Sunday, November 29, 2015

December Will be a Busy Month

Upcoming Events:

December 2 - Piper Jaffray Healthcare Conference Presentation
                       1 pm EST
                        http://investor.sangamo.com/events.cfm

December 3 GeneticRx Edward Lanphier
                     https://bbbiotechconference.com/conference-details.php?id=35
                     Not Webcast

December 5-8 57th Annual Meeting of the American Society of Hematology (ASH)
                     Non-human primate data from its proprietary In Vivo Protein Replacement Platform                             program for hemophilia B, and data from its ZFP Therapeutic® hemoglobinopathy                               program
                       Abstracts: http://sangamodomain.blogspot.com/2015/11/ash-abstracts-for-sangamo-2015.html

December 8-11 HIV Persistence during Therapy Conference
                          Gene editing CCR5 in HIV subjects CD4 T cells Dale Ando, Sangamo,

December 10 Life Science Disruptors Conference
                       http://sangamodomain.blogspot.com/2015/11/lanphier-speaking-at-life-science.html

December 31 Deadline for announcement of IND acceptance for Hemophilia B and Hurlers
                       http://sangamodomain.blogspot.com/2015/11/jefferies-still-waiting.html

Thursday, November 26, 2015

Very Promising Type 1 Diabetes Research (not Sangamo)

End of daily injections for diabetes as scientists restore insulin production

Injecting billions of immune cells back into the body boosts insulin production, preventing the need for daily injections

 The end of daily injections for diabetes sufferers could be in sight after scientists showed it is possible to restore insulin production for up to a year by boosting the immune system.

Hundreds of thousands of people in Britain suffer from Type 1 diabetes and need to inject themselves daily to keep blood sugar levels under control.
The disease attacks insulin-secreting cells in the pancreas. Healthy people have billions of ‘peacekeeping’ cells called ‘T-regs’ which protect insulin-making cells from the immune system but people suffering Type 1 diabetes do not have enough.

"The T-reg intervention frees people like me from the daily grind of insulin therapy and lifelong fear of complication"
Mary Rooney, Type 1 diabetes patient
Now researchers at the University of California and Yale have shown that the ‘T-regs’ can be removed from the body, increased by 1,500x in the laboratory and infused back into the bloodstream to restore normal function.
An initial trial of 14 people has shown that the therapy is safe, and can last up to a year.
"This could be a game-changer," said Dr Jeffrey Bluestone, Professor in Metabolism and Endocrinology at the University of California, San Francisco (UCSF).
“By using T-regs to 're-educate' the immune system, we may be able to really change the course of this disease.
"We expect T-regs to be an important part of diabetes therapy in the future."
Read the entire article here:
 http://www.telegraph.co.uk/news/science/science-news/12016532/End-of-daily-injections-for-diabetes-as-scientists-restore-insulin-production.html

Tuesday, November 24, 2015

Sangamo Biosciences (SGMO) Short Interest Declines 660,000 Shares, Low Volume Increases Days to Cover to 14

Sangamo Short Interest (SGMO)
DATE                                   SHORT INTEREST
11/13/15                              11,088,163 (13.91 days to cover)
10/30/15                              11,668,561 
10/15/15                              12,821,197    
9/30/15                                12,619,596
9/15/15                                10,882,795
8/31/15                                10,150,146
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

Monday, November 23, 2015

Sangamo BioSciences Announces Presentation At The 2015 Piper Jaffray Healthcare Conference

Sangamo BioSciences Announces Presentation At The 2015 Piper Jaffray Healthcare Conference
Edward Lanphier to Also Participate in the Genetic Rx Conference in Boston


RICHMOND, Calif., Nov. 23, 2015 /PRNewswire/ -- Sangamo BioSciences, Inc. (NASDAQ: SGMO), a leader in therapeutic genome editing, announced today that Edward Lanphier, Sangamo's president and chief executive officer, will provide an update on the progress of Sangamo's ZFP Therapeutic® development programs and an overview of the company's business strategy at 1:00pm ET on Wednesday, December 2, 2015, at the 27th Annual Piper Jaffray Healthcare Conference. The conference is being held in New York.

Sangamo BioSciences, Inc.
The presentation will be webcast live and may be accessed via a link on the Sangamo BioSciences website in the Investor Relations section under Events and Presentations. The webcast will be archived on the Sangamo website for two weeks after the event.
Edward Lanphier will also participate in the Genetic Rx conference taking place at Harvard Medical School on Thursday, December 3, 2015. Genetic Rx, a networking conference of the Boston Biotech Conferences series, will bring together prominent academics, leaders of the biopharmaceutical industry and key investors to discuss the present and future of genetic therapies, pricing, reimbursement, the regulatory landscape and the treatment of patients with rare and ultra-rare genetic diseases. The event will not be webcast.
About SangamoSangamo BioSciences, Inc. is focused on Engineering Genetic CuresTM for monogenic and infectious diseases by deploying its novel DNA-binding protein technology platform in therapeutic genome editing and gene regulation. The Company has a Phase 2 clinical program to evaluate the safety and efficacy of novel ZFP Therapeutics® for the treatment of HIV/AIDS (SB-728). Sangamo's other therapeutic programs are focused on monogenic and rare diseases. The Company has 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 also 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.

Friday, November 20, 2015

Another Bio Bites the Dust

Celladon, run by Sangamo director, Paul Cleveland.
PALO ALTO, Calif. and SAN DIEGO, Nov. 18, 2015 (GLOBE NEWSWIRE) -- Celladon Corporation (Nasdaq:CLDN) and Eiger BioPharmaceuticals, Inc., a privately-held biopharmaceutical company, today announced that they have entered into a definitive merger agreement under which the stockholders of Eiger would become the majority owners of Celladon, and the operations of Celladon and Eiger would be combined. The proposed merger remains subject to certain conditions, including the approval of Celladon stockholders.  If approved, upon closing of the transaction, Celladon will be renamed Eiger BioPharmaceuticals, Inc.
An investor syndicate has committed to invest $39.5 million in the combined company including new investors HBM Healthcare Investments, RA Capital Management, Sabby Management, Sphera Global Healthcare, Perceptive Advisors and Monashee Capital Partners together with existing long-time Eiger stockholders, ViVo Capital and InterWest Partners.  Six million dollars of the financing has already been funded, and the remaining $33.5 million is expected to close contemporaneously with the completion of the proposed merger.  The total cash balance of the combined company upon the closing of the proposed merger and the financing is expected to be greater than $60 million.
The proposed merger will create a clinical-stage company with a diversified development portfolio of product candidates addressing novel targets for four distinct orphan diseases: Hepatitis Delta Virus, bariatric surgery-induced hyperinsulinemic hypoglycemia, pulmonary arterial hypertension, and lymphedema.
"The proposed merger and concurrent financing are expected to be sufficient to allow the combined company to obtain Phase 2 clinical trial results from at least two of the four planned development programs by late 2016," said David Cory, President and Chief Executive Officer of Eiger.  "We have a dedicated, experienced management team in place and executing on these four novel programs, all of which address potentially significant markets with unmet medical needs."
Paul Cleveland, Celladon's departing President and Chief Executive Officer added, "Following an extensive review of strategic alternatives and a thorough process, we have chosen to combine with Eiger because we believe the proposed merger provides Celladon shareholders with an attractive opportunity for value appreciation. I'm also pleased that Mr.Wiklund will assume the President and Chief Executive role at Celladon."
About the Proposed Merger
Existing Eiger stockholders as well as investors in the contemporaneous financing will receive newly issued shares of Celladon common stock in connection with the proposed merger.  Celladon is expected to issue approximately 85.0 million new shares of its common stock to Eiger stockholders and participants in the financing.  On a pro forma basis for the combined company, current Celladon shareholders are expected to own approximately 22%, current Eiger shareholders approximately 45% and the new Eiger investor syndicate approximately 33%, each on a fully-diluted basis.
The transaction has been unanimously approved by the boards of directors of both companies, and a majority of Eiger stockholders have agreed to vote in favor of the transaction. The proposed merger is expected to close in the first half of 2016, subject to the approval of the stockholders of each company as well as regulatory approval and other customary conditions.  The merger agreement contains further details with respect to the proposed merger.
Celladon's exclusive financial advisor in the transaction is Wedbush PacGrow Healthcare.  Jefferies LLC is acting as lead financial advisor to Eiger and Piper Jaffray is acting as financial advisor.  Pillsbury Winthrop Shaw Pittman LLP served as legal counsel to Celladon and Cooley LLP served as legal counsel to Eiger.
Management and Organization
The directors and executive officers of Celladon will resign from their positions with Celladon upon the closing of the proposed merger, and the combined company will be under the leadership of Eiger's current executive management team with David Cory serving as President and Chief Executive Officer. Following the closing of the proposed merger, the Board of Directors of the combined company is expected to consist of seven members all of whom will be designated by Eiger.  The corporate headquarters will be located in the San Francisco Bay Area.
Celladon CEO Transition
Celladon also announced that, as part of a further reduction in force implemented in connection with the signing of the merger agreement, Paul Cleveland, President and Chief Executive Officer, is leaving the company effective November 19, 2015 to pursue other industry opportunities. Fredrik Wiklund, Celladon's Vice President of Corporate Development and Investor Relations has been named Celladon's President and Chief Executive Officer effective the same date.

