Tuesday, August 1, 2017

Big Trouble for uniQure - Chiesi Drops collaboration - Hemo B

uniQure Reacquires Development and Commercialization Rights for its Gene Therapy Candidate in Hemophilia B

~ Company now owns full global rights to late-stage program with clinical proof-of-concept ~

LEXINGTON, Mass. and AMSTERDAM, the Netherlands, July 31, 2017 (GLOBE NEWSWIRE) -- uniQure N.V. (NASDAQ:QURE), a leading gene therapy company advancing transformative therapies for patients with severe medical needs, today announced that it has entered into an agreement with Chiesi Group to reacquire the rights to co-develop and commercialize its hemophilia B gene therapy in Europe and other select territories and to terminate their co-development and license agreement.
"We are very pleased to reach an agreement with Chiesi to acquire back European and other territorial rights to our lead gene therapy program in hemophilia B," stated Matthew Kapusta, Chief Executive Officer of uniQure.  "By regaining unencumbered, global rights to a late-stage program that has demonstrated significant clinical benefit for patients with hemophilia B, we believe uniQure is better positioned to accelerate the global clinical development plan, maximize shareholder return on our pipeline and take advantage of new potential opportunities related to the program. We are grateful for the substantial investments that Chiesi has made in AMT-060, and we have been fortunate to have them as a collaboration partner over the years."
"As we recently announced, we have made significant progress in preparing for a late-stage clinical program in hemophilia B and look forward to providing additional updates this fall," added Mr. Kapusta.
"Chiesi's decision was driven by recent changes in our strategic priorities," stated Ugo Di Francesco, Chief Executive Officer of Chiesi.  "We greatly appreciate the advances uniQure has made in the development of AMT-060 over the years and sincerely wish them the best as they advance this potentially exciting gene therapy to patients.  We will continue to support the transition and expect it will be relatively quick and seamless."
In 2013, uniQure and Chiesi entered into an agreement for the co-development and commercialization of a hemophilia B gene therapy in Europe and other select territories, including an equal sharing of all development related costs.  Under the terms of the agreement announced today, uniQure will be responsible for all future development costs related to its hemophilia B program, including approximately $3 million of expenses in 2017 that would have otherwise been shared with Chiesi.  The Company does not expect the transaction will impact its previous cash guidance, and continues to anticipate cash on hand will be sufficient to fund operations into 2019.
As a result of the transaction, uniQure expects to recognize in the third quarter of 2017 the remaining deferred revenue of approximately $14 million from non-refundable payments received from Chiesi in 2013.

Monday, July 24, 2017

Xenotransplantation !

Sangamo job opening describes areas of interest to include Xenotransplantation.

Title: Product Development Sr. Program Manager
 
Description
Sangamo Therapeutics, Inc. (Nasdaq: SGMO) is the world’s leading developer of customized DNA-binding proteins for targeted gene regulation and genome engineering.  We are applying this technology in diverse therapeutic areas including HIV / AIDS, hemophilia, Huntington's disease, hemoglobinopathies, and lysosomal storage diseases.  Sangamo also has active programs and collaborations in a variety of other areas including crop engineering, gene-edited stem cells, and the development of humanized donor animals for xenotransplantation.
 
The Product Development function at Sangamo leads product strategy and execution from IND through clinical development and approval. The senior Project Manager in Product Development will be a highly-organized and versatile team player responsible for supporting two or more core teams and alliance management. The Project Manager supports the core team and core team leader in all aspects of product development (strategic, operational, and technical), from the time of IND filing to product launch. The Project Manager is focused on leading the operational aspects of the program and the team like program strategy and planning; timelines; budgets, meeting agendas, facilitation, minutes, and action-item follow-up) while also supporting and influencing the team in making complex cross-functional decisions.
The primary responsibility will be to provide project management expertise and support to cross-functional core teams and core team leader, ensuring that project priorities and plans are developed and executed in accordance with corporate goals.  This position will eventually lead to core team leadership roles.
 
