles, might be used to target suitable Env epitopes to appropriate B cell inductive pathways.
In parallel to these efforts, genetic studies at the population level could provide critical information on the most promising paths to follow. In particular, the recent completion of the International HapMap project now permits whole genome associated studies to be conducted with a minimum number of single nucleotide polymorphism (SNP) tags. This powerful new technology could be used to identify genes that are associated with the wide variation in neutralizing antibody responses in HIV-1-infected individuals and in vaccine recipients. A critical question to ask is whether the potent neutralizing antibody response in a small subset of infected individuals is due to unique viral epitopes or to host genetic polymorphisms. Current evidence suggests that both might make a substantial contribution in the context of combined epitope and allelic representations.
45 Report of Activities | 2005 – 2007
Other means to improve the B cell response to Env might reside in the Env molecule itself. Results of a recent study suggest that Env exerts a suppressive effect on CD4+ T cells that is mediated by CD4 binding and might explain why Env is a poor immunogen. The fact that Gag-specific but not Env-specific antibody responses decline in parallel with CD4+ T cell loss suggests that Env is mostly a T cell-independent immunogen in infected individuals. Chronic activation of B cells through a CD40-independent pathway involving the up regulation of a B cell activating factor of the TNF family (BAFF) was recently proposed as a mechanism that contributes to the impairment of T cell-dependent antibody responses to Env. Continued investigations along similar lines, when combined with new information about B cell regulatory pathways, could give rise to a new blueprint for improved vaccine design and delivery strategies (e.g., adjuvants, vectors, routes of administration).
To date most studies of the humoral responses in AIDS virus infections have investigated immunoglobulins, the final product of B cells responses. Relatively few studies have examined B cell immunopathogenesis. A number of basic questions are still unanswered (e.g. extent and reason for perturbation of B cell subset changes, including memory B cells and plasma cells in peripheral blood and tissues). Questions also remain about other potential functional contributions of B cells to AIDS virus infections (e.g. role as antigen presenting cells). In vivo studies should be performed in the non-human primate animal model to determine the emergence of pathologic events in the B cell compartment, in particular in lymphatic and gastrointestinal tissues of naïve and vaccinated animals that get challenged with pathogenic SIV or SHIV. These investigations should be done in parallel to detailed analyses of the magnitude and function of AIDS virus-specific immunoglobulin responses determined in plasma and tissue secretions, and of AIDS virus-specific B cells on a single cell basis.
Participantsin themeetingmadethefollowingrecommendations:
New information is needed to identify relevant epitopes to target with vaccines. Epitopes responsible ++for generating potent autologous virus neutralization as well as broadly neutralizing activity in sera from infected individuals appear to be suitably immunogenic and antigenic for this purpose. These epitopes need to be identified and characterized for possible use as monovalent and polyvalent vaccine immunogens. Increased effort is needed to develop sophisticated, high-throughput methods for screening epitopes and antibodies using the latest technologies,
Crystal structures of monomeric gp120 and gp120-gp41 trimer complexes in their native unliganded ++form need to be elucidated as the natural targets for neutralizing antibodies. This information is needed for multiple genetic subtypes of the virus and for transmitted strains of the virus. Coupled with this effort should be a program to make necessary improvements in electron tomography technology to gain a higher resolution of native Env spikes as they exist on virus particles.
Antibody effector functions that mediate complement activation and FcR engagement on macrophages, ++DCs, NK cells, and other cell types need to be evaluated to determine their relevance to HIV-1 vaccines. Assays that measure these antiviral antibodies should be standardized and used to assess biologic relevance in passive protection experiments in animal models using antibodies that exhibit the different effector functions in vitro,
Additional effort is needed to standardize and compare neutralizing antibody assays and to decide ++which assay or combination of assays should be used for standardized assessments of vaccine-elicited
Section II: 2. Humoral responses to HIV46 Global HIV Vaccine Enterprise
neutralizing antibody responses. A major priority is to strengthen the standardization of the PBMC assay as the only assay that has been at least partially validated in passive antibody experiments in animal challenge models. Similar validation experiments in animals models are needed to determine the potential correlative value of new assay technologies that rely on the use of genetically engineered cells lines and Env-pseudotyped viruses,
New and better SHIVs are needed that contain non-clade B envelope glycoproteins and that more ++closely approximate the neutralization phenotype, cellular tropism and pathology of HIV-1. These SHIVs are needed for studies of the biologic relevance of in vitro assays and to decide which antibodies and assays are most relevant for HIV-1 vaccine design and testing,
Additional efforts are needed to support studies in fundamental B cell biology as it relates to HIV-1 vac++cines. A program could be structured in a way that asks key scientific questions about B cell regulatory pathways that modulate Env immunogenicity, including new adjuvant development,
More detailed B cell immunopathogenesis studies are needed to correlate pathologic changes in the ++B cell compartment in peripheral blood and organs including lymphatic and gastrointestinal tissues to AIDS virus-specific immunoglobulin responses. In particular, the non-human primate animal model should be utilized to study B cell immunopathogenesis in naïve and vaccinated monkeys that get challenged with pathogenic AIDS viruses,
Ultimately, the design of new immunogens either with stabilizing mutations in gp120, scaffolds of con++served neutralization epitopes on other proteins, or other structurally-based approaches (e.g., Env-CD4 chimeras or mimics of them) may lead to more promising products. Unmistakably, significantly improving HIV vaccine design is still of the highest priority.