Thursday, November 19, 2015

FDA Approves AquAdvantage® Salmon (genetically engineered)

MAYNARD, Massachusetts, 19 November 2015 – AquaBounty Technologies, Inc. (AIM: ABTU; OTC: AQBT), a biotechnology company focused on enhancing productivity in aquaculture, and a majority-owned subsidiary of Intrexon Corporation (NYSE: XON), announces today that the U.S. Food and Drug Administration (FDA) has approved the Company’s New Animal Drug Application for the production, sale, and consumption of its AquAdvantage® Salmon, an Atlantic salmon that has been genetically enhanced to reach market size in less time than conventional farmed Atlantic salmon.
Ronald L. Stotish, Ph.D., Chief Executive Officer of AquaBounty, commented, “AquAdvantage Salmon is a game-changer that brings healthy and nutritious food to consumers in an environmentally responsible manner without damaging the ocean and other marine habitats. Using land-based aquaculture systems, this rich source of protein and other nutrients can be farmed close to major consumer markets in a more sustainable manner.”
The U.S. currently imports over 90% of all the seafood, and more specifically over 95% of the Atlantic salmon, it consumes. AquAdvantage Salmon will offer the opportunity for an economically viable domestic aquaculture industry while providing consumers a fresh and delicious product.
Through greater efficiency and localized production, AquaBounty’s AquAdvantage Salmon increases productivity while reducing costs and the environmental impacts associated with current salmon farming operations. Land-based aquaculture systems can provide a continuous supply of fresh, safe, traceable, and sustainable AquAdvantage Salmon to communities across the U.S. and do it with a reduced carbon footprint. Importantly, it offers an alternative approach to fish farming that does not exploit the oceans.
Jack A. Bobo, Senior Vice President and Chief Communications Officer at Intrexon, stated, “The U.S. Dietary Guidelines Advisory Committee encourages Americans to eat a wide variety of seafood—including wild caught and farmed—as part of a healthy diet rich in healthy fatty acids. However, this must occur in an environmentally friendly and sustainable manner. FDA’s approval of the AquAdvantage Salmon is an important step in this direction.”

AquaBounty Technologies, Inc. is a publicly traded company whose largest shareholder is Intrexon Corporation (NYSE: XON). Intrexon is a synthetic biology company with diverse interests in medicine, food and agriculture, and fuels and the environment. AquaBounty is an aquaculture company focused on improving productivity in commercial aquaculture, a $144 billion industry and the fastest-growing segment of the worldwide food industry. The Company’s objective is the application of biotechnology to ensure the availability of high quality seafood to meet global consumer demand. AquaBounty is developing products to address critical production constraints in the most popular farmed species, focusing initially on salmon, trout, and tilapia. Its AquAdvantage® fish program is based upon a single, specific molecular modification in fish that results in more rapid growth in early development.

Look what Shkreli is up to now. (no it does not involve Sangamo)

5:26 am KaloBios Pharma comments on acquisition of shares by group comprised of Martin Shkreli and associates, says in discussions with the group regarding possible direction for the company to continue in operation (KBIO) :
The co announced that it has been informed that an investor group comprised of Martin Shkreli and associates together have acquired more than 50% of the outstanding shares of KaloBios, and that the company is in discussions with Mr. Shkreli regarding possible direction for the company to continue in operation. Mr. Shkreli is the founder and chief executive officer of Turing Pharmaceuticals

Shkreli became a poster boy of sorts for high-priced pharmaceuticals when The New York Times wrote about Turing earlier this year, noting the company had bought an infectious-disease drug that previously sold for $13.50 a pill and increased that price to $750. Shkreli said after an uproar that he would cut the price of the drug. 

KBIO closed at $2.07 yesterday, indicated at $16.07 premarket

Wednesday, November 18, 2015

Genetic Rx Conference 12-3-15

Prominent academics, leaders of the biopharmaceutical industry and key investors will gather on December 3rd at Genetic Rx, a networking conference of the Boston Biotech Conferences series. Genetic Rx will take place at the Joseph B. Martin Conference Center at Harvard Medical School and will discuss the present and future of genetic therapies, pricing, reimbursement, the regulatory landscape as well as the treatment of patients with rare and ultra-rare genetic diseases.
Boston, MA (PRWEB) November 18, 2015
Prominent academics, leaders of the biopharmaceutical industry and key investors will gather on December 3rd at Genetic Rx, a networking conference of the Boston Biotech Conferences series. Genetic Rx will take place at the Joseph B. Martin Conference Center at Harvard Medical School and will discuss the present and future of genetic therapies, pricing, reimbursement, the regulatory landscape as well as the treatment of patients with rare and ultra-rare genetic diseases.
Speakers include Jörn Aldag (uniQure), George Church (Harvard Medical School), JC Gutierrez Ramos (Synlogic Therapeutics), Kathy High (Spark Therapeutics), Edward Kaye (Sarepta), Henrik Ørum (Roche), Mene Pangalos (Astrazeneca), Peter Saltonstall (NORD), Akshay Vaishnaw (Alnylam), James Wilson (University of Pennsylvania) among others. Leading investors from Bain Capital, Wellington Management Company, Pioneer Investments, Deerfield Partners, Fidelity Investments, Sofinnova Ventures, and Third Rock Ventures will also be in attendance.
“After attending Genetic Rx 2014, I’m looking forward to this year’s conference. As discoveries in a broad range of genetic therapies continue to develop, this conference is an excellent opportunity to catch up on progress and network with fellow thought leaders focused on the advancement of novel treatments and potential cures for multiple diseases” says Edward Lanphier, President and CEO of Sangamo BioSciences. “This intimate and off-the-record forum allows for high quality of exchanges and I am happy to be a part of it.”
Mark Pykett, President and CEO of Agilis Biotherapeutics says of the event: "The momentum of the field of genetic medicine is mirrored in the energy of BBC's Genetic Rx Conference. As the field confronts the consequences of increasing success, leaders from genetic medicine gather at the Genetic Rx Conference to exchange ideas and address the challenges and opportunities they face, and to collaborate on capturing the promise of this important, emerging area of medicine."
“During the last 100 years we treated signs and symptoms with small molecules. Antibodies and proteins have been part of the armamentarium to treat disease for the last 40 years. Now we are seeing the explosion of therapeutic opportunities in genetic medicines becoming a reality: gene therapy, gene editing, RNA silencing and synthetic biology. The intense and fast pace approach of the GeneticRX conference allows a deep dive to participants and helps to propel the field forward”, says Belen Carrillo-Rivas, Head of R&D Innovation Projects at Pfizer.
Genetic Rx is sponsored by Alexandria Real Estate Equities, H.C. Wainwright, Alnylam Pharmaceuticals, Agilis Biotherapeutics, Isis Pharmaceuticals, Mintz Levin, Pfizer and the BioPharma Executive Council.
For more information, please visit: http://bbbiotechconference.com/conference-agenda.php?id=35
ABOUT THE BOSTON BIOTECH CONFERENCES (BBC)