RESPONSIBILITIES
  • Support identification, planning, and drive execution of program strategy and key program initiatives
  • Support alliance management and joint collaboration efforts with Pharma partners
  • Prepare agendas, facilitate discussions, prepare minutes, and follow up on action items for core team and join collaboration meetings
  • Support internal and external meetings related to patient events, publications, congresses, senior management meetings
  • Familiarize with the scientific and operational aspects of the program
  • Support the core team and core team leader in making complex cross-functional recommendations / decisions with data collection, scenario planning, risk assessment and strategic prioritization
  • Establish collaborative relationships with core team members and functional stakeholders
  • Ensure that timely and consistent communications to the team, partners and to other stakeholders, including senior management
  • Support core team and department initiatives such as strategic off-sites, team-building events
EDUCATION
A Bachelor of Science required. PhD in a scientific discipline, MBA or other advanced degree, highly preferred. Certification or professional training in project management is a plus.
 
SKILLS / EXPERIENCE
  • 5+ years' experience in a Biotech or Pharma in drug development, including project management experience
  • Working knowledge of core teams, functions and overall drug development process
  • Demonstrated excellence in supporting clinical-stage drug development programs
  • Attention to detail, curiosity, adaptability, and willingness/ability to learn quickly
  • Strong interpersonal and influencing skills and oral and written communication skills
  • Excellent analytical skills and Excel, PowerPoint, Project, and Word skills
 

Tuesday, July 11, 2017

uniQure Presents New Clinical Data in Hemophilia B Patients Demonstrating Therapeutic Efficacy of AAV5 Gene Therapy in the Presence of Pre-Existing Neutralizing Antibodies

-- Findings Further Support Expanding the Eligibility of AAV5 Gene Therapies to Nearly All Patients with Hemophilia B --

LEXINGTON, Mass. and AMSTERDAM, the Netherlands, July 11, 2017 (GLOBE NEWSWIRE) -- uniQure N.V. (NASDAQ:QURE), a leading gene therapy company advancing transformative therapies for patients with severe medical needs, today presented new clinical data demonstrating that the presence of pre-existing anti-AAV5 neutralizing antibodies (NABs) does not predict the potential efficacy of AAV5-mediated gene transfer in patients with hemophilia B. Clinically meaningful factor IX (FIX) activity levels from the ongoing Phase I-II trial of AMT-060 were observed at NAB titers up to 1:341, determined as corresponding up to the 90th percentile of a healthy control population. NABs were quantified in the blood sera of these patients using a highly sensitive assay. These clinical data were presented today in a poster presentation at the 26th Biennial Congress of the International Society on Thrombosis and Hemostasis (ISTH), taking place this week in Berlin, Germany.
The presence of pre-existing NABs to adeno-associated virus (AAV) vectors has long posed a critical challenge for the clinical application of gene therapies, as patients who currently screen positive for NABs are generally excluded from treatment. Researchers from uniQure recently presented data in non-human primates suggesting that AAV5 could successfully mediate gene transfer in the presence of NABs at levels as high as 1:1031.   
In a poster presentation at the ISTH meeting, a re-analysis was described of pre-gene transfer screening samples from the 10 patients who have been treated in the ongoing Phase I/II trial of AMT-060 for hemophilia B. The patients had tested negative for pre-existing anti-AAV5 NAbs using a green fluorescent protein-based (GFP) assay before receiving treatment. These samples were later re-assessed using a highly sensitive luciferase-based (LUC) NAB assay.  Anti-AAV5 NABs were detected retrospectively in three patients who had been treated with the low dose (5x1012 gc/kg) of AMT-060. However, all three patients presented increases in FIX expression and, especially, the patient with the highest NAB level (titer 1:341) had the highest FIX-activity (steady-state FIX 6.8% of normal; latest FIX measurement 10.7% of normal) among all five patients treated in the low-dose cohort. None of the three patients who tested positive for NAB titers, experienced over time elevations in liver enzymes post gene transfer, FIX activity loss, or clinically relevant T-cell responses to the capsid.
"These clinical data show that hemophilia B patients presenting with neutralizing antibodies may be considered eligible for AAV5-mediated gene transfer," stated Matthew Kapusta, chief executive officer at uniQure. "This development potentially expands the applicability of AAV5 gene therapies to nearly all hemophilia B patients. We believe these factors contribute to making AAV5 a potential best-in-class vector for delivering gene therapies more effectively and safely to a greater portion of patients in need of treatment."

