Tuesday, May 10, 2016

Sangamo BioSciences Announces Participation At Upcoming Investor Conferences




RICHMOND, Calif., May 10, 2016 /PRNewswire/ -- Sangamo BioSciences, Inc. (NASDAQ: SGMO), the leader in therapeutic genome editing, announced today that Edward Lanphier, Sangamo's president and chief executive officer, will participate in the following upcoming conferences.
  • Piper Jaffray GenomeRx Symposium, New York, NY, May 17-18, 2016Mr. Lanphier will participate in two panel discussion entitled "Approaches to Gene Integration: Lenti, AAV & More" and "The Cutting/Editing Edge" on May 17, 2016.
  • 11th Annual World Stem Cells & Regenerative Medicine Congress, London, UK, May 18-20, 2016Mr. Lanphier will lead a roundtable discussion focused on infectious diseases on May 19, 2016.
About Sangamo
Sangamo BioSciences, Inc. is focused on Engineering Genetic Cures® for monogenic and infectious diseases by deploying its novel DNA-binding protein technology platform in therapeutic genome editing and gene regulation. The Company's proprietary In Vivo Protein Replacement Platform™ (IVPRP) approach is focused on monogenic diseases, including hemophilia and lysosomal storage disorders. Based on its proprietary IVPRP approach, Sangamo is initiating Phase 1/2 clinical trials for hemophilia B, the first in vivo genome editing application cleared by the FDA, and MPS I. In addition, Sangamo has a Phase 2 clinical program to evaluate the safety and efficacy of novel ZFP Therapeutics® for the treatment of HIV/AIDS (SB-728). The Company has also formed a strategic collaboration with Biogen Inc. for hemoglobinopathies, such as sickle cell disease and beta-thalassemia, and with Shire International GmbH to develop therapeutics for Huntington's disease. It has established strategic partnerships with companies in non-therapeutic applications of its technology, including Dow AgroSciences and Sigma-Aldrich Corporation. For more information about Sangamo, visit the Company's website at www.sangamo.com.

Tuesday, May 3, 2016

Sciencemag.org: The gene editor CRISPR won’t fully fix sick people anytime soon. Here’s why

http://www.sciencemag.org/news/2016/05/gene-editor-crispr-won-t-fully-fix-sick-people-anytime-soon-here-s-why

This week, scientists will gather in Washington, D.C., for an annual meeting devoted to gene therapy—a long-struggling field that has clawed its way back to respectability with a string of promising results in small clinical trials. Now, many believe the powerful new gene-editing technology known as CRISPR will add to gene therapy’s newfound momentum. But is CRISPR really ready for prime time? Science explores the promise—and peril—of the new technology.

How does CRISPR work?

Traditional gene therapy works via a relatively brute-force method of gene transfer. A harmless virus, or some other form of so-called vector, ferries a good copy of a gene into cells that can compensate for a defective gene that is causing disease. But CRISPR can fix the flawed gene directly, by snipping out bad DNA and replacing it with the correct sequence. In principle, that should work much better than adding a new gene because it eliminates the risk that a foreign gene will land in the wrong place and turn on a cancer gene. And a CRISPR-repaired gene will be under the control of that gene’s natural promoter, so the cell won’t make too much or too little of its protein product.

What has CRISPR accomplished so far?

Researchers have published successes with using CRISPR to treat animals with an inherited liver disease and muscular dystrophy, and there will be more such preclinical reports at this week’s annual meeting of the American Society of Gene and Cell Therapy (ASGCT). The buzz around CRISPR is growing. This year’s meeting includes 93 abstracts on CRISPR (of 768 total), compared with only 33 last year. What’s more, investors are flocking to CRISPR. Three startups, Editas Medicine, Intellia Therapeutics, and CRISPR Therapeutics, have already attracted hundreds of millions of dollars.

So why isn’t CRISPR ready for prime time?

CRISPR still has a long way to go before it can be used safely and effectively to repair—not just disrupt—genes in people. That is particularly true for most diseases, such as muscular dystrophy and cystic fibrosis, which require correcting genes in a living person because if the cells were first removed and repaired then put back, too few would survive. And the need to treat cells inside the body means gene editing faces many of the same delivery challenges as gene transfer—researchers must devise efficient ways to get a working CRISPR into specific tissues in a person, for example.
CRISPR also poses its own safety risks. Most often mentioned is that the Cas9 enzyme that CRISPR uses to cleave DNA at a specific location could also make cuts where it’s not intended to, potentially causing cancer.

With these caveats, do you even need CRISPR?

