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.
Alan Bernstein re: Mexico City conf -
Statement from Alan Bernstein,
Executive Director of the Global HIV Vaccine Enterprise,
following the XVII International AIDS Conference:
http://www.hivvaccineenterprise.org/_dwn/news/2008_mexicocity_closing_statement.pdf
The AIDS 2008 conference, which ended last week, brought renewed focus to HIV prevention
efforts. This represents a dramatic shift, as increased access to treatment has dominated the
HIV/AIDS agenda for much of the past 20 years. While it is essential that treatment reach all
those who need it, it is clear that effective prevention is the only way to end the HIV pandemic.
The anti‐retroviral (ARV) drugs developed over the past two decades have been spectacularly
successful at keeping HIV in check – far beyond what anyone optimistically could have hoped
for. Yet despite these powerful new drugs and expanded access through programs like PEPFAR,
new infections continue to outpace the availability of treatment. There are still five new
infections for every two people who gain access to ARVs. ARVs, used in combinations to limit
resistance, can control HIV in most individuals, but they are not a long‐term solution to the HIV
pandemic. As Anthony Fauci, Director of the US National Institute of Allergy and Infectious
Diseases (NIAID), said during the conference, even these powerful drugs have never truly cured
anyone of HIV/AIDS – in the sense that they no longer require continued therapy.
When a person infected with HIV stops using ARVs, a latent reservoir of the virus, probably in
the gut, becomes activated. People living with HIV are therefore dependent on life‐long
combination therapy. Given that there are still almost 3 million new cases of HIV each year, the
cost of universal access to ARVs is astronomically high and will continue to grow unless new
infections fall. As Mark Harrington, Executive Director of the Treatment Action Group, stressed
in his conference presentation, "We must seek a cure and a vaccine."
Mark and other conference speakers (myself included) emphasized the need for a
comprehensive prevention strategy that includes short, medium and long term goals, and that
emphasizes both a cure and a vaccine. In the short term, we need to expand access to therapy,
which we know works to save lives. In the medium term, the world must intensify efforts to
develop and use social and biological interventions (such as condoms, behavior change and
circumcision), microbicides and other HIV prevention methods. ARVs themselves may offer
some hope in this regard: AIDS 2008 saw considerable discussion and some data on the concept
of Pre‐Exposure Prophylaxis (PrEP), including data from the BC Centre for Excellence in
HIV/AIDS in Vancouver that suggests that aggressive treatment prior to exposure may
significantly reduce the probability of viral transmission. While “Treatment as Prevention” holds
promise, further study is needed.
In the long term, the best way to stop HIV is with an effective, preventative vaccine. Finding an
HIV vaccine is the greatest scientific challenge of our time, and the effort will not be easy. But
history has shown that an effective vaccine is the best way to prevent the spread of a virus.
With a vaccine as our long‐term goal, we must ensure continued scientific momentum. We
should work to renew the current generation of distinguished scientists by attracting and
nurturing the next generation of HIV vaccine researchers. Young scientists from across the
globe will bring the creativity and energy required to surmount HIV's unique challenges. They
will also bring knowledge of the cutting edge technologies that are revolutionizing other areas
of biomedical research.
During AIDS 2008, in partnership with many other organizations, the Enterprise sponsored an
exciting Satellite Symposium entitled "New Minds, New Ideas: Attracting the Next Generation
of Investigators to HIV Vaccine Research." Panelists included José Esparza from the Bill &
Melinda Gates Foundation, Peggy Johnston from NIAID, Giuseppe Pantaleo from Centre
Hospitalier Universitaire Vaudois, and two distinguished young investigators: Dan Barouch from
Beth Deaconess Medical Center and Thumbi Ndung’u from the University of KwaZulu Natal. The
discussion was extremely stimulating and constructive, and resulted in a number of good ideas
and programs that the Enterprise will expand on in the coming months.
Later in the meeting, I chaired a panel discussion with four young prize‐winning investigators
working on different aspects of HIV/AIDS, including vaccine research. In both these sessions, I
was astounded by the skills and passion of the field’s young investigators – they are bright,
energetic, articulate and full of great ideas about how to move the field forward.
Today’s HIV vaccine scientists must be sure to mentor these young researchers and encourage
new investigators to enter the field. Their success will be our success. At the same time, young
scientists themselves must come together and form a coherent vision of the future of vaccine
research.
I believe strongly that it the responsibility of all Enterprise members – researchers young and
old, funders, host institutions, governments and advocates – to provide the opportunities,
environment and resources to foster creative ideas and advance the HIV vaccine field. This
effort must also cut across sectors. During AIDS 2008, Richard Horton, Editor of Lancet, noted
the decline in the number of scientists and science sessions at this year's meeting. He expressed
a concern that I share: that we are creating two solitudes, with social scientists and activists on
one side, and biomedical researchers on the other. This is not healthy for the field.
Coming out of this meeting, it essential that we ensure meaningful dialogue with the global
activist community, and that all of us agree that an effective HIV vaccine is an integral part of
any long‐term, comprehensive HIV prevention strategy. At the same time, the conference
reinforced my conviction that, with dedication and global collaboration, we will get a vaccine –
and that it is the new minds and new ideas that will get us there. AIDS 2008 provided new
impetus to global efforts to prevent the spread of HIV. It is up to all of us now to live up to its
promise.
---------
j
KT,
re: "I will find it interesting to see the nature and extent of the PPHM anti-PS technology platform that is discussed by Duke in October."
Yes, me too.
and while it will be discussed/presented by Haynes of Duke,
Haynes represents not simply Duke research, he'll be speaking for the two largest collaborative HIV research efforts ever. One which he himself is the Director of- CHAVI,
and the other which he is one of the principle researchers,The Gates Foundation's CAVD.
j
moby,
Yes, of course, in as much as we are both PPHM shareholders with similar hopes and expectations for the stock and the tech. You and I share a common desire to see Peregrine succeed.
j
what paper?
jess,
re: Haynes paper in Virology -
It's true - Haynes discovery that during early HIV infection a quick accumulation of apoptotic debris (PS-exposing microparticles) stifles antibody production by B cells! was a VERY important discovery.
It changes everything. It's why Tony Fauci, (present head of NIAID, allocator of mucho HIV research moneys, and world-famous HIV researcher from the beginning), mentioned his new optimistic outlook... - basically, while it "raised the bar" in terms of what a successful vaccine will need to do, it also finally much more clearly elucidated what a successful vaccine will need to do. They now know what they're up against. That's what the paper discussed.
(Personally, I think Haynes recent findings were important in Fauci's July decision to ditch the PAVE-100 vaccine trial, since Haynes findings, regarding exposed PS, predict that only vaccines that (somehow :) "deal" with that exposed PS will work...).
-------------------
Here's an excerpt by Marilyn Chase who writes for the Wall St. Journal-
NIH’s Fauci Finds Hope Amid Challenges in AIDS Research
by Marilyn Chase
The NIH’s Anthony Fauci, who once cautioned that there might never be a traditional vaccine to prevent HIV infection and recently pulled the plug on a troubled vaccine trial, sounded a cautiously optimistic note at the 17th International AIDS Conference yesterday.
“The future for AIDS research looks bright and promising,” said Fauci, singling out recent work by Barton Haynes of Duke and Robert Siliciano of Johns Hopkins in illuminating how the virus hides inside the body and suppresses the immune system within days of infection.
-------------------
Here's Fauci's verbatim quote in Mexico City which Marilyn Chase was referring to -
NIAID Director Anthony Fauci, QUOTE from his Mexico City speech -
"A recent paper just a few days ago from Bart Haynes’ group in CHAVI and Duke showed that not only is a reservoir formed early, but byproducts of CD4 positive T-cell deaths increase significantly within days and are capable of suppressing the human immune response to the virus. So, we have a double whammy. We have a reservoir that almost immediately is formed and we have products of the death of cells suppressing the immune response that would hopefully prevent the establishment of that reservoir.
---------------------
By the way - in the paper (in the journal Virology) Haynes also cites and seems to agree with previous researchers (the ones I've been citing for two years! :) who have shown how exposed PS changes macrophage behavior, the cytokine environment, and T cells.
What remains to be seen is the work being done by Dr's Borrow and Bhardwaj into how the massive apoptotic debris (PS-exposing microparticles) in early HIV infection affects DENDRITIC CELLS.... (I'd expect to hear about their work soon ;).
(but I'll tell you, that Joel Shilyansky has already proven that PS-exposing microparticles screw up dendritic cells antigen-presenting abilities...)
----------------
Meanwhile... Some Haynes patent applications recently became public on the WIPO and USPTO databases. They discuss HOW to do what we now know (thanks to Haynes Virology paper) needs to be done! :)
* Haynes now says that successful vaccines will need to BLOCK PS-MEDIATED IMMUNOSUPPRESSIVE SIGNALING. He suggests inducing anti-PS abs...
* In another patent applicaiton, Haynes now also suggests anti-PS mabs as HIV THERAPY for people infected with HIV!, - what looks to obviously be PEREGRINE'S anti-PS mab. (IS1, from Pojen Chen)...
-------------------------
To sum up the present situation -
Haynes says that HIV vaccines will need to induce anti-PS abs.
Haynes says that anti-PS mabs should be successful THERAPY for people already infected with HIV.
Haynes specifically mentions IS1 as the safe therapeutic anti-PS mab for people already infected with HIV.
hang on a sec -
Q: THIS IS THE DIRECTOR OF CHAVI AND GATES RIGHT HAND MAN SAYING THIS????
A: YES, THAT'S RIGHT :).............
In the course of his recent papers and patent applications, Haynes mentions the following anti-PS mabs:
*Bavi
*Humanized Bavi
*Tarvi
*2AG4
*3G4
* IS1
*IS2
* IS6
They ALL seem to be Peregrine's.........
from both Thorpe, and Chen, who both contribute "CORE TECHNOLOGY" to the company....
-------------------------
As for PAPERS with mentions of Peregrine's mabs, I expect one around the time of the Global HIV Vaccine Enterprise conference in October.
moby,
I expect it to mention, quite clearly, Peregrine's anti-PS, not as simply a testing reagent, but as a promising DRUG / THERAPY / TREATMENT FOR PEOPLE INFECTED WITH HIV, (as so clearly discussed in Haynes recent patent application).
It's happening :)
but hey -
Who's gonna pay for it?
Where will clinical trials take place?
LOL!
:)
j
Barton Haynes Recently Published Papers and Patent Applications
Haynes recent paper in the August issue of the journal Virology has generated much discussion in the HIV research community, as well as several popular news articles.
Examples of follow-on popular news stories:
HIV Conquers Immune System Faster Than Previously Realised
http://www.docguide.com/news/content.nsf/news/852571020057CCF68525748A00661CD7
HIV Overpowers Immune System Quicker than Previously Thought
http://www.genengnews.com/news/bnitem.aspx?name=38938669
These popular articles do a nice job of summarizing the basic insights found by the Haynes team which shed light on what is responsible for the rapid immune cell death found in early HIV infection. Haynes paper as well as his several patent applications that have recently appeared in the various US and international patent databases provide the all-important details. Haynes work discusses exposed phosphatidylserine (PS) on microparticles overwhelming immune cells, facilitating the rise in viral counts. The following information details this fascinating area of research, and points to this PS-induced immune suppression pathway as being an important mechanism involved in the pathogenesis of a broad range of diseases.
First, some background
Recent insights into enveloped virus infection, from the journal Science
In the April 2008 journal Science, authors Mercer and Helenius discuss data showing how vaccinia viruses (the pox family of viruses), utilize phosphatidylserine (PS) exposed on its surface to facilitate entry into cells. Besides PS aiding viral entry, the authors also discuss recent research showing that exposed PS also prevents an immune response to the virus. They use the term "apoptotic mimicry", (masquerading as a dying native cell), a term showing up more often in recent research involving how various unrelated pathogens use similar means to evade the cells of the immune system, thereby surviving and thriving in the body.
Vaccinia Virus Uses Macropinocytosis and Apoptotic Mimicry to Enter Host Cells
http://www.ncbi.nlm.nih.gov/pubmed/18436786?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
short quotes from the paper in Science -
“The induction of blebs, the endocytic event, and infection were all critically dependent on the presence of exposed phosphatidylserine in the viral membrane, which suggests that vaccinia virus uses apoptotic mimicry to enter cells.”
“The Mature Virion membrane has been shown to be enriched in PS and PS is required for infectivity”
The Mercer and Helenius paper spawned several well-written layman's articles on the new insight into viral infection. For instance:
Trojan Horse Of Viruses Revealed
http://www.sciencedaily.com/releases/2008/04/080425065354.htm
excerpts:
ScienceDaily (Apr. 25, 2008) — Viruses use various tricks and disguises to invade cells. Researchers have now discovered yet another strategy used by viruses: the vaccinia virus disguises itself as cell waste, triggers the formation of evaginations in cells and is suspected to enter the cell interior before the immune defense even notices.
The invasion strategy In order to infiltrate a cell, the vaccinia virus exploits the cellular waste disposal mechanism. When a cell dies, other cells in the vicinity ingest the remains, without needing waste disposal experts such as macrophages. The cells recognize the waste via a special molecule, phosphatidylserine, which sits on the inner surface of the double membrane of cells.
This special molecule is pushed out as soon as the cell dies and is broken into parts. The vaccinia virus itself also carries this official waste tag on its surface.
"The substance accumulates on the shell of vaccinia viruses", Jason Mercer explained. The pathogen disguises itself as waste material and tricks cells into digesting it, just as they normally would with the remains of dead cells. As the immune response is simultaneously suppressed, the virus can be ingested as waste without being noticed.
Dirty Rotten Poxviruses
http://sciencenow.sciencemag.org/cgi/content/full/2008/425/1
excerpts:
"poxviruses have engineered a way to sneak into cells through the garbage chute."
"the virus disguises itself as junk"
"the team wondered if the virus was playing dead. They found that the virus's surface was studded with phosphatidylserine, a lipid that also flags dead cells as garbage.
Removing lipids from the virus's surface stopped infection, and recoating the virus with phosphatidylserine
restarted it.
The results suggest that the virus is "more clever than originally thought" because it exploits a garbage-collection process found in almost all cells, says Mercer.
before proceeding further, two words to understand -
Phosphatidylserine: A phospholipid found on the inside of healthy cell walls. As our cells die, they lose the ability to maintain phosphatidylserine (PS) on the inside of the cell wall. PS becomes exposed on our dying cells, and is perceived by cells of the immune system as native / "self" debris, not as a foreign pathogen.
Microparticles: Small vesicles shed from dying cells. Microparticles are made of cell membrane phospholipid molecules. Microparticles expose PS.
A brief primer on phosphatidylserine's role in cell death and immune clearance
As multicellular organisms, we need an immune system that is flexible enough to react differently to foreign invaders as compared to dying native cells. Over the past decade, researchers have shed light on the "fork in the road" which dictates which general type of response our immune system pursues. Phosphatidylserine (PS) is a lipid typically found lining the interior of all healthy cell walls. PS flips to the exterior of the cell membrane as a result of various stresses, and is an early and universal sign of a dying native cell. It is not surprising that exposed PS has recently been shown to provide the fundamental signal for an immune response steered toward homeostasis, a self / maintenance type of immune response. Recent data now illustrates that exposed PS is the impetus that alters the behavior of all the major cell types of the immune system into NOT mounting a specific attack.
Nature finds a way
Scientists have also recently discovered a commonality between cancer, viral, and protozoan parasitic infections. These distinctly different pathogens all exploit exposed PS to aid their survival and proliferation in the body. This common strategy is no surprise when considered with the new understanding of the evolution of metazoan PS mediated immune suppression. Taken together, these recent discoveries suggest that successful pathogens independently evolved to exploit a similar evasion mechanism because it provides the crucial advantage (exposed PS) needed to avoid the immune system's attack.
Where does Barton Haynes fit in?