Friday, July 7, 2017

Biocompare.com Editorial - Gene Editing: Not Just CRISPR

http://www.biocompare.com/Editorial-Articles/339420-Gene-Editing-Not-Just-CRISPR/

The impact of gene editing on biomedical research has been similar in breadth and depth to that of PCR 30 years ago. Readers can find excellent summaries of the principal editing techniques, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR), transcription activator-like effector nuclease (TALEN), and zinc finger nuclease (ZFN), through any search engine.
ZFN led the surge chronologically, followed by TALEN, then CRISPR. Each succeeding development eclipsed the previous one by virtue of easier use and greater familiarity with gene editing. CRISPR is generally credited with being significantly easier to use, hence its wide adoption among basic researchers.
Yet despite sales of ZFN reagents tailing off to baseline, and TALEN use dropping nearly as precipitously, those methods still hold promise for what is arguably the most value-driven gene editing application of all—therapy. ZFN and TALEN have reputations for being more precise than CRISPR. A recent paper by Schaefer et al., in Nature Methods that described deep sequencing to uncover off-target CRISPR effects, reinforced this belief. The authors noted that “concerns persist regarding secondary mutations in regions not targeted by the single guide RNA...We found an unexpected number of single-nucleotide variants...compared with the widely accepted assumption that CRISPR causes mostly indels at regions homologous to the sgRNA.”
ZFN and TALEN have reputations for being more precise than CRISPR.
“The simplicity and accessibility of CRISPR to researchers has democratized genome editing and changed the face of disease research to the benefit of scientists and, in fact, the world,” says Martha S. Rook, Ph.D., head of gene editing and novel modalities at MilliporeSigma. “However, mature gene-editing technologies such as ZFN and TALEN remain the methods of choice for critical research where clear intellectual property rights are desirable.”

Back to ZFNs?

Where CRISPR relies on a gene-targeting guide RNA to bind a chosen gene sequence, ZFNs have the advantage of being a protein-only construct that may be optimized to increase precision and specificity, particularly for clinical gene editing.
According to Michael Holmes, Ph.D., vice president of research at Sangamo, ZFNs demand a higher level of protein engineering knowledge than CRISPR. “That’s why when researchers think of gene editing they first consider CRISPR, but when you’re thinking therapeutics what’s easiest may not be best.
In December 2015, FDA approved an Investigational New Drug Application for SB-FIX, Sangamo’s hemophilia B treatment, that inserts a therapeutic gene into the albumin gene locus in liver cells.
Sangamo’s target, the endogenous albumin locus in the liver, is highly expressed. “When we insert a therapeutic transgene into this very powerful promoter we can exploit the liver as a protein production factory, a technique that requires editing a very small number of hepatocytes to produce therapeutic levels of protein,” Holmes says.
Sangamo’s optimized ZFNs provide high specificity with levels of off-target modification below the detection limits of state-of-the-art oligo-capture and deep sequencing assays. “This ability, to obtain editing efficiencies of greater than 80% at the intended on-target site in the genome with no detectable off-targets is where you want to be with therapeutic gene editing,” Holmes adds.
The edits made to the genome by Sangamo’s zinc finger platform are genetically stable, and the process does not appear to affect the inherent stability of the genome in either primary or transformed cells. The reagents act transiently but the traits they confer are permanent, even with rapidly dividing transformed cells—many of which are genomically unstable. “There are of course natural evolutionary forces, which cause cells after many passages to not divide as well or to lose chromosomes over time, which is to be expected” Holmes explains. “But this is neither more nor less likely than in the unedited genome.”

