Monday, August 31, 2015

Sangamo IVPR approach for Hemophilia will avoid problems described in this approach

Hemophilia Gene Therapy: Caught Between a Cure and an Immune Response

http://www.nature.com/mt/journal/v23/n9/full/mt2015135a.html

Roland W. Herzog
Editor-in-Chief, Molecular Therapy—Methods & Clinical Development
Gene therapy for the X-linked bleeding disorder hemophilia B—factor IX (FIX) deficiency—by means of in vivo gene transfer with adeno-associated viral (AAV) vectors has been in clinical trials for the past 16 years, cycling between partial successes in the clinic and further development in the laboratory. At the recent World Congress of the International Society on Thrombosis and Haemostasis (Toronto, Canada, June 2015), Paul Monahan and colleagues presented data from a patient who has achieved FIX levels of 20–25% that have been sustained for more than 6 months since undergoing hepatic gene transfer with an AAV8 vector (administered via peripheral vein). This result, representing another milestone in gene therapy for hemophilia, was in part accomplished by incorporating a missense mutation into a codon-optimized FIX sequence, which improves FIX enzymatic activity by 5- to 10-fold. However, similarly treated patients lost therapeutic expression because of immune responses to virally transduced hepatocytes or for other, yet-to-be-determined reasons.
In the past decade, gene therapy for hemophilia B has focused on expression and secretion of FIX by hepatocytes into the circulation. Sustained correction from severe to mild hemophilia (>5% of normal coagulation activity) using an AAV serotype 8 vector has been documented in recent years. Further improvements so as to sustain levels of >10% would essentially represent a cure for most patients, preventing spontaneous bleeds and limiting the need for intravenous factor infusions to surgery and treatment of severe trauma. The ”Padua” mutation was discovered when analyzing a case of X-linked juvenile thrombophilia, and has since been rigorously tested for improved gene therapy in murine and canine models of hemophilia B. For the clinical trial sponsored by Baxalta and conducted by Monahan and colleagues at the University of North Carolina–Chapel Hill, an FIX expression cassette was incorporated into a self-complementary AAV (scAAV), a vector system initially developed by McCarty and Samulski to eliminate the need for second-strand synthesis, which limits traditional AAV vectors. Seven patients have been treated in the trial thus far, and the results vary widely.
It has been known for 10 years that the immune system may limit the duration of therapeutic gene expression from AAV vectors in the human liver. The discovery of a hepatotoxic CD8+ T-cell response against AAV capsid was surprising at the time because none of the animal models had shown anything similar. Several articles in Molecular Therapy have since uncovered differences between murine and human, and even human and nonhuman primate, T-cell responses to capsid. More recently, a murine model was developed that mimics transaminitis and loss of FIX expression after ex vivo expansion followed by adoptive transfer of capsid-specific CD8+ T cells. At the highest vector dose in the current trial by Monahan et al. (3 × 1012 vector genomes/kg), both patients showed even higher levels of expression than those mentioned above (up to >50% of normal) but subsequently lost expression concomitant with transaminitis and a T-cell response to capsid. It remains remarkable that these responses may emerge two months after vector administration. No immune response against FIX (or the Padua variant) was found. In efforts to counter the destructive T cells, elevation of liver enzyme levels has previously been established as a biomarker that warrants initiation of immune suppression, and the steroid drug prednisone has been successfully applied to stop the T-cell response to AAV8 in its tracks and preserve FIX expression.
Although the same strategy was adopted in the trial by Monahan et al., efforts to preserve gene expression upon onset of transaminitis were unsuccessful. One drawback to this approach is that drug administration must be initiated very soon after the first signs of hepatotoxicity. Additionally, transaminitis may not be a sufficiently sensitive biomarker, and steroid drugs may not be effective against T-cell responses in all patients. Hence, the field continues to wrestle with the question of whether a prophylactic immunosuppression protocol should be incorporated into hepatic AAV gene transfer, and how such a regimen should be designed. Preclinical studies suggest a requirement for innate immune sensing of the AAV genome by Toll-like receptor 9 (TLR9) for CD8+ T-cell activation. Development of vectors devoid of TLR9-activating CpG motifs has been proposed. Using scAAV vectors may, on the one hand, increase or accelerate responses because of stronger TLR9 signaling, which, on the other hand, could be an advantage by providing a more defined target for immune suppression. No changes in liver enzymes or capsid-specific T cells were detected at a mid-dose of 1 × 1012 vector genomes/kg, reinforcing the conclusion that the T-cell response is vector dose–dependent. However, differences in vector production, purification, design (such as promoter), and measurement of titers complicate a direct comparison between trials. Interestingly, the three patients treated with the mid-dose experienced very diverse outcomes.
As mentioned, one patient continues to express at curative levels of >20%, whereas the others showed therapeutic levels initially but then spontaneously lost expression in the absence of any evidence for an immune response. Although minor changes in persistence of gene transfer or expression may have been amplified by the highly active Padua mutation, this observation remains unsettling because it adds another layer of complexity. Hence, AAV gene transfer to the human liver is caught somewhere between a cure, cellular immune responses, and additional factors that have yet to be determined.