Over the past couple years, in his work looking into the pathogenesis of HIV, Barton Haynes has been following the hypothesis that the reason HIV manages to proliferate so quickly after infection, and the reason no vaccines have yet been effective, have both been due to a massive amount of "apoptotic debris" (microparticles from dying cells) which occurs early in HIV infection. This debris contains exposed phosphatidylserine (PS), and is responsible for the depletion of CD4+ T cells, as well as an overall blunting of immune response to the virus. Haynes puts forth the idea (documented below) that the fundamental culprit is phosphatidylserine (PS). He cites recent research (Hoffman et al, Interaction between Phosphatidylserine and the Phosphatidylserine Receptor Inhibits Immune Responses In Vivo, The Journal of Immunology, 174: 1393-1404), showing how exposed PS alters the function of macrophages, dendritic cells, and T cells. In the August Journal of Virology he also provides new data showing how microparticles with exposed PS alter the fuctions of antibody producing B cells.
here are earlier excerpts illustrating Haynes/CHAVI overall working hypothesis:
Haynes / CHAVI / Gates hypothesis example 1:
"Their [Haynes group] current hypothesis is that HIV induces a massive apoptosis before and during viral ramp up, and that plasma microparticles (fragments of apoptotic CD3 and T cells) have a suppressive effect on Ab generation, thereby amplifying the apoptotic cascade. "
May 22, 2007
http://www3.niaid.nih.gov/research/topics/HIV/vaccines/advisory/avrs/PDF/AVRS_May07_Summary.pdf
Haynes / CHAVI / Gates hypothesis example 2:
Conclusion: These results demonstrate that at the time of viral load ramp-up in acute HIV-1 infection, there are elevations in plasma levels of TRAIL, FAS Ligand, and TNFR2 that were associated with the presence of microparticles from apoptotic T cells. The presence of these apoptotic markers suggests apoptosis occurring at the time of initial HIV-1 viral load ramp-up. That 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-1 immune responses."
August 20, 2007
http://www.hivvaccineenterprise.org/_dwn/poster_sessions.pdf
Haynes recent publication
The following are excerpts from Bart Haynes' new paper in the August 2008 Journal of Virology, which discuss detailed data showing how exposed PS on microparticles is responsible for the weak immune response to HIV.
Induction of plasma (TRAIL), TNFR-2, Fas ligand, and plasma microparticles after human immunodeficiency virus type 1 (HIV-1) transmission: implications for HIV-1 vaccine design.
http://jvi.asm.org/cgi/content/abstract/82/15/7700
Excerpts:
("MP" = MicroParticles)
“In this study, we raise the hypothesis that in addition to gut CD4 T cell loss, delay in HIV-1 protective immune responses early on after HIV-1 transmission may involve the production of elevated levels of immunosuppressive moieties such as TRAIL, TNFR2 and Fas ligand as well as plasma microparticles.
Microparticles (MPs) are small membrane-bound vesicles that are released from the surface of apoptotic cells by exocytic or budding processes; as such, MPs bear cell surface markers and can bind annexin V because of the expression of phosphatidylserine (32-44, 39).
MPs, which circulate in the blood in many clinical conditions, are part of a spectrum of subcellular structures that are released from cells and can be distinguished from exosomes which are released from multivesicular bodies during activation.
“MPs have immunomodulatory activities and can promote immune cell death; exosomes are also immunologically active, can suppress immune responses (20,34,42,55), and have been reported elevated in chronic HIV-1 (4).”
“suppression of immune responses can be mediated by T cell MPs (32,34,35). CXCR4+ and CCR5+ MPs can transfer co-receptors to coreceptor negative cells, making them susceptible to infection by HIV-1 (48,57).
Phagocytosis of MPs by macrophages releases TGF-beta, prostaglandin E2 and IL-10 that can inhibit antigen specific T and B cell responses (20,35,42). In this regard, Estes et al. have shown dramatic increases in lymph node TGF-beta and IL-10 on day 12 following SIV infection (22).
Importantly, we have demonstrated that PBMC and tonsillar cell MPs can directly inhibit memory B cell activation (Figure 8).”
“it is likely that MPs are responsible for the observed B cell suppressive activity seen in vitro in Figure 8.
In the setting of HIV-1 infection where both activation and apoptosis occur, however, MPs and exosomes may act concomitantly, with exosomes suppressing immune responses (2,7,15,61), and MPs contributing to both immune suppression and cell death (20,32,34,35,39,42,55).”
August 2008, Journal of Virology
http://jvi.asm.org/cgi/content/abstract/82/15/7700
Where Does Haynes Go From Here? Patents Point The Way...
We can see where Haynes is going in his work by looking at two recent patent applications.
In one recent patent application, in the World Intellectual Property Organization database, Haynes discusses what a future successful HIV vaccine must do. He specifically states that any future successful vaccine must stop exposed PS from blunting immune response. He states that an essential goal of a future vaccine is to induce anti-PS antibodies, which would bind/block the PS from downregulating the immune response.
In Haynes second recent patent application, in the US database, he discusses how monoclonal anti-PS antibodies can be safe therapy in people already infected with HIV.
excerpts and links to the two recent patent applications:
The “Vaccine” Patent -
- Thus, HTV virions and HIV envelope can directly induce T cell death in AHI, soluble TRAIL can bind to uninfected cells and induce death in AHI, and with both HTV infection of cells and with massive apoptosis, high levels of phosphatidylserine containing cells and particles likely abound in AHI.
Phosphatidylserine (PS) on the surface of HIV infected cells and virions has been found (Figure 7) and Callahan et al have found PS is a cofactor for HIV infection of monocytes (Callahan, J. Immunol 170:4840 (2003)).
PS-dependent ingestion of apoptotic cells promotes TGF-βl secretion (Huynh et al, J. Clin. Invest. 109:41 (2002)) and interaction between PS and PS receptor inhibits antibody responses in vivo (Hoffman et al, J. Immunol. 174:1393 (2005)).
There are increases in PS+ shed membrane particles in chronic HIV infection (Aupeix et al, J. Clin. Invest. 99:1546 (1997)), and apoptotic microparticles modulate macrophage immune responses (Distler et al, Apoptosis 10:731 (2005)).
The present invention relates to a multicomponent vaccine that addresses problems resulting from the diversity of HIV by the use consensus and/or mosaic HIV genes coupled with strategies designed to break immune tolerance to allow for induction of the desired specificity of neutralzing antibodies at mucosal sites (e.g., through the use of T regulatory cell inhibition and/or TLR-9 agonist adjuvants), and strategies designed to overcome HIV-I induced apoptosis (e.g., induction of anti- phosphatidylserine (PS) antibodies, anti-CD36 antibodies, and/or antitat antibodies).
WHAT IS CLAIMED IS:
1. A method of inducing the production of an immune response against HIV-I in a mammal comprising administering to said mammal: i) a centralized HIV-I gene sequence, ii) an agent that breaks mammalian immune tolerance, and iii) an agent that inhibits HIV-I -induced apoptosis or an immunosuppressive effect of HIV-I -induced apoptosis, wherein (i), (ii) and (iii) are administered in amounts sufficient to effect said production.
22. The method according to claim 1 wherein said agent that inhibits HIV-1-induced apoptosis induces anti phosphatidylserine (PS) antibodies, anti- CD36 antibodies, or anti-HIV tat antibodies.
http://www.wipo.int/pctdb/en/wads.jsp?IA=US2007024122&LANGUAGE=EN&ID=id00000006427540&VOL=87&DOC=0084d3&WO=08/063 586&WEEK=22/2008&TYPE=A2&DOC_TYPE=PAMPH&PAGE=1
The “therapeutic” patent
Here Haynes discusses the use of anti-phospholipid monoclonal antibodies as HIV therapy. Haynes has moved from seeing the lipid binding characteristics of the well-known broadly-neutralizing HIV abs (2F5, 4E10) as unfortunate, and now sees the lipid-binding characteristics as “the key” to broad and safe HIV neutralization. Haynes goes so far as to say that there is no need for an HIV neutralizing antibody to bind a viral epitope. The idea is presented that binding a host-cell 'tag-along' phospholipid results in successful HIV neutralization.
WHAT IS CLAIMED IS:
1. A method of treating HIV comprising administering to a patient in need thereof an antibody derivable from a normal subject or from an autoimmune disease subject that binds to a lipid on the surface of HIV or on the surface of HIV-infected cells and thereby neutralizes HIV-
1, wherein said antibody is administered in an amount sufficient to effect said treatment.
2. The method according to claim 1 wherein said antibody is derivable from an anti-phospholipid syndrome subject.
3. The method according to claim 1 wherein said antibody is non-pathogenic.
4. The method according to claim 1 wherein said antibody is IS1, IS4 or IS6, or binding fragment thereof.
5. The method according to claim 1 wherein said antibody is IS1, or binding fragment thereof.
[0105] ....That IS1 neutralized HIV evidences the facts that: a) humans can make non-pathogenic anti-lipid antibodies that neutralize HIV, and b) IS1 is an antibody that can be safely used as a therapeutic Mab for treatment of HIV infected subjects or in the setting of postexposure prophylaxis of subjects following needle, sexual or other exposure to HIV or HIV infected materials.
http://appft1.uspto.gov/netacgi/nphParser?Sect1=PTO2&Sect2=HITOFF&u=%2Fnetahtml%2FPTO%2Fsearchdv.html&r=2&p=1&f=G&l=50&d=PG01&S1=%28%22haynes+barton%22.IN.%29&OS=in/%22haynes+barton%22RS=IN/%22haynes+barton%22
The Big Picture
In the Haynes "therapeutic patent" above, one additional quote stands out:
[0051]" It will be appreciated from a reading of the foregoing that if HIV has evolved to escape the host immune response by making the immune system blind to it, other infectious agents may have evolved similarly. That is, this may represent a general mechanism of escape. That being the case, approaches comparable to those described herein can be expected to be useful in the treatment of such other agents well."
In their recent paper in Science, (the top of this post) Mercer and Helenius also speculate on this broad-based pathogenic immuneevasion mechanism:
Also, in the same issue of Science, Fairn and Grinstein discuss the broader implications of Mercer and Helenius's findings:
A One-Sided Signal
http://www.sciencemag.org/cgi/content/full/320/5875/458
Gregory D. Fairn and Sergio Grinstein
excerpts:
"Because the infected cells undergo apoptosis, and thus experience scrambling of plasma membrane lipids, the budding virus also acquires an envelope that exposes phosphatidylserine on its external surface"
"the presence of exofacial phosphatidylserine is required for viral entry. "
"The involvement of phosphatidylserine may not be limited to infection by vaccinia viruses."
"It is therefore conceivable that HIV similarly requires phosphatidylserine for infection"
"The role of phosphatidylserine in the entry of HIV and other viruses will surely be explored in greater detail now”
ADDENDUM - JULY 24, 2008:
On July 24, 2008, another Haynes patent application went public on the World Intellectual Property Organization database.
Pertinent excerpts follow:
"The time of appearance of antibodies in the development of acute HIV infection has been recently mapped and it has been shown that most of the antibodies arise after a delay in the peak response to HIV envelope epitopes of approximately two to three weeks. Indeed, the most protective antibodies, those that neutralize autologous virus, can be delayed for up to a year."
"To begin to understand the "delay" in induction of antibodies at the time of HIV transmission, the first question to be addressed was whether there are immunosuppressive events, such as massive apoptosis, with release of phosphatidylserine microparticles at the time of viral load ramp up during acute HIV infection."
"Apoptotic microparticles are the products of either activated or apoptotic cells, that are increased in the plasma of a number of diseases, including autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis, Crohn's disease, coronary artery disease and other forms of heart disease, and chronic HIV-I infection."
"Apoptotic microparticles can bind to non-apoptotic cells and induce apoptosis, are procoagulant, proinflammatory, and can be immunosuppressive for T and B cell responses to specific antigen."
"Thus, the massive apoptosis that occurs with acute HIV infection with resulting release of TRAIL, mediation of apoptosis via FAS-FASL interactions, and release of PS containing viral and other particles, all conspire to initially immuno suppress the host, preventing rapid protective B cell responses."
http://www.wipo.int/pctdb/en/wads.jsp?IA=US2008000412&LANGUAGE=EN&ID=id00000006622149&VOL=89DOC=00fca1&WO=08/088747&WEEK=30/2008&TYPE=A2&DOC_TYPE=PAMPH&PAGE=1
-------------------
j
flgf4 -
from one of several extremely pertinent and fascinating Barton Haynes recently published patent applications:
WHAT IS CLAIMED IS:
1. A method of treating HIV comprising administering to a patient in need thereof an antibody derivable from a normal subject or from an
autoimmune disease subject that binds to a lipid on the surface of HIV or on the surface of HIV-infected cells and thereby neutralizes HIV-
1, wherein said antibody is administered in an amount sufficient to effect said treatment.
2. The method according to claim 1 wherein said antibody is derivable from an anti-phospholipid syndrome subject.
3. The method according to claim 1 wherein said antibody is non-pathogenic.
4. The method according to claim 1 wherein said antibody is IS1, IS4 or IS6, or binding fragment thereof.
5. The method according to claim 1 wherein said antibody is IS1, or binding fragment thereof.
[0105] ....That IS1 neutralized HIV evidences the facts that: a) humans can make non-pathogenic anti-lipid antibodies that neutralize HIV,
and b) IS1 is an antibody that can be safely used as a therapeutic Mab for treatment of HIV infected subjects or in the setting of postexposure
prophylaxis
---------
j
Michael,
"all the collaborators" is referring to all the institutions doing the work with Peregrine's anti-PS. (Duke, Harvard, NIH, etc.)
As for more info from Georgia, they said they'll be putting out an update in mid October, which is right around the time of the Global Vaccine Enterprise's conf., where none other than Bart Haynes will be presenting data on Peregrine's anti-PS against HIV / SIV / SHIV / whatever. They don't have to put out any updates. They could just run the cancer trial and report on it when it's complete, but they announced, in advance, that they would be putting out an update in mid October when all patients in the initial cohort had been out at least six months, and King specifically said they'd be talking about progression-free data, duration of response, etc...
Looks to me like the world will hear about a promising new HIV treatment - Peregrine's anti-PS, (from a very famous and the number 1 most NIH-funded research scientist, who happens to be the Director of the NIH's $350MM CHAVI), and then- news will also come out around the same time regarding Peregrine's anti-PS being used as a cancer treatment...
j
Upcoming Global HIV Vaccine Enterprise conference -------
Question to SK-
"The AIDS conference in Africa - is that going to be a comprehensive presentation of everything that Duke has been doing?"
SK: "It's probably not comprehensive, but it will certainly be laying out an awful lot of the story of what was learned with the anti-PS platform technology, and what we think is its great promise as potentially playing a role in treating or developing an AIDS vaccine. So it's going to be a lot of data. Bottom line is we've generated more data than we could possible present in one simple presentation."
Q: "Who will be presenting it?"
SK "That will be presented by Bart Haynes, who's the head of the group there, and obviously very well-known in the HIV research area. So there'll be a lot of data presented there, but there's a lot more coming, so i would see this being sort of the kick-off if you will of a number of potential presentations and publications in this area for the program in our collaboration with Duke and the other institutions involved."
Q: "Could a scientific publication precede this conference or-"
SK: "It possibly could. It's a little harder to predict. Once things are submitted, the review process - sometimes they sail through, sometimes they want you to make some adjustments to the manuscript, so... but yeah, I think there's a lot of - we've generated so much data we probably have enough for quite a few future activities"...
----------
j
Duke / Peregrine anti-PS quote ----------
SK: "We're also continuing multiple preclinical collaborations in the anti-viral area. In particular, our collaboration with researchers at Duke and other institutions testing our anti-PS antibodies continues to yield valuable insights into the potential of this technology platform in HIV, and I am happy to report that data from this collaboration will be highlighted this fall at the prestigeous AIDS Vaccine 2008 conference to be held in Cape Town South Africa from October 13th to the 16th. This will mark the first time that data from our collaboration has been presented, and we are very pleased with the opportunity to share the information with the broader scientific community. These collaborations have allowed us to make considerable progress in understanding the potential of our anti-PS technology platform for the treatment and possible prevention of serious virus infections, and have only served to heighten our excitement over the antiviral potential of this platform."