Improving a good idea

Despite its disadvantages CRISPR retains distinct advantages for many applications, Rook explains that CRISPR’s programmable RNA-to-DNA targeting provides the design simplicity that makes CRISPR whole-genome libraries cost-effective and accessible to all researchers off-the-shelf. “Further, the increased cleavage efficiency of CRISPR compared with ZFN and TALENs enhances its application to both whole-genome screens and single target genome-editing projects.”
MilliporeSigma recently announced an improvement to CRISPR that makes the editing tool more efficient, flexible, and specific. Proxy-CRISPR, described in a recent Nature Communications paper, will accelerate drug development and gene therapy by accessing “previously unreachable areas of the genome,” according to the company.
Biocompare’s CRISPR Products Search Tool
Find, compare and review CRISPR
products from different suppliers Search
The Proxy-CRISPR method employs two CRISPR binding events for high-efficiency cutting. The first binding event is from a CAS9 that lacks DNA endonuclease (i.e. cutting) activity; the second binding event arises through a CRISPR system that similarly cannot, on its own, cut human DNA. “We discovered that targeting dead Cas9 next to other CRISPR systems can greatly enhance their activity,” Rook says. This strategy mimics what occurs in nature, where many different CRISPR systems bind to different DNA sequences.
Proxy-CRISPR allows greater diversity of DNA targets as might be seen in DNA sequences implicated in disease that often differ among individuals. “To address any possible disease-related mutation, to correct it, or to model it into healthy cells requires an editing technique that can target almost any DNA sequence,” Rook says. “Proxy-CRISPR method opens the door for using many additional CRISPR variants capable of unraveling the significance of genomic differences we can observe through next-generation sequencing.”

Greater flexibility

Poseida Therapeutics also shows that improving CRISPR technology can overcome off-target effects. The company recently reported preclinical data demonstrating its high-fidelity genome-editing system, NextGEN™ CRISPR, for producing allogeneic “universal donor” chimeric antigen receptor T-cells (CAR-T), an important platform for cancer immunotherapy.
Poseida has accumulated multiple gene-editing tools. In addition to NextGEN CRISPR, Poseida employs the piggyBac™ DNA Modification System, XTN™ TALEN site-specific nuclease, and Footprint-Free™ Gene Editing.
“For any given cell type there is not always one superior technology, depending on what you want to do,” says Eric Ostertag, Poseida’s CEO. “CRISPR reagents are easier and faster to design, and slightly cheaper to create than TALEN. But if you’re experienced with TALEN and are making a therapeutic, the cost and design differences amount to a rounding error.”
An advantage of NextGEN CRISPR is its application to both activated (i.e. dividing) and resting (quiescent) T cells, which confers greater flexibility over editing approaches that work only on activated cells. “The more you manipulate T cells, the greater the likelihood that the end product will exhibit an undesirable phenotype,” Ostertag explains. “Having a stem-like phenotype is critical for product efficacy and durability, which is why we prefer to engineer resting cells.” NextGEN CRISPR works well in resting T cells, whereas TALEN does not.
Wild-type CRISPR uses the Cas9 protein and a guide RNA to cut DNA. The natural ribonucleoprotein is known for high-efficiency cutting of target sequences but “is quite sloppy in the way it causes undesirable off-target mutations,” Ostertag says. Poseida’s NextGEN CRISPR uses nuclease enzymes that are more typical for zinc finger and TALEN in that they require both half-sites to be present at the same time and at the correct location in the genome, thereby resulting in exquisite on-target specificity.