Nature Methods: Functional footprinting of regulatory DNA

Journal name:
Nature Methods
Year published:
DOI:
doi:10.1038/nmeth.3554
Received
Accepted  30 July 2015
Published online

Saturday, August 29, 2015

Sangamo Short Interest (SGMO)

Sangamo Short Interest (SGMO)
DATE                                   SHORT INTEREST
8/14/15                                  9,488,474
7/31/15                                  9,207,150
7/15/15                                  9,205,202
6/30/15                                  9,387,951
6/15/15                                  9,393,825
5/29/15                                  8,938,987
5/15/15                                  8,668,559
4/30/15                                  8,198,983
4/15/15                                  8,050,307
3/31/15                                  8,285,803
3/13/15                                  8,441,291
2/27/15                                  8,939,000
2/13/15                                  9,268,065
1/30/15                                  9,082,814
1/15/15                                  9,387,913

Days to cover as of 8/14/15 is 9.15

Thursday, August 27, 2015

Dow Agro Enters into Rice Collaboration Agreement in China - Royalty Free????

Royalty free research and commercialization license for EXZACT with ICS-CAAS.

NYSE:
DOW
  
BEIJING--(BUSINESS WIRE)--Dow AgroSciences LLC, a wholly owned subsidiary of The Dow Chemical Company (NYSE: DOW), has entered into a collaboration agreement with the Institute of Crop Sciences of the Chinese Academy of Agricultural Sciences (ICS-CAAS). Under the agreement, Dow AgroSciences grants ICS-CAAS a royalty-free, non-transferable research and commercialization license for its proprietary EXZACT™ Precision Genome Editing Technology to be used in rice in China. Dow AgroSciences and ICS-CAAS scientists will collaboratively develop an industry-leading rice genome editing technology platform.
The EXZACT Precision Genome Editing Platform will enable ICS-CAAS scientists to capitalize on their significant investment and technical expertise in rice genomics, transcriptomics, proteomics, and cell biology as well as accelerate integration of the scientific knowledge generated in rice to rapidly develop valuable products for China. Dow AgroSciences has developed the EXZACT Precision Technology platform under an exclusive license and collaboration agreement in plants with Sangamo BioSciences, Inc. Dow AgroSciences and ICS-CAAS scientists will work together to make sure that the expertise and strengths of both parties are best combined to accelerate rice research and product development in China. This is part of Dow AgroSciences’ commitment to providing innovative and sustainable solutions to bolster food security and food safety in China.
ICS-CAAS indicates this collaboration is a milestone, which will accelerate the development of rice genome editing technology platform in China. It will have significant impact in developing new agronomic traits in rice.
“Our collaboration with ICS-CAAS, a world-renowned agricultural science research organization, is one with great promise,” said Tim Hassinger, President and CEO of Dow AgroSciences. “We have a long-term commitment in China as a strategic partner for agricultural sustainability and best practices. The EXZACT Precision Technology collaboration with CAAS is a strong example of this commitment, which will significantly enable Chinese scientists to improve rice research and product development, thus benefiting China’s long term food security.”
http://newsroom.dowagro.com/press-release/dow-agrosciences-and-ics-caas-partner-accelerate-rice-research-and-product-development

Friday, August 21, 2015

Blood Journal | In vivo genome editing of the albumin locus as a platform for protein replacement therapy

http://www.bloodjournal.org/content/early/2015/08/20/blood-2014-12-615492

Key points


  • AAV and ZFN mediated targeting of albumin locus corrects disease phenotype in mouse models of hemophilia A and B.
  • Robust expression from the albumin locus provides a versatile platform for liver directed protein replacement therapy.