While each of the above research objectives needs to be better supported, structuring the support in a manner that fosters collaboration, coordination, and results sharing would be equally important.
free,
- and here's the reason the DC's were able to do their thing:
super important slide/evidence.
----
REQUIRED READING #2 of 5 ------
Phosphatidylserine Regulates the Maturation of Human Dendritic Cells
The Journal of Immunology, 2004
http://www.jimmunol.org/cgi/content/full/173/5/2985
-----------
Phosphatidylserine Regulates the Maturation of Human Dendritic Cells
Abstract
Phosphatidylserine (PS), which is exposed on the surface of apoptotic cells, has been implicated in immune regulation. However, the effects of PS on the maturation and function of dendritic cells (DCs), which play a central role in both immune activation and regulation, have not been described. Large unilamellar liposomes containing PS or phosphatidylcholine were used to model the plasma membrane phospholipid composition of apoptotic and live cells, respectively. PS liposomes inhibited the up-regulation of HLA-ABC, HLA-DR, CD80, CD86, CD40, and CD83, as well as the production of IL-12p70 by human DCs in response to LPS. PS did not affect DC viability directly but predisposed DCs to apoptosis in response to LPS. DCs exposed to PS had diminished capacity to stimulate allogeneic T cell proliferation and to activate IFN-{gamma}-producing CD4+ T cells. Exogenous IL-12 restored IFN-{gamma} production by CD4+ T cells. Furthermore, activated CTLs proliferated poorly to cognate Ag presented by DCs exposed to PS. Our findings suggest that PS exposure provides a sufficient signal to inhibit DC maturation and to modulate adaptive immune responses.
Introduction
Dendritic cells (DCs),3 which are the most potent professional APCs of the immune system, play an important role in the initiation of adaptive immunity (1). DCs acquire, process, and present Ags derived from pathogenic organisms to activate naive T cells, which mount a specific anti-pathogen immune response. DCs also continuously acquire self-Ags from cells that undergo apoptosis as part of normal cell turnover. However, pathological autoimmune conditions occur rarely, suggesting that the ability of DCs to stimulate self-reactive T cells is tightly controlled. The mechanisms permitting DCs to activate vigorous immune responses to most pathogens and to maintain tolerance to tissue-derived "self" Ags are not fully understood (1, 2). There has been a growing body of evidence suggesting that phagocytosis of apoptotic cells under steady state conditions leads to impaired DC maturation and may induce tolerance (3, 4).
The mechanisms by which apoptotic cells inhibit DC maturation or induce tolerizing DCs are not fully understood. CD36, {alpha}v{beta}5, {alpha}v{beta}3, and other surface molecules have been proposed as possible receptors used by DCs to recognize and phagocytose apoptotic cells (5, 6, 7, 8). A recent study using mAbs directed against scavenger receptor CD36 and {alpha}v integrins suggested that these receptors could also modulate DC function (9). Furthermore, Plasmodium falciparum-infected erythrocytes have been shown to inhibit human DC maturation, reportedly through CD36 and CD51 (10). Receptors for iC3b, an opsonin present on the surface of apoptotic bodies, have also been shown to mediate engulfment of apoptotic bodies by DCs and to modulate DC maturation (11, 12).
Recognition and removal of apoptotic cells by phagocytes, including DCs, is mediated by changes in the expression of membrane-associated markers on the dying cells. One of the most striking and consistent changes on the cell surface of apoptotic cells is the exposure of phosphatidylserine (PS). PS is an anionic aminophospholipid restricted mostly to the inner leaflet of plasma membrane in live cells (13). However, when cells undergo apoptosis, PS molecules are exposed on the cell surface (14, 15). Exposure of PS has been shown to play a central role in the recognition and phagocytosis of apoptotic cells by macrophages (16, 17, 18, 19). Recent studies demonstrate that PS-dependent recognition and ingestion of apoptotic cells by macrophages triggers the release of anti-inflammatory cytokines and inhibits the production of proinflammatory cytokines (20, 21, 22). These studies offer a link between the recognition of apoptotic cells and the physiological consequences of their phagocytosis. Whereas the role of PS in the interaction of apoptotic cells with macrophages and the immunological consequence of PS ligation have been broadly delineated, the role of PS in regulating the phagocytosis of apoptotic cells by DCs and the immunological implication of such interactions are not known. Because of the close lineage relationship between macrophages and myeloid DCs, we hypothesized that PS might play a role in the inhibition of myeloid DC maturation. To test this hypothesis, we prepared large unilamellar liposomes containing PS as a simplified model of cell membrane alterations associated with apoptosis. We examined the effect of PS containing liposomes on DC maturation and immunostimulatory capacity. Our results suggest that PS inhibits the ability of DCs to undergo maturation, secrete IL-12, activate T cells, and stimulate IFN-{gamma}-producing CD4+ T cells.