---------------
HIV TREATMENT......
Who's been paying for this work so far?
The Gates Foundation has.
j
jake,
Agree. Response rate looks great, but the most important stat will be median progression-free survival, duration of response, etc.
They'll be seeing more clearly whether the Bavi patients beat Avastin after they've passed the 7 month mark. As patients pass that point in time, if more than half are still progression-free, that's big.
The first patient(s) were dosed around Feb 12th. It has now been 30 weeks, to the day, since feb 12th. The patients go OFF CHEMO after 6 four-week chemo cycles, (24 weeks). So some of the earliest treated patients are on just Bavi now, but we don't know if they've been scanned post-chemo yet. Now, at the 30 week point, the earliest patients dosed are just passing the point of Avastin median progression-free survival...
j
9 out of`14 with objective responses -----------
(previous update was 7 out of 14)
j
HIV & DC's - Nina Bhardwaj of NYU -------
(in work funded by Gates & CHAVI)
http://www.jci.org/articles/view/34823
j
CMV burden / Immunosenescence / Aging
Biology of longevity: role of the innate immune system.
Rejuvenation Res. 2006
Candore G, Colonna-Romano G, Balistreri CR, Di Carlo D, Grimaldi MP, Listì F, Nuzzo D, Vasto S, Lio D, Caruso C.
Immunosenescence Unit, Department of Pathobiology and Biomedical Methodology, University of Palermo, Palermo, Italy.
Genetic factors play a relevant role in the attainment of longevity because they are involved in cell maintenance systems, including the immune system. In fact, longevity may be correlated with optimal functioning of clonotypic and natural immunity. The aging of the immune system, known as immunosenescence, is the consequence of the continuous attrition caused by chronic antigenic overload. The antigenic load results in the progressive generation of inflammatory responses involved in age-related diseases. Most of the parameters influencing immunosenescence appear to be under genetic control, and immunosenescence fits with the basic assumptions of evolutionary theories of aging, such as antagonistic pleiotropy. In fact, by neutralizing infectious agents the immune system plays a beneficial role until reproduction and parenting. However, by determining chronic inflammation, it can be detrimental later in life, a period largely unforeseen by evolution. In particular, the data coming from the long-lived male population under study show that genetic polymorphisms responsible for a low inflammatory response might result in an increased chance of long lifespan in an environment with a reduced pathogen burden. Such a modern and healthy environment also permits a lower grade of survivable atherogenic inflammatory response.
http://www.ncbi.nlm.nih.gov/pubmed/16608411?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_Discovery_RA
Human immunosenescence: is it infectious?
Immunol Rev. 2005 Jun
Pawelec G, Akbar A, Caruso C, Solana R, Grubeck-Loebenstein B, Wikby A.
University of Tübingen Medical School, Center for Medical Research, ZMF, Germany.
Morbidity and mortality due to infectious disease is greater in the elderly than in the young, at least partly because of age-associated decreased immune competence, which renders individuals more susceptible to pathogens. This susceptibility is particularly evident for novel infectious agents such as in severe acute respiratory syndrome but is also all too apparent for common pathogens such as influenza. Many years ago, it was noted that the elderly possessed oligoclonal expansions of T cells, especially of CD8(+) cells. At the same time, it was established that cytomegalovirus (CMV) seropositivity was associated with many of the same phenotypic and functional alterations to T-cell immunity that were being reported as biomarkers associated with aging. It was discovered that CMV was the prime driving force behind most of the oligoclonal expansions and altered phenotypes and functions of CD8 cells. Independently, longitudinal studies of a free-living population of the very old in Sweden over the past decade have led to the emerging concept of an 'immune risk phenotype' (IRP), predicting mortality, which was itself found to be associated with CMV seropositivity. These findings support our hypothesis that the manner in which CMV and the host immune system interact is critical in determining the IRP and hence is predictive of mortality. In this sense, then, we suggest that immunosenescence is contagious.
http://www.ncbi.nlm.nih.gov/pubmed/15882359?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_Discovery_RA
Human immunosenescence: does it have an infectious component?
Ann N Y Acad Sci. 2006 May
Pawelec G, Koch S, Franceschi C, Wikby A.
University of Tübingen Medical School, Center for Medical Research, ZMF, Germany.
The rate of acceleration of the frequency of death due to cardiovascular disease or cancer increases with age from middle age up to around 75-80 years, plateauing thereafter. Mortality due to infectious disease, however, does not plateau, but continues to accelerate indefinitely. The elderly are particularly susceptible to novel infectious agents such as SARS, as well as to previously encountered pathogens. Why is this? The elderly commonly possess oligoclonal expansions of T cells, especially of CD8 cells, which, surprisingly, are associated with cytomegalovirus (CMV) seropositivity. This in turn is associated with many of the same phenotypic and functional alterations to T cell immunity that have been suggested as biomarkers of immune system aging. We suggest that, in fact, CMV, not age per se, is the prime driving force behind many or most of the oligoclonal expansions and altered phenotypes and functions of CD8 cells in the elderly. Thus, the manner in which CMV and the host immune system interact (over which period? on which genetic background? with which co-infections?) is critical in determining the "age" of adaptive immunity and hence human longevity. In this respect, immunosenescence is infectious.
http://www.ncbi.nlm.nih.gov/pubmed/16803971?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_Discovery_RA
Immunological biomarkers of ageing in man: changes in both innate and adaptive immunity are associated with health and longevity.
Biogerontology. 2006 Oct-Dec
DelaRosa O, Pawelec G, Peralbo E, Wikby A, Mariani E, Mocchegiani E, Tarazona R, Solana R.
Immunology Unit, Department of Cellular Biology, Physiology and Immunology, University of Córdoba, Avda. Menéndez Pidal, s/n. 14071, Cordoba, Spain.
Scientific and clinical advances in the last century have led to increased numbers of individuals living to older ages. Thus a major concern is how to live these years with a high quality of life. The ageing immune system is less well able to cope with infectious diseases than the youthful immune system probably as a consequence of altered immune response to pathogens. Thus, both innate and adaptive immune responses show age-related changes that could be decisive for healthy ageing and survival. Longitudinal studies in healthy elderly have allowed the definition of the ''immune risk phenotype" (IRP) a predictor of mortality in elderly individuals that is based on several parameters of the adaptive immune response. Here, we hypothesize that failures in innate immunity observed in frail elderly are related to those alterations described in adaptive immunity defined as the IRP. It will be important to include assays of NK cell markers and functions in future longitudinal studies in order to investigate this point in detail as well as to consider the trace element zinc as an essential co-factor for optimal NK cell activity.
http://www.ncbi.nlm.nih.gov/pubmed/16957868?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_Discovery_RA
Immunity and ageing in man.
Exp Gerontol. 2006 Dec
Pawelec G.
University of Tübingen Center for Medical Research, Waldhörnlestr. 22, D-72072 Tübingen, Germany.
Immunosenescence resulting in decreased ability to control infectious disease contributes to morbidity and mortality not only in the very elderly, but in all likelihood already from middle age. Studying immunity in humans is therefore essential for developing treatments to restore dysregulated immune responses and assure healthy longevity. The past year has seen many significant advances in our knowledge of age-associated alterations to immunity in elderly people, only some of which can be briefly reviewed here.
http://www.ncbi.nlm.nih.gov/pubmed/17118601?ordinalpos=20&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
Analyses of immunosenescent markers in patients with autoimmune disease.
Clin Immunol. 2007 May
Thewissen M, Somers V, Venken K, Linsen L, van Paassen P, Geusens P, Damoiseaux J, Stinissen P.
Hasselt University, Biomedical Research Institute and Transnationale Universiteit Limburg, School of Life Sciences, Agoralaan, Diepenbeek, Belgium, and Department of Clinical and Experimental Immunology, University Hospital Maastricht, The Netherlands.
The objective of this study was to evaluate the degree of immunosenescence in patients with autoimmune disease. T cell receptor excision circles (TREC) and the percentage of CD4+CD28null T cells were studied as markers of immunosenescence in 175 patients with chronic autoimmune arthritis, other connective tissue autoimmune diseases, multiple sclerosis and 60 healthy controls. In both the rheumatoid arthritis (RA) and multiple sclerosis patient group, TREC numbers were age-inappropriately declined which points to an accelerated thymic output. Furthermore, enhanced percentages of CD4+CD28null T cells could be detected in a significant proportion of patients included in this study. These immunosenescent phenomena seemed to be present already early in the disease process. High percentages of CD4+CD28null T cells were associated with the presence of RA linked HLA DR4 alleles and with plasma reactivity to cytomegalovirus. Further analysis of CD4+CD28null T cells provided indications for a restricted T cell receptor (TCR) BV gene expression and cytoplasmic stores of various cytotoxic molecules. This study indicates that the immune system of patients with autoimmune diseases shows signs of an accelerated aging. Both genetic factors, such as HLA DR4, and environmental factors, like CMV infection, might speed up this immunosenescence and contribute in this way to disease pathogenesis.
http://www.ncbi.nlm.nih.gov/pubmed/17317320?ordinalpos=15&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
Healthy aging and latent infection with CMV lead to distinct changes in CD8+ and CD4+ T-cell subsets in the elderly.
Hum Immunol. 2007 Feb
Weinberger B, Lazuardi L, Weiskirchner I, Keller M, Neuner C, Fischer KH, Neuman B, Würzner R, Grubeck-Loebenstein B.
Immunology Division, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria.
Despite general acceptance that immunologic changes are associated with aging and latent infection with Cytomegalovirus (CMV), no clear-cut distinction has so far been made between strictly age-related and CMV-induced changes. We therefore compared CD4+ and CD8+ naïve (CD45RA+CD28+), memory (CD45RA-CD28+), and effector (CD28-) T cells in CMV-positive (n = 164) and CMV-negative (n = 87) elderly persons and correlated CD8+ and CD4+ effector T cells with other T-cell subpopulations. Percentages of CD8+ as well as CD4+ effector T cells were higher, but percentages of naïve and memory cells were lower in CMV-positive compared to CMV-negative elderly persons. Negative correlations within CD8+ T-cell subsets were found to be present in both CMV-positive and CMV-negative elderly individuals. In contrast, correlations within CD4+ T-cell subpopulations and a positive correlation between CD8+ and CD4+ effector T cells were found in CMV-positive individuals only. Our results demonstrate that (a) in the elderly different T-cell subsets compete for space within the CD8+, but not the CD4+ T-cell population; (b) CMV induces changes in the CD4+ compartment that differ from the solely age-related changes seen in CMV-negative elderly population; and (c) the CMV-status of a population has to be taken into account before a conclusion on the effect of aging on the composition of the T-cell pool can be reached.
http://www.ncbi.nlm.nih.gov/pubmed/17321897?ordinalpos=14&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
Role of persistent CMV infection in configuring T cell immunity in the elderly.
Immun Ageing. 2007 Mar
Vasto S, Colonna-Romano G, Larbi A, Wikby A, Caruso C, Pawelec G.
Gruppo di Studio sull'Immunosenescenza, Dipartimento di Biopatologia e Metedologie Biomediche, University of Palermo, Italy.
Ageing is associated with declines in many physiological parameters, including multiple immune system functions. The rate of acceleration of the frequency of death due to cardiovascular disease or cancer seems to increase with age from middle age up to around 80 years, plateauing thereafter. Mortality due to infectious disease, however, does not plateau, but continues to accelerate indefinitely. The elderly commonly possess oligoclonal expansions of T cells, especially of CD8 cells, which, surprisingly, are often associated with cytomegalovirus (CMV) seropositivity. This in turn is associated with many of the same phenotypic and functional alterations to T cell immunity that have been suggested as biomarkers of immune system aging. Thus, the manner in which CMV and the host immune system interact is critical in determining the "age" of specific immunity. We may therefore consider immunosenescence in some respects as an infectious state. This implies that interventions aimed at the pathogen may improve the organ system affected. Hence, CMV-directed anti-virals or vaccination may have beneficial effects on immunity in later life.
http://www.ncbi.nlm.nih.gov/pubmed/17376222?ordinalpos=13&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
The cytomegalovirus-specific CD4+ T-cell response expands with age and markedly alters the CD4+ T-cell repertoire.
J Virol. 2007 Jul
Pourgheysari B, Khan N, Best D, Bruton R, Nayak L, Moss PA.
C.R. U.K. Institute for Cancer Studies, Vincent Drive, Edgbaston, University of Birmingham, Birmingham B15 2TA, United Kingdom.
Immune function in the elderly is associated with a number of phenotypic and functional abnormalities, and this phenomenon of immune senescence is associated with increased susceptibility to infection. The immune response to pathogens frequently declines with age, but the CD8(+) T-cell response to cytomegalovirus (CMV) is unusual, as it demonstrates a significant expansion over time. Here we have documented the CD4(+) T-cell immune response to CMV in healthy donors of different ages. The magnitude of the CMV-specific CD4(+) T-cell immune response increases from a mean of 2.2% of the CD4(+) T-cell pool in donors below 50 years of age to 4.7% in donors aged over 65 years. In addition, CMV-specific CD4(+) T cells in elderly donors demonstrate decreased production of interleukin-2 and less dependence on costimulation. CMV seropositivity is associated with marked changes in the phenotype of the overall CD4(+) T-cell repertoire in healthy aged donors, including an increase in CD57(+) expression and a decrease in CD28 and CD27 expression, a phenotypic profile characteristic of immune senescence. This memory inflation of CMV-specific CD4(+) T cells contributes to evidence that CMV infection may be damaging to immune function in elderly individuals.
http://www.ncbi.nlm.nih.gov/pubmed/17409149?ordinalpos=9&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
No Immune Risk Profile among individuals who reach 100 years of age: findings from the Swedish NONA immune longitudinal study.
Exp Gerontol. 2007 Aug
Strindhall J, Nilsson BO, Löfgren S, Ernerudh J, Pawelec G, Johansson B, Wikby A.
Department of Natural Science and Biomedicine, School of Health Sciences, Jönköping University, Box 1026, 551 11 Jönköping, Sweden.
In the present NONA immune longitudinal study, we investigate the previously identified Immune Risk Profile (IRP), defined by an inverted CD4/CD8 ratio and associated with persistent cytomegalovirus infection and increased numbers of CD8+CD28- cells, relative 6-year survival and age in NONA individuals. These subjects have now reached age 92, 96, and for the first time in this study, 100 years at follow-up. A 55 year old middle-aged group was used for comparison. Immunological monitoring included the analysis of numbers of lymphocytes and neutrophils, the T-cell subsets CD3+CD4+, CD3+CD8+, CD8+CD28+, CD8+CD28-, and the CD4/CD8 ratio. Longitudinal data were analysed by multivariate analyses of variance (MANOVA) from four measurement occasions at 2-year inter-intervals. One-way ANOVA was used for cross-sectional comparisons at baseline and the 6-year follow-up. The results confirmed the importance of the IRP as a major predictor of mortality in this population of very old. Moreover, the results suggested that survival to the age of 100 years is associated with selection of individuals with an "inverted" IRP that was stable across time, i.e., maintenance of a high CD4/CD8 ratio and low numbers of CD8+CD28- cells. The results underlines the importance of a longitudinal study design in dissecting immune parameters predictive of survival and show for the first time that centenarian status is associated with avoidance of the IRP over at least the previous 6 years and probably throughout life.
http://www.ncbi.nlm.nih.gov/pubmed/17606347?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
Massive load of functional effector CD4+ and CD8+ T cells against cytomegalovirus in very old subjects.
J Immunol. 2007 Sep 15
Vescovini R, Biasini C, Fagnoni FF, Telera AR, Zanlari L, Pedrazzoni M, Bucci L, Monti D, Medici MC, Chezzi C, Franceschi C, Sansoni P.