Abstract

Site specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting of the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: Zinc Finger Nuclease (ZFN)-mediated site-specific integration of therapeutic transgenes within the albumin gene. Employing adeno-associated viral vector (AAV) delivery in vivo, we achieved long-term expression of human factors VIII and IX (hF.VIII and hF.IX) in mouse models of hemophilia A and B at therapeutic levels. Using the same targeting reagents in wild type mice, we expressed lysosomal enzymes that are deficient in Fabry and Gaucher diseases, as well as in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and non-genetic diseases.
  • Submitted December 3, 2014.
  • Accepted August 8, 2015.

The Economist: Genome Editing Review with Sangamo (SGMO) mention

Heartbreaking discussion of MPS-3 patient really clarifies the need for SGMO to get their program into the clinic. Also spends most of the article talking CRISPR's.
Sangamo Excerpt:

The easiest sorts of gene therapy will be those that can be done outside the body—ex vivo, in lab speak. The appeal of ex vivo work is the level of control; cells can be extracted, have their genes manipulated, and have their new genes tested before being put back. To see the sort of things that this makes possible take a look at the work being done by Sangamo Biosciences, based in Richmond, California, which has been working for a decade on an earlier, more cumbersome gene-editing technology that makes use of what are known as “zinc fingers”. It is trying to apply that technology to beta-thalassaemia, sickle-cell disease, haemophilia and HIV infection.
In clinical trials of its HIV treatment, Sangamo takes the immune cells that the virus infects out of the patient’s bloodstream and edits in a mutation that makes them highly resistant to infection. It then grows up a large number of the edited cells and infuses them back into the patient, where it is hoped they will flourish. A similar sort of approach can also be used in blood disorders such as beta-thalassaemia and sickle-cell disease which are caused by mutations in the globin gene. The idea is to extract blood stem cells from bone marrow, edit them so as to switch on the production of fetal haemoglobin (which the body stops producing shortly after birth, even if it cannot make the adult stuff) and return the stem cells to the body. It would be like a bone-marrow transplant—except that since the new genetically improved cells come from the patient’s own body there is no danger of rejection.

http://www.economist.com/news/briefing/21661799-it-now-easy-edit-genomes-plants-animals-and-humans-age-red-pen

Tuesday, August 18, 2015

Xconomy - Excellent Review of Competitive Landscape in Sickle Cell

http://www.xconomy.com/national/2015/08/18/100-years-but-only-one-drug-sickle-cell-patients-wait-for-help/

Sangamo Excerpt:
Another gene editing program for sickle cell is in the works from Sangamo Biosciences (NASDAQ: SGMO) of Richmond, CA. Sometime in the second half of 2016, Sangamo and its development partner Biogen (NASDAQ: BIIB) will ask FDA permission to start human trials with its program.
To do its gene editing, Sangamo uses a system called zinc finger proteins, which it owns. No one else can use zinc fingers without a license, and Sangamo, with 20 years of development under its belt, is the only company to advance a gene-editing product into human trials, for HIV.

CRISPR Excerpt:
CRISPR/Cas9 hasn’t been around as long as zinc finger proteins, and the technology has a major hurdle to overcome: making sure the molecular “scissors” it uses are making DNA cuts in the right places. Right now, the methods used to detect off-target cuts simply aren’t sophisticated enough. And all it takes is one cut in the wrong place to trigger a tragic unintended consequence. The fear dates back to gene therapy experiments fifteen years ago, in which genes meant to heal kids with severe combined immunodeficiency—the so-called “bubble boy disease”—inserted themselves in the wrong place and triggered cancer. Being more precise with gene editing tools, like CRISPR/Cas9, is still a goal, not a reality.
“Our ability to find off targets isn’t great right now,” says Corn. “No matter how bullish you are, the field [of gene therapy] has been bitten by kids getting leukemia. That should keep everyone in the hematopoietic field up at night.”

Saturday, August 15, 2015

Recap of Fred Hutch CGT4HIVCURE conference / Great news for Matt Sharp (5 years after treatment)

Matt Sharp veteran AIDS activist was treated with Sangamo's SB-728-T in 2010. He is still reaping the benefits.