Thursday, July 6, 2017

Shire Submits IND for Gene Therapy Treatment of Hemophilia A

Shire submits investigational New Drug Application to FDA for Gene Therapy candidate SHP654 for treatment of Hemophilia A
SHP654 aims to deliver sustained protection against bleeds for patients with hemophilia A
Lexington, Mass.  July 6, 2017 – Shire plc (LSE: SHP, NASDAQ: SHPG), the leading biotechnology company focused on serving people with rare diseases, today announced the submission of an investigational new drug (IND) application to the U.S. Food and Drug Administration (FDA) for SHP654, also designated as BAX 888, an investigational factor VIII (FVIII) gene therapy for the treatment of hemophilia A. SHP654 aims to protect hemophilia A patients against bleeds through the delivery of a long-term, constant level of factor expression.1 The IND filing for SHP654 represents the latest step forward for Shire’s gene therapy program, which shows promise for both hemophilia A and B populations.
“Shire is leveraging decades of scientific leadership in hemophilia to advance research in gene therapy for this community,” said Paul Monahan, M.D., Senior Medical Director, Gene Therapy, Shire. “Drawing from our rich heritage, Shire is well equipped to sustainably support the development of gene therapies that aim to advance current standards of care and minimize the burden of this disease. SHP654 uses a proprietary technology platform designed to produce sustained levels of factor similar to the natural mechanisms of the body. Our goal with gene therapy for hemophilia is to uphold the highest standards for safety and efficacy.”
Shire’s gene therapy program for hemophilia A uses a recombinant adeno-associated virus serotype 8 (rAAV8) vector, which selectively targets the liver.1,2 It involves the delivery of a functional copy of FVIII to the body’s liver to enable its own production of FVIII, rather than relying on a factor-based treatment.1 SHP654 uses the rAAV8 vector to deliver a codon-optimized, B-domain deleted FVIII (BDD-FVIII) specifically to a patient’s liver, where FVIII would then be produced and used to manage bleeds.1 The FVIII expression is further controlled in patients by incorporating the liver-specific transthyretin (TTR) promoter/enhancer.1
The IND filing for SHP654 was based on the results of pre-clinical and phase 1 studies demonstrating the potential utility of this candidate, including the following that will be presented at the International Society on Thrombosis and Haemostasis (ISTH) 26th Biennial Congress in Berlin, Germany, from July 8 – 13, 2017:
  • Development of SHP654, a highly efficient AAV8-based BDD-FVIII gene therapy vector for treatment of hemophilia A. Session Title: Gene Therapy for Hemophilia: Clinical. Oral # OC 13.6.10th July, 17:45-19:00 CEST; Hall B1
  • Integration site analysis in mice demonstrates excellent biosafety profile of a recombinant (r) FVIII adeno-associated virus (AAV8) gene therapy product. Session Title: Poster Session. Poster # PB 1094. 11th July, 12:00-13:15 CEST; Exhibition Hall 4.23
  • Dose response and long-term expression of a human FVIII gene therapy construct in hemophilia A mice. Session Title: Poster Session. Poster # PB 1101. 11th July, 12:00-13:15 CEST; Exhibition Hall 4.22
  • Nonclinical safety evaluation of a human FVIII gene therapy construct in mice. Session Title: Poster Session. Poster # PB 1099. 11th July, 12:00-13:15 CEST; Exhibition Hall 4.24
An IND is a request for FDA authorization to administer an investigational drug to humans.5 Following the FDA’s acceptance of the IND for SHP654, Shire will study SHP654 in a global multi-center study evaluating safety and examining the SHP654 doses required to boost factor VIII activity levels and affect hemophilic bleeding and will pursue bringing this innovation to markets worldwide.
About SHP654
Shire is developing SHP654 (BAX 888), which includes technology acquired from Chatham Therapeutics, LLC, a spin-out of Asklepios Biopharmaceutical, Inc. SHP654 is an investigational factor VIII (FVIII) gene therapy intended to treat hemophilia A using a recombinant adeno-associated virus serotype 8 (rAAV8) vector to deliver a codon-optimized, B-domain deleted FVIII (BDD-FVIII) specifically to a patient’s liver, where FVIII would then be produced and used to manage bleeds.1,2
About Hemophilia A
Hemophilia A, the most common type of hemophilia, is a rare bleeding disorder that causes longer-than-normal bleeding due to lack of clotting factor VIII in the blood.6 The severity of hemophilia A is determined by the amount of factor in the blood, with more severity associated with lower amounts of factor.7 More than half of patients with hemophilia A have the severe form of the condition.7 Approximately 25-30% of individuals with severe hemophilia A develop inhibitors.8 Inhibitors are a serious medical problem that can occur when a person with hemophilia has an immune response to treatment with clotting factor concentrates.9 Hemophilia primarily affects males, with an incidence of one in 5,000 male births.7,10
References
  1. Falkner et al. “Development of SHP654 a highly efficient AAV8-based BDD-FVIII gene therapy vector for treatment of hemophilia A.” International Society on Thrombosis and Haemostasis Congress. Berlin, Germany July 8-13, 2017. Available at: http://onlinelibrary.wiley.com/doi/10.1111/rth2.2017.1.issue-S1/issuetoc
  2. Hoellriegl et al. “Dose response and long-term expression of a human FVIII gene therapy construct in hemophilia A mice.” International Society on Thrombosis and Haemostasis Congress. Berlin, Germany July 8-13, 2017. Available at: http://onlinelibrary.wiley.com/doi/10.1111/rth2.2017.1.issue-S1/issuetoc
  3. Hoellriegl et al. “Integration site analysis in mice demonstrates excellent biosafety profile of a recombinant ® FVIII adeno-associated virus (AAV8) gene therapy product.” International Society on Thrombosis and Haemostasis Congress. Berlin, Germany July 8-13, 2017. Available at: http://onlinelibrary.wiley.com/doi/10.1111/rth2.2017.1.issue-S1/issuetoc
  4. Hoellriegl et al. “Nonclinical safety evaluation of a human FVIII gene therapy construct in mice.” International Society on Thrombosis and Haemostasis Congress. Berlin, Germany July 8-13, 2017. Available at: http://onlinelibrary.wiley.com/doi/10.1111/rth2.2017.1.issue-S1/issuetoc
  5. U.S Food and Drug Administration. “Investigational New Drug (IND) or Device Exemption (IDE) Process (CBER).” U.S Food and Drug Administration website. https://www.fda.gov/biologicsbloodvaccines/developmentapprovalprocess/investigationalnewdrugindordeviceexemptionideprocess/default.htm. Accessed June 28, 2017.
  6. World Federation of Hemophilia. “What is hemophilia?” World Federation of Hemophilia website. http://www.wfh.org/en/page.aspx?pid=646. Accessed June 23, 2017. 
  7. National Hemophilia Foundation. “Hemophilia A.” National Hemophilia Foundation website. https://www.hemophilia.org/Bleeding-Disorders/Types-of-Bleeding-Disorders/Hemophilia-A.  Accessed June 23, 2017.
  8. World Federation of Hemophilia. “Who is at risk of developing inhibitors?” World Federation of Hemophilia website. http://www.wfh.org/en/page.aspx?pid=653. Accessed June 23, 2017.
  9. World Federation of Hemophilia. “What are inhibitors?” World Federation of Hemophilia website. http://www.wfh.org/en/page.aspx?pid=651. Accessed June 23, 2017.
  10. Centers for Disease Control and Prevention. “Hemophilia.” Centers for Disease Control and Prevention website. http://www.cdc.gov/ncbddd/hemophilia/facts.html. Accessed June 29, 2017.