Results
Exposure to PS liposomes inhibited the phenotypic maturation of DCs
To determine whether PS exposure plays a role in modulating DC function, we first examined the effects of PS liposomes on the immunophenotype of DCs undergoing maturation. DCs, generated from CD14+ PBMCs in the presence of GM-CSF and IL-4, exhibited the typical immature phenotype: CD1ahigh, CD80low, CD83low, CD86low, HLA-DRint, and CD14 negative. DCs underwent maturation after treatment with LPS, a TLR-4 ligand, as evidenced by significantly increased expression of CD83, MHC, and costimulatory molecules. Immature DCs were exposed to medium, latex beads (0.1 µm), or PS and PC liposomes for 18 h before LPS was added to induce maturation. DCs exposed to PS failed to significantly increase the expression of HLA-DR, HLA-ABC, CD40, CD80, CD86, and CD83 in response to LPS, resembling iDCs (Fig. 1, A and D). In contrast, LPS induced significant increase in the expression of surface molecules associated with maturation by DCs exposed to medium alone, latex beads (data not shown), or PC liposomes (Fig. 1, A and C). Inhibition of the LPS-induced DC maturation by PS was dose dependent (Fig. 1B). Whereas PS exposure prevented DC maturation, PS alone, without LPS treatment, had no observable effect on DCs, which retained an immature phenotype (Fig. 1D). To confirm that the inhibitory effect of PS liposomes was mediated by PS and not by a minor contaminant in the natural PS preparation, we prepared pure synthetic POPS and POPC liposomes and tested their effect on DC maturation. Synthetic pure POPS liposomes (150 µM) were equipotent to natural PS-containing liposomes in inhibiting the phenotypic maturation of DCs, whereas synthetic pure POPC liposomes at the same concentration did not inhibit DC maturation (Fig. 1E). The findings show that PS liposomes altered the ability of DCs to respond effectively to inflammatory or "danger" signals, such as LPS, failing to up-regulate the expression of surface molecules that are critical for effective Ag presentation and T cell stimulation.
Effect of PS liposomes on TLR-4 expression by DCs
LPS is recognized by TLR-4, which activates several intracellular signaling cascades in DCs, leading to maturation. We examined the effect of PS exposure on TLR-4 expression using Western blot analysis and flow cytometry. Western blot analysis demonstrated that PS did not alter TLR-4 expression by DCs (Fig. 2A). Flow cytometry analysis shown that surface TLR-4 staining of iDCs was very faint, a finding consistent with a previous report (26). However, intracellular staining of iDCs suggested that, in contrast with monocytes, most of the TLR-4 was intracellular. Exposure of iDCs to PS altered neither surface nor intracellular expression of TLR-4 (Fig. 2B). Interestingly, treatment with LPS led to increased surface but not total TLR-4 expression, which was blocked by PS exposure (Fig. 2B). These data suggest that PS does not interfere with TLR-4 expression but prevents the maturation-associated up-regulation of the receptor on the surface of DCs.
PS liposomes are nontoxic but predispose DCs to apoptosis
We next determined the effect of PS liposomes on DC viability. DCs staining positively for activated caspase-3, a marker of early apoptosis, comprised <2% of all DCs and did not increase after exposure to 150 µM PS for up to 66 h (Fig. 3, I and II). We also assessed DC viability by trypan blue exclusion and found no differences among DCs exposed to PS, PC, or medium alone (data not shown). These results demonstrated that PS liposomes were not toxic to DCs. We then examined the effect of LPS on DC viability. DCs were exposed to PS liposomes, PC liposomes, or medium alone for 18 h and were matured with LPS for 48 h. DCs exposed to PC or medium alone were resistant to apoptosis, whereas DCs exposed to PS were susceptible to LPS-inducedapoptosis (Fig. 3, III–V). Decreased resistance to the proapoptotic effects of LPS by DCs exposed to PS could lead to a reduction in their ability to stimulate an immune response.