Matt Sharp who has been living with HIV since 1988, is an example of that benefit. In 2010, he took part in a small clinical trial in San Francisco, one of the first to test gene therapy for HIV. His blood was drawn and his T cells were filtered out and genetically engineered to have the CCR5 mutation, then returned to his body. The goal of the trial was simply to see if altering and returning the genes worked and could be done safely, but Sharp received an unexpected benefit: His persistently low T-cell count more than doubled and remains high, relieving him of the regular bouts of pneumonia he used to suffer despite being on antiretroviral therapy.
One of Friday’s presenters, Dr. Joumana Zeidan, a postdoctoral research fellow from Case Western Reserve University, presented an analysis of that San Francisco trial, saying that the single infusion of CCR5-modified T cells helped shrink the reservoir.
Sharp, who attended the conference, raised his hand.
“Thank you,” he said. “I was a participant in that trial. I’ve been waiting for six years to understand what happened.”  

Read the article here:






http://www.fredhutch.org/en/news/center-news/2015/08/hiv-cure-research-today.html

Tuesday, August 11, 2015

Transient differential expression of CD44 after TCR triggering in CD4 Central Memory cells as a marker of asymmetric fate of daughter cells

http://cgt4hivcure2015.org/wp-content/uploads/2015/08/CGT4HIVCure-2015-Conference-Booklet-Final.pdf

Zeidan1, G Lee2, R Fromentin3, M Aid1, J Lalezari4, R Mitsuyasu5, S Wang2, W

Tang2, S Deeks6, D Ando2, RP Sekaly1

1Case Western Reserve University, Cleveland, OH; 2Sangamo Biosciences Inc. Richmond, CA, 3Centre

hospitalier de l’Université de Montréal, Montreal, CA; 4Quest Clinical Research, San Francisco, CA;

5University of California Los Angeles, Los Angeles, CA; 6University of California San Francisco, San



Francisco, CA.
 
Background: Nine aviremic HIV+ subjects on ART received 10-30 billion
 
SB-728-T cells. CCR5-modified cells expanded and persisted in PB up to 3.5

yrs post infusion (median = 2.1%), suggesting the presence of long lived CD4

populations, such as T memory stem cells (TSCM), within the product.
 

Methods: TSCM phenotyping was performed on PBMCs pre- and postinfusion.

Total HIV-DNA was measured using ddPCR. CD4 T-cell subsets were

sorted in the product and post-infusion and the levels of CCR5 modification

and integrated HIV-DNA were determined by qPCR. Gene array analysis was

performed to determine pathways involved in long-term persistence.
 

Results: We have previously identified a novel TSCM–like CD4 subset,



characterized by co-expression of intermediate levels of CD45RA and

CD45RO and high levels of CD95 and CD58, that expanded post-infusion and

correlated with long-term CD4 reconstitution (p=0.0279). Gene array analysis

suggested that persistence of this novel TSCM subset could be mediated by

up-regulation of genes involved in self-renewal (Notch pathway) and metabolic

pathways associated with cellular longevity (Fatty Acid Oxidation, OXPHOS,

etc...). These cells originated from the product, as they were highly enriched in

CCR5 modification (23.2% ± 17.6 modified alleles at 3yrs). Six out of 9 subjects

displayed a significant decrease in levels of total HIV-DNA over time (range

of decay from 0.5 to 3.6 log). Expansion of TSCM-like CD4 T-cells at month

6 correlated with the long-term decay of the HIV reservoir, which may be

explained by their lower levels of integrated HIV-DNA (mean of 172 copies/1e6

cells vs 1133 and 2415 copies/1e6 for central (TCM) and effector (TEM) memory

cells). Detection of up to 3% CCR5-modified TEM at 3yrs suggests that TSCM

have the capacity to differentiate into other memory populations over time.
 

Conclusions: A single infusion of CCR5-modified SB-728-T led to significant
 
decay in the HIV reservoir. An HIV-resistant CD4 TSCM–like population
can reduce the latent reservoir by selection and/or dilution, as well as by

differentiating into effector memory cells and limiting reservoir replenishment.

Our results suggest that targeting TSCM as a source of cells for genetic
 

manipulation of lymphocytes can enhance efficacy of adoptive T-cell therapy.
 