Department of Internal Medicine and Biomedical Sciences, University of Parma, Parma, Italy.
A progressive, systemic, and low-grade proinflammatory status is one of the major characteristics of immunosenescence. Emerging data suggest a possible contribution of CMV, known to chronically infect a large proportion of humans, lifelong from newborns to centenarians. To test this hypothesis, we evaluated functional T cell responses to two CMV immunogenic proteins, pp65 and IE-1, in 65 chronically infected subjects aged 25-100 years. PBMC were stimulated with mixtures of peptides spanning the entire sequence of both proteins, and Ag specificity and magnitude of intracellular IFN-gamma- and TNF-alpha-positive cells were then analyzed within both CD4+ and CD8+ T cells. Results indicate that pp65 and, to a lesser extent, IE-1 constitute major Ags against which aged people target functionally efficient T cell effector responses with massive production of Th1 cytokines and exhibition of CD107a degranulation marker. As a result, the production of IFN-gamma induced in T cells by both Ags was seven to eight times greater in very old than in young subjects. The comparative analysis of pp65-specific responses in these very long-term carriers revealed a reciprocal relationship between CD4+ and CD8+ producing IFN-gamma in the same individuals. These results indicate that CMV represents an important pathogen responsible for a strong immune activation in human aging. Such a remarkable burden of effector CD4+ and CD8+ T cells may be necessary to protect the elderly from CMV endogenous reactivation, but can turn detrimental by giving a substantial contribution to the proinflammatory status that accompanies the main age-related diseases.
http://www.ncbi.nlm.nih.gov/pubmed/17785869?ordinalpos=5&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
Dynamics of T cell memory in human cytomegalovirus infection.
Med Microbiol Immunol. 2008 Feb
Waller EC, Day E, Sissons JG, Wills MR.
Department of Medicine, Level 5, Addenbrookes Hospital, University of Cambridge, Hills Rd, Cambridge, CB2 2QQ, UK.
Primary human cytomegalovirus (HCMV) infection of an immunocompetent individual leads to the generation of a robust CD4+ and CD8+ T cell response which subsequently controls viral replication. HCMV is never cleared from the host and enters into latency with periodic reactivation and viral replication, which is controlled by reactivation of the memory T cells. In this article, we discuss the magnitude, phenotype and clonality of the T cell response following primary HCMV infection, the selection of responding T cells into the long-term memory pool and maintenance of this memory T cell population in the face of a latent/persistent infection. The article also considers the effect that this long-term surveillance of HCMV has on the T cell memory phenotype, their differentiation, function and the associated concepts of T cell memory inflation and immunosenescence.
http://www.ncbi.nlm.nih.gov/pubmed/18301918?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
The immune system in extreme longevity.
Exp Gerontol. 2008 Feb
Sansoni P, Vescovini R, Fagnoni F, Biasini C, Zanni F, Zanlari L, Telera A, Lucchini G, Passeri G, Monti D, Franceschi C, Passeri M.
Dipartimento di Medicina Interna e Scienze Biomediche, Università di Parma, Via Gramsci 14, 43100 Parma, Italy.
Recent observations indicate that immunosenescence is not accompanied by an unavoidable and progressive deterioration of the immune function, but is rather the result of a remodeling where some functions are reduced, others remain unchanged or even increased. In addition, it appears that the ancestral/innate compartment of the immune system is relatively preserved during aging in comparison to the more recent and sophisticated adaptive compartment that exhibit more profound modifications. The T-cell branch displays an age-dependent decline of the absolute number of total T-cells (CD3+), involving both CD4+ and CD8+ subsets, accompanied by an increase of NK cells with well-preserved cytotoxic function and by a reduction of B-cells. One of the main characteristics of the immune system during aging is a progressive, age-dependent decline of the virgin T-cells (CD95-), which is particularly profound at the level of the CD8+ subpopulation of the oldest old subjects. The progressive exhaustion of this important T-cell subpopulation dedicated primarily to the defense against new antigenic challenges (viral, neoplastic, bacterial ones), could be a consequence of both the thymic involution and the lifelong chronic antigenic stimulation. The immune function of the elderly, is therefore weakened by the exhaustion of CD95- virgin cells that are replaced by large clonal expansions of CD28- T-cells. The origin of CD28- cells has not been completely clarified yet, but it is assumed that they represent cells in the phase of replicative senescence characterized by shortening telomers and reduced proliferative capacity. A major characteristic of the immune system during aging is the up-regulation of the inflammatory responses which appears to be detrimental for longevity. In this regard, we have recently observed a progressive age-dependent increase of type 1(IL-2, IFN-gamma, TNF-alpha) and type 2 (IL-4, IL-6, IL-10) positive CD8+ T-cells; in particular, type 1 cytokine-positive cells significantly increased, with age, in all CD8+ subsets particularly among effector/cytotoxic and memory cells. A major force able to drive a chronic pro-inflammatory state during aging may be represented by persistent viral infections by EBV and CMV. Therefore, we have determined the frequency and the absolute number of viral antigen-specific CD8+ T-cells in subjects older than 85 years, who were serologically positive for CMV or EBV. In the majority of these subjects we detected the presence of T lymphocytes positive for epitopes of CMV or EBV. In all subjects the absolute number of CMV-positive CD8+ cells outnumbered that of EBV-positive ones. In addition, the majority of CMV+ T cells were included within the CD28- subpopulation, while EBV+ T cells belonged mainly to the CD28+ subset. These data indicate that the chronic antigenic stimulation induced by persistent viral infections during aging bring about important modifications among CD8+ subsets, which are particularly evident in the presence of CMV persistence. The age-dependent expansions of CD8+CD28- T-cells, mostly positive for pro-inflammatory cytokines and including the majority of CMV-epitope-specific cells, underlines the importance of chronic antigenic stimulation in the pathogenesis of the main immunological alterations of aging and may favour the appearance of several pathologies (arteriosclerosis, dementia, osteoporosis, cancer) all of which share an inflammatory pathogenesis.
http://www.ncbi.nlm.nih.gov/pubmed/17870272?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_Discovery_RA
The immunological burden of human cytomegalovirus infection.
Arch Immunol Ther Exp (Warsz). 2007
Khan N.
Division of Immunology, School of Infection and Host Defence, University of Liverpool, UK.
Cytomegalovirus (CMV) is a persistent DNA virus that has evolved with humans to establish a finely balanced host-virus relationship. This balance is maintained by host immune surveillance since deficiencies in these processes can result in life-threatening disease, as observed in immunologically immature neonates and pharmacologically immunosuppressed transplant recipients. Both T cells and natural killer cells are intimately involved in maintaining asymptomatic infection by specific and non-specific recognition of infected cells. Under pressure from such host immune responses, CMV appears to have evolved elaborate strategies to subvert these responses in order to persist in the host. CMV target antigens are well characterized, with many CD8 T cell and CD4 T cell epitopes reported. This information is now being exploited to treat immunocompromised patients in order to boost virus-specific immunity. This review also discusses our current understanding of how virus carriage may skew lymphocyte populations in immunocompetent subjects and the association of CMV-seropositivity with immunosenescence.
http://www.ncbi.nlm.nih.gov/pubmed/18219760?ordinalpos=9&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum
---------
j
As for blocking "pathways"..............
Scientists are saying that PS is the impetus that shifts the immune response away from an antigen-specific response.
I often post papers from the growing body of evidence that discusses this. If I had to pick one that best points to the new "grand unification" perspective of pathogenesis, I suppose this paper by Peter Henson sums it up well.
Immunological Consequences of Apoptotic Cell Phagocytosis
http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pubmed&pubmedid=17591947
Immunological Consequences of Apoptotic Cell Phagocytosis
Abstract
Cells undergo apoptosis in development, tissue homeostasis, and disease and are subsequently cleared by professional and nonprofessional phagocytes. There is now overwhelming evidence that phagocyte function is profoundly altered following apoptotic cell uptake, with consequences for the ensuing innate and adaptive immune response. Pathogens and tumors exploit the changes in macrophage function following apoptotic cell uptake. Here, we will outline the consequences of apoptotic cell phagocytosis and illustrate how apoptotic cells could be used to manipulate the immune response for therapeutic gain.
....................
Exploitation of Apoptotic Cells by Tumors and Pathogens
In some ways, the most convincing evidence for the anti-inflammatory consequences of apoptotic cell phagocytosis is the exploitation of these immune inhibitory signals by pathogens and tumors to aid their survival. Plasmodium falciparum-infected erythrocytes inhibit the maturation of DCs by binding to CD36, a known recognition receptor for apoptotic cells. Infected DCs still secrete tumor necrosis factor α but fail to activate T cells and secrete interleukin-10.56 This response can be mimicked by antibodies to CD36 or apoptotic cells and suggests that the pathogen and apoptotic cells engage the same pathway regulating DC function. It seems that plasmodium almost inadvertently profits from using the same entry mechanism as apoptotic cells, whereas other pathogens not only exploit recognition mechanism but also profit from the microenvironment created by apoptotic cell phagocytosis. Intense lymphocyte apoptosis occurs in Chagas disease, a debilitating cardiac illness caused by the protozoan Trypanosoma cruzi. In a mouse model of the disease, interaction of apoptotic but not necrotic T lymphocytes with macrophages infected with T. cruzi fuels parasite growth in a manner dependent on prostaglandins, TGF-β, and polyamine biosynthesis.57 Work by Freire-de-Lima et al57 further show that the vitronectin receptor is critical in both apoptotic-cell binding to phagocytes and the induction of prostaglandin E2/TGF-β release and ornithine decarboxylase activity in macrophages. These results suggest that continual lymphocyte apoptosis and phagocytosis of apoptotic cells by macrophages have a role in parasite persistence in the host.
A blunted immune response to rapidly growing tumors is frequently observed and thought to be at least partly mediated by the immune inhibitory effects of apoptotic cell phagocytosis. Reiter et al58 showed that exposure of bone marrow-derived macrophages to apoptotic tumor cells (but not necrotic) tumor cell inhibits their cytotoxicity and nitric oxide production in response to interferon γ and lipopolysaccharide. Furthermore, unstimulated bone marrow-derived macrophages exposed to apoptotic tumor cells enhanced growth of live tumor cells by 40%. Therefore, treatment of cancers with chemotherapy or radiation, which leads to massive tumor cell apoptosis, is likely to inhibit macrophage-mediated antitumor responses.
These examples clearly illustrate the profound effects of apoptotic cell recognition on the outcome of the immune response to pathogens and tumors. It shows that pathogens and tumors use endogenous anti-inflammatory pathways to aid their survival, suggesting possibilities for developing similar avenues to treat inflammatory disease. A recent article by Rossi et al59 establishes that we are already in position to apply this principle to treat experimental lung and joint inflammation. They show that human neutrophils contain functionally active cyclin-dependent kinases (CDKs) and that structurally diverse CDK inhibitors induce caspase-dependent apoptosis and override powerful anti-apoptosis signals from survival factors such as granulocyte-macrophage colony-stimulating factor. Furthermore, the CDK inhibitor R-roscovitine markedly enhances resolution of established neutrophil-dependent inflammation in carrageenan-elicited acute pleurisy, bleomycin-induced lung injury, and passively induced arthritis in mice. In the pleurisy model, the caspase inhibitor zVAD-fmk prevents R-roscovitine-enhanced resolution of inflammation, indicating that this CDK inhibitor augments inflammatory cell apoptosis. Thus, they show that CDK inhibitors enhance the resolution of established inflammation by promoting apoptosis of inflammatory cells.
.....................
Conclusions
Resolution of inflammation is not a passive process but rather an active response to terminate the immune response.60 We show here that the effective recognition and clearance of apoptotic cells is critically important in this process and that this important endogenous mechanism of controlling the immune response is exploited by pathogens and tumors. The challenge for the future is to manipulate effectively and coordinately the clearance of dying cells to develop new therapies for inflammatory and autoimmune disease and prevent inappropriate immune inhibition in the context of pathogens and cancer.
---------------
Evolution has favored pathogenesis that resembles apoptosis.
j
Dranoff/Harvard/cancer, Haynes/Duke/HIV --------
Dranoff is suggesting/patenting more of a "cocktail", but, as with Bart Haynes recent insights into HIV pathogenesis, blocking PS-mediated immunosuppressive signaling is an absolutely essential part of it all.
Both Dranoff from Harvard with regard to cancer,
and Haynes from Duke with regard to HIV,
are saying that exposed PS must be blocked to stimulate the proper immune response to fight the cancer and the virus.
Both Dranoff from Harvard with regard to cancer,
and Haynes from Duke with regard to HIV,
specifically mention using Peregrine's anti-PS to do the job.
j
jake, The Harvard anti-tumor patent suggests using Peregrine's Bavi as the PS-blocker which shifts the immune response to recognize and destroy the tumor, (any tumor).
I thought it fit in nicely with your theme this morning.
j
carbon nanotubes + Bavi, to COOK tumors -----------
WIPO patent application published August 28, 2008 -
Targeting carbon nanotubes to exposed PS on tumor vasculature, and then cooking the tumor(s) by heating up the nanotubes..
Making specific mention of joining the Single-walled Nano-Tubes (SWNT) to a monoclonal anti-PS mab, (citing all Bavi/Thorpe patents) -
"The linking protein may be a phosphatidylserine-specific or other anionic phospholipid-specific monoclonal antibody to which the SWNT is complexed, conjugated or adsorbed or otherwise physically associated with methods known to those of ordinary skill in the art, for example using functionalized SWNTs. Examples of PS-specific monoclonal antibodies include those described in U.S. Patent Nos. 6,406,693; 6,818,213; 6,312,694; 6,783,760; 7,247,303; and PCT application WO2004/006847."
"After treatment with the protein-SWNT complex or peptide-SWNT complex of the present invention, the tumor having the SWNTs bound thereto is then selectively exposed to electromagnetic radiation, for example, near-infrared (NIR) radiation. NIR radiation causes excessive local heating of SWNTs but does not otherwise affect biological systems which are not associated to the SWNTs"
http://www.wipo.int/pctdb/en/fetch.jsp?ELEMENT_SET=B&IDB=0&LANG=ENG&IDOC=1508411&C=10&TOTAL=2978&START=1&QUERY=%28DE%2F11.31%29+&FORM=SEP-0%2FHITNUM%2CB-ENG%2CDP%2CMC%2CAN%2CPA%2CABSUM-ENG&TYPE_FIELD=256&SEARCH_IA=US2008002214&DISP=25&SERVER_TYPE=19-10&IA=US2008002214&RESULT=2&DBSELECT=PCT&SORT=41245759-KEY&DISPLAY=DESC
---------
j
jake, did you read the Dranoff patent I posted?
http://investorshub.advfn.com/boards/read_msg.aspx?message_id=31783692
j
Dr. Alan Bernstein, Aug 4, 2008 ------------
Alan Bernstein: Executive Director, Global HIV Vaccine Enterprise
An HIV Vaccine, Where Do We Go From Here?
(paste into address bar for ppt presentation)
www.aids2008.org/Pag/ppt/MOSY0102.ppt
j
Peregrine Pharmaceuticals to Report First Quarter FY 2009 Financial Results
Wednesday September 3, 7:00 am ET
TUSTIN, Calif., Sept. 3 /PRNewswire-FirstCall/ -- Peregrine Pharmaceuticals, Inc. (Nasdaq: PPHM - News) today announced that it will release its financial results for the first quarter of fiscal year 2009 on September 9, 2008 at 7:00 a.m. EDT and will host a conference call and webcast to discuss the results at 11:30 a.m. EDT on the same day.