DCs specifically recognized PS-containing liposomes
We next examined the specificity of DC interaction with PS, which marks cells for phagocytosis. Rho-PE was incorporated into PS or PC liposomes. DCs were exposed to fluorescently labeled liposomes for 60 min, and uptake of dye was examined by flow cytometry and fluorescent microscopy. DCs phagocytosed PS liposomes better than did PC liposomes, as demonstrated by significantly higher fluorescent labeling (Fig. 4, A and B). The head groups of the PS molecules likely were responsible for the difference in phagocytosis of PC and PS liposomes because the lipid portions were identical in both preparations. DCs failed to incorporate Rho-PE-labeled PS liposomes at 4°C or in the presence of 10 µM cytochalasin D (Fig. 4C), which inhibit phagocytosis by interfering with cell metabolism and cytoskeletal rearrangement, respectively. The findings suggested that PS liposomes were phagocytosed as opposed to directly incorporated into, or bound to, cell membranes. Unlabeled liposomes containing PS but not PC alone also blocked uptake of Rho-PE-labeled liposomes, suggesting that the interaction was saturable and specific (Fig. 4D). Finally, a monoclonal IgM Ab directed against PSR, but not anti-TNP monoclonal IgM control Ab or anti-CD36 mAb, blocked the phagocytosis of PS liposomes (Fig. 4D). Taken together, the findings demonstrated specific recognition and preferential phagocytosis of PS containing liposomes by DCs via PSR.
DCs exposed to PS stimulated allogeneic T cell proliferation poorly
Because PS inhibited the phenotypic maturation of DCs, we examined its effects on the immunostimulatory capacity of DCs. Immature DCs were exposed to liposomes for 18 h, treated with LPS for 48 h, and used to stimulate allogeneic T cells. As shown in Fig. 5, A and B, DCs exposed to PS liposomes, but not to PC liposomes or medium alone, had an impaired capacity to stimulate the proliferation of allogeneic T lymphocytes at DC-to-T cell ratios of both 1:100 and 1:500. The diminished immunostimulatory capacity of DCs exposed to PS liposomes may reflect the lower cell surface expression of MHC and costimulatory molecules. Interestingly, when the DC-to-T cell ratio was increased to 1:20 or greater, allogeneic T cell proliferation was restored to the level achieved against control DCs (Fig. 5A). The presence of a large number of allogeneic DCs may have overcome the effects of reduced expression of MHC and costimulatory molecules by DCs exposed to PS liposomes. Our findings suggest that PS can impair the activation of T cells even when responding to abundant high-affinity Ags such as alloantigens.
DCs exposed to PS stimulated Ag-specific activated CD8+ T cell proliferation poorly
We next examined whether DCs exposed to PS liposomes would extinguish a secondary Ag-specific response, which is thought to be less dependent on costimulatory molecules. DCs were exposed to liposomes, treated with LPS, and pulsed with a peptide derived from a melanoma-associated Ag, melan-A/MART-1. DCs were then used to stimulate activated melan-A/MART-1-specific CD8+ T cells. When compared with control DCs, DCs exposed to PS liposomes had a significantly diminished capacity to stimulate melan-A/MART-1-specific CD8+ T cell proliferation at DC-to-T cell ratios of 1:10 and 1:50 (Fig. 5C). At higher DC-to-T cell ratios, the level of proliferation of melan-A/MART-1-specific CTLs in response to DCs exposed to PS was not different from the levels achieved against control DCs. Diminished proliferation of activated T cells stimulated with DCs exposed to PS liposomes supports the hypothesis that PS can modulate immune responses.
DCs exposed to PS liposomes had impaired ability to induce IFN-{gamma} production
Next, we investigated whether exposure of DCs to PS liposomes affected T cell differentiation. IFN-{gamma} production by T cells is a hallmark of Th1 responses and is required for effective cellular immune responses. IL-4 production is a hallmark of Th2 responses and is required for humoral immune responses. Purified CD4+ T cells were stimulated with DCs that were exposed to PS or PC liposomes and were treated with LPS. As noted above, allogeneic T cell proliferation was not greatly diminished when stimulated at a high DC-to-T cell ratio (1:10). The proportion of CD4+ T cells producing IFN-{gamma} or IL-4 was determined by intracellular cytokine staining. IFN-{gamma} and IL-4 production by CD4+ T cells was determined by cytokine bead array on days 3, 5, and 7 of culture. Compared with control DCs, DCs exposed to PS stimulated a 1.9-fold lower frequency of IFN-{gamma}+, CD4+ T cells (p = 0.005; Fig. 6A). Correspondingly, secretion of IFN-{gamma} by CD4+ T cells stimulated with DCs exposed to PS was markedly reduced (Fig. 6C). Although in some experiments the frequency of IL-4-producing T cells was increased in response to DCs exposed to PS (Fig. 7A), the effect was variable, possibly due to the differences between donors. As a result, we did not demonstrate a significant increase in the average frequency of IL-4-producing CD4+ T cells (Fig. 6B) or in IL-4 secretion (Fig. 6D). The ratios of IFN-{gamma}+ to IL-4+CD4+ T cells induced by DCs exposed to PC and PS were 3.1 ± 0.7 and 1.05 ± 0.8, respectively (p = 0.002), further highlighting the effect of PS on DCs and reflecting its modest effect on IL-4 production. These experiments show that DCs exposed to PS liposomes had a significantly diminished capacity to induce IFN-{gamma}-producing CD4+ T cells, even at a high DC-to-T cell ratio, strengthening our hypothesis that PS modulates the immune response by modifying DC function.