Cross-Clade Inhibition of HIV on Primary Cells by CXCR4 or or CCR5 Fused to the C34 Peptide from gp41 HR2

Michael Holmes presenting at CGT4HIVCURE 2015
http://cgt4hivcure2015.org/wp-content/uploads/2015/08/CGT4HIVCure-2015-Conference-Booklet-Final.pdf

Wang1, GJ Leslie2, J DeClercq1, MW Richardson2, APO Jordon2, PD Gregory1, JL

Riley2, JA Hoxie2, MC Holmes1

1Sangamo Biosciences Inc. Richmond, CA, USA; 2University of Pennsylvania, Philadelphia, PA, USA



HIV-1 entry into CD4+ T cells requires binding to CD4 and either the CCR5 (R5)

or CXCR4 (X4) co-receptor. Thus, strategies that disable productive co-receptor

(CoR) engagement should provide potent protection from HIV infection.

Previously we described a 34 amino acid peptide from the C-terminal heptad

repeat-2 domain of gp41 (C34) which, when fused to the amino terminus (NT)

of either R5 or X4, inhibits HIV-1 infection in transformed cells in vitro. Moreover,

our initial studies suggested that C34-R5 or C34-X4 fusions provided transdominant

resistance to infection irrespective of viral tropism (i.e. either C34-R5

or C34-X4 could inhibit entry of R5, X4 or dual-tropic isolates).

Here we demonstrate that C34-R5 or C34-X4 expression by lentiviral

transduction in primary CD4 T-cells from multiple donors results in almost

complete inhibition (>98%) of HIV-1 infection based on intracellular p24 levels

and RT activities. GFP-only and C34-CD4 expressing cells were infected at

levels similar to untransduced T-cells. C34/CoR expression was >90% on Day

0 and stable during the 14 days of culture (>85%). Trans-dominant inhibition by

C34-R5 or C34-X4 occurred for X4, R5 and dual-tropic primary isolates from

clades B and A/E. Remarkably, when C34-CoR transduced and untransduced

cells were mixed (1:4, respectively) and challenged with diverse HIV isolates, a

condition that provides a more sustained exposure to HIV, selective enrichment

of C34-CoR expressing cells occurred from the expected starting levels of ~25%

up to 60% C34-CoR+ cells during viral replication. Lastly, PMA/ionomycin and

anti-CD3/CD28 stimulation of C34-R5 and C34-X4 expressing T-cells resulted

in the expected robust expansion of these T cells which produced levels of

intracellular IFNg, MIP-1b, TNFa, and IL-2 that were indistinguishable from

untransduced cells. This novel method of engineering HIV-resistant, functional

CD4 T-cells that can be expanded ex vivo and adoptively reinfused represents a

promising and innovative approach with the potential to control HIV infection in

humans.
 

David Baltimore & Paula Cannon "Can Gene Therapy Cure HIV?"

On Wednesday, August 12th at 7 PM, Nobel laureate David Baltimore will join stem cell expert Paula Cannon to tell us why they think gene therapy will be vital to curing and controlling HIV.
Get some answers about gene therapy and its potential to cure HIV and prevent its transmission during a moderated Q&A with two of the brilliant minds working to achieve the impossible.

This FREE community event will be held in Pelton Auditorium, which is located in the Thomas Building on the campus of
Fred Hutch, Seattle, WA (1100 Fairview Ave N, Seattle, WA 98109).
Joining Baltimore will be a woman whose lab has been at the forefront of this field of science.
Her work has brought us the first clinical trial of genetically modified blood stem cells in human beings as well as science talks with saucy titles, such as "Giving HIV the [Zinc] Finger Using Targeted an HIV-Resistant Immune System"
Her name is Dr. Paula Cannon.
Dr. Cannon’s research is focused on gene therapy, hematopoietic stem cells, and HIV. Her expertise in the laboratory lies in gene editing and humanized mice
Using zinc finger nucleases (ZFNs) to genetically alter CD4 cells is proving safe and effective in treatment studies. Laboratory work conducted by Cannon suggests it might be possible to do the same thing with stem cells to rebuild entire immune systems resistant to HIV, thus curing the virus.
Cannon is part of a “Disease Team’ of researchers awarded by the California Institute for Regenerative Medicine (CIRM), who gave out more than $250 million to 14 multidisciplinary teams of researchers in California, the UK and Canada to develop stem cell-based therapies for 11 diseases.[raw]
Cannon’s Team seeks to treat HIV using a gene therapy approach to modify the patient’s blood-forming stem cells. When transplanted back, those cells will produce T cells that are resistant to HIV infection
The Phase I clinical trial, which is sponsored by City of Hope and funded by Sangamo BioSciences and CIRM, has started to enroll participants.
Since this is the first study of its kind involving modified blood stem cells, the FDA has set the bar for safety precautions very high.  How did they convince the FDA that there was a reasonable expectation of safety in moving this technique into humans?  That there would be no impact on stem cell function and no tumors developing from this treatment?

