Innovation Grants 2017

PI: Andrew Badley, MD
Mayo Clinic College of Medicine, Rochester, MN

$200,000 (#109593)
Ixazomib to reduce HIV reservoir size: Cells have a recycling mechanism that disposes of or reuses old proteins—the proteasome. If the proteasome is disrupted, these damaged proteins persist, clogging up the cell and eventually leading to cell death. Ixazomib is a drug that is currently used in multiple myeloma, a form of blood cancer. Dr. Andrew Badley proposes to conduct a clinical study to test the ability of the drug to reduce the viral reservoir, the main barrier to a cure. 

PI: Benjamin Burwitz, PhD
Oregon Health and Science University, Portland, OR
$199,948 (#109596)
Creation of CCR5 knockout Mauritian cynomolgus macaques for stem cell transplants:
The Berlin patient, the first and only patient so far to have been cured of HIV, received a stem cell transplant from a donor who was genetically resistant to HIV because the donor lacked a key HIV receptor, the protein CCR5.  The Berlin patient’s difficult medical history and complications have made it difficult to determine exactly what led to his cure: was it the stem cell transplant, the complications presented by his new immune system attacking HIV-infected cells as part of a transplant complication known as graft vs. host disease, the chemotherapy and radiation used in the transplant, or some combination of these? Dr. Benjamin Burwitz proposes to answer these questions by generating a monkey model lacking CCR5, enabling him to test the multiple hypotheses concerning the Berlin patient’s cure.

PI: Andrew Henderson, PhD
Boston University School of Medicine, Boston, MA
$200,000 (#109603)
Disabling HIV provirus by promoting chromatinization: CRISPR/Cas9, a protein complex recently discovered in bacteria, has revolutionized biology because of the flexibility and ease with which scientists can use it to target and edit DNA, cutting out unwanted pieces, including the DNA form of HIV. Dr. Andrew Henderson is proposing to use this protein complex to silence the HIV DNA in a so-called “Block & Lock” approach. Unlike “Shock & Kill”, which requires the cell to wake up from latency- a formidable challenge - “Block & Lock” would permanently silence HIV and prevent the emergence of virus when ART is stopped.

PI: Brad Jones, PhD
The George Washington University, Washington, DC
$199,998 (#109606)
HLA-E specific TCR-like Antibodies for the Universal Targeting of Persistent HIV Reservoirs: Broadly neutralizing antibodies that target the viral protein Env—the only viral protein expressed on the surface of infected cells - must circumvent the high mutation rate of Env in order to be effective. On the other hand, viral proteins present inside the cell are much less subject to mutation but are poorly accessible to our body’s antibody making machinery. Because of a newly discovered immune mechanism, scientists have now found that the internal viral proteins may be digested and displayed on the surface of cells in a molecule called HLA-E. Dr. Brad Jones proposes to engineer antibodies that will recognize the digested protein/HLA-E complex and make the cell susceptible to death by Natural Killer cells.

PI: Fabio Romerio, PhD
University of Maryland, Baltimore, MD
$199,999 (#109612)
Permanent Silencing of HIV-1 Expression through the Polycomb Repressor Complex 2 epigenetic pathway: Dr. Fabio Romerio has uncovered a unique mechanism through which HIV drives its own latency, namely by making a molecule called Ast. He proposes that Ast participates in actively preventing the viral DNA from making virus. Dr. Romerio aims to determine how Ast asserts its effect and whether it can be delivered to all HIV infected cells to permanently and specifically block viral DNA. In contrast to the curative approach “Shock & Kill”, this “Block & Lock” approach aims to silence HIV and prevent the emergence of virus when ART is stopped.

PI: Joshua Schiffer, MD
Fred Hutchinson Cancer Research Center, Seattle, WA
$200,000 (#109614)
Anti-proliferative therapy for eradication of the HIV reservoir: Antiretroviral therapy (ART) controls viral load because the virus is prevented from infecting new cells. However, the reservoir persists even under ART through mechanisms that are still being uncovered. One possibility is that normal cell division of latent HIV infected cells maintains the reservoir even in the absence of viral replication. Dr. Joshua Schiffer aims to determine if CellCept, a drug that reduces cell replication – normally used to prevent organ transplant rejection - can also eliminate the persistence of the reservoir. His clinical trial will span 2 years, after which participants will discontinue their ART and determine whether the curative intervention worked.