PS suppressed IL-12p70 secretion by DCs
IL-12 plays a central role in driving the development of Th1 immune responses and IFN-{gamma} production (27, 28, 29). Because PS liposomes significantly inhibited the ability of DCs to induce IFN-{gamma}-producing T cells, we examined IL-12 secretion by DCs. We also examined the effect of PS on the production of the anti-inflammatory cytokines TGF-{beta} and IL-10 by DCs, because macrophages were shown to produce TGF-{beta} in response to PS and DCs were shown to produce IL-10 in response to apoptotic cells (9, 20, 22, 30). IL-12p40-, IL-12p70-, IL-10-, and TGF-{beta}-specific ELISAs were used to assay supernatants from DCs exposed to liposomes or medium alone and stimulated with LPS for 48 h to induce maturation. As shown in Fig. 8A, DCs treated with PS liposomes produced significantly less IL-12p70 in response to LPS, compared with DCs exposed to PC liposomes or medium alone (p < 0.01). There was not a statistically significant difference for IL-12p70 production between DCs exposed to medium and PC liposomes (p = 0.09). IL-12p40 secretion was also diminished, but to a lesser extent (data not shown). Interestingly, we did not observe significantly enhanced production of IL-10 or TGF-{beta} (Fig. 8, B and C). Instead, IL-10 production, which is stimulated by LPS, was inhibited by PS exposure.
Exogenous IL-12 restored IFN-{gamma} production by CD4+ T cells stimulated with DCs exposed to PS liposomes
Because PS dramatically inhibited IL-12p70 secretion, we examined the effect of restoring IL-12 on the CD4+ T cell response. The addition of exogenous IL-12 (10 ng/ml) restored IFN-{gamma} production by T cells stimulated with DCs exposed to PS liposomes (Fig. 7, A and B). The findings suggest that DCs exposed to PS liposomes fail to secrete sufficient IL-12p70 and, as a result, have a diminished ability to stimulate IFN-{gamma}-producing CD4+ T cells.
Discussion
An emerging paradigm suggests that under steady state conditions, apoptotic cells modulate DC function, inducing immune tolerance to self-Ags and preventing autoimmunity (3, 4, 9). Early apoptosis is characterized by loss of membrane asymmetry and exposure of PS on the cell surface (13). PS recently has been shown to play a central role in the recognition of apoptotic cells by macrophages and may be a signal to secrete anti-inflammatory cytokines (17, 20). However, the effects of PS on the maturation and function of human monocyte-derived DCs, whose lineage is closely related to macrophages, have not been well described. We found that PS, which is recognized specifically by DCs, triggers phagocytosis and plays a key role in modulating DC maturation and function.
To study the effects of PS on DCs, we used large unilamellar liposomes containing PS or PC as a simplified model of cell membranes, avoiding the heterogeneity of signals likely provided by cells induced to undergo apoptosis. We found that DCs specifically recognized and preferentially phagocytosed PS liposomes (Fig. 4, A and B). Inhibiting cellular metabolism or cytoskeletal rearrangement prevented phagocytosis and eliminated incorporation of PS liposomes (Fig. 4C). Excess unlabeled PS, but not PC liposomes, blocked the incorporation of fluorescently labeled PS (Fig. 4D). Phagocytosis was likely triggered through PSR, because anti-PSR Ab, which blocks PS binding to the PSR but not mAb to CD36, a scavenger receptor type B, prevented the phagocytosis of PS liposomes (Fig. 4D). In the current study, PS alone was sufficient to trigger phagocytosis and to modulate DC function. This is in contrast with macrophages, where PS alone was reportedly insufficient to promote phagocytosis of erythrocytes or to modulate cytokine production (21). A second tethering interaction with the phagocyte was necessary to trigger either event in macrophages. Our divergent results in DCs may stem from the difference in DC and macrophage function. Alternatively, it is possible that the requirements for phagocytosis of liposomes and erythrocytes are different or that the density of PS on liposomes is greater than on erythrocytes loaded with PS. Although PSR appears to be essential for DC phagocytosis of PS liposomes, we did not rule out a potential role for other receptors, such as {alpha}v{beta}3 integrins, that could interact with PS (5, 6, 9). Our findings, for the first time, demonstrate that PS is specifically recognized via PSR expressed on the surface of DCs and triggers phagocytosis.