8:30-9:10 amGenetic Therapies: 2015 Milestones and 2016 GoalsKatherine High, Spark Therapeutics; Eirene Kontopoulos, Fidelity; Edward Lanphier, Sangamo BioSciences; Ken Mills, Regenxbio; Joshua Schimmer, Piper Jaffray
Moderator: Jon Kravetz, Mintz Levin

Jefferies..... Still Waiting


Tuesday, November 17, 2015

Zinc finger nuclease-based double-strand breaks attenuate malaria parasites and reveal rare microhomology-mediated end joining

For all author emails, please log on.
Genome Biology 2015, 16:249  doi:10.1186/s13059-015-0811-1
Published: 17 November 2015

 

Abstract

Background

Genome editing of malaria parasites is key to the generation of live attenuated parasites used in experimental vaccination approaches. DNA repair in Plasmodium generally occurs only through homologous recombination. This has been used to generate transgenic parasites that lack one to three genes, leading to developmental arrest in the liver and allowing the host to launch a protective immune response. While effective in principle, this approach is not safe for use in humans as single surviving parasites can still cause disease. Here we use zinc-finger nucleases to generate attenuated parasite lines lacking an entire chromosome arm, by a timed induction of a double-strand break. Rare surviving parasites also allow the investigation of unconventional DNA repair mechanisms in a rodent malaria parasite.

Results

A single, zinc-finger nuclease-induced DNA double-strand break results in the generation of attenuated parasite lines that show varying degrees of developmental arrest, protection efficacy in an immunisation regime and safety, depending on the timing of zinc-finger nuclease expression within the life cycle. We also identify DNA repair by microhomology-mediated end joining with as little as four base pairs, resulting in surviving parasites and thus breakthrough infections.

Conclusions

Malaria parasites can repair DNA double-strand breaks with surprisingly small mini-homology domains located across the break point. Timely expression of zinc-finger nucleases could be used to generate a new generation of attenuated parasite lines lacking hundreds of genes. 

Friday, November 13, 2015

Sigma-Aldrich CompoZr ZFN Ad

Published on Nov 13, 2015
CompoZr® Zinc Finger Nucleases are a class of engineered DNA-binding proteins that facilitate genome editing by creating a double-stranded break in DNA at a user-specified location. A double-stranded break is important for site-specific mutagenesis in that it stimulates the cell’s natural DNA-repair processes, namely homologous recombination and Non-Homologous End Joining (NHEJ). Using well-established and robust protocols, these cellular processes can be harnessed to generate precisely targeted in vivo genomic edits with targeted gene deletions (Knockouts). Here, Professor Peter Currie (Monash University, Australia) speaks about the application of this technology in studying neuromuscular disease

Open this link in a new window:
https://youtu.be/4idrX2_W1QA

Thursday, November 12, 2015

Lanphier Speaking at Life Science Disruptors December 10th

We’re one month away from our annual San Francisco Bay Area biotech forum on December 10, and we’re excited to share the lineup with you. This year, we’re focusing on the most disruptive biotechnologies to explore their future implications, not to mention the profound effects they’re triggering already in healthcare, business, and society.
One highlight will be gene editing, which has taken the world by storm for its ease of use—in the form of CRISPR-Cas9—its medical potential, and its ethical and environmental cautions. No company has brought gene editing closer to treating human disease than Sangamo Biosciences, so it’s apt that Sangamo CEO Edward Lanphier will kick off our event with an update on Sangamo and an overview of the field. Lanphier and colleagues wrote one of the two commentaries earlier this year that helped galvanize a huge public conversation about gene editing. At our Dec. 10 event, Lanphier and others will be just back from an international summit on the topic. We look forward to getting a report from the front.
To discuss the latest in CRISPR-Cas9, Andy May of Caribou Biosciences and Jenny Rooke, an early investor in Caribou, will discuss the evolution of the technology from Jennifer Doudna’s Berkeley lab to Caribou, which Doudna cofounded, to Intellia Therapeutics, which has exclusive use of Caribou’s platform for certain human therapeutics.
The human microbiome is another fast-moving field, with new revelations arriving seemingly every day about the connections between our health and the trillions of microbes in and on our bodies. How to harness that progress into a viable company is a tough nut to crack; only a few startups have emerged in the human therapeutics space. We’ll have two Bay Area CEOs, Peter DiLaura of Second Genome and Colleen Cutcliffe of Whole Biome, talk about their breakthroughs, insights, and frustrations.
To explain the pace of innovation and disruption, look no farther than digital technology, which is now intertwined with practically every aspect of the life sciences. Bioinformatics is changing healthcare, or threatening to change it, everywhere we look. Sequencing our DNA grows cheaper by the year, and researchers have just scratched the surface when it comes to sharing and analyzing it.
If it is indeed inevitable that we’ll all be sequenced and analyzed in the future, how can we ensure that we benefit from it? And it is OK that, along the way, companies and organizations profit from our data, as well? We’ll discuss those questions and more with Atul Butte, who leads UCSF’s new Institute for Computational Health Sciences as well as the University of California’s statewide precision medicine initiative; Randy Scott, CEO of San Francisco gene-testing company Invitae, which has ambitions to collect huge pools of genomic data for better health and new businesses; and veteran healthcare VC Nina Kjellson of Canaan Partners.
With the life sciences moving so fast, it’s easy to lose sight of the most important goal: our well-being. We’ll wrap up the Dec. 10 forum with a discussion dedicated to the ethical and social implications of all this disruption, which we hope spills over into more conversation at the post-event reception. If you want to be part of that conversation, we hope you can join us next month (register here), and stay tuned for news about a few more speakers to round out the agenda.
Confirmed Speakers
Atul Butte, Director, Institute of Computational Health Sciences, UCSF
Colleen Cutcliffe, CEO, Whole Biome
Peter DiLaura, CEO, Second Genome
Nina Kjellson, General Partner, Canaan Partners
Edward Lanphier, CEO, Sangamo Biosciences
Andy May, CSO, Caribou Biosciences
Jenny Rooke, Managing Director, Five Prime Ventures
Randy Scott, CEO, Invitae

Wednesday, November 11, 2015

Local Time for Jefferies Conference 11/18/2015

This presentation will take place at 5:40 AM EST Wednesday November 18th.

Sangamo BioSciences Announces Presentation At The Jefferies Autumn 2015 Global Healthcare Conference

RICHMOND, Calif., Nov. 11, 2015 /PRNewswire/ -- Sangamo BioSciences, Inc. (NASDAQ: SGMO), a leader in therapeutic genome editing, announced today that Geoffrey Nichol, M.B., Ch.B., Sangamo's executive vice president, research and development, will provide an update on the progress of Sangamo's ZFP Therapeutic® development programs and an overview of the company's business strategy at 10:40am GMT on Wednesday, November 18, 2015, at the Jefferies Autumn 2015 Global Healthcare Conference. The conference is being held in London, UK.
Sangamo BioSciences, Inc.
The presentation will be webcast live and may be accessed via a link on the Sangamo BioSciences website in the Investor Relations section under Events and Presentations. The webcast will be archived on the Sangamo website for two weeks after the event.
About SangamoSangamo BioSciences, Inc. is focused on Engineering Genetic CuresTM for monogenic and infectious diseases by deploying its novel DNA-binding protein technology platform in therapeutic genome editing and gene regulation. The Company has a Phase 2 clinical program to evaluate the safety and efficacy of novel ZFP Therapeutics® for the treatment of HIV/AIDS (SB-728). Sangamo's other therapeutic programs are focused on monogenic and rare diseases. The Company has 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 also 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, November 10, 2015

Sangamo Biosciences (SGMO) Short Interest - Down 9% from 10/15/15

Sangamo Short Interest (SGMO)
DATE                                   SHORT INTEREST
10/30/15                              11,668,561 (9.38 days to cover)
10/15/15                              12,821,197    
9/30/15                                12,619,596
9/15/15                                10,882,795
8/31/15                                10,150,146
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