Conventional gene addition treatments for some diseases are so far along that it may not make sense to start over with CRISPR. In Europe, where one gene therapy is already approved for use for a rare metabolic disorder, regulators are poised to approve a second for an immune disorder known as adenosine deaminase–severe combined immunodeficiency (SCID). And in the United States, a company this year expects to seek approval for a gene transfer treatment for a childhood blindness disease called Leber congenital amaurosis (LCA).
At the ASCGT meeting, researchers working with the company Bluebird Bio will present interim data for a late-stage trial showing that gene addition can halt the progression of cerebral adrenoleukodystrophy, a devastating childhood neurological disease. Final results could help pave the way for regulatory approval. Bluebird will also report on trials using gene transfer for two blood disorders, sickle cell disease and β-thalassemia, bringing these treatments closer to the clinic.
Except for LCA, in which gene-carrying viruses are injected directly into eyes, these diseases are treated by removing bone marrow cells from patients, adding a gene to the cells, and reinfusing the cells back into the patient. New, safer viral vectors have reduced risks of leukemia seen in a few patients in some early trials for immunodeficiency diseases. Researchers are seeing “excellent clinical responses,” says Donald Kohn of the University of California, Los Angeles.
Although Kohn and other researchers have used an older gene-editing tool known as zinc finger nucleases to repair defective genes causing sickle cell disease and a type of SCID in cells in a dish, only a tiny fraction of immature blood cells needed for the therapy to work end up with the gene corrected—far below the fraction altered by now standard gene transfer methods. One reason is because the primitive blood cells aren’t dividing much (more on this below). Because gene-editing methods such as CRISPR are so much less efficient than gene addition, for several diseases, “I don’t think there will be a strong rationale for switching to editing,” says Luigi Naldini of the San Raffaele Telethon Institute for Gene Therapy in Milan, Italy.

CRISPR also has other issues

Using CRISPR to cut out part of a gene—not correct the sequence—is relatively easy to do. In fact, this strategy is already being tested with zinc finger nucleases in a clinical effort to stop HIV infection. In this treatment, the nucleases are used to knock out a gene for a receptor called CCR5 in blood cells that HIV uses to get into cells.
But when CRISPR is used to correct a gene using a strand of DNA that scientists supply to cells, not just to snip out some DNA, it doesn’t work very well. That’s because the cells must edit the DNA using a process called homology-directed repair, or HDR, that is only active in dividing cells. And unfortunately, most cells in the body—liver, neuron, muscle, eye, blood stem cells—are not normally dividing. For this reason, “knocking out a gene is a lot simpler than knocking in a gene and correcting a mutation,” says Cynthia Dunbar, president-elect of ASGCT and a gene therapy researcher at the National Heart, Lung, and Blood Institute in Bethesda, Maryland.
Researchers are working on ways to get around this limitation. The genes for HDR are present in all cells, and it’s a matter of turning them on, perhaps by adding certain drugs to the cells, says CRISPR researcher Feng Zhang of the Broad Institute in Cambridge, Massachusetts. Another avenue is to find alternatives to the Cas9 system that don’t rely on the HDR process, Zhang says.
But the low rate of HDR in most cells is one reason why the first use of CRISPR in the clinic will likely involve disrupting genes, not fixing them. For example, several labs have shown in mice that CRISPR can remove a portion of the defective gene that causes Duchenne muscular dystrophy, so that the remaining portion will produce a functional, albeit truncated protein. Editas hopes to start a clinical trial next year to treat a form of LCA blindness by chopping out part of the defective gene. One proposed gene-editing treatment for sickle cell disease would similarly snip out some DNA, so that blood cells produce a fetal form of the oxygen-carrying protein hemoglobin.

And CRISPR still has big safety risks

The most-discussed safety risk with CRISPR is that the Cas9 enzyme, which is supposed to slice a specific DNA sequence, will also make cuts in other parts of the genome that could result in mutations that raise cancer risk. Researchers are moving quickly to make CRISPR more specific. For example, in January, one lab described a tweak to Cas9 that dramatically reduces off-target effects. And in April in Nature, another team showed how to make the enzyme more efficient at swapping out single DNA bases.
But immediate off-target cuts aren’t the only worry. Although it’s possible to deliver CRISPR’s components into cells in a dish as proteins or RNA, so far researchers can usually only get it working in tissue inside the body by using a viral vector to deliver the DNA for Cas9 into cells. This means that even after Cas9 has made the desired cuts, cells will keep cranking it out. “The enzyme will still hang around over 10, 20 years,” Zhang says. That raises the chances that even a very specific Cas9 will still make off-target cuts and that the body will mount an immune response to the enzyme.
This may not truly be a problem, Zhang suggests. His team created a mouse strain that is born with the gene for Cas9 turned on all the time, so it expresses the enzyme in all cells for the animal’s entire life. Even after interbreeding these mice for about 20 generations, the mice “seem to be fine” with no obvious abnormal health effects, Zhang says. All the same, “the most ideal case is, we want to shut off the enzyme.” And that may mean finding nonviral methods for getting Cas9 into cells, such as ferrying the protein with lipids or nanoparticles—delivery methods that biologists have long struggled to make work in living animals.
Other long-standing obstacles to gene therapy will confront efforts using CRISPR, too. Depending on the disease, any gene-edited cells may eventually die and patients could have to be treated multiple times. Researchers using gene transfer and editing approaches are also both hindered by limits on how much DNA a viral vector can carry. Right now CRISPR researchers often must use two different viruses to get CRISPR’s components into cells, which is less efficient than a single vector.