------
j
HAYNES: DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to methods of determining the degree of immune system destruction in HIV, of determining the prognosis and the course of the disease in AHI, and of determining the need for treatment in AHI. The present invention further relates to a method of monitoring the intensity of HIV infection and predicting the time to progression of AIDS. The methods are based on plasma tests of immune activation and/or apoptosis plus phenotypic and quantitative assays of plasma microparticles. In preferred embodiments, the methods comprise monitoring plasma levels of TRAIL, Fas Ligand and/or TNFR2. Other plasma markers of HIV-I destruction of the immune system can be used either alone or in combination with these tests (one such marker is nuclear protein HMGB-I that is released in plasma from apoptotic cells (Nowak et al, Cytokine 34:17-23 (2006), epub May 11, 2006)).
Microparticles can come from any immune or non-immune cells. Phenotypic characterization of microparticles has diagnostic use. HIV-I virion microparticles are CD45- and thus CD45 can be used to distinguish virions from immune cell microparticles (Esser et al, Virol. 75:6173-6182 (2001)). In general, elevated T cell microparticles indicate destruction of T cells in vivo.
It is important to note that there is heterogeneity in the degree of increase in TNFR2 and Fas Ligand. This indicates that there are patients with massive immune system apoptosis and those with little immune system apoptosis at the time of AHI. Thus, identification of patients who need treatment with anti-HIV drugs (i.e., those with elevated Fas Ligand, TNFR2, microparticles and TRAIL levels) versus those who do not (no or minimal Fas Ligand, TNFR2, TRAIL and CD45, CD3 T cell microparticle levels) is important for directing treatment in AHI.
The data provided in the Examples that follow demonstrate that, at the time of viral load ramp-up in AHI, when a protective antibody needs to be made, there are high levels of microparticles, high TRAIL, high Fas Ligand, and high TNFR2 levels in plasma in many patients. That there is heterogeneity in the levels of these proteins and microparticles indicates that measurement of these parameters individually or in combination provides a prognostic test suitable for use in the setting of AHI to predict the course of HIV infection and time to AIDS. It will be appreciated that one or more of these parameters can also be considered with other markers of apoptosis and cell death and/ or activation (e.g., soluble HMGB-I, CD25, CD69, or other soluble molecules of T cell activation/apoptosi s) .
While the details provided in the Examples below relate specifically to HIV, the invention includes within its scope methods of monitoring the progression and/or clinical course of other infectious diseases as well that are associated with apoptosis during acute infection (Bahl et al, J. Immunol. 176:4284-4295 (2006)) using these same parameters.
Certain aspects of the invention are described in greater detail in the non- limiting Examples that follows. This application is related to US Prov. Appln. No. 60/859,496, filed November 17, 2006, the entire content of which is incorporated herein by reference.
EXAMPLE 1
Several plasma markers of apoptosis have been studied in Blood Bank panels, including Fas Ligand, TNFR2, and TRAIL. TRAIL has been implicated in the activation induced cell death in HIV infection (Katsikis et al, J. Exp. Med. 186:1365-1372 (1997)). Fas Ligand has been found to be elevated in AHI
(Figs. 4 and 5). Elevations of Fas Ligand were observed in the plasma of 1 1/13 patients while it was not elevated in 2/13 — implying no or less apoptosis in some
patients and not others. Similarly, TNFR2 was elevated in 11/13 patients, and again, the same two patients with low Fas Ligand levels had no increases in plasma TNFR2 (Figs. 6 and 7). Finally, TRAIL levels were elevated in all thirteen patients tested, with the two patients that were low in the other markers of apoptosis, low in TRAIL (Figs. 8, 9).
Thus, the massive apoptosis that occurs with acute HIV infection with resulting release of TRAIL, mediation of apoptosis via FAS-FASL interactions, and release of PS containing viral and other particles, all conspire to initially immuno suppress the host, preventing rapid protective B cell responses. Next, flow cytometry phenotypic analysis of the microparticles in AHI sample 6246-15 was performed (Fig. 10). Fig. 11 shows the forward and side scatter of background with phosphate buffered saline, pH 7, purified microparticles from staurosporine treated Jurkat T cells, and the microparticles in patient 6246 plasma on day 11 at the time of peak viremia. Fig. 11 shows the microparticles in plasma with each panel counted for 2 minutes. Fig. 12 shows the phenotype of either purified Jurkat microparticles (pMP) or patient 6246 MP and shows that the microparticles can be phenotypically analyzed and that the AHI patient microparticles are 78.6 % CD3+, 53% CD45 +. CD45 is a surface molecule of T and monocyte cells that is not incorporated into HIV virions (Esser et al, J. Virol. 75:6173-6182 (2001)). Thus, about one half of the microparticles are likely virions in Fig. 12 and one half of the particles are apoptotic particles from HIV-uninfected cells or do not contain HIV genetic material. Of importance, most of the microparticles from patient 6246 are phosphatidylserine (PS) positive (Fig. 13). Similarly, HIV-infected cells are PS+, as shown in Fig. 14.
EXAMPLE 2
A Surface Plasmon Resonance (SPR) based proteomics assay has been developed for characterization of T cell derived microparticles. The assay allows characterization of micorparticles released from immune cells. Figs. 17 and 18 illustrate the application of the SPR assay for the characterization of T cell microparticles. The developed assay described here can be used to monitor the status of antigen-specific T cell responses in terms of TCR specificity, protein phosphorylation and functional markers of activation and apoptosis.
Characterization of antigen specific T cell microparticles using MHC class I and MHC class II tetramers.
Microparticles are released from immune cells like T or B cells or antigen presenting cells upon activation or apopotosis (Distler et al, Athritis & Rheumatism 52:3337-3348 (2005)). However, these microparticles differ quantitatively and qualitatively and vary depending upon the inducing stimulus (Jimenez et al, Thromb. Res. 109:175-180 (2003); Distler et al, Proc. Natl. Acad. Sci. 102:2892-2897 (2005); Kolowas et al, Scand. J. Immunol., 61 :226-233 (2005)). Since these particles carry membrane derived from the parental cell, characterization of surface antigens can be used to identify the source of the microparticles. Microparticles have been reported to contain surface proteins of the immunoglobulin family (TCR, BCR), glycosylphosphatidylinositol-(GPI-) anchored molecules and members of the tetraspan family (Denzer et al, J. Cell Sci. 113:3336-3374 (2000); Koopman et al, Blood 84:1415-1420 (1994); Heijnen et al, 94:3791-3799 (1999)).
The SPR assay described here is a highly sensitive assay for use in detecting antigen specific T cells using peptide-MHC tetramers. In the study described here, T cell micro vesicles were isolated from murine OTl T cells,
which express Vα2Vβ5 TCR specific for SIINFEKL-Kb (Ova-Kb) complex. These microvesicles are heterogenous in size and as assessed by dynamic light scattering and transmission electron microscopy their hydrodynamic radius vary form 400nm to 70nm (Fig. 17A). Microvesicles have been characterized that are spontaneously released in culture, upon cell lysis and following sucrose density centrifugation to harvest detergent resistant microdomains (DRM). The DRM have been reported to be enriched in TCR/CD3 complex and co-receptors (CD8/CD4) molecules (Montixi et al, The EMBO J. 17:5334-5348 (1998)). Reported here is the development of a SPR based assay in which the microvesicles were first anchored on a lipid linker immobilized on the surface of Ll sensor chip (BIAcore Inc.) (Fig. 17 1C). About 1600 Response Unit (RU) of OTl microvesicles were anchored on the Ll sensor chip and, following a brief period of stabilization of the surface, BSA (0.5 mg/mL) was injected for 5 min to block non-specific binding. Injection of Ova-Kb complex gave specific binding to the anchored OTl microvesicles when compared to the control, VSV-Kb complex (Fig. 18). This demonstrates that the SPR assay can be used to monitor the presence of microparticles released from T cells upon activation or apoptosis. Additionally, the capture of microvesicles on the Ll sensor surface in RU units can be quantitated using standard synthetic microvesicles of known size and total phospholipid content. Thus, it is possible to monitor the release of microvesicles from T cells in samples from HIV infected individual and determine the antigen specificity of the TCR on the T cells undergoing apopotosis. The T cell microvesicles will be selectively captured on anti-CD3 immobilized surface and then the antigen specificity of the captured microparticles will be determined using MHC class I or MHC class II tetramers.
The assay described above is not restricted to T cell microparticles but includes characterization of microparticles from B cells as well. B cell specific
tetramers have been developed by (see U.S. Prov. Appln. 60/840,423) and have been tested for specificity in SPR binding assays and by flow cytometry.
Characterization of protein phosphorylation status of T cell microparticles . Microparticles consist of shed plasma membrane fragments and include cytoplasmic elements (Distler et al, Athritis & Rheumatism 52:3337-3348 (2005)). For T cell microparticles, Src kinases, Lck and Fyn, and the adapter protein LAT are associated with lipid microdomains and are the key components involved in T cell signaling (Zhang et al, Immunity, 9:239-246 (1998); Resh et al, Nature, 387:617-620 (1997); Schade & Levine, Biochem. Biophys. Res.
Commun., 296:637-643 (2002)). Thus the methodologies described will serve to study the correlation of protein phosphorylation status identified in microparticles with immune T cell activation or HIV infection.
The strategies employed here include first the characterization of T cell microparticles in terms of antigen specificity using MHC tetramers as described above, and then to define the phoshorylation status of the identified antigen specific microparticles. The captured microparticles will be eluted from the BIAcore sensor surface using the BIAcore 300 recovery capability. The eluted particles will then be lysed and then the phosphorylation status determined by: a) immunoblotting with anti-signaling molecule antibodies (Src kinases, Lck, Fyn, LAT); and b) 2D-liquid chromatography followed by identification by mass spectrometry. Eluted material will be separated using 2D-LC and individual phosphorylated proteins will be analyzed using a MALDI-TOF/TOF. Proteins identified using this approach will be verified to be phosphorylated using conventional immunoblotting with anti-phospho antibodies. This application allows characterization of released microparticles in order to identify the activation state and signaling pathways in immune cells and their changes upon exposure to HIV infection.
Characterization of microparticles to determine the functional status of the parental cell.
As with HIV-I viruses that incorporate cell specific membrane proteins into its viral envelope in the process of budding from infected cells (Hioe et al, J. Virol. 75: 1077-1088 (2001); Laio et al, AIDS Res. Hum. Retro. 16:355-366 (2000)), budding microparticles carry on their cell surface protein markers derived from the parental cell. These surface markers are tell-tale sign of the functional status of the T cells from which they came from. In addition to flow cytometric based pheno typing of microparticles, the capture of microparticles from T cells using the SPR assay described above is a novel methodology for determining the activation status of the T cells. The functional status of the T cells is determined by monitoring the expression of activation markers (CD69, CD25), apopotosis markers (PDl, TRAIL receptors, FAS, Fas L).
EXAMPLE 3
A critical event in HIV-I and SIV infection is virus-induced massive CD4+, CCR5+ T cell loss that is severe in gut associated lymphoid tissues (GALT) (Guadalupe et al, J. Virol. 77:11708-11717 (2003), Brenchley et al, J. Exp. Med. 200:749-759 (2004), Mehandru et al, J. Exp. Med. 200:761-770 (2004)). Depletion of GALT CD4 T cells has been documented at peak viral load in acute SIVmac239 infection (Veazey et al, Science 280:427-431 (1998), Haase, Nat. Rev. Immunol. 5:783-792 (2005), Li et al, Nature 434:1148-1152 (2005), Mattapallil et al, Nature 434:1093-1097 (2005)), as well as within weeks of HIV- 1 transmission in man (Guadalupe et al, J. Virol. 77:11708-11717 (2003), Brenchley et al, J. Exp. Med. 200:749-759 (2004), Mehandru et al, J. Exp. Med. 200:761-770 (2004)). During acute SIV infection, 30-40% of memory CD4+ T cells are infected (Veazey et al, Science 280:427-431 (1998), Haase, Nat. Rev.
Immunol. 5:783-792 (2005), Li et al, Nature 434:1148-1152 (2005), Mattapallil et al, Nature 434:1093-1097 (2005)). The mechanisms of immune cell death during acute HIV-I infection are not known, but may involve induction of apoptotic pathways by HIV Tat, Vpr or gpl20 proteins (Badley et al, Blood 96:2951-1964 (2000), Chase et al, Trends Pharmacol. Sci. 27:4-7 (2006), Boya et al, Biochim. Biophys. Acta 1659:178-189 (2004)), HIV-I infection of CD4+ T cells (Guadalupe et al, J. Virol. 77:1 1708-11717 (2003), Mehandru et al, J. Exp. Med. 200:761-770 (2004), Veazey et al, Science 280:427-431 (1998), Li et al, Nature 434: 1148-1152 (2005)), and induction of uninfected cell death due to killing by molecules such as tumor necrosis factor related apoptotis inducing ligand (TRAIL) (Lum et al, J. Virol. 75:11128-11136 (2001), Herbeuval et al, Clin. Immunol. 123:121-128 (2007)).
The time from HIV-I transmission to establishment of the latently infected pool of CD4 T cells has been termed the window of opportunity within which a preventive HIV-I vaccine has to extinguish HIV-I (Johnston et al, N. Engl. J.
Med. 356:2073-2081 (2007, Wong et al, Biology of Early Infection and Impact on Vaccine Design, pgs. 17-22 (Caister Academic Press, Norfolk, UK (2007)). The latent pool is established at least by the time of symptomatic acute HIV-I infection at the time of seroconversion (-25 days after transmission), although the exact earliest time of establishment of the latent CD4 T cell pool is not known (Wong et al, Biology of Early Infection and Impact on Vaccine Design, pgs. 17- 22 (Caister Academic Press, Norfolk, UK (2007), Chun et al, Proc. Natl. Acad. Sci USA 95:8869-8873 (1998)). Adaptive CD4, CD8 and B cell antibody responses to HIV-I do not appear during the eclipse or viral load ramp-up phases of HIV-I infection, but rather appear coincident with the fall in viral load (VL) and appearance of acute infection symptoms at the end of the window of opportunity (Reynolds et al, J. Virol. 79:9228-9235 (2005), Abel et al, J. Virol. 79:12164-12172 (2005), Fiebig et al, AIDS 17:1871-1879 (2003)). Thus, study
of the events that transpire from transmission until the onset of plasma viremia (the eclipse phase) and during the viral load ramp-up phase of acute HIV-I infection are critical to understanding why immune responses do not occur earlier after HIV-I transmission, and to define what a successful vaccine must overcome to extinguish HIV-I.
In the study described below, the hypothesis is raised that, in addition to gut CD4 T cell loss, delay in HIV-I protective immune responses early on after HIV-I transmission may involve the production of elevated levels of immunosuppressive moieties such as TRAIL, TNFR2 and Fas ligand as well as plasma mi croparticles. If elevations in immunosuppressive molecules, coupled with early CD4+ T cell death, occur early on after HIV-I transmission, then this would define a protected time for HIV-I to replicate while anti-HIV-1 T or B cell responses were suppressed.
To study the eclipse and early viral load ramp-up phases of acute HIV-I infection, archived plasma of plasma donors with samples available before, during, and after HIV-I viral load ramp-up have been used (Fiebig et al (AIDS 17:1871-1879 (2003)). An initial burst of soluble TRAIL was found in plasma soon after the appearance of HIV-I in plasma, corresponding to ~17 days following transmission. Also observed were later elevated plasma TNFR2, Fas ligand and plasma microparticles (MP) levels around the peak of plasma VL. These data implicate TRAIL as an early mediator of cell death in acute HIV-I infection, and demonstrate a narrow window of opportunity in which a HIV-I vaccine must extinguish the transmitted virus.
Experimental Details Plasma Samples. Seroconversion panels (HIV- 1+/HCV-/HBV-, n=30,
HIV-1-/HCV-/HBV+, n=10, and HIV-I-, HCV+/HCV-, n=10) were obtained from ZeptoMetrix Corporation (Buffalo, NY). Each panel consisted of sequential
aliquots of plasma (range 4-30) collected approximately every 3 days during the time of acute infection with HIV-I (Huang et al, J. Immunol. 177:2304-1313 (2006)). HIV-I -/HC V-/HBV-human plasmas (n=25) were obtained from Innovative Research (Southfield, MI). All studies were approved by the Duke University human subjects institutional review board.