Friday, June 23, 2017

Article - Fixing Crispr

http://www.genengnews.com/gen-exclusives/fixing-crispr/77900928

  • Elegant in its simplicity and easily accessible to most laboratories, CRISPR/Cas9 technology has been widely adopted as a DNA-editing tool in research labs, and a group of investigators in China has begun to use it in a clinical trial to disable the PD-1 gene in lung cancer patients to slow or stop the cancer’s progression. But the technology’s off-target effects have raised serious concerns about its ultimate applicability to human therapies.
    CRISPR proponents say it’s less clunky to use that other recently emerged genome-editing technologies, including zinc-finger nucleases (ZFNs) and transcription activator–like effector nucleases (TALENs). ZFNs and TALENs work by using a strategy of tethering endo­nuclease catalytic domains to modular DNA-binding proteins for inducing targeted DNA double-stranded breaks (DSBs) at specific genomic loci.
    In contrast, Cas9 is a nuclease guided by small RNAs through Watson-Crick base pairing with target DNA, described by F.A. Ran et al. as a system that is “markedly easier to design, highly specific, efficient, and well-suited for high throughput and multiplexed gene editing for a variety of cell types and organisms.”
    CRISPR/Cas9 nucleases consist of the Cas9 bacterial enzyme and a short, 20-nucleotide RNA molecule that matches the desired target DNA sequence. In addition to the RNA/DNA match, the Cas9 enzyme must recognize a specific nucleotide sequence called a protospacer adjacent motif (PAM) adjacent to the target DNA.
    The most commonly used form of Cas9, derived from the bacteria Streptococcus pyogenes and known as SpCas9, recognizes PAM sequences in which any nucleotide is followed by two guanine DNA bases. This limits the DNA sequences that can be targeted using SpCas9 only to those that include two sequential guanines. 
  • Off Target Effects