ARCHE Gene Therapy Grants

960-arche.jpg

July 2017

Hildegard Büning, Ph.D. – principal investigator
Hannover Medical School, Hannover, Germany
$386,578 (109570)
Vector-mediated in vivo targeting of HIV reservoir cells or provirus elimination: To maximize safety of gene therapy, methods to specifically target reservoir cells are required. The HIV reservoir consists mainly of CD4+ T cells, with certain subsets more commonly harboring latent HIV. Dr. Büning plans to design an adeno-associated viral vector that specifically targets one of these main subsets, the central memory T cells. The vector will be designed to carry a gene editing tool called Brec1, previously shown to suppress HIV. One reported advantage of Brec1 over other gene editing tools is the apparent lack of off-target effects. 

Keith Jerome, M.D., Ph.D. – principal investigator
University of Washington, Seattle, WA
$399,955 (109575)
Subcutaneous administration of DARPin-modified adeno-associated virus vectors for selective targeting of CD4+ T cells: Curing a diffuse disease such as HIV, as opposed to a localized tissue-specific disease, requires a way to target the gene therapy to the target cells of interest, which in HIV are dispersed throughout the body. Dr. Jerome plans to use adeno-associated viral virus vectors engineered to utilize small, antibody-like  proteins to target the vectors and their gene-editing cargo directly to CD4+ T cells, which comprise the majority of the HIV reservoir. He will also experiment with ways of infusing the vectors such that they preferentially reach the lymph nodes, where most reservoir cells are found.

Hans-Peter Kiem, M.D., F.A.C.P. – principal investigator
Fred Hutchinson Cancer Research Center, Seattle, WA
$200,000 (109608)
Engineering blood cells to produce broadly neutralizing anti-HIV antibodies: Antibodies have been the focus of increasing optimism in the HIV cure research field due to their ability to not only neutralize virus particles, but also to target and help kill infected cells. Dr. Kiem plans to genetically modify immune system progenitor cells to serve as a constant source of antibodies. By engineering cells that will differentiate into a variety of types of immune cells, Dr. Kiem hopes that the progeny cells will migrate to various parts of the body, including lymph nodes and brain known to be important reservoirs of HIV.

Scott Kitchen, Ph.D. – principal investigator
University of California, Los Angeles, Los Angeles, CA
$400,000 (109577)
Optimized efficacy and persistence of engineered HIV-specific cellular immunity: Chimeric antigen receptor (CAR) cells have shown remarkable promise in their ability to clear some cancers in patients, and additionally have shown potential in HIV cure. Dr. Kitchen plans to improve the ability of CAR cells to kill HIV-infected cells by increasing their ability to detect the appropriate target cells and prolonging their survival. He also plans to engineer improvements to minimize the loss of CAR cells to attack by the immune system, and to design a mechanism to “switch off” CAR cells should they become unsafe or are no longer needed.

Yasuhiro Takeuchi, Ph.D. – principal investigator
University College London, London, United Kingdom
$360,034 (109584)
LentiStim: Mass production of lentiviral vectors for in vivo gene delivery: Curing HIV by gene therapy may require long-term persistence of the gene editing tools delivered by lentiviral vectors, which are closely related to HIV. Because the reservoir that harbors persistent HIV consists almost entirely of resting cells, such vectors cannot gain entry into the cells to deliver their cargo. Dr. Takeuchi plans to modify lentiviral vectors with a cocktail of stimulatory molecules that induce an optimal level of activation in the target cell, thus allowing entry of the vector. The effectiveness of lentiviral vectors is also plagued by immune responses raised against the vector, so the team will produce vectors that are resistant to destruction by the immune system.

Drew Weissman, M.D., Ph.D. – principal investigator
University of Pennsylvania, Philadelphia, PA
$400,000 (109587)
Targeting of nucleic acid therapeutics to cure HIV: Gene therapy approaches to cure HIV include efforts to cut the virus out of the human DNA, or to imbue the human host cells with characteristics that protect them from infection. Dr. Weissman plans to employ nucleic acid therapeutics as the tools to edit DNA to achieve either or both of these goals. One advantage of nucleic acid therapeutics is the ability to administer them repeatedly without raising an immune response that would destroy them. He plans to optimize their delivery to the appropriate cells by packaging them inside lipid nanoparticles that can be engineered with surface particles that guide them to specific targets.