Viral Load Testing. Viral load testing of HIV-I plasma plasma donor panels was performed by Quest Diagnostics (Lyndhurst, NJ) (HIV-I RNA PCR Ultra). HCV and HBV viral loads were preformed by Zeptometrix; select HCV viral loads were provided by Philip Norris, Blood Systems Research Institute, San Francisco, CA.
ELISAs For Plasma Markers ofApoptosis. ELISAs for Fas, Fas Ligand, TRAIL, (Diaclone, Besancon Cedex, France), and TNFR2 (Hycult Biotechnology, Uden, The Netherlands) were performed according to the manufacturer's directions. Plasma was assayed undiluted (TRAIL), diluted 1:10 (TNFR2) or diluted 1 :2 (Fas Ligand). Increases in plasma analytes over time were defined as >20% increase of values after TO versus before TO.
Apoptotic Microparticle (MP) Quantification. The number of MP in each plasma sample was determined with flow cytometry. All flow cytometry analyses were performed on the LSRII Flow Cytometer (BD Biosciences, San Jose, CA), and data analyses were performed using FlowJo software (Ashland, OR). All buffers (PBS without calcium and magnesium (Cellgro, Herndon, VA) and formaldehyde (Sigma, St. Louis, MO)) were filtered with a 0.22 μm filter
(Millipore, Billerica, MA), before use in any MP experiment. The buffer used to dilute plasma samples (1% Formaldehyde in PBS without calcium and magnesium), was used to define the background MP count (-150 events counted
in 60 seconds on the flow cytometer). To define the MP gate, FluoSpheres Fluorescent Microspheres (Molecular Probes, Eugene, OR), ranging in size from 0.1 μm to 1 μm, were analyzed on the flow cytometer. The MP gate was drawn around the beads, encompassing the 0.1 μm, 0.2 μm, 0.5 μm, and 1.0 μm beads. Each plasma sample was diluted 1 : 100 and 1 : 1000 in 1 % formaldehyde/PBS, and data acquired for 60 seconds. Optimal sample dilutions were determined experimentally, with the acceptance criteria being the dilution of plasma with abort counts < 5%, and noise to signal ratios < 0.1 (noise to signal ratio ^background MP count in PBS/experimental plasma MP count).
Microparticle Phenotypic Analysis. MP were analyzed by flow cytometry for cell surface markers as described (Hosaka et al, J. Infect. Dis. 178:1030-1039 (1998), Stacey et al and the NIAID Centre for HIV/AIDS Vaccine Immunology. Elevations in plasma levels of innate cytokines prior to the peak in plasma viremia in acute HIV-I infection (2007), Clark et al, N. Engl. J. Med. 324:954- 960 (1991)). Plasma samples (l-2ml) were diluted in 5 ml of filtered saline, and then filtered through a 5 μm filter (Pall Corporation, East Hills, NY). The diluted samples were then centrifuged (lhr at 200,000xg at 40C) (Sorvall RC Ml 50 GX, Thermo Fisher Scientific, Waltham, MA). The top 2.5 ml of supernatant was removed, 2.5 ml of fresh saline added, and samples were centrifuged again (lhr at 200,000xg at 4°C). The pellet was washed X2 in ImI of filtered saline; after the last wash, 900 μl of the supernatant was removed, and the pellet resuspended in the remaining 200 μl of saline. Ten μl of MP suspension was incubated with an antibody and/or annexin V (total volume of 100 μl x 20 minutes, 20° C, in the dark). Saline with 1% BSA (Sigma) was used as staining buffer for incubation with antibodies, and 2.5 mM CaCl2 added to the buffer for annexin V staining. For annexin V controls, 50 mM EDTA was added to the buffer. After incubation,
the volume was adjusted to 500 μl with saline/formaldehyde, and analyzed by flow cytometry within 24 hours. Conjugated antibodies included mouse anti- human CD45-PE, CD3-PE, CD61-PerCp, CCR5-PE, and isotype controls (BD Biosciences, San Jose, CA) and annexin V conjugated to AlexaFluor 647 (Molecular Probes, Eugene, OR).
Electron Microscopy of Plasma Microparticles. Eight ml of plasma was diluted 1 :5 in filtered saline and MP were pelleted (200,000xg x 1 hr, 4°C). Pellets were washed x2 (100,000xg x 30 min.) in ImI of saline. The MP pellet was resuspended in 500 μl of saline and overlaid onto ImI of a 40% sucrose solution (in saline) and MP were centrifuged (100,000xg x 90 min.). The pellets were fixed (1% formaldehyde, 4°C overnight), pelleted (100,00xg x 60 min.), then fixed in 1 % osmium tetroxide. Ultrathin sections were cut and post stained with uranyl acetate and examined on a Philips CMl 2 electron microscope (FEI Co., Hillsboro, OR).
In Vitro Mucosal B cell Culture Model. Tonsils were obtained from pediatric and adult patients who underwent tonsillectomy at the Duke University Medical Center. Tonsils were placed in transport media (RPMI 1640 with L- Glutamine (Gibco, Carlsbad, CA), supplemented with 200U/ml penicillin G, 200 μg/ml streptomycin, 50 μg/ml gentamicin, and 1 μg/ml amphotericin B (Sigma) upon excision and transported to the laboratory for processing within 4 hours. The tonsils were washed extensively in RPMI 1640 with L-Glutamine supplemented with 100 U/ml penicillin G, 100 μg/ml streptomycin, 50 μg/ml gentamicin, and 2 μg/ml amphotericin B to prevent bacterial and fungal contamination. To isolate tonsillar lymphocytes, the specimens were mechanically minced and teased with sterile forceps, and the resulting single-cell
suspension was processed through a 70 μm nylon cell strainer (BD Biosciences). Lymphocytes were separated with lymphocyte separation media (Fisher Scientific, Pittsburg, PA) and washed twice. Cell number and viability were determined with a Guava EasyCyte Mini (Hayward, CA) per the manufacturer's instructions. Cells were cultured in RPMI 1640 supplemented withlOO U/ml penicillin G, 100 μg/ml streptomycin, 25 μg/ml gentamicin, 1 μg/ml amphotericin B, and 10% FBS (Gemini Bioproducts, West Sacramento, CA), at a density of 1x106 cells/ml in total volumes of ImI in polystyrene 5ml round-bottom tubes (BD Biosciences). To stimulate antibody production, cells were cultured with an optimized cocktail of oCpG (6 μg/ml) (Coley Pharmaceutical Group, Wellesley, MA), and pokeweed mitogen extract (PWM) 1/2000 (Crotty et al, J. Immunol. Methods 286:111-122 (2004)). After 5 days of culture, supernatants were harvested and total IgG and IgA present in culture supernatants was quantified by ELISA. Briefly, 96-well ELISA plates (Corning, Corning, NY) were coated with purified murine anti-human IgG Fc (HRL, Baltimore, MD) or purified murine anti-human IgA (BD Pharmingen) in 0.1 M NaHCO3 overnight. Wells were washed x3 with wash buffer (PBS with 0.1% Tween (Sigma)). Wells were blocked with dilution buffer (1% FBS, 0.5% BSA (Sigma) and 0.05% Tween) for 2 hours at 200C. Wells were washed x3, and supernatants were added diluted 1 :2 with dilution buffer. Plates were incubated overnight at 4°C, and washed x3.
Biotinylated murine anti-human IgG Fc (HRL) or biotinylated murine anti-human IgA (BD Pharmingen) was added, and plates were incubated for 2 hours at 200C. After washing x3, horseradish peroxidase streptavidin (Vector Laboratories, Burlingame, CA) was added and plates incubated for 45 minutes at 200C. Plates were washed x3, and the assay was developed using a 3,3',5,5' tetra- methylbenzidine (TMB), substrate and stop solution system (KPL, Gaithersburg,
MD). Standard curves were constructed using serial dilutions of purified human IgG and IgA (Sigma).
Microparticles were generated by treatment of normal donor PBMC or tonsil cells with staurosporine Bell et al, Am. J. Physiol. Cell Physiol. 291 :C1318- Cl 325 (2006)). Cell viability and cell counts were performed on the Guava EasyCyte Mini, and 5x107 cells were cultured in 5 ml RPMI 1640 with L- glutamine supplemented with 25 μg/ml of gentamicin, 10% FBS, and l μM staurosporine (Sigma). After culture overnight, the cells and supernatant were collected, and centrifuged X2 (5min at 400xg at 40C). The cell-free supernatant was then centrifuged in an ultracentrifuge to harvest the MP (30 min at 200,000xg at 4°C) and MP were washed Xl with RPMI 1640, resuspended in 1000 μl of fresh RPMI, and 100 μl of MP suspension was added to select tonsil cell cultures (or varying volumes to determine dose dependency).
Statistical Analyses. Statistical Analysis was performed as outlined in the figures and descriptions thereof using boxplot analyses, Student's t tests, and Wilcoxon Rank Sum analyses. To establish a reference point throughout all the plasma seroconversion panels, Time 0 (TO) was defined as the date when viral load reached 100 copies/ml for HIV-I, 600 copies/ml for HCV, and 700 copies/ml for HBV. To determine the percent increase in plasma markers of apoptosis during HIV-I, HBV, and HCV infections, the mean TRAIL, TNFR2, or Fas Ligand level before Day 0 was compared to the mean level after Day 0, and percent increase was calculated ([(mean after day 0 - mean before day 0)/mean after day 0] x 100).
Results
TRAIL, TNFR2 and Fas ligand plasma levels are elevated during acute HIV-I infection.
A timepoint (TO) was determined for each of 30 HIV-I, 10 HCV and 10 HBV patients, defined as the lower limits of detection for each viral load determination (Fig. 19). Soluble TRAIL, TNFR2, and Fas Ligand were next assayed in sequential plasma samples of each plasma donor (Fig. 20A). The percent change in plasma plasma TRAIL, TNFR2 and Fas ligand levels were determined by comparing the mean analyte level before T0 to the mean level after T0; 27/30 demonstrated increases in TRAIL, 26/30 had increased TNFR2, and 22/30 had increased Fas ligand levels by these criteria (Fig. 20B).
Hepatitis B and C infections have been reported to induce cell death of hepatocytes (Chase et al, Trends Pharmacol. Sci. 27:4-7 (2006), Chou et al, J. Immunol. 174:2160-2166 (2005)). As controls, HCV and HBV acutely infected subjects demonstrated a > 20% rise in TRAIL, TNFR2 or Fas ligand only in 0/10, 3/10, and 2/10 HBV acutely infected subjects, and in 1/10, 6/10 and 7/10 HCV acutely infected subjects, respectively (Figs. 2OC, 21B).
Second, cell death plasma analyte levels at the time of peak viral load were compared to samples drawn from subjects before viral load ramp-up, and as well, compared with uninfected plasmas. The mean TRAIL, TNFR2, and Fas ligand levels at the time of peak viral load were significantly different from the earliest plasma sample drawn from each acute HIV-I infected patient before To (p=0.0075 for TRAIL, p=1.18xlθ-5 for TNRF2 and p=3.88xlθ"6 for Fas ligand) (Fig. 21A). The peak TRAIL, TNFR2 and Fas ligand levels were also significantly different from the levels of TRAIL, TNFR2, and Fas ligand in uninfected plasma sample controls (p=2.16xlθ"8, p=6.16xlθ"9, and p=1.64xlθ"6 , respectively) (Fig. 21A).
Third, to investigate the timing of peak levels of TRAIL, TNFR2 and Fas ligand compared to peak plasma VL, a determination was made of the temporal relationship between an apoptotic analyte peak compared to the peak plasma VL. Also determined was the number of subjects that had peaks in plasma cell death analytes occurring before, coincident with or following the peak in HIV-I viral load (Fig. 21B). The majority of acute HIV-I infection subjects (30/30 for TRAIL, 27/30 for TNFR2, and 26/30 for Fas ligand) demonstrated peak analyte levels occurring within a 30-day time frame (i.e., 15 days before, at the time of, or within 15 days after the viral load peak). Of particular interest, the majority of subjects' TRAIL levels (21 /30) peaked before the peak viral load, while TNFR2 and Fas ligand levels more often peaked coincident with viral load (Fig. 21B).
Fourth, to analyze the timing of peak analyte levels relative to the rate of viral expansion during viral load ramp-up, paired Wilcoxon rank tests were performed (Fig. 21 C). The day of the peak viral expansion rate indicates the day following To on which the virus was replicating at the maximum rate. In 24 plasma donors in which the rate of VL expansion could be calculated, the peak viral expansion rate occurred on mean day 5.5 following T0 (Fig. 21C). Plasma TRAIL levels peaked 1.7 days after maximal viral expansion rate (day 7.2 after To) while TNFR2 levels peaked 7.5 days (day 13 after To), and Fas ligand levels peaked 9.8 days (day 15.3 after T0) after the time of maximal rate of viral expansion. Plasma donor HIV-I VL reached its peak an average of 13.9 days after TO (median 13 days, interquartile range 3 days), indicating that TRAIL levels peaked well before VL peaked, while TNFR2 and Fas ligand reached peak levels close to the time of highest VL levels. The mean of the peak plasma TRAIL levels was 201 lpg/ml with a range of 886-4138pg/ml. This level of TRAIL is well within the biologically relevant concentration range of an activity for induction apoptosis in immune cells21.
Quantitative flow cytometry analysis of plasma microparticles. Plasma MP are a normal by-product of a variety of types of activated or apoptotic cells, or are derived from multivesicular bodies (exosomes) (Distler et al, Autoimmunity 39:683-690 (2006), Piccin et al, Blood Rev. 21 :157-171 (2007) (Fig. 25). Eighteen of thirty (60%) plasma donors demonstrated peak MP levels near (within 15 days before or 15 days after T0) the peak in viral load, and 1 1 of these 18 peaks occurred immediately before the peak in viral load, while 4/18 peaked at the time of VL peak (Table 1 and Fig. 22A). Five often (50%) HCV donors had similar elevations in MP, but only two often (20%) HBV panels studied had elevations in MP levels near the peak VL (Fig. 22B).
Table 1
The morphology of MP at the time of peak viral load was studied from acute HIV-I infection patient 6244 on sucrose gradient purified MP from the time of both peak VL and peak MPs (day 10 in Fig. 22A) and found to be heterogeneous in size ranging from IOnm to lOOOnm (Fig. 23A).
Phenotypic characterization of plasma microparticles. Most studies of phenotypic analysis of MP used fresh plasma that was processed within hours (Jy
et al, J. Thromb. Haemost. 2:1842-1851 (2004)), whereas the plasma donor samples in this study have been frozen/thawed at least twice. It was found that two freeze/thaw cycles markedly decreased the percentage of CD3, CD45+, CD61+ and annexin+ MPs (Fig. 26). Thus, unable to accurately quantitate the 5 phenotype of plasma MP, MP was analyzed for annexin and CCR5 expression in a qualitative manner to determine if MP expressing annexin or CCR5 were present at the time of peak MP in five plasma donor peak MP samples vs. uninfected first samples. Both annexin+ and CCR5+ MP were present in the plasma donor plasmas; for annexin, the mean %+ at MP peak was 12%, range l o 2.3% - 38.0%, and for CCR5+ MP, %+ mean at the MP peak was 5.7,%, range 1.1% - 12.6% (Figs. 23B, 23C). For CCR5+ but not for annexin+ MP, there was a trend in higher MP numbers at the time of peak MP compared to the first panel sample (Fig. 23C).
While the average peak HIV-I VL level was 1,421,628 copies/ml, the
15 average peak of total MPs was 606,881,733/ml. Thus, there was an average of 427-fold more MP than virions present in plasma at their peaks during acute HIV- 1 infection.
Table 2
*Corrected MP levels - Quantitation of MP levels in fresh plasma vs. 2x frozen /thawed plasma showed a ~20% increase in MP levels in 2x frozen/thawed plasma. Thus, the mean MP level was corrected by -20%.