DC apoptosis may play a central role in immune regulation in vivo (3). We examined the effect of PS on DC viability. Exposure to PS liposomes alone did not induce apoptosis, but increased the susceptibility of DCs to apoptosis 48–72 h after treatment with LPS (Fig. 3). We speculate that PS may prevent the activation of DC survival pathways induced by LPS, permitting proapoptotic pathways to predominate (31, 32, 33). Induction of apoptosis may lead to decreased number of DCs migrating to draining lymph nodes and diminished T cell activation. Alternatively, DCs undergoing apoptosis may amplify a tolerogenic response as they carry Ags to secondary lymph nodes where they can be phagocytosed by resident DCs, preventing their maturation (3, 34). By predisposing DCs to apoptosis in response to proinflammatory signals, PS may suppress the activation of adaptive immunity.
Previous studies have demonstrated that exposure to PS or apoptotic cells modulated the activation of macrophages and microglial cells by LPS (13, 20, 22, 25, 35, 36, 37). We examined the effect of PS on the expression of surface molecules associated with DC maturation. Compared with control groups, DCs exposed to PS liposomes expressed significantly lower levels of HLA-ABC, HLA-DR, CD80, CD86, CD40, and CD83 in response to LPS. Our findings show that although PS alone did not alter the phenotype of DCs (Fig. 1D), it inhibited the LPS-triggered maturation of DCs in a dose-dependent fashion (Fig. 1B). Lack of DC maturation was not the result of reduced DC viability, because in our studies PS liposomes were not directly toxic to DCs. Phagocytosis alone was not responsible for inhibiting maturation, because DCs that took up latex beads with the same size as liposomes acquired a mature phenotype in response to LPS (data not shown). PS also did not directly interfere with LPS receptor expression. Both total and cell surface expression of TLR-4 remained unaffected after exposure to PS alone. Furthermore, apoptosis of DCs exposed to PS occurred only after treatment with LPS, suggesting that TLR-4 remained functional. PS exposure also inhibited DC maturation in response to other maturation signals such as anti-CD40 agonist Ab (our unpublished results), suggesting that the inhibitory effects of PS were not the result of direct interference with LPS binding, but of modulation of signaling associated with maturation. Indeed, we found that, similar to its effect on MHC and costimulatory molecules, PS inhibited the increase of cell surface TLR-4 expression in response to LPS. Surface TLR-4 expression was low, however, and it has not been established whether such an increase in surface TLR-4 expression is required for DC maturation.
It is tempting to speculate that the effect of PS on DC maturation is mediated by the PSR. PSR ligation has been previously shown to inhibit the activation of macrophages by LPS (22, 35), and in the current study it was required for DCs to recognize and take up PS liposomes. However, we have not excluded the possibility that an alternative receptor mediated the immunomodulatory effects of PS. We could not use the 217 mAb to block the immune effects of PS because it is a PSR agonist (35). The role and function of PSR in DCs continue to be the subjects of active investigation in our laboratory.
Previous reports have suggested that iDCs can induce immune unresponsiveness in vitro and in vivo, possibly due to reduced Ag presentation and T cell costimulation (38, 39, 40, 41). Inhibition of DC maturation by exposure to PS is a potential mechanism for inducing immune unresponsiveness or tolerance. We found that DCs exposed to PS had impaired ability to activate allogeneic T cells (Fig. 5, A and B) and to stimulate IFN-{gamma} production (Fig. 6, A and C). Increasing the ratio of DCs to responders restored the proliferation of allogeneic T cells; however, IFN-{gamma} production by allogeneic CD4+ T cells was still impaired. Increased susceptibility to apoptosis by DCs exposed to PS after LPS treatment could not explain the inability to stimulate IFN-{gamma}-producing T cells in vitro, because IL-4-producing T cells were still induced. Furthermore, exogenous IL-12, which does not affect DC viability, restored IFN-{gamma} production by T cells. The findings suggest that PS did not simply inhibit the ability to activate T cells, but selectively modulated the ability to induce IFN-{gamma}-producing T cells.