    But almost as soon as the technology was introduced, scientists raised concerns about off target effects. Said Xiao-Hui Zhang et al. of the College of Veterinary Medicine, South China Agricultural University and coauthors at MIT in a 2015 Molecular Therapy—Nucleic Acids article, “The high frequency of off-target activity (≥50%)—RGEN (RNA-guided endonuclease)-induced mutations at sites other than the intended on-target site is one major concern, especially for CRISPR technology therapeutic and clinical applications.”
    The growth of any new technology, the authors note, including CRISPR/Cas9, demands progressive enhancement. And while research in CRISPR/Cas9 has made “huge strides in the evolution of gene editing,” it and other RGENs, which include ZFNs and TALENs, have more severe off-target effects than other nucleases due to their inherent structure and mechanism.
    At an American Society of Hematology workshop on genome editing, CRISPR pioneer J. Keith Joung, M.D., Ph.D., of Massachusetts General Hospital said, “In the early days of this field, algorithms were generated to predict off-target effects and [made] available on the web miss a fair number of off-target effects. He added, “These tools are used in a lot of papers, but they really aren’t very good at predicting where there will be off-target effects,” according to STAT.
    Observations in the recent literature have raised more alarms among CRISPR cognoscenti, all of whom would agree than technical improvements are needed. In one of the most blogged-about papers on CRISPR, “Unexpected mutations after CRISPR–Cas9 editing in vivo,” Kellie A. Schaefer and colleagues at Stanford concluded that “More work may be needed to increase the fidelity of CRISPR/Cas9 with regard to off-target mutation generation before the CRISPR platform can be used without risk, especially in the clinical setting.”
    The authors had, they reported in a 2016 study by W.H. Wu et al. in Molecular Therapy, used CRISPR/Cas9 for sight restoration in blind rd1 mice by correcting a mutation in the Pde6b gene. Mice homozygous for the rd mutation have hereditary retinal degeneration and have been considered a model for human retinitis pigmentosa.
    Citing persistent concerns about secondary mutations in regions not targeted by a single guide RNA (sgRNA)—concerns also expressed by a number of other scientists—Kellie A. Schaefer, Ph.D., at Stanford University and colleagues at Howard Hughes and Massachusetts General Hospital performed whole genome sequencing (WGS) on DNA isolated from two CRISPR-repaired mice (F03 and F05) and one uncorrected control.
    CRISPR/Cas9-treated mice were sequenced at an average depth of 50×, and the control to 30× to identify all off target mutations. The sequencing the authors said identified an “unexpectedly high” number of single nucleotide variations (SNVs), contrary to the widely accepted assumption that CRISPR causes mutations mostly at regions homologous to the sgRNA.
    CRISPR’s penchant for promiscuous behavior has spawned an entirely new research field focused on fixing it. Patents have already been filed on the fixes and the developers believe that these advances will incrementally enable more reliable CRISPER performance. Most efforts have concentrated on modifying CRISPR nuclease Cas9 using structure-design based changes in the enzyme, chemical modifications, and amino acid substitutions at critical sights to better predict and control its function.
    Writing in Nature in 2015, Benjamin P. Kleinstiver, Ph.D., of the Molecular Pathology Unit and Center for Cancer Research at the Massachusetts General Hospital and colleagues noted that although CRISPR/Cas9 nucleases are widely used for genome editing, the range of sequences that Cas9 can recognize is constrained by the need for a specific PAM.
    The investigators reported that they could modify Cas9 to recognize alternative PAM sequences using structural information, bacterial selection-based directed evolution, and combinatorial design. The altered PAM specificity variants, they said, could edit endogenous gene sites in zebrafish and human cells that are not targetable by wild-type SpCas9. Further, they said, the variants’ genome-wide specificities are comparable to wild-type SpCas9 as judged by GUIDE-seq analysis and establish the feasibility of engineering a wide range of Cas9s with altered and improved PAM specificities.
    Kleinstiver and other investigators working in Joung’s also developed the unique endonuclease SpCas9-HF1, which they describe as a high-fidelity enzyme variant with alterations designed to reduce non-specific DNA contacts. The scientists hypothesized that reducing iCas9 and the target DNA interactions might help eliminate off-target effects while still retaining the desired on-target interaction.
    Since certain portions of the Cas9 nuclease can itself interact with the backbone of the target DNA molecule, the team altered four of these Cas9-mediated contacts by replacing the long amino acid side-chains that bind to the DNA backbone with shorter ones that could not bind.
    SpCas9-HF1 retained on-target activities comparable to wild-type SpCas9 with >85% of single-guide RNAs (sgRNAs) tested in human cells. Notably, with sgRNAs targeted to standard nonrepetitive sequences, SpCas9-HF1 rendered all or nearly all off-target events undetectable by genome-wide break capture and targeted sequencing methods.
    Jiang and Doudna pointed out in a 2017 piece in Annual Review of Biophysics how Cas9 locates specific 20-base-pair (bp) target sequences within the genomes that are millions to billions of base pairs long and, subsequently, how it induces sequence-specific double-stranded DNA (dsDNA) cleavage remain critical questions, not just in CRISPR biology, but in the efforts to develop more precise and efficient Cas9-based tools.
    Molecular insights from biochemical and structural studies such as those describe above will provide a framework for rational engineering aimed at altering catalytic function, guide RNA specificity and PAM requirements, and reducing off-target activity for the development of Cas9-based therapies against genetic diseases.