MP-Induced B Cell Suppression In Vitro. While plasma MPs have potent known suppressive effects on macrophages and DCs (Hoffmann et al, J. Immunol. 174:1393-1404 (2005), Huynh et al, J. Clin. Invest. 109:41-50 (2002)), only one study has suggested MP may inhibit B cell activation (Koppler et al, Eur. J. Immunol. 36:648-660 (2006)). There was particular interest in MP effects on human memory B cell activation, since what is desired is a rapid virus-induced memory B cell response after transmission. To determine if PBMC-derived or tonsil leukocyte-derived MP could be suppressive for memory B cell activation, a memory B cell Ig induction assay was used using pokeweed mitogen (PWM) + class B oCpG (Crotty et al, J. Immunol. Methods 286:11 1-122 (2004)). The addition of MP in PWM-stimulated tonsil cell cultures reduced total IgG and IgA production by 70.8 % +/- SEM for IgG (p=0.0064) and 94.2% +/- SEM for IgA (p=0.00004) (Fig. 24A); B cell suppression by MPs was dose-dependent (Fig. 24B). Similar results were observed when MP were generated from autologous tonsil leukocytes or from the Jurkat T cell line (data not shown).
In summary, a major finding in this study is the early appearance of a peak of TRAIL at 17 days of transmission in plasma donors, and implies the TRAIL/DR5 in a key pathway in HIV-I induced cell death immediately following transmission. An IFN-α, TRAIL, DR5 pathway of CD4+ T cell apoptosis has been proposed for chronic HIV-I infection based on in vitro studies and on studies in HIV-1+ progressor tonsillar tissues (Lum et al, J. Virol. 75:11128- 11136 (2001), Herbeuval et al, Clin. Immunol. 123:121-128 (2007), Herbeuvel et al, Blood 106:3524-3531 (2005)). CD4+ T cells in infected subjects are more sensitive to TRAIL-mediated apoptosis than are CD4+ T cells from uninfected subjects due to upregulated TRAIL receptor DR5 (Lum et al, J. Virol. 75:11128- 11136 (2001), Herbeuval et al, Clin. Immunol. 123:121-128 (2007), Herbeuvel et al, Blood 106:3524-3531 (2005), Jeremias et al, Eur. J. Immunol. 28:143-152
(1998)). In vitro, HIV-I gpl20 (Herveuval et al, Blood 105:2458-2464 (2005)) induces monocyte and plasmacytoid dendritic cell IFN-α, which in turn induces CD4+ T cell and monocyte/macrophage TRAIL (Lum et al, J. Virol. 75:11128- 11136 (2001), Herbeuval et al, Clin. Immunol. 123:121-128 (2007), Herbeuvel et al, Blood 106:3524-3531 (2005)). HIV-I Tat has also been reported to induce TRAIL as a mechanism of bystander killing of CD4+ T cells (Yang et al, J. Virol. 77:6700-6708 (2003)).
An important question is why do plasma TRAIL levels peak earlier after HIV-I transmission than do plasma Fas ligand, TNFR2 and MP? Plasma elevations of TRAIL, Fas ligand and TNFR2 occur in chronic HIV-I, and can be induced by immune cell activation, cell death, or both (Herveuval et al, Blood 105:2458-2464 (2005), Aukrust et al, J. Infect. Dis. 169:420-424 (1994), Hober et al, Infection 24:213-217 (1996), Hosaka et al, J. Infect. Dis. 178:1030-1039 (1998)). Stacy et al (Stacey et al and the NIAID Centre for HIV/ AIDS Vaccine Immunology. Elevations in plasma levels of innate cytokines prior to the peak in plasma viremia in acute HIV-I infection (2007)) have found a burst of IFN-α in the same plasma donors that coincides with the timing of the TRAIL peak seen in this study . Thus, the plasma TRAIL peak that precedes the VL plasma peak may be due either to early apoptosis, but may also result from immune activation and pDC production of IFN-αin response to rising VL. It is hypothesized that the later appearance of elevated plasma Fas ligand, TNFR2 and microparticles maybe the result of, or in response to, massive cell death, as this peak comes at an analogous time to the cell death peak documented in experimental SIV infection in rhesus macaques (Veazey et al, Science 280:427-431 (1998), Haase, Nat. Rev. Immunol. 5:783-792 (2005), Li et al, Nature 434:1148-1152 (2005), Mattapallil et al, Nature 434:1093-1097 (2005)).
Veazey (Mattapallil et al, Nature 434:1093-1097 (2005)) noted the onset of CD4+ gut T cell loss as early as 7 days after SIV infection . In humans,
Guadalupe et al (J. Virol. 77:11708-11717 (2003), Mehandru et al, J. Exp. Med. 200:761-770 (2004) and Mehandru and colleagues (Brenchley et al, J. Exp. Med. 200:749-759 (2004)) have studied 2, 1 and 9 patients, respectively, during the first month of HIV-I infection and found depletion of gut CD4+ T cells. The eclipse phase of HIV-I infection is the time from transmission until the appearance of plasma viremia, and is estimated to be 10 days with a range of 7-21 days (Clark et al, N. Engl. J. Med. 324:954-960 (1991), Gaines et al, BMJ 297:1363-1368 (1988), Littl et al, J. Exp. Med. 190:841-850 (1999), Schacker et al, Ann. Intern. Med. 125:257-264 (1996)). The time from appearance of HIV-I viremia until the first antibody response and symptomatic HIV-I infection (and therefore establishment of the latent pool) is approximately 14 days (Cooper et al, J. Infect. Dis. 155:1113-1118 (1987), Daar et al, N. Engl. J. med. 324:961-964 (1991), Gaines et al, Lancet 1 :1249-1253 (1987)). Thus, the maximal window of opportunity for preventive HIV-I vaccine efficacy without cell death-induced immune suppression is approximately 24 days. With mediators of apoptosis and immune suppression present as early as day 17 following transmission (10 days average eclipse phase + onset of TRAIL 7 days after T0), the window of opportunity is narrowed to ~14-17 days.
The presence of TRAIL, TNFR2 and elevated MP during this early period of acute HIV-I infection suggests at least four potential mechanisms of immunosuppression. First, direct HIV-I infection results in loss of a substantial proportion of CD4+ T cells, although the numbers of infected cells does not account for all CD4+ T cell depletion ((Guadalupe et al, J. Virol. 77:11708-11717 (2003), Brenchley et al, J. Exp. Med. 200:749-759 (2004), Mehandru et al, J. Exp. Med. 200:761-770 (2004), Fiebig et al, AIDS 17:1871-1879 (2003)). Second, in uninfected CD4+ T cells, TRAIL induces bystander killing ((Lum et al, J. Virol. 75:11128-1 1136 (2001), Herbeuval et al, Clin. Immunol. 123:121-128 (2007), Herveuval et al, Blood 105:2458-2464 (2005)). In this regard, Miura et al (J. Exp.
Med. 193:651-660 (2001)) have shown that administration of an anti-TRAIL mAb in HIV-I infected hu-PBL-NOD-SCID mice markedly reduces CD4+ T cell apoptosis.
Third, suppression of immune responses can be mediated by T cell MP (Huang et al, J. Immunol. 177:2304-1313 (2006), Distler et al, Arth. Rheum.
52:33337-3348 (2005), Krysko et al, Apoptosis 11:1709-1726 (2006)). CXCR4+ and CCR5+ MP can transfer co-receptors to co-receptor negative cells making them susceptible to HIV-I (Mack et al, Nat. Med. 6:769-775 (2000), Rozmyslowicz et al, AIDS 17:33-42 (2003)). Phagocytosis of MP by macrophages releases TGF- βprostaglandin E2 and IL-10 that can inhibit antigen-specific T and B cell responses (Huang et al, J. Immunol. 177:2304-1313 (2006), Hoffmann et al, J. Immunol. 174:1393-1404 (2005), Huynh et al, J. Clin. Invest. 109:41-50 (2002)). In this regard, Estes et al (J. Infect. Dis. 193:703-712 (2006)). have demonstrated dramatic increases in lymph node TGF-β and IL-10 on day 12 following SIV infection. Importantly, it has been directly shown that PBMC and tonsillar cell MP can directly inhibit memory B cell activation (Fig. 24).
Finally, both Fas ligand and TRAIL are incorporated into MP (Huynh et al, J. Clin. Invest. 109:41-50 (2002), Koppler et al, Eur. J. Immunol. 36:648-660 (2006), Crotty et al, J. Immunol. Methods 286: 111-122 (2004)). Fas ligand expressing MP can directly induce apoptosis in nearby cells (Huang et al, J. Immunol. 177:2304-1313 (2006), Jodo et al, J. Biol. Chem. 276:39938-39944 (2001), Monleon et al, J. Immunol. 167:6736-6744 (2001)) activated T cells can be the target of Fas ligand mediated proapoptotic microvesicles (Monleon et al, J. Immunol. 167:6736-6744 (2001)). Salvato et al (Clinical and Developmental Immunology (2008)) have recently suggested that treatment of SIV-infected macaques with a mAb against Fas ligand attenuates disease and may lead to elevated antibody responses to SIV.
Thus, the production of high levels of biologically active plasma mediators and byproducts of cell death during the first two to three weeks of HIV- 1 transmission raises the notion that the window of opportunity for a preventive vaccine to work may be shorter than previously thought, ie within the first 14-17 days of transmission, placing considerable constraints on the time available for development of robust anti-HIV-1 immunity following transmission. Preventive vaccine candidates may need to target HIV-I molecules that induce cell death and be designed to induce protective immune responses to HIV-I that will either be at maximum inhibitory levels at the time of transmission, or be boosted within hours to days as a secondary immune response to extinguish HIV-I before HIV-I- induced immunosuppression occurs.
Inhibition of cell death and immunosuppressive MP mediated effects by a vaccine for HIV or other infectious agents may be important as well. This could be accomplished, for example, by an HIV vaccine component inducing anti-lipid antibodies or antibodies against other components of microparticles to facilitate clearance of microparticles and/or to block microparticle toxic effects.
Another use of the data herein is as a rationale for the treatment of HIV-I. For example, antibodies against TNFR or TNF-α, antiphosphatidylserine antibodies or other inhibitors of cell death (Fas-Fc as an inhibitor of FAS-FAS ligand interactions and DR5-Fc as an inhibitor of TRAIL DR5 interactions) can be used to inhibit cell death in HIV as a therapy.
All documents and other information sources cited above are hereby incorporated in their entirety by reference.
-----------
j
jess,
I looked back, sorry - it wasn't you who downplayed Bavi's role in the work. It was "moby", saying it "only referred to Bavi in the context as a stain control."
As for Bavi, yes, it was indeed bavi/2AG4 that was used by Haynes in the microparticle work that was published in Virology, which showed how the exposed PS on microparticles is responsible for blunting the function of the immune cells that would ideally fight the virus, and that Bavi (2AG4) itself masks/binds the exposed PS.
Interestingly.....Bavi was not mentioned in the paper, but reading the details of the same work in the patent application shows that it was indeed Bavi. Here are those details -
The following graphs (re: microparticle counts vs: viral load) were in the paper, and the paper discussed the microparticles being responsible..., and counting how the microparticles ramped up right before (allowing) the virus to ramp up..., since the microparticles expose PS...
The patent application contains the 'same' graph (re: microparticle counts vs: viral load) - with the following caption:
Figure 15. Microparticles expressing phosphatidylserine in AHI Plasma. Various time points of panel 6246 (21 days before viral load was detected, day - 21, and 7, 11 and 14 days after viral load was measured to be > 100 copies/ml) were assayed by flow cytometry for the presence of microparticles stained with anti-phosphatidylserine antibodies (2aG4). As a control, plasma from a normal, uninfected blood bank donor was also assayed in the same manner.
..........
more info -
Thus, the massive apoptosis that occurs with acute HIV infection with resulting release of TRAIL, mediation of apoptosis via FAS-FASL interactions, and release of PS containing viral and other particles, all conspire to initially immuno suppress the host, preventing rapid protective B cell responses. Next, flow cytometry phenotypic analysis of the microparticles in AHI sample 6246-15 was performed (Fig. 10). Fig. 11 shows the forward and side scatter of background with phosphate buffered saline, pH 7, purified microparticles from staurosporine treated Jurkat T cells, and the microparticles in patient 6246 plasma on day 11 at the time of peak viremia. Fig. 11 shows the microparticles in plasma with each panel counted for 2 minutes. Fig. 12 shows the phenotype of either purified Jurkat microparticles (pMP) or patient 6246 MP and shows that the microparticles can be phenotypically analyzed and that the AHI patient microparticles are 78.6 % CD3+, 53% CD45 +. CD45 is a surface molecule of T and monocyte cells that is not incorporated into HIV virions (Esser et al, J. Virol. 75:6173-6182 (2001)). Thus, about one half of the microparticles are likely virions in Fig. 12 and one half of the particles are apoptotic particles from HIV-uninfected cells or do not contain HIV genetic material. Of importance, most of the microparticles from patient 6246 are phosphatidylserine (PS) positive (Fig. 13). Similarly, HIV-infected cells are PS+, as shown in Fig. 14.
-----------
Now that I've gone back to clarify this, it's become obvious to me why Haynes didn't mention Bavi, or even that the counting of the microparticles discussed in the paper were counted via anti-PS / (Bavi), while it IS mentioned, repeatedly, in the patent applications.
IMHO, because it's an important topic for a separate paper. I base that opinion on all the recently published patent applications containing the BAVI / HUMANIZED BAVI / TARVI / IS1 discussions/info...
j
jess,
re:"Haynes never mentions bavi"
Actually, Haynes mentions Bavi (as Bavi, Humanized Bavi, Tarvi, 3G4, 2AG4) several times in his recent patent applications. Bavi was also used in the work described in Haynes paper in the August issue of Virology.
Haynes used Bavi (2AG4) to bind and count the microparticles in patient # 6246. You previously tried to "dis" this work referring to Bavi as performing a minor role as a reagent, but the truth is Bavi played a role in proving how the exposed PS on microparticles is responsible for blunting the function of the immune cells that would ideally fight the virus, and that Bavi itself masks/binds the exposed PS.
In the WIPO patent applications Haynes discusses in great detail how exposed PS needs to be dealt with to give the immune system the chance to fight off the virus.
I conclude from these several patent applications, as well as the important paper in the August Virology (important because Haynes, besides corroborating the immunosuppressive function of exposed PS on macrophages, dendritic cells and resultant T cells, he also for the first time illustrates how exposed PS stifles antibody-producing B cells) that Haynes and the other scientists involved will be publishing the work which is discussed in the patent applications.
It seems obvious from his patent applications that anti-PS is a very promising therapeutic for those already infected with HIV,as well as anti-PS being an important goal of a vaccine immunogen.
Basically, Haynes, (CHAVI, and CAVD) seem to be focussing in on exposed PS as the immunosuppressive culprit, as well as anti-PS being the fix, and the only anti-PS mentioned in patent applications in the WIPO & USPTO databases, various recent Haynes CAVD conf. presentation abstracts, and the paper in Virology have been:
2AG4
3G4
Bavi
Tarvi
IS1
IS2
IS4
IS6
it's happening,
j
in one sense we already have...
PER.C6 ups the mab yield bigtime. I'm not talking about a decent percentage increase. I'm talking multi-fold increase in product.
j
re:A government agency recently asked Peregrine for permission to test the drug against some 30 viruses considered to be potential agents of bioterror.
How did that work out for us?
I'd say darn good, judging by the presentation that Thorpe then made at the NIH Biodefense conf., in which he reported Bavi completely inhibiting H5N1.