Monday, August 10, 2015

Conference on Cell and Gene Therapy for HIV Cure 2015 at the Fred Hutch

defeatHIV, in collaboration with the Fred Hutchinson Cancer Research Center, the University of Washington Center for AIDS Research (UW CFAR) and the UW Virology Division, will host the second Conference on Cell and Gene Therapy for HIV Cure in Seattle, WA on August 13-14, 2015. The event will be held on the campus of the Fred Hutchinson Cancer Research Center. Born out of the desire to bring much needed attention to HIV cell and gene therapies, this conference aims to unite an international roster of esteemed scientists to share and discuss novel treatments and research using these innovative modalities.  View more on the DETAILS page

Sangamo is a Corporate Sponsor, Dr Michael Holmes, Sangamo BioSciences is speaking on Friday August 14th.

Sangamo collaborators Paula Cannon, Rafik-Pierre Sekaly, Joumana Zeidan, Keith Jerome and Hans-Peter Kiem also speaking.

Thursday, August 6, 2015

Sangamo Presenting at Stem Cell on the Mesa October 7-9 2015

The Alliance for Regenerative Medicine (ARM), the international advocacy organization representing the interests of the advanced therapies and regenerative medicine community, today announced the initial slate of companies selected to present at its upcoming Stem Cell Meeting on the Mesa (SCMOM) Partnering Forum.
The meeting will feature presentations from more than 60 leading companies in the fields of cell therapy, gene and gene-modified cell therapy and tissue engineering. The Partnering Forum will take place October 7 and 8, 2015 at the Estancia Hotel in La Jolla, California as part of the full SCMOM conference, which also features a Scientific Symposium on October 9, 2015.
Collectively, the initial slate of presenting companies are currently conducting 125+ clinical trials with an additional 130 studies in the pre-clinical or research phase and 15+ products on the market. Altogether, these companies are developing 165+ unique products across a range of possible therapeutic applications.
Initial slate of presenting companies:
Abeona Therapeutics, Argos Therapeutics, Asterias Biotherapeutics, Avalanche Biotech, BioLife Solutions, bluebird bio, BrainStorm Cell Therapeutics, Caladrius Biosciences, Capricor Therapeutics, Cellectis, Cell Therapy Ltd., Cellular Dynamics International, Celyad, Cesca Therapeutics, Cyfuse, DiscGenics, Editas Medicine, Fate Therapeutics, Fibrocell Science, GenVec, HemoStemix, Histogenics, Inovio Pharmaceuticals, InVivo Therapeutics, jCyte, Juventas Therapeutics, Kiadis Pharma, MaxCyte, MEDIPOST America, Mesoblast Ltd., MiMedx, MolMed, Opexa Therapeutics, Organovo, Orthocell, Osiris Therapeutics, Oxford BioMedica, Pluristem Therapeutics, Regenerative Patch Technologies, REGENXBIO, ReNeuron, RepliCel, Sangamo BioSciences, StemBioSys, StemCells, Inc., TaiwanBio, Terumo BCT, TiGenix, TissueGene, uniQure, Vericel, ViaCyte, Voyager Therapeutics


Read more: http://www.digitaljournal.com/pr/2636604#ixzz3i2Ob3mZ5

Monday, August 3, 2015

CIRM recap of HIV Matters Town Hall

Last week CIRM joined with the AIDS Project Los Angeles to hold a Town Hall event in West Hollywood called HIV Matters: Countdown to a Cure: California Leads the Way. Around 120 people showed up to listen to stem cell scientists from City of Hope, University of Southern California (USC), Calimmune, and Sangamo Biosciences all of whom are using CIRM funding to develop new treatments, hopefully even cures, for HIV/AIDS.

http://blog.cirm.ca.gov/