We also demonstrated that activated CTLs proliferated poorly in response to cognate Ag presented by DCs exposed to PS. Stimulation of activated CD8+ T cells is thought to be independent of CD80 and CD86 costimulation or IL-12 production (42, 43). Impaired proliferation by CTLs could be the result of reduced expression of MHC and Ag presentation or of direct inhibition by DCs exposed to PS. We did not directly examine the effect of PS on the expression of costimulatory molecules such as 4-1-BB ligand that are thought to enhance the proliferation of activated CTLs (44, 45, 46, 47). Taken together, these findings suggest that DCs exposed to PS would fail to initiate and may extinguish cellular immune responses.
Cytokines produced by DCs play an important role in defining the T cell response. The immunostimulatory form of IL-12, IL-12p70 heterodimer consisting of p35 and p40 subunits, is required to induce Th1 responses (27, 28, 29). We found significantly reduced secretion of IL-12p70 by DCs exposed to PS, providing a mechanism for the diminished capacity to stimulate IFN-{gamma} production by T cells. The addition of exogenous IL-12 restored IFN-{gamma} production in response to DCs exposed to PS. The findings suggest that diminished capacity to stimulate IFN-{gamma}-producing T cells by DCs exposed to PS resulted from diminished IL-12p70 secretion. IL-12p40 secretion was also diminished, but to a lesser extent. IL-12p40 is usually produced in great excess and can form homodimers, which may serve as natural antagonists of biologically active IL-12 and have been shown to induce tolerance and to prevent autoimmunity in murine models (48). Although the effect in humans is controversial, unopposed IL-12p40 has been thought to induce Th2 differentiation or anergy (49).
We also examined the effect of PS on secretion of IL-10 and TGF-{beta}, which could modulate DC function and have been associated with resolution of inflammation (50, 51, 52). Macrophages exposed to PS or apoptotic cells produce TGF-{beta}, which in turn inhibits TNF-{alpha} secretion in response to LPS (22, 35). Notably, we did not observe an increase in TGF-{beta} secretion by DCs exposed to PS. These findings suggest that despite the close lineage relationship between macrophages and DCs, the responses to PS by DCs and macrophages appear substantially different, possibly because they play distinct immunological roles. The mechanisms responsible for modulating DC maturation by apoptotic cells are still not clear. Urban et al. (9) reported that human DCs exposed to apoptotic cells and treated with LPS secreted IL-10, but the inhibition of DC maturation was not IL-10 dependent. Takahashi and Kobayashi (53) did not find an increase in either IL-10 or TGF-{beta} secretion by murine DCs exposed to apoptotic cells. Stuart et al. (30) and Morelli et al. (12) also examined the effect of apoptotic cells on murine DC function. They found that inhibition of DC maturation was not dependent on either IL-10 or TGF-{beta}. The discrepancy between studies may arise from differences in the source of DCs, species examined, and type of serum or medium used. In addition, apoptotic cell preparations are heterogeneous and may include live, necrotic, early apoptotic, and late apoptotic cells. Such cells may express an array of surface molecules and secrete a myriad of cytokines that could affect DC maturation. We found that PS did not stimulate IL-10 secretion by DCs; instead, production of IL-10 in response to LPS was inhibited by PS. These findings are consistent with a recent report suggesting that PS inhibits the secretion of proinflammatory cytokines by microglial cells, without altering IL-10 or TGF-{beta} production (37). Although PS did not induce IL-10 or TGF-{beta} production by DCs in our in vitro model, TGF-{beta} produced by tissue macrophages in response to PS and IL-10 produced by regulatory T cells may play important roles in modulating DC maturation in vivo (41, 50, 52, 54, 55, 56, 57).
PS, externalized on the surface of apoptotic cells, tumor cells, and cells infected with some intracellular pathogens, represents a possible mechanism for tolerance to self-Ags and for immune evasion (10, 22, 57, 58, 59, 60, 61, 62, 63, 64). Our findings suggest that PS exposure directly inhibits DC maturation and modulates their ability to activate T cell responses. PS is specifically recognized by DCs and triggers phagocytosis via the PSR. Our findings suggest that PS modulates the adaptive immune response at three different levels: 1) PS inhibits survival, Ag presentation, and costimulation by DCs, leading to ineffective T cell proliferation; 2) PS inhibits IL-12p70 secretion by DCs, diminishing the ability to stimulate IFN-{gamma}-producing T cells characteristic of Th1 responses; and 3) DCs exposed to PS have reduced ability to stimulate the proliferation of activated CD8+ T cells, even in the presence of abundant Ags. PS may synergize with additional signals, such as CD200, IL-10, and TGF-{beta}, expressed by apoptotic, inflammatory, or stromal cells (20, 22, 50, 51, 56, 65, 66). Further understanding of the mechanisms by which PS modulates DC function may lead to the development of effective immunotherapies for autoimmune diseases and for cancer.