Monday, November 9, 2015

Sangamo BioSciences Announces Publication Of Improved Method For Efficient Targeted Integration In Hematopoietic Stem Cells And T-cells

RICHMOND, Calif., Nov. 9, 2015 /PRNewswire/ -- Sangamo BioSciences, Inc. (Nasdaq: SGMO), a leader in therapeutic genome editing, announced today the publication in Nature Biotechnology of data demonstrating efficient zinc finger nuclease (ZFN)-mediated, targeted gene insertion in human hematopoietic stem and progenitor cells (HSPCs). In addition, data published earlier this month in Nucleic Acids Research, demonstrate that similar efficiencies can be obtained in primary human T-cells. Sangamo's genome editing method provides a precisely targeted and therefore potentially safer approach than traditional randomly integrative gene therapies, such as lentiviral vectors, for the treatment of human diseases of the blood and immune system, and the generation of T-cell based cancer immunotherapies.
Sangamo BioSciences, Inc.
"These studies present new and substantial data affirming that Sangamo's ZFN genome-editing platform provides precise, robust targeted insertion of therapeutic genes, in contrast to conventional integrating vector approaches, such as lentiviral vectors that insert genes randomly," stated Edward Lanphier, Sangamo's president, and chief executive officer. "Our extensive clinical experience and a substantial body of research, documents the accuracy and efficiency of our ZFN technology, and positions it as a more versatile and potentially safer therapeutic strategy than other gene therapy and genome editing approaches. Our technology also provides an alternative manufacturing platform for cell therapies."
Researchers capitalized on the fact that ZFN-mediated genome editing requires only transient expression of the ZFNs to effect a permanent change in the genome.  They used messenger RNA and electroporation to deliver the ZFNs and a non-integrating vector, an adeno-associated virus (AAV) serotype 6, to provide a therapeutic DNA sequence, a so-called "donor template", to  HSPCs and CD4 and CD8 T-cells. The "donor template" can encode an entire gene that integrates at the targeted site of ZFN action.  This delivery approach achieved efficiencies of gene transfer and targeted integration of the therapeutic DNA sequence, particularly in more primitive longer-lasting stem cells and T-cells, to levels that can be used therapeutically to potentially treat a range of monogenic diseases.
"Efficient targeted integration of a therapeutic gene into both hematopoietic stem cells and T-cells represents a major step forward in the quest for more precise and safer gene-based therapies," stated Michael Holmes, Ph.D., Sangamo's vice president, research, and a corresponding author on both papers. "Importantly, in our hematopoietic stem cell work with our scientific collaborators at USC, we successfully modified a class of the most pluripotent stem cells, leading to long-term production of ZFN-modified cells across all blood cell lineages following transplantation into immune deficient mice. Our results in these two studies provide a robust strategy for additional therapeutic applications of ZFN-mediated genome editing in HSPCs and T-cells."
The stem cell study, "Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors" was carried out in collaboration with scientists in the laboratory of Paula Cannon, Ph.D., professor of molecular microbiology and immunology, pediatrics, biochemistry and molecular biology, and stem cell biology and regenerative medicine at the University of Southern California, and was published as an Advance Online Publication in Nature Biotechnology.http://www.nature.com/articles/doi:10.1038/nbt.3408

The T-cell study, "Highly efficient homology-driven genome editing in human T-cells by combining zinc finger nuclease mRNA and AAV6 donor delivery" was published as Advance Access in Nucleic Acids Research on November 2, 2015.
http://nar.oxfordjournals.org/content/early/2015/11/01/nar.gkv1121.full.pdf+html

Sunday, November 8, 2015

Zinc Finger Sensor Could Detect Viruses, Kill Cancer Cells

Awesome application here.
This system can be customized to detect any DNA sequence in a mammalian cell and then trigger a desired response, including killing cancer cells or cells infected with a virus, the researchers say.
"There is a range of applications for which this could be important," says James Collins, the Termeer Professor of Medical Engineering and Science in MIT's Department of Biological Engineering and Institute of Medical Engineering and Science (IMES). "This allows you to readily design constructs that enable a programmed cell to both detect DNA and act on that detection, with a report system and/or a respond system."

cancer cells
Collins is the senior author of a Sept. 21 Nature Methods paper describing the technology, which is based on a type of DNA-binding proteins known as zinc fingers. These proteins can be designed to recognize any DNA sequence.
"The technologies are out there to engineer proteins to bind to virtually any DNA sequence that you want," says Shimyn Slomovic, an IMES postdoc and the paper's lead author. "This is used in many ways, but not so much for detection. We felt that there was a lot of potential in harnessing this designable DNA-binding technology for detection."
Sense and respond
To create their new system, the researchers needed to link zinc fingers' DNA-binding capability with a consequence -- either turning on a fluorescent protein to reveal that the target DNA is present or generating another type of action inside the cell.
The researchers achieved this by exploiting a type of protein known as an "intein" -- a short protein that can be inserted into a larger protein, splitting it into two pieces. The split protein pieces, known as "exteins," only become functional once the intein removes itself while rejoining the two halves.
Collins and Slomovic decided to divide an intein in two and then attach each portion to a split extein half and a zinc finger protein. The zinc finger proteins are engineered to recognize adjacent DNA sequences within the targeted gene, so if they both find their sequences, the inteins line up and are then cut out, allowing the extein halves to rejoin and form a functional protein. The extein protein is a transcription factor designed to turn on any gene the researchers want.
In this paper, they linked green fluorescent protein (GFP) production to the zinc fingers' recognition of a DNA sequence from an adenovirus, so that any cell infected with this virus would glow green.
This approach could be used not only to reveal infected cells, but also to kill them. To achieve this, the researchers could program the system to produce proteins that alert immune cells to fight the infection, instead of GFP.
"Since this is modular, you can potentially evoke any response that you want," Slomovic says. "You could program the cell to kill itself, or to secrete proteins that would allow the immune system to identify it as an enemy cell so the immune system would take care of it."
The MIT researchers also deployed this system to kill cells by linking detection of the DNA target to production of an enzyme called NTR. This enzyme activates a harmless drug precursor called CB 1954, which the researchers added to the petri dish where the cells were growing. When activated by NTR, CB 1954 kills the cells.
Future versions of the system could be designed to bind to DNA sequences found in cancerous genes and then produce transcription factors that would activate the cells' own programmed cell death pathways.

cancer cells
Research tool
The researchers are now adapting this system to detect latent HIV proviruses, which remain dormant in some infected cells even after treatment. Learning more about such viruses could help scientists find ways to permanently eliminate them.
"Latent HIV provirus is pretty much the final barrier to curing AIDS, which currently is incurable simply because the provirus sequence is there, dormant, and there aren't any ways to eradicate it," Slomovic says.
While treating diseases using this system is likely many years away, it could be used much sooner as a research tool, Collins says. For example, scientists could use it to test whether genetic material has been successfully delivered to cells that scientists are trying to genetically alter. Cells that did not receive the new gene could be induced to undergo cell death, creating a pure population of the desired cells.

It could also be used to study chromosomal inversions and transpositions that occur in cancer cells, or to study the 3-D structure of normal chromosomes by testing whether two genes located far from each other on a chromosome fold in such a way that they end up next to each other, the researchers say.
http://www.agriarticle.com/2015/11/sensor-could-detect-viruses-kill-cancer.html

Friday, November 6, 2015

Press Reports

The Cellectis news from yesterday and subsequent Sangamo mention has Genomeweb.com placing SGMO is some heady company.