I'd also say it went VERY well based on the prominent inclusion of Bavi in Dr. Kurilla's NIH biodefense mab meeting -
http://www3.niaid.nih.gov/topics/BiodefenseRelated/Biodefense/PDF/tarvacin.pdf
Finally, seeing that the govt just committed to up to 44 million to do more advanced testing with Bavi on ebola. marburg, lassa, hanta, rift, etc., I would think the work has gone great!
j
jess, it's MRK,
Here's some info-
Note that Besides increased CD4 & CD8 T cells, the drug is also increasing T reg cells.....
j
---------------------------------
Characterization of cellular immune-responses in cancer patients treated with the immunocytokine selectikine, an IL2/anti-DNA fusion protein
J. Laurent, P. Romero, R. Stupp, and D.E. Speiser
Selectikine (EMD 521873) is a fusion protein consisting of a human antibody fused to two molecules of a genetically modified human interleukin-2 (hIL2). Preclinical data suggest that immunocytokines exert antitumor-activity via CD8 T cell mediated mechanisms. Experiments in mice with the wild-type-IL2 containing immunocytokine ch.14.18-IL2 demonstrated CD8 T cell responses against tumors. Other experiments with ch14.18.IL2 showed that immunocytokine-treatment induced clonal expansion of T cells. Since 2007, 20 patients have been treated with escalating doses of selectikine from 0.075 mg/kg to 0.6 mg/kg. Analysis of T cell populations revealed an increase of proliferation and activation state of CD8 and CD4 T cells, but no significant effect on natural killer cells as expected. No dose depending effect has been observed between the different groups, showing that T cells react well already at low doses. Analysis of sub-populations of CD4 T cells revealed that T-reg cells are also sensitive to selectikine, likely due to their expression of the high avidity IL2 receptor CD25. As a consequence, T-reg cells proliferate considerably upon treatment with selectikine. Functional analysis of T cells, including T-reg cells are ongoing (Study Sponsored by Merck KGaA, Darmstad and EMD Pharmaceuticals Durham, USA).
http://www.cancer-chuv.ch/ccl_home/ccl_recherche/annual_scientific_report.htm
---------------
j
WHO's USING PER.C6 ?
-----------
j
Bill Gates:
June 26, 2008, on his plans for 'retirement' from Microsoft:
"I'll be learning more about the aspects of biology that affect the [Gates] foundation's work," he said. For example, he said, solving key problems in immunology could provide the kind of breakthroughs needed to prod pharmaceutical companies to invest more in long-neglected problems of the developing world such as tuberculosis or malaria.
SP, re: Crucell's PER.C6 & Peregrine/Avid ---------
IMHO,
If the govt funded work goes well and the govt were considering "stockpiling" Bavi or any of its future anti-PS offspring, (or even simply expected needing a lot of it for the 5 yr's of experiments), the best current expression system to make the stuff - by far - would be PER.C6.
It's INCREDIBLY high-yield.
and-
It's been through the wringer... it's been around a long long time. It's as safe as it gets.
It sure does make one wonder why Avid wound up being the FIRST U.S. production plant using Crucell PER.C6 to crank out mabs etc...
Personally, I think it could be related to either the govt expecting to order a lot of bavi/humanized anti-PS (if the work goes AS WELL AS IT ALREADY HAS in the Pichinde and CMV experiments), or CHAVI /Gates expecting to order a lot of the stuff.
or both.
here- this is a brief pdf with a little info on PER.C6 -
http://www.crucell.com/page/downloads/Factsheet_PER.C6_June2008.pdf
j
Haynes patent application --------
1. A method of treating HIV comprising administering to a patient in need thereof an antibody derivable from an normal subject or from an autoimmune disease subject that binds to a lipid on the surface of HIV or on the surface of HIV-infected cells and thereby neutralizes HIV-1, wherein said antibody is administered in an amount sufficient to effect said treatment.
2. The method according to claim 1 wherein said antibody is derivable from an anti-phospholipid syndrome subject.
3. The method according to claim 1 wherein said antibody is non-pathogenic.
4. The method according to claim 1 wherein said antibody is IS1, IS4 or IS6, or binding fragment thereof.
5. The method according to claim 1 wherein said antibody is IS1, or binding fragment thereof.
[0108] Alving and colleagues have made a mouse mab against phosphatidyl inositol phosphate and have shown that it neutralizes HIV in a PBMC assay. What the present studies show is that humans can spontaneously make anti-lipid antibodies and that these antibodies can broadly neutralize HIV in an unprecedented manner.
[0109] Summarizing, autoimmune disease patients can make antibodies that bind to virus-infected cells and, presumably, to budding HIV virions by virtue of their reactivity to HIV membranes and host membranes. Certain anti-lipid antibodies from autoimmune disease patients can also react with the Envelope trimer (such as IS6) but not all of the antibodies react also with the trimer (i.e., IS1 and IS4 do not react). Therefore, reactivity with the HIV envelope is not a prerequisite for neutralization in these antibodies.
AND MY FAVORITE SNIP!!!!!!!!!!! -
[0051] It will be appreciated from a reading of the foregoing that if HIV has evolved to escape the host immune response by making the immune system blind to it, other infectious agents may have evolved similarly. That is, this may represent a general mechanism of escape. That being the case, approaches comparable to those described herein can be expected to be useful in the treatment of such other agents well.
---------
Fauci NEJM full text ---------
http://content.nejm.org/cgi/content/full/359/9/888
j
NIAID describes challenges, prospects for an HIV vaccine
Aug 27 2008, 6:00 PM EST
http://www3.niaid.nih.gov/news/newsreleases/2008/HIV_vax_NEJM08.htm
NIH/National Institute of Allergy and Infectious Diseases
WHAT: Events of the past year in HIV vaccine research have led some to question whether an effective HIV vaccine will ever be developed. In the August 28 edition of the New England Journal of Medicine, officials from the National Institute of Allergy and Infectious Diseases (NIAID), part of the National Institutes of Health, examine the extraordinarily challenging properties of the virus that have made a vaccine elusive and outline the scientific questions that, if answered, could lead to an effective HIV vaccine.
In recent years, the most extensively studied HIV vaccines have aimed to mobilize immune cells called T cells, write Anthony S. Fauci, M.D., NIAID director, and Margaret I. Johnston, Ph.D., director of the Vaccine Research Program in NIAID's Division of AIDS. T-cell vaccines are not expected to prevent HIV infection. Rather, they could potentially reduce the level of virus (but not eliminate it) following infection, limit the number of immune cells that HIV destroys, and thus delay the progression to AIDS. There is no evidence yet that T-cell HIV vaccines work in humans, however. If the vaccines ultimately do, their effectiveness may vary greatly depending on the genetic make-up of each individual, given that T-cell immunity is dependent on genetic factors. Furthermore, because the virus would persist in the blood of vaccinated individuals, T-cell vaccines would likely generate only transient "herd immunity"--that is, population-wide protection from disease conferred by vaccination of a percentage of the community.
In response to the failure last September of a T-cell vaccine for which many people had high hopes, the HIV vaccine field has undergone a self-reexamination and has determined that the balance between fundamental discovery research and product development should shift toward discovery. In particular, future research must intensify the study of broadly neutralizing antibodies to HIV, why most HIV-infected people do not make them, and the design of novel strategies to induce them with a vaccine. Also, studying the earliest stages of HIV infection may shed light on ways to manipulate innate and mucosal immune responses to widen the window of opportunity for viral eradication, to prevent the virus from advancing to gut-associated lymphoid tissue, or both.
The authors conclude with cautious optimism that an effective HIV vaccine will be developed, but will depend on the significant growth of scientific understanding of HIV disease and human responses to the virus.
----------
Haynes Haynes Haynes,
j
jake,
read it.
j
anti-PS at Harvard as cancer treatment ----------
You may remember my posting the recent paper by Glen Dranoff of Harvard, who has done some amazingly effective tumor treatments, by blocking PS-mediated immunosuppression. Basically, they've been using autologous tumor cells as 'vaccine' combined with a PS-signalling blocker. GREAT TO SEE this happening, with such great results, at such a prestigeous place.
quick review
http://investorshub.advfn.com/boards/read_msg.aspx?message_id=27868131
http://investorshub.advfn.com/boards/read_msg.aspx?message_id=27891121
http://investorshub.advfn.com/boards/read_msg.aspx?message_id=28129169
Well,
This WIPO patent application appeared recently ----------
http://www.wipo.int/pctdb/en/wo.jsp?WO=2008043018&IA=US2007080446&DISPLAY=DESC
IT'S HAPPENING :)
j
jake, if you look at a particular Thorpe patent application a few years ago just before CHAVI/Gates CAVD got rolling, he goes into big detail into methods for generating mabs isolated from auto-immune patients to be used as therapy.
I posted it here when it came out.
Basically, Thorpe, (and Sophia Ran), were the first to really look into developing anti-phospholipid mabs AS THERAPY. It went (and still goes) against traditional immunological common sense. When it is finally accepted, (like for instance if some top doc like Bart Haynes ;) published a paper in a major journal about using anti-phospholipid mabs therapeutically, it will signify the beginning of the new paradigm.
While Thorpe and Ran were into therapeutics, others (like Pojen Chen), were primarily interested in understanding auto-immune disease, and what exactly makes anti-phospholipid abs pathogenic. They weren't really thinking about using anti-phospholipid mabs therapeutically.
Thorpe then listed all the ways to find them / produce them in a therapeutic patent application, which he filed a few years ago.
j
abstract from the Seattle Gates HIV vaccine conf last fall. Work that was done by Haynes Duke team, along with - Pojen Chen.
P10-22
Neutralization of HIV-1 primary isolates in PBMC assays
by monoclonal anti-lipid antibodies derived from a
patient with anti-phospholipid syndrome.
MA Moody1, D Montefiori1, MK Plonk1, H Liao1, S Xia1, TC Gurley1,
SM Alam1, P Chen2 and BF Haynes1
1 Duke University Medical Center, Durham, NC, USA;
2 UCLA School of Medicine, Los Angeles, CA, USA
Background: Rare human anti-envelope monoclonal antibodies (Mabs) that
broadly neutralize HIV-1 have been made, but HIV-1+ or vaccinated subjects
rarely make broadly reactive neutralizing antibodies. The observations
that 2F5, 4E10 and 1b12 are polyspecific antibodies and that AIDS may
be rare in patients with primary autoimmune diseases have prompted the
hypothesis that patients with autoimmune diseases who have defects in
tolerance mechanisms may be able to make antibodies that have anti-HIV-1
activity. Thus, we have assayed Mabs derived from anti-phospholipid
syndrome patients for their ability to neutralize HIV-1.
Methods: IS1 and IS4 are human Mabs derived from a patient with antiphospholipid
syndrome. Both are autoantibodies that react with cardiolipin
and phosphatidylserine. IS4 promotes thrombosis in a murine pinch-induced
thrombosis model while IS1 is not prothrombotic and, therefore, is nonpathogenic.
Neutralization assays used were pseudovirus assays in TZM/bl
cells and whole virus assays in peripheral blood mononuclear cells (PBMC).
Interaction of antibodies with Env and lipids was determined by surface
plasmon resonance (SPR) and flow cytometry.
Results: IS1 and IS4 Mabs had no neutralizing activity against HIV-1 primary
isolate pseudoviruses in the TZM/bl assay. In the PBMC assay, however,
both IS1 and IS4 neutralized 7/7 primary isolates tested (Torno, PAVO,
6535, DU123, DU156, DU151, and DU172; IC80s ranged from 0.06-4.1ug/
mL). Against SHIV and SIV viruses IS1 and IS4 neutralized SHIV SP162P3
at IC80s of 0.06 and 0.2μg/mL, respectively, while neither Mab neutralized
SHIV 89,6P or SIVmac239. Using SPR, neither IS1 nor IS4 reacted with
recombinant HIV-1 wild-type Envs. By flow cytometry, neither Mab labeled
viable uninfected T cell lines or PBMC; however, both IS1 and IS4 Mabs
reacted strongly with the surface of virus infected T cells.
Conclusion: Select anti-membrane polyspecific antibodies bind to HIV-1
infected but not uninfected cells, are non-pathogenic, and have anti-HIV-1
neutralizing activity in vitro in PBMC but not pseudovirus assays. It will be
of interest to determine if such non-pathogenic antibodies are protective in
passive therapy trials in vivo in non-human primates. Study of the mechanism
of neutralization of these antibodies and their B cells of origin may provide
clues to novel ways to safely induce protective antibodies to HIV-1.
Supported by the Center for HIV/AIDS Vaccine Immunology AI067854,
CHAVI 005 protocol.
http://www.hivvaccineenterprise.org/_dwn/Late_Breaker_Abstracts.pdf
------
j
James Robinson (at Tulane) has been the CHAVI hybridoma go-to-guy from day 1. Any interesting abs found in patients from CHAVI 005 or 006 protocols go to Robinson to make mabs for further study. So far all I've seen mentioned in Haynes' patent applications (and presentations) has been: bavituximab, tarvacin, 3G4, 2AG4, IS1, IS2, IS6. If Haynes is submitting patent applications that say:
IS1 can be safely used as a therapeutic Mab for treatment of HIV infected
subjects.
and can "broadly neutralize HIV in an unprecedented manner"
I'm very confident that we'll see a paper discussing WHY he said that in his patent application.
Also, obviously, you'd expect the work to 'continue'....
- funded by whom?....
Well, who's been funding it so far?....
j
Duke Human Vaccine Institute: HIV/TB/Malaria vaccine project ---------
Vaccine Development
HIV | Orthopoxvirus | TB
Oral Vaccines
TB
Richard Frothingham, MD
Hua-Xin (Larry) Liao, MD, PhD
Jae-Sung Yu, PhD
Barton Haynes, MD
Over the past two years, DHVI investigators have expanded their expertise in Mycobacterium tuberculosis (MTB) and Plasmodium falciparium (malaria) research. Working with the support of a grant from the National Institutes of Health, our investigators have teamed with Dr. William Jacobs at Albert Einstein College of Medicine, Dr. Louis Miller of the NIH, and Dr. Norman Letvin of Harvard, to form an inter-institutional, interdisciplinary effort to develop an HIV, TB and malaria trivalent vaccine for children.
A major hurdle in the development of a successful TB vaccine for adults is being addressed through this DHVI program. Adults have pre-existing immune responses to environmental mycobacteria, which interfere with the immune responses and the protection generated by the current TB vaccine, BCG. These pre-existing responses may interfere with novel TB vaccine candidates. Dr. Richard Frothingham and coworkers have developed a murine model of latent M. avium infection that mimics many aspects of latent human infection with environmental mycobacteria. His lab is using this model to characterize the CD4 and CD8 components of immune responses to environmental mycobacteria that interfere with protective responses to TB vaccines, and to evaluate several proposed mechanisms for this interference. The ultimate goal is the development of TB vaccines which are effective in adults. This work is relevant to the development of vaccines against other bacteria, including Bacillus anthracis and Yersinia pestis.
http://humanvaccine.duke.edu/modules/develop/index.php?id=3
---------
j
Passage of PEPFAR
US President George Bush recently signed into law a revised version of the President’s Emergency Plan for AIDS Relief (PEPFAR) authorizing US$48 billion in funding over the next five years to expand existing HIV/AIDS prevention, treatment, and care efforts worldwide. The original five-year, $15 billion plan, which has supported the provision of life-saving antiretroviral (ARV) treatment for approximately 1.7 million HIV-infected people, was due to expire in September.
The revised version more than doubles the amount of funding for HIV/AIDS prevention, treatment, and care programs, and also authorizes $9 billion in funding for malaria and tuberculosis programs.
A section of the new PEPFAR bill also contains provisions related specifically to facilitating the development of vaccines, including those against HIV/AIDS, tuberculosis, and malaria. The US President is required to report to Congress within one year on a strategy for accelerating the development of these vaccines, including details on creation of economic incentives for research, development, and manufacture, as well as the efforts taken by the US to support clinical trials of vaccines in developing countries and to prepare these countries for the introduction of new vaccines. —Jonathan Grund, contributing writer
http://www.iavireport.org/vax/VAXAugust2008.asp#3
--------
j
Wow, short shares are down again.
Now down to about four tenths of one percent of the float....
j