--------
j
j
KT, LOL, thanks :)
only on my first cup...
free,
I see it as dealing with our understanding/perspective on pathogenesis in general, in that it appears that in the evolution of metazoans, certain 'compromises'/loopholes had to happen between recognition of host/invader and their respective subsequent clearance processes. The invaders and disease states that now seem to be the most successful in causing the most serious problems in us are the ones that have exploited this.
in a nut shell -
Evolution has favored pathogenesis that resembles apoptosis.
and Bavi addresses this loophole.
It is indeed a paradigm shift, and everybody should read "the book"....
j
you forgot tuberculosis, atherosclerotic plaque, autoimmune diseases, sickle cell anemia, diabetes.
j
short info is coming out bi-monthly now, and the next glimpse we get will be tomorrow after 4:00
http://www.nasdaqtrader.com/trader/defincludes/nasdshortint_def.stm#pubnew
j
yes, it is amazing, and it's bavi's MOA, and it is applicable to any tumor type. It's the most important work I've seen.
Here's the abstract from AACR, it might answer some of your questions.
Abstract Number: 4091
Enhancing the immunogenicity of glioma cells with anti-phosphatidylserine antibody.
Jin He, Troy A. Luster, Philip E. Thorpe. UT Southwestern Medical Ctr., Dallas, TX
Malignant gliomas grow aggressively and infiltrate surrounding normal brain. None of the traditional therapies prevents the lethal growth of infiltrating tumor cells. In a previous study, we showed that phosphatidylserine (PS) becomes exposed on the surface of tumor vascular endothelium and F98 glioblastoma cells after exposure to irradiation. Rats having orthotopic glioblastomas were treated with single fractionated radiation combined with 2aG4, a mouse monoclonal antibody against PS. The treatment resulted in a marked prolongation of survival time and some tumor cures (15%). Surviving animals were immune to intracerebral challenge with live, untreated F98 cells. These findings suggested that PS on tumor cells might be suppressing host immune response to the tumor cells and that blocking PS with 2aG4 restored immunogenicity. In the present study, we explored the possibility of enhancing the immunogenicity of irradiated F98 cells by treating them with 2aG4 in vitro. Fisher/344 rats received biweekly s.c. injection of 107 irradiated F98 cells premixed with 2aG4, control antibody C44, or PBS for three weeks. One week after the last injection, rats were challenged with intracerebral injection of 105 F98 cells. A 57% long-term survival rate was observed in the 2aG4 group, versus a 16% and 0% for rats immunized in the control antibody group and PBS group, respectively. The superior survival in the 2aG4 group was statistically significant (P < 0.01). 2aG4-coated F98 cells were efficiently phagocytosed by DCs through FcgR on DCs. Co-culture of tumor-loaded DCs and T-cells demonstrated that antibody-coated F98 glioma cells are able to elicit specific T-cell responses, as judged by enhanced production of tumor-reactive IFN-g producing T cells and increased cytotoxicity of T cells on F98 tumors. These data suggest that anti-PS antibody treatment of tumor cells can enhance cross-presentation of tumor antigens and the generation of glioma-specific T cells by dendritic cells. Anti-PS antibody treatment might be combined with radiosurgery in clinical settings to treat brain tumor patients.
This work was conducted with the support of a grant from the Gillson Longenbaugh Foundation and a sponsored research agreement with Peregrine Pharmaceuticals Inc.
j
unpathedhaunt,
how long would you speculate it would take to produce anti-viral bavi?
Well, bavi is bavi, I'm not sure exactly how long it takes to make a batch. I think that in the past year or two they've (Avid) sped up the process, and increased efficiency/yield.
Is the DOD working on H5N1 in particular?
That's a good question. Don't know.
As for the animal rule, as Bavi demonstrates safety in more humans, it gets closer to satisfying the requirements.
There are other requirements - you've got to demonstrate efficacy in animals, you've got to have a good grasp on the MOA('s), you've got to be able to predict a good human dose.
j
free,
What you're looking at is evidence that covering the PS on the irradiated tumor cells restored the immunogenicity of the tumor cells, that DC's were successful in training T cells to go after tumor-specific antigens.
j
OT: ............
http://www.cnbc.com/id/21083662
j
free,
Who knows. We'll see when we see clinical data.
In general, the ability to target negative host cell phospholipids is looking incredibly promising.
As for adding an interleukin, interferon, etc., we'll see when we see clinical data.
Personally I'd expect that down the road, the receptorbodies, and betabodies, and the double beta-2 glycoprotein will be great stuff as well.
Then there would also be the possibility of small (peptide) PS-targeting molecules.
It'll be a huge field, if Bavi simply keeps performing as it already has been, which is why the Dept of Defense has already funded our PS-targeting abs for breast cancer, prostate cancer, tumor imaging, and is now negotiating for a very large project using the same PS-targeting ab platform as viral therapeutics.
pretty amazing platform :)
j