(snip)
Still, the Journal notes that a number of companies like Pfizer, AstraZeneca, Novartis, and Sangamo BioSciences and academic researchers are exploring gene editing, including using zinc finger nucleases and CRISPR.

https://www.genomeweb.com/scan/gene-editing-remission


Seeking Alpha Article that Relates to Sangamo HIV Manufacturing program


http://seekingalpha.com/article/3651256-victory-in-the-car-t-race-may-be-pyrrhic-juno-the-biggest-loser-cellectis-too-late-for-the-spoils?page=2


Summary

"T cell manufacturing is robust and dependable...A handful of academic centers have successfully established manufacturing procedures that have proven to be reproducible and dependable" (Sadelain, 2015).
"The field ... has a dearth of data as to whether centralized manufacturing of patient-derived CAR T cells is broadly feasible and does not result in ruinous costs..." (Cooper, 2015).
"Amateurs talk about tactics, but professionals study logistics" applies to military ops but also to clinical ops being transitioned to commercial, centralized manufacturing.
CEOs who think they can take this brilliant science commercial without a road map through the daunting logistics prove themselves amateurs.
Avoid them all until you at least see a road map, especially the biotechs with largest market cap. Consider small-cap contract manufacturers who demonstrate expertise in logistics.
A victory is pyrrhic when even the victor counts the cost too high, and that is how the crowded race for autologous CAR-T cells could end.
Genetic engineering of CARs into T cells is one of the most complex processes known to modern medicine. Central as opposed to point-of-care manufacturing of CAR-T cells for autologous transfusion adds logistical complexity and treatment risk to an intense therapy that already carries great risk to a vulnerable patient group, those with cancer refractory to conventional therapies.
Experts in commercial scale manufacturing warn against the scaling up of immune cell production without a transition period of close collaboration between manufacturer and the academic facilities in which patient-specific, genetically engineered T cell therapies originate: the science behind CAR-T is brilliant but scaling up central manufacturing requires a vastly different skill set (see Preti, 2015 and O'Donnell, 2015).

No one should know more about failed cell manufacturing models than Juno (NASDAQ:JUNO) CEO Hans Bishop who was EVP and COO at Dendreon (OTCPK:DNDNQ) from January 2010 to September 2011. Dendreon attempted to manufacture its own cellular therapeutic. DNDNQ entered Chapter 11 in November 2014.
Dendreon's PROVENGE® sipuleucel-T is a patient-specific but centrally manufactured immunocellular therapy for autologous transfusion in prostate cancer. Despite a favorable impact on overall survival (Kantoff, 2010), the medical community did not think the benefits of Sipuleucel-T justified the $93,000 price tag, sales were disappointing, production costs high - a Dendreon website video shows why: logistics are a potential nightmare.
Harvesting cells from a patient, shipping them to a manufacturer for immunological manipulation, and sending them back for transfusion into the same (we hope) patient is not only risky business, it is risky medicine: Dendreon had difficulty maintaining quality control amidst the complexities of manufacture and distribution. I am not saying that cancer therapy with CAR-T will be as unimpressive as Provenge has been. T cell therapies are poised to become part of the standard-of-care treatments for patients with cancer (Wang and Rivière, 2015). But manufacturing genetically engineered CAR-T cells is longer (2 weeks) and more complex than for Provenge (not genetically modified and takes 3 days) by an order of magnitude.
In a white paper, titled "Manufacturing Cell Therapies: Development Strategies for Commercial Vision", authors Richard Grant and Brian Hampson, VP of Manufacturing Development and Engineering for Progenitor Cell Therapy ((NASDAQ:CLBS)), offer a fairly detailed road map which I have summarized:
  1. Client (e.g. Juno) creates a Quality Target Product Profile for future labeling that follows FDA guidelines,
  2. Long-term collaboration begins with manufacturing system developer (e.g. PCT/CLBS) visiting client to observe processes currently in use
  3. Client formally communicates to manufacturer expectations for numbers of patients to be treated, costs of products, handling and shipping of products, and other requirements
  4. client gains a clearer strategic vision to communicate to investors and others
  5. CMO (e.g. Invetech/CLBS) builds prototype for small-scale production, produces cells for quality testing by client
I searched JUNO's latest 10K, 10Q and earnings conference calls for evidence that Hans Bishop had some learned some important lessons from the DNDNQ failure, and communicated to investors (as in step 4 above) his road map for success. This is what I do see:
"We've transitioned from an academic process to a commercially scalable one."
How?
"The company entered into a lease for an approximately 68,000 square foot manufacturing facility in Bothell, Washington, which lease commenced in March 2015."
Already?
"We plan to complement the use of one or more CMOs by establishing our own cGMP manufacturing facility to be brought on-line after the first CMO. As described...we have entered into a ten-year lease for a facility that we plan to remodel to support our clinical and commercial manufacturing activities."
Headlong into uncharted waters is what that sounds like to me. Meanwhile, Novartis has already moved into the NJ cell manufacturing plant that Mr. Bishop equipped, and his successor at DNDNQ had to liquidate.
Novartis is too big a company for me to study and present, but I took note of its collaboration with U Penn when the CART19 story made big news in NEJM (Porter, 2011). NVS surely has the liquidity to stay in this CAR-T race longer than JUNO could stay solvent without the aid of Celgene (NASDAQ:CELG). And what an advantage having a ready-made (courtesy of Hans Bishop's Dendreon) cell manufacturing plant when all JUNO has to show its investors is a lease agreement!
David Chang, Kite's (NASDAQ:KITE) chief medical officer, told a Reuters reporter that Kite will minimize overhead and manufacturing costs by building individual modules that can each handle a patient's cells and then will then "build out" with more factory modules as demand increases. That sounds like a good idea, and I think Kite's plan for a commercial manufacturing facility adjacent to Los Angeles International Airport also shows they are logistically minded. Meanwhile, KITE will leave responsibility for cell manufacture to Progenitor Cell Therapy, a subsidiary of Caladrius Biosciences. CLBS bioprocesses and manufactures but does not genetically engineer cells. CLBS operating expenses exclusive of R&D rose faster than revenues in FY 2013 and 2014, and its own cell therapy programs are not doing well. President of this company, Dr. Robert Preti, has posted an article in DDNews, September 2015, provocatively entitled "Building a problem or a solution?" Dr. Preti articulates concerns about the state of his industry and the direction it must go. KITE's dependence on this tiny company with only $21M in PP&E is of some concern - CLBS will need capital to build capacity, and that will be more difficult with its stock price way down this year. Perhaps CLBS will be acquired.
There are some sub-$1B market cap companies that are engineering features into T cells to make them safer and more effective, but for whom logistics is not yet a critical concern.
Bellicum (NASDAQ:BLCM) has a variety of T lymphocyte and other immune cell platforms. Its most advanced product in phases 1 and 2 is BPX-501, an allogeneic, partially matched T cell removed from donor bone marrow prior to transplantation, engineered with a suicide switch, then used to reconstitute the immune system of the recipient of the transplant. Genetically adding suicide switches (Stasi, 2011; Zhou, 2014) allow for these cells to be shut down if they attack the patients own healthy tissues. Bellicum does research in partnership with scientists at Baylor. BPX-201 is a preclinical cancer vaccine that uses a prostate cancer patient's own dendritic white cells for autologous infusion after genetic engineering of a suicide switch and expression of a prostate cancer antigen that will stimulate and immune attack on the cancer. BPX-601 and -401 are preclinical CAR-T cells with suicide switches. Both are for autologous transfusion, so centralized manufacturing for wide distribution will face the same logistical issues.
Adaptimmune (NASDAQ:ADAP) plans to build its manufacturing plant in The Navy Yard in Philadelphia for use in 2016. Ships are known to move slowly, so logistics was probably not a consideration in selecting that location. The ADAP platform is entirely autologous and not off-the-shelf. ADAP has 8 phase 1 or 1/2 trials registered at ClincialTrials.gov, all using T cells with genetically engineered TCR, not CARs. One of these trials which uses anti-NY-ESO-1 TCR for synovial sarcoma is in collaboration NCI. GlaxoSmithKline (NYSE:GSK) is not named as co-sponsor on any of the registered trials but has options on the T cells with TCR engineered against NY-ESO. ADAP form 10-K indicates (page 56) that current CMO is Progenitor Cell Therapy.
Pity that Cellectis (NASDAQ:CLLS) (OTCPK:CMVLF), the only company developing a universal, off-the shelf CAR T product that would be logistically simpler to manufacture and distribute, has registered nary a single clinical T cell trial among the numerous already registered at ClinicalTrials.gov (see Table below). Not only that, but in reviewing a large amount of the literature on the subject, I came across not a single paper authored by Cellectis scientists, and no mention of them by peers in the field. When I first discovered Cellectis in a review of the 2014 ASH abstracts, I thought I had discovered a gem - and it may indeed be a gem. But Cellectis promised a clinical trial in 2015 which as of November has not started or been further discussed. Lead product is another anti-CD19 CAR, but the race against CD19-bearing B-cell malignancies is getting crowded, and Cellectis may have difficulty finding a market by the time its CRO has gotten UCART19 through trials and approved. The "barrier to entry" into the universal donor CAR-T race is not high: it requires that the lymphocytes native TCR be knocked out, and that technology is currently being widely adopted by others.
Conclusions.
  • Central bioprocessing and manufacturing of genetically engineered T cells for autologous transfusion is an unproven business model. Those who try it should learn from Dendreon's failure with a much simpler, genetically un-modified cell therapy product.
  • Even industry experts admit (Kaiser, 2015):
"It is conceivable to see a future where gene-modified T cells are manufactured at the point-of-care in a facility in close proximity, associated with, or at the hospital."
...that's quite an understatement
  • The cost of manufacturing an off-the-shelf, allogeneic rather than autologous, cancer-fighting T cell will be less ruinous.
  • Beyond the economic fundamentals, identification the most important and safe molecular targets, and the ability to edit T cell genes translate competitive edge (June, 2015).
Recommendations:
  • $5B JUNO is a STRONG SELL
  • $3B KITE is a SELL
  • $0.9B Cellectis is very late to enter the race particularly for a CD19 CAR-T, but has a platform for allogeneic use. Speculative HOLD.
  • $0.8B market cap Adaptimmune, new to the market in May, was noticed by another SA author in September and is worth following.
  • $0.3B BLCM is worth watching
  • $68 Million market microcap CLBS is also worth watching. CLBS does have the skill set for manufacturing, and its stock will soar if BLCM or ADAP go commercial before they are fully cGMP-capable.