Richard Wyatt, Ph.D. – principal investigator
The Scripps Research Institute, La Jolla, CA
$199,362 (109618)
Vector-mediated in vivo targeting of HIV reservoir cells or provirus elimination: Chimeric antigen receptor (CAR) cells have shown remarkable promise in their ability to clear some cancers in patients, and additionally have shown potential in HIV cure. One advantage is their ability to overcome a cancer or virus-infected cell’s ability to hide from the immune system. Dr. Wyatt plans to generate CAR T cells with the ability to kill HIV infected cells just as they begin to produce virus. Additionally, the CAR T cells will be engineered to produce antibodies that can neutralize any virus that is produced.

Shocking Latent HIV, Enhancing Immune Defense with a Single Drug

Last month we highlighted the work of scientists from the amfAR Institute for HIV Cure Research at the University of California, San Francisco, and their identification of a new pathway to induce HIV out of its latent state. Such activation renders the virus vulnerable to attack by the immune system. Unfortunately, many of the drugs currently being studied as such latency reversing agents work much better in the test tube than in patients.

Dr. Ole Søgaard

Dr. Ole Søgaard

Writing in the June issue of the journal Clinical Infectious Diseases, amfAR-funded scientist Dr. Ole Søgaard of Aarhus University in Denmark, with colleagues from there and the University of Copenhagen, joining an international team from Barcelona, Berlin, Belgium, Boston, San Francisco, and Philadelphia, report on an experimental drug with the capacity not only to activate latent HIV, but also to enhance the patient’s innate immune defenses against the activated virus.

Lefitolimod, also known as MGN1703, activates a protein known as TLR9, found on the surface of many types of immune cells. It belongs to a class of agents known as “immune surveillance reactivators,” which induce production of immune hormones, such as interferon-alpha, and enhance the function of dendritic cells, B cells, and natural killer cells. All of these cell types form part of our innate defense against HIV. Lefitolimod is currently in advanced stages of testing in colon cancer patients.

Based on promising studies in the test tube with cells from HIV-infected patients, Søgaard and associates sought to test the drug’s effects in patients themselves.

Fifteen adults taking effective antiretroviral therapy (ART) for at least one year were enrolled in the study. Lefitolimod was injected under the skin twice a week for four weeks. In 40% of the participants, HIV levels dramatically increased, from undetectable (less than 20 copies) to over 1500 copies, consistent with the role of a latency reversing agent. In addition, the researchers observed an enhancement of all immune responses evaluated.

The authors noted that the use of another drug to enhance TLR7, a related immune surveillance reactivator, in monkeys infected with the simian form of HIV also showed promising results, but only when combined with a therapeutic vaccine.

They concluded that their research is “the first clinical trial using a single drug in HIV-1-infected individuals on ART with the aim of both enhancing innate immunity and activating the HIV-1 reservoir.”

Further studies are underway in this promising area of cure research.

Dr. Laurence is amfAR’s senior scientific consultant.

 

Gene-Editing Tool Reduces HIV in Infected Mice

Dr. Marcella Flores, associate director of research at amfAR

Dr. Marcella Flores, associate director of research at amfAR

Researchers at Temple University and the University of Pittsburgh have been able to cut a fraction of HIV out of infected mice using CRISPR gene-editing technology, according to a new study in Molecular Therapy. The study builds upon previous findings by the research team, including one where CRISPR was used to cut HIV out of transgenic mice (genetically modified mice with HIV DNA inserted into their genomes—as opposed to having been infected with HIV).

In this study, researchers reduced HIV RNA (a measure of HIV) in transgenic mice by 60−95%. They were able to achieve a 96% reduction when they used the method in mice with EcoHIV, a mouse equivalent of HIV, during acute infection.

Employing a more widely used humanized BLT (bone marrow, liver, and thymus) mouse model, the researchers found that CRISPR was able to cut a fraction of HIV out of latently infected cells in some organs.

“It’s a good step forward,” said Dr. Marcella Flores, amfAR’s associate director of research. “It would be important to repeat the studies in monkeys and ensure that CRISPR is able to excise HIV from reservoir cells.”