So what’s the bottom line?

Gene therapists remain excited by CRISPR, in part because it could tackle many more inherited diseases than can be treated with gene transfer. Among them are certain immune diseases where the amount of the repaired protein must be precisely controlled. In other cases, such as sickle cell disease, patients won’t get completely well unless a defective protein is no longer made by their cells, so just adding a gene isn’t enough. “It opens up a lot of diseases to gene therapy because gene addition wasn’t going to work,” Dunbar says.
After more than 2 decades of seeing their field through ups and downs, veterans of the gene therapy field are wary of raising expectations about CRISPR for treating diseases. “Whenever there’s a new technology, there’s a huge amount of excitement and everybody thinks it will be ready tomorrow to cure patients,” says gene therapy researcher Mark Kay of Stanford University in Palo Alto, California. “It’s going to take some time.”

Monday, May 2, 2016

First Quarter 2016 Earnings Report

"During the first quarter of 2016 we focused on activities to enable initiation of the first clinical trials of our IVPRP approach in hemophilia B and MPS I, and I am pleased to report that we will meet our stated timelines for the opening of both trials," said Edward Lanphier, Sangamo's president and chief executive officer. "We look forward to generating and presenting clinical data that support the application of this highly leverageable platform while continuing to progress our programs in hemophilia A and several lysosomal storage disorders toward clinical development."
Recent Highlights
  • Presentation of new data from Sangamo's proprietary In Vivo Protein Replacement Platform™ (IVPRP) programs for MPS I and MPS II at the 12th Annual WORLDSymposium™ Meeting. Sangamo scientists and academic collaborators from the University of Minnesota presented new preclinical data in disease models of the Company's IVPRP-based MPS I (Hurler syndrome) and MPS II (Hunter syndrome) programs at the 2016 WORLDSymposium™ Annual Meeting. In animal models of disease, the data demonstrated the production of stable, therapeutic levels of replacement enzyme from the liver into the circulation and secondary tissues, including the brain, resulting in significant reduction in biomarkers of the disease, and notably, statistically significant improvements in cognitive function in treated animals.
  • Publication in the New England Journal of Medicine (NEJM) highlighting Sangamo's IVPRP approach for hemophilia B. In March, NEJM published a review authored by Dr. Amit Nathwani, a key opinion leader in the field of gene therapy approaches to hemophilia, highlighting Sangamo's IVPRP-based SB-FIX program for the one-time, permanent treatment of hemophilia B. The article details the significant advantages of Sangamo's ZFN-mediated, targeted integration of the Factor IX (FIX) gene at the albumin locus over conventional non-integrating gene therapy approaches that use adeno-associated virus (AAV) to deliver the FIX gene and other therapeutic genes to liver cells.
  • Sangamo augments clinical expertise with the appointment of Matthew Spear, M.D. as Vice President and Head of Clinical Development. Dr. Spear joins Sangamo with more than 20 years of experience in all stages of biopharmaceutical research and development and has led the development of over 15 therapeutic products. Most recently, Dr. Spear served as a Vice President in Clinical Development at Incyte Corp.
  • Presentation of immunological data from Sangamo's SB-728-T Phase 2 clinical program in HIV/AIDS at the 2016 Annual Conference on Retroviral and Opportunistic Infections (CROI 2016). Sangamo's collaborators from Case Western Reserve University presented analyses of immunological data from the Company's clinical trials of SB-728-T, a ZFP Therapeutic® designed to provide functional control of HIV/AIDS. The presentation outlined potentially interrelated mechanisms for viral load (VL) control in SB-728-T-treated subjects during a treatment interruption (TI) from their antiretroviral therapy. This suggests a model mechanism of action for SB-728-T and enables identification of patients who will most benefit from Sangamo's ZFN-mediated CCR5 knock-out approach.
Upcoming Events in 2016
  • Initiation of IVPRP-based Phase 1/2 clinical trials SB-FIX-1501 (hemophilia B) and SB-318-1502 (MPS I / Hurler syndrome) by the end of the second quarter and mid-2016, respectively.
  • Presentation of data from several ZFP Therapeutic programs by Sangamo scientists and collaborators at the upcoming 19th Annual Meeting of the American Society of Gene & Cell Therapy (ASGCT), which will be held in Washington D.C. from May 4-7, 2016.
  • Submission of investigational new drug (IND) applications in the first half of 2016 for Sangamo's IVPRP-based SB-913 program for MPS II (Hunter syndrome), and beta-thalassemia program in collaboration with Biogen Inc. (Biogen).
  • Presentation of new clinical data from Cohort 3* of Sangamo's SB-728-1101 Phase 1/2 trial in T-cells for the functional control of HIV/AIDS in the second half of 2016.
First Quarter 2016 ResultsFor the first quarter ended March 31, 2016, Sangamo reported a consolidated net loss of $16.5 million, or $0.23 per share, compared to a net loss of $5.3 million, or $0.08 per share, for the same period in 2015. As of March 31, 2016, the Company had cash, cash equivalents, marketable securities and interest receivable of $189.0 million.
Revenues for the first quarter of 2016 were $3.9 million, compared to $13.5 million for the same period in 2015. First quarter 2016 revenues were generated from the Company's collaboration agreements with Biogen and Shire International GmbH (Shire), enabling technology agreements and research grants. The revenues recognized for the first quarter of 2016 consisted of $3.7 million from collaboration agreements and $0.2 million from research grants, compared to $12.7 million and $0.8 million, respectively, for the same period in 2015.
The decrease in collaboration agreement revenues was primarily a result of an amendment to the Company's collaboration and license agreement with Shire in the third quarter of 2015, which returned the rights to the hemophilia programs to Sangamo, and a decrease in revenue under the Company's collaboration agreement with Sigma.
In the first quarter of 2016, Sangamo recognized $2.0 million of revenues related to research services performed under the collaboration agreement with Biogen, and $0.4 million of revenues related to research services performed under the collaboration agreement with Shire. In addition, Sangamo received upfront payments of $13.0 million and $20.0 million pursuant to the agreements entered into with Shire in 2012 and Biogen in 2014, respectively. The Shire payment is being recognized as revenue on a straight-line basis over the initial six-year research term. Beginning in January 2016, the Biogen payment will be recognized over approximately 42 months which reflects the revised service period related to Sangamo's deliverables under the Biogen agreement. The Company recognized $0.5 million of the Shire upfront payment and $0.6 million of the Biogen upfront payment as revenue for the first quarter of 2016.
Research and development expenses were $15.3 million for the first quarter of 2016, compared to $15.0 million for the same period in 2015. Research and development expenses were primarily comprised of manufacturing expenses, research expenses associated with Sangamo's clinical and preclinical programs, and personnel-related expenses, including stock-based compensation.
General and administrative expenses were $5.4 million for the first quarter of 2016, compared to $4.7 million for the same period in 2015.
Total operating expenses for the first quarter of 2016 were $20.6 million, compared to $19.7 million for the same period in 2015.
Financial Guidance for 2016 The Company reiterates its earlier guidance as follows:
  • Cash and Investments: Sangamo expects that its cash, cash equivalents and marketable securities will be at least $150 million at the end of 2016, inclusive of research funding from existing collaborators but exclusive of funds arising from any additional new collaborations or partnerships, equity financings or other new sources.
  • Revenues: Sangamo expects that revenues will be in the range of $20 million to $25 million in 2016, inclusive of research funding from existing collaborations.
  • Operating Expenses: Sangamo expects that operating expenses will be in the range of $85 million to $95 million for 2016. 