Thursday, November 5, 2015

Sangamo BioSciences To Present Data From ZFP Therapeutic Programs At Annual Meeting Of The American Society Of Hematology

Five Presentations Include Non-Human Primate Data from Sangamo's In Vivo Protein Replacement Platform™ Approach for Hemophilia B, and Data from ZFN-Mediated Genome Editing Approach for Hemoglobinopathies

RICHMOND, Calif., Nov. 5, 2015 /PRNewswire/ -- Sangamo BioSciences, Inc. (Nasdaq: SGMO), a leader in therapeutic genome editing, announced today that non-human primate data from its proprietary In Vivo Protein Replacement Platform (IVPRP™) program for hemophilia B, and data from its ZFP Therapeutic® hemoglobinopathy programs in collaboration with Biogen, will be presented at the 57th Annual Meeting of the American Society of Hematology (ASH). The 2015 ASH meeting will be held in Orlando, FL from December 5-8, 2015.
Sangamo BioSciences, Inc.
"Our presentations at this year's ASH meeting highlight the breadth of our highly specific genome editing platform in both in vivo and ex vivo therapeutic applications," said Edward Lanphier, Sangamo's president and chief executive officer. "Data from non-human primates generated in our Factor IX program for hemophilia B demonstrate the potential of our IVPRP strategy to produce clinically beneficial levels of therapeutic protein from a single treatment. We, and our collaborators at Biogen, will also present data from our hemoglobinopathy programs in beta-thalassemia and sickle cell disease, which use an efficient ZFN-mediated knockout approach in hematopoietic stem cells to elevate functional globin to provide a potentially life-long therapeutic effect." 
Sangamo's hemophilia B program is the first therapeutic application of its IVPRP strategy, an in vivo targeted integration strategy that can be leveraged across multiple monogenic diseases that are currently treated using protein or enzyme replacement therapy. Sangamo remains on track to file Investigational New Drug (IND) applications for hemophilia B (Factor IX) and Hurler syndrome (MPS I) by the end of 2015, and several more IND applications, including hemophilia A, Hunter syndrome (MPS II), Gaucher disease and other lysosomal storage disorders in 2016.
Sangamo is collaborating with Biogen to develop a ZFP Therapeutic approach to beta-thalassemia and sickle cell disease (SCD) that replaces deficient expression of the mutant, disease-causing form of beta-globin with expression of functional fetal globin. The companies expect to file IND applications for beta-thalassemia in the first half of 2016 and for SCD in the second half of 2016.
The following presentations are scheduled at the ASH Meeting sessions:
IVPRP
  • ZFN-Mediated Gene Targeting at the Albumin Locus in Liver Results in Therapeutic Levels of Human FIX in Mice and Non-Human Primates - Abstract #200
    Session: 801. Gene Therapy and Transfer: Gene Therapy for Hemoglobinopathies and Inherited Bleeding DisordersOral Presentation - Sunday, December 6, 2015: 7:45 AM
    Presenter - Michael C. Holmes, Ph.D., Sangamo BioSciences
Hemoglobinopathies
  • Genome Editing of the Bcl11A Erythroid Specific Enhancer in Bone Marrow Derived Hematopoietic Stem and Progenitor Cells for the Treatment of Sickle Cell Disease - Abstract #203
    Session: 801. Gene Therapy and Transfer: Gene Therapy for Hemoglobinopathies and Inherited Bleeding DisordersOral Presentation - Sunday, December 6, 2015: 8:30 AM
    Presenter - Siyuan Tan, Ph.D., Biogen
  • Clinical-Scale Genome Editing of the Human BCL11A Erythroid Enhancer for Treatment of the Hemoglobinopathies - Abstract #204
    Session: 801. Gene Therapy and Transfer: Gene Therapy for Hemoglobinopathies and Inherited Bleeding DisordersOral Presentation - Sunday, December 6, 2015: 8:45 AM
    Presenter - Fyodor, D. Urnov, Ph.D., Sangamo BioSciences
  • Clonal Analysis of Human Bone Marrow CD34+ Cells Edited by BCL11A-Targeting ZFNs Reveals Clinically Relevant Levels of Gamma Globin Expression in Edited Erythroid Cells - Abstract #3234Session: 801. Gene Therapy and Transfer: Poster IIPoster Session - Sunday, December 6, 2015: 6:00 - 8:00 PM
    Presenter - Kai-Hsin Chang, Ph.D., Biogen
Other Applications
  • In Vivo Genome Editing in Neonatal Mouse Liver Preferentially Utilizes Homology Directed Repair - Abstract #4422Session: 801. Gene Therapy and Transfer: Poster IIIPoster Session - Monday, December 7, 2015: 6:00 - 8:00 PM
    Presenter - Xavier M. Anguela, Ph.D., Children's Hospital of Philadelphia
All abstracts for the ASH meeting are available online at 2015 ASH Annual Meeting Abstracts.
About Sangamo's IVPRPThe IVPRP approach makes use of the albumin gene locus, a highly expressing and liver-specific genomic "safe-harbor site", that can be edited with zinc finger nucleases (ZFNs) to accept and express any therapeutic gene. The platform enables the patient's liver to permanently produce therapeutic levels of a corrective protein product such as factor VIII or IX to treat hemophilia, or replacement enzymes to treat lysosomal storage disorders. With such a large capacity for protein production (approximately 15g/day of albumin), which is in excess of the body's requirements, targeting and co-opting only a very small percentage of the albumin gene's capacity is sufficient to produce the needed replacement protein at therapeutically relevant levels with no significant effect on albumin production.
About Sangamo's ZFP Therapeutic Approach to HemoglobinopathiesSangamo's proprietary ZFN genome editing technology enables the correction of SCD and beta-thalassemia. Both diseases manifest after birth, when patients switch from producing functional fetal gamma-globin to a mutant form of adult beta-globin, which causes their condition. Naturally occurring increased levels of therapeutic fetal hemoglobin have been shown to reduce the severity of both SCD and beta-thalassemia disorders in adulthood. In hematopoietic stem and progenitor cells (HSPCs), Sangamo's genome editing technology can be used to precisely disrupt a key DNA sequence that acts as a powerful tissue and developmental stage "Enhancer" of BCL11A expression. BCL11A is a key transcriptional regulator of the switch from fetal to adult globin production. Knockout of the Enhancer results in the disruption of that switch leading to elevation of fetal globin and reduction in the expression of adult globin.
A bone marrow transplant (BMT) of HSPCs from a "matched" related donor (allogeneic BMT) is curative for both diseases. However, this therapy is limited by the scarcity of matched donors and the significant risk of graft versus host disease (GvHD) after transplantation of the foreign cells. By performing genome editing in HSPCs that are isolated from and subsequently returned to the same patient, an autologous HSPC transplant, Sangamo's approach eliminates both the need for a matched donor and the risk of acute and chronic GvHD. The ultimate goal of this approach is to develop a one-time, life-long treatment for SCD and beta-thalassemia.
About SangamoSangamo BioSciences, Inc. is focused on Engineering Genetic CuresTM for monogenic and infectious diseases by deploying its novel DNA-binding protein technology platform in therapeutic genome editing and gene regulation. The Company has a Phase 2 clinical program to evaluate the safety and efficacy of novel ZFP Therapeutics® for the treatment of HIV/AIDS (SB-728). Sangamo's other therapeutic programs are focused on monogenic and rare diseases. The Company has 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 also 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.
ZFP Therapeutic® is a registered trademark of Sangamo BioSciences, Inc.