Read more about the study here: http://bit.ly/gene-editing-tool

 

Using Mice to Detect 'Undetectable' HIV

A major part of amfAR’s “Countdown to a Cure for AIDS” initiative, which is aimed at developing the scientific foundations for a cure by 2020, is to determine the best method to detect and measure latently infected cells from individuals on antiretroviral therapy (ART).

Dr. Ramesh Akkina

Dr. Ramesh Akkina

This is critical to assessing the outcome of a cure intervention. Writing in the April issue of the journal Virology, amfAR-funded scientist Dr. Ramesh Akkina and colleagues at Colorado State University, along with researchers from the amfAR Institute for HIV Cure Research and Harvard University, reported a very novel approach.

The current “gold standard” for detecting persistent HIV is a test-tube technique known as qVOA (quantitative viral outgrowth assay). It is cumbersome and involves chemicals and antibodies in an attempt to activate virus in latently infected cells. One of its greatest drawbacks, however, is its inability to detect every virus that is capable of growing out of infected cells, estimated at approximately 60 per million cells. Akkina and colleagues sought a living alternative, using mice with human immune systems.

To “humanize” the mice and thus enable HIV to grow in these animals, the researchers used genetically immune-deficient mice transplanted with human stem cells or with bone marrow, liver, and thymus gland cells. They injected these animals with varying numbers of CD4+ T cells—from 100,000 to 20 million cells per mouse—obtained from 11 HIV-positive donors, all on ART. Five of these donors had undetectable viral loads by the standard qVOA.

The injected mice were followed for eight weeks. At weekly intervals blood was obtained in an attempt to detect, by a very sensitive molecular test known as RT-PCR, virus that wasn’t detectable using qVOA. Of the five qVOA-negative samples, four proved positive in the mice. The one sample that failed to produce virus was said to be of “poor quality”—donor samples had been frozen and don’t always thaw out well.

The authors concluded that the higher sensitivity of their humanized mouse models over qVOA and other types of mice used in similar experiments may ultimately lead to avoiding ART interruption as the only means to definitively assess the effectiveness of a potential HIV cure.

Let the mice do the work.

Dr. Laurence is amfAR’s senior scientific consultant.

Lab Mice Photo By Maggie Bartlett, NHGRI. - http://www.genome.gov/pressDisplay.cfm?photoID=5006, Public Domain, https://commons.wikimedia.org/w/index.php?curid=19805359

 

Dock and Block: Scientists Develop Technique that Makes Cells Resistant to HIV

Dock and Block: Scientists Develop Technique that Makes Cells Resistant to HIV

Scientists at The Scripps Research Institute (TSRI) have discovered a way of creating HIV- resistant cells by using antibodies to block HIV directly on the cell surface. Interestingly, in lab experiments, the resistant cells largely replaced the susceptible cells, potentially leading to long-term HIV protection.

Aging with HIV: Concerns About Accelerated Heart Disease

Aging with HIV: Concerns About Accelerated Heart Disease

In prior publications, these researchers have documented that heart attacks, sudden death due to heart failure, and stroke are more frequent among HIV-infected individuals, despite complete viral suppression by antiretroviral therapy (ART). These conditions are particularly worrisome as people with HIV are aging.

New amfAR Grants Enlist Help of Bioengineers to Overcome Obstacles to Curing HIV

New amfAR Grants Enlist Help of Bioengineers to Overcome Obstacles to Curing HIV

In a novel approach to conquering HIV, amfAR, The Foundation for AIDS Research, is pairing HIV researchers with bioengineers to address the main barrier to a cure for HIV: the persistent reservoirs of virus not cleared by antiretroviral therapy. A new round of Investment grants, totaling $1.2 million, will support six new research projects aimed at bringing to bear highly advanced technologies that until recently might have belonged in the pages of a science fiction novel.   

Understanding the ‘Interferon Paradox:’ A Novel Approach to Eradicating HIV

Understanding the ‘Interferon Paradox:’ A Novel Approach to Eradicating HIV

In an editorial in the January issue of The Journal of Clinical Investigation, amfAR-funded scientist Dr. Steven Deeks of the University of California, San Francisco (UCSF), working with colleagues from UCSF and Case Western Reserve University in Cleveland, Ohio, outline another approach involving interferon that may be superior at achieving HIV suppression.