Sunday, May 1, 2016

New Session at ASGCT this Year - Industry Matchmaking

http://www.asgct.org/meetings-educational-programs/asgct-annual-meetings/2016-annual-meeting/attendee/special-sessions

Industry Matchmaking Sessions

Wednesday, May 4 – Friday, May 6, 2016
Marriott Wardman Park Hotel
Individually scheduled meetings
New this year, ASGCT’s Industry Matchmaking Sessions will provide an opportunity for abstract presenters at the 19th Annual Meeting to meet one-on-one with a senior industry representative and give a short 5-10-minute presentation of their work to discuss potential new collaborative research agreements, technologies, and products with industry representatives.
Participating companies include:
  • Audentes Therapeutics
  • Bayer
  • BioMarin
  • bluebird bio
  • CRISPR Therapeutics
  • GlaxoSmithKline
  • Lentigen Technology Inc., A Miltenyi Biotec Company
  • Merck
  • Pfizer
  • Precision BioSciences
  • Precision Nanosystems
  • Sangamo BioSciences
  • Sanofi
  • Spark Therapeutics
  • Tocagen
As the abstract deadline for the 19th Annual Meeting has already passed, the Industry Matchmaking Sessions have been filled at this point. The abstract review committees will match each applicant with one company, and the ASGCT office will schedule individual meetings between applicants and company representatives to take place throughout the Annual Meeting.
If you are planning on submitting an abstract at next year’s 20th Annual Meeting, please consider signing up for this unique opportunity in 2017 during the abstract submission process!