ASH Abstracts for Sangamo 2015

200 ZFN-Mediated Gene Targeting at the Albumin Locus in Liver Results in Therapeutic Levels of Human FIX in Mice and Non-Human Primates

Hemophilia is an attractive target for gene therapy, since activity levels as low as 1% to 2% of normal are beneficial and levels of ~5% prevent spontaneous bleeding. Our goal was to provide a single treatment that permanently enables hepatic production of therapeutic levels of hFIX activity to decrease or potentially eliminate the need for prophylactic treatment in hemophilia B patients. We performed targeted in vivo genome editing using 1) two zinc finger nucleases (ZFNs) targeting intron 1 of the albumin locus, and 2) a human F9 donor template construct. The ZFNs and donor template are encoded on separate hepatotropic adeno-associated virus serotype 2/6 (AAV2/6) vectors injected intravenously, resulting in targeted insertion of a corrected copy of the hF9 gene into the albumin locus in a proportion of liver hepatocytes. The albumin locus was selected as a “safe harbor” as production of this most abundant plasma protein exceeds 10 g/day, and moderate reductions in those levels are well-tolerated.  These genome edited hepatocytes produce normal hFIX in therapeutic quantities, rather than albumin, driven by the highly active albumin enhancer/promoter, to treat hemophilia B; the genetic modification is expected to be sustained even in the face of hepatocyte turnover, making this approach attractive for treating young children with hemophilia before the appearance of significant organ damage. Transformed and primary human hepatocytes transduced in vitro with AAV2/6 encoding human albumin ZFNs and a promoterless hF9 transgene were shown to secrete hFIX. Extensive molecular analyses demonstrated that this was due to targeted integration of the hF9 transgene at the albumin locus and splicing of this gene into the albumin transcript. By employing AAV2/6 delivery of murine-specific ZFNs in vivo, stable levels of hFIX were observed in blood of mice injected with the albumin ZFNs and hF9 transgene donor. C57BL/6 mice were administered vehicle (n=20) or AAV2/6 vectors (n=25) encoding mouse surrogate reagents at 1.0 x1013 vector genome (vg)/kg via tail vein injection. ELISA analysis of plasma hFIX in the treated mice showed peak levels of 50-1053 ng/mL that were sustained for the duration of the 6-month study. Analysis of FIX activity from mouse plasma confirmed bioactivity commensurate with expression levels. Next, we report the feasibility of this approach in non-human primates (NHPs), showing that a single intravenous co-infusion of AAV2/6 vectors encoding the NHP targeted albumin-specific ZFNs and a human F9 donor at 1.2x1013 vg/kg (n=5/group) resulted in >50 ng/mL (>1% of normal) in this large animal model. The use of higher AAV2/6 doses (up to 1.5x1014 vg/kg) yielded plasma hFIX levels up to 1000 ng/ml (or 20% of normal) in several animals and up to 2000 ng/ml (or 50% of normal) in a single animal, for the duration of the study (3 months). The treatment was well tolerated in mice and NHPs, with no significant toxicological findings related to AAV2/6 ZFN + donor treatment in either species at therapeutic doses. Together, these data support a clinical trial to determine if a single co-administration of ZFN and donor AAV vectors is sufficient to enable therapeutic and potentially lifelong production of the clotting factor for the treatment of Hemophilia B.


204 Clinical-Scale Genome Editing of the Human BCL11A Erythroid Enhancer for Treatment of the Hemoglobinopathies

We describe here a fundamentally novel way to develop a disease therapeutic: combining genome-wide association studies (GWAS) with targeted genome editing to create, in a clinically compliant setting, a disease-ameliorating genotype in the patient’s own cells. In β-thalassemia, elevated levels of fetal hemoglobin (HbF) lessen or eliminate disease symptoms, thus making a reversal of HbF silencing in patients an appealing therapeutic strategy. Loss-of-function variants in the erythroid-specific enhancer of the fetal globin repressor, BCL11A, elevate HbF; rare individuals carrying a monoallelic knockout of BCL11A exhibit no known hematologic abnormality and up to 30% circulating HbF. We previously reported de novo knockout of BCL11A using targeted genome editing with engineered zinc finger nucleases (ZFNs) yielding up to 40% HbF in erythroid progeny of edited human CD34 cells in vitro. We now find that the targeted ablation of a single, specific GATAA motif in the BCL11A intronic enhancer does not affect in vitro erythroid differentiation, but reproducibly (n=6) activates fetal globin transcription in erythroid progeny of modified CD34 cells; importantly, at similar levels of on-target marking in CD34+ cells, these effects on fetal globin mRNA are comparable to those resulting from ZFN-driven coding knockout of BCL11A itself. We demonstrate reproducible (n=8), high-efficiency (up to 82%; average, 69%) ZFN-driven marking at the enhancer in peripheral blood mobilized human CD34 cells at clinical production scale (>1e8 cells) in a GMP-compliant setting for which we use a clinical-grade electroporation device to deliver nuclease-encoding transcribed mRNA ex vivo. Using erythroid colony assay genotyping we find that up to 70% of the cells in the resulting population are biallelically modified at the target locus, while ~10% remain wild-type, and find comparably high levels of marking in research-scale preparations of CD34 cells from patients with β-thalassemia. We observe robust long-term (18-24 week) engraftment and multilineage differentiation of genome-edited cells in immunodeficient mice, similar to control cells, and equivalent modification at the targeted enhancer locus  at all timepoints in both differentiated (CD19+, CD3+, CD33+) and more primitive progenitor (CD34+CD38low) cells of human origin purified from bone marrow of long-term-engrafted animals. Our findings support clinical development of enhancer editing as a treatment of the β hemoglobinopathies with autologous hematopoietic stem cell transplant.

 

3234 Clonal Analysis of Human Bone Marrow CD34+ Cells Edited By BCL11A-Targeting Zinc Finger Nucleases Reveals Clinically Relevant Levels of Fetal Globin Expression in Edited Erythroid Progeny

Sickle cell disease (SCD) is one of the most common inherited blood disorders and is caused by a mutation at the adult beta globin gene resulting in substitution of valine for glutamic acid at position 6 in the encoded protein. While SCD can be cured by hematopoietic stem cell transplant (HSCT), complete donor chimerism is not required to achieve clinical benefits. Stable mixed chimerism of 10-15% in bone marrow or peripheral blood nucleated cells with >70% donor-derived RBCs has been reported to achieve transfusion independence and a symptom-free state in a SCD patient. It has also been proposed that SCD can be treated by reactivating developmentally silenced fetal gamma globin to form fetal hemoglobin (alpha2gamma2, HbF), which inhibits polymerization of HbS. The effect of HbF is predicted to be maximal when HbF content per cell exceeds 10 pg (~30% of total Hb). Furthermore, pathology is prevented when protective F cells (>30% HbF per cell) constitute >70% of total RBCs. We hypothesize that in a gene therapy setting, if >15% of SCD patients’ autologous HSCs are programmed to produce protective F cells during erythropoiesis, it will translate into >70% protective F cells in circulation and provide significant alleviation of clinical symptoms. Genome wide association studies have identified BCL11A as a major modifier of HbF levels. Subsequent studies have shown that BCL11A plays a critical role in the fetal to adult globin developmental switch and in repressing fetal globin expression in adult erythroid cells. Conditional inactivation of BCL11A in adult erythroid cells leads to high levels of pan-cellular fetal globin expression and correction of hematologic and pathologic defects in a humanized SCD mouse model. Previously, we have reported that zinc finger nucleases (ZFNs) targeting BCL11A either in the coding region or the GATAA motif in the erythroid-specific enhancer efficiently disrupt the BCL11A locus in human primary CD34+ cells following electroporation of ZFN-encoding mRNA. Elevated fetal globin expression in bulk erythroid cultures was observed following disruption. To determine what percentage of HSPCs have been modified and whether the HbF/F cell content has reached the hypothesized therapeutic level, we analyzed erythroid cells clonally derived from ZFN-transfected CD34+ cells. Genotype of each clonal culture was determined by deep sequencing and globin production was analyzed by a highly sensitive UPLC method. We found that up to 80% of the BFU-Es had both BCL11A alleles edited, half of which had KO/KO alleles (either out of frame mutations for coding region or elimination of the GATAA motif in the enhancer). BCL11A coding KO/KO cells expressed on average 79.1% ± 12.2% fetal globin (Mean ± SD) whereas GATAA motif enhancer region KO/KO cells expressed approximately 48.4% ± 14.1% fetal globin, in comparison with 14.5% ± 9.6% in WT/WT cells .  These levels of fetal globin should be sufficiently high to confer protection against HbS polymerization in sickle cells. WT/KO cells in both coding and enhancer editing experiments showed an intermediate phenotype with fetal globin averaging 26.9%± 9.9% and 25.79% ± 12.6%, respectively. Interestingly, when background (WT/WT) fetal globin level was subtracted, the fetal globin levels in WT/KO cells are comparable to those observed in patients with BCL11A haploinsufficiency, which average 14.6%± 10.3%. Together, our data demonstrate that genome editing of BCL11A using highly efficient ZFNs can lead to clinically relevant levels of fetal globin expression in KO/KO erythroid cells. If the frequency of KO/KO BFU-Es we observed in vitro reflects the frequency of KO/KO HSCs in bone marrow after autologous transplantation, genome editing of BCL11A has the potential to provide significant clinical benefit for patients with SCD.


4422 In Vivo Genome Editing in Neonatal Mouse Liver Preferentially Utilizes Homology Directed Repair

Genome editing has the potential to provide long-term therapeutic gene expression in vivo. We have previously demonstrated efficient editing in a mouse model of hemophilia B through liver-directed adeno-associated viral vector (AAV) delivery of a zinc finger nuclease (ZFN) pair and a corrective donor. We determined that homology is not necessary to achieve efficient levels of genome editing in adult mice, consistent with the fact that quiescent cells, including adult hepatocytes, are not thought to be amenable to homology directed repair (HDR). As a consequence of the donor containing a splice acceptor, both HDR and homology independent vector integration are capable of driving human factor 9 (hF.IX) expression. In this study we sought to determine whether hF.IX expression in mice treated as neonates, undergoing substantial hepatocyte proliferation, is predominantly the result of HDR or homology independent genome editing. Provided the efficacy is not substantially reduced, an HDR dependent approach would impose additional constraints on targeting.

Treatment of neonatal hF9mut mice (harboring the ZFN target site) with 1x1011 vg AAV8-ZFN and 5x1011 vg AAV8-Donor via retro-orbital injection resulted in a drastic difference in hF.IX expression between donors with and without homology 10 weeks post injection (Homology: 1531 ± 174.5 ng/mL vs. No-homology: 146.1 ± 5.8 ng/mL; n=12 and 7, respectively). We next asked whether HDR could be stimulated even more specifically through the induction of DNA single strand breaks at the target site. We treated neonatal mice with homologous or non-homologous donors, as well as ZFNs or ZFNickases (in which one FokI nuclease domain was inactivated with the D450A mutation). ZFNickases were indeed active, resulting in ~250 ng/mL hF.IX 4 weeks post injection (Figure 1). Interestingly, we could not detect hF.IX in mice treated with ZFNickase and no-homology donor (LOD: 15ng/mL). To rule out the possibility that this was simply due to the lower efficacy of ZFNickases compared to ZFNs, we increased the ZFNickase dose 4 fold. Four weeks post treatment, we observed substantial levels of hF.IX in mice treated with homologous donor (2041 ± 269 ng/mL) and were again unable to detect hF.IX in mice treated with the non-homologous donor (n=10 and 7, respectively). These data point to homology directed repair as the primary mechanism of protein production for genome editing in neonatal mouse liver, and suggest improvements in both efficacy and specificity can be made through deeper understanding of the molecular requirements of this approach.

ASH%202015%20Abstract%20Figure%201.jpg
  1. Genome Editing of the Bcl11A Erythroid Specific Enhancer in Bone Marrow Derived Hematopoietic Stem and Progenitor Cells for the Treatment of Sickle Cell Disease

Ablation of Bcl11A could be a viable approach for the treatment of β-hemoglobinopathies such as β-thalassemia and sickle cell disease (SCD), since patients with Bcl11A haploinsufficiency have persistently high levels of fetal hemoglobin (HbF) (up to 30%), which are associated with development of minimal to no disease symptoms.  Genome editing by engineered zinc-finger nucleases that target either the exon 2 (exon ZFN) or the GATA motif of the erythroid specific enhancer (enhancer ZFN) of Bcl11A has been shown to increase HbF level in erythroid progeny from mobilized peripheral hematopoietic stem and progenitor cells (PB-CD34+ HSPCs).  However, peripheral mobilization of CD34+ cells is associated with high risk and currently is not an option for SCD patients.  Therefore, we investigated the efficacy of genome editing of Bcl11A in bone marrow derived CD34+ cells (BM-CD34+ HSPCs). We first established a clinically compatible large-scale process to isolate CD34+ HSPCs from human bone marrow aspirates and to transiently express the ZFN protein by mRNA electroporation.  The CD34+ isolation process resulted in ~ 95% pure CD34+ cells with greater than 90% viability.  Both the exon and the enhancer ZFN drove 50-60% Bcl11A gene editing, resulting in a robust elevation of HbF in the erythroid progeny. Notably, the BM-CD34+ HSPCs were found to contain a small population (10 to 25%) of CD34+CD19+ pro-B cells that were refractory to ZFN transfection under our current electroporation condition.  Since CD34+CD19+ pro-B cells are not expected to contribute to reconstituting the hematopoietic system other than B-cell lineage, the Bcl11A editing efficiency in the multipotent BM-CD34+ HSPC could be even higher.
The engraftment abilities of Bcl11A edited BM-CD34+ cells were then investigated in an immunodeficient NOD/scid/gamma (NSG) mouse model.  At a dose of 1 million cells per mouse, treatment with either the exon ZFN or the enhancer ZFN did not detectably impact engraftment or multi-lineage reconstitution compared with untreated cells.  However, Bcl11A marking in engrafted human cells was found to be markedly higher in the mice treated by the enhancer ZFN than that by the exon ZFN.   The exon ZFN resulted in a strong bias towards in-frame mutations across multi-lineages with the strongest effect observed in the B-cell lineage, suggesting that a threshold level of Bcl11A is required for efficient hematopoietic reconstitution and that cells fully lacking it due to disruption of the coding sequence are at a disadvantage.  In contrast, the enhancer ZFN resulted in comparable Bcl11A marking across all lineages with no apparent selection for cells with a functional GATA sequence.  Collectively, these data indicate that genome editing of the erythroid specific enhancer of Bcl11A in BM-CD34+ promotes HbF reactivation in the erythroid progeny while maintaining the engraftment and multi-lineage repopulating activities of edited BM-CD34+ HSPCs, which supports further clinical development of this approach for the treatment of SCD.