Thursday, June 22, 2017

Hemophilia B Article - Genetic Literacy Project

https://geneticliteracyproject.org/2017/06/22/blood-disease-plagued-europes-royal-families-might-treatable-using-gene-editing/
It was once known as the disease that plagued royal families in the 19th and 20th centuries. Hemophilia B famously struck dynasties in England, Spain, Germany, and Russia, beginning with diminutive Queen Victoria, who ruled England from 1837 to 1901. The Queen’s life was depicted in the first season of a recent Masterpiece Theater series.
Queen Victoria’s parents had no family history of hemophilia, so she was likely a carrier due to a spontaneous mutation. She passed her mutation onto her son, Prince Leopold (a hemophiliac who died from his mutation at age 30 after a minor fall, but not before passing it along to his only daughter, Princess Alice of Albany), and to two of her daughters, Princess Alice and Princess Beatrice, who were carriers, who carried the mutation he mutation into the Spanish, German and Russian royal families.
Victoria’s granddaughter Alix married Tsar Nicholas, resulting in son Alexei. The child was executed at age 13 along with the rest of his family, but in 2009 his charred bones yielded DNA that revealed him to have the bleeding disorder.
The last living relative of the lineage was Prince Waldemar of Prussia, a cousin of the ill-fated Alexei, who died as World War II was ending and blood supplies were sent to the emptying concentration camps. He was denied a transfusion.

Today, researchers are looking to genome editing to provide a more robust treatment option for the disease sufferers. And for at least one company, those efforts involve harnessing the power of albumin – the stuff of egg white and also the most abundant protein in blood plasma. Albumin controls fluid distribution in the body, but researchers can borrow it’s genetic controls to mass-produce selected proteins.
Sangamo Therapeutics is using genome editing to hitch the gene for the clotting factor that’s missing in hemophilia B (factor IX, aka FIX) to the controls of the albumin gene, rather than manipulate the FIX gene directly. And instead of using the media darling CRISPR-Cas9, it relies on an older, perhaps more controllable editing tool, zinc finger nucleases (ZFNs). The strategy works in mice, and a clinical trial is just getting underway, with FDA giving fast track designation to the company’s “SB-FIX in vivo genome editing treatment for hemophilia B.”
 “Our ultimate goal is to treat children young enough so they will have a solution that will last for a lifetime and they will not have to face the consequences of the disease,” said Sandy Macrae, president and CEO of Sangamo. He calls the technology “beautiful Star Trek type science.”
Normally, in response to a puncture injury, the body stanches blood flow by knitting long, feathery strands of the protein fibrin into clots as the culmination of a cascade of interacting proteins, the clotting factors. FIX and factor VIII join in midway through the action. About 16,000 people in the U.S. have hemophilia A (factor VIII deficiency) and 4,000 have hemophilia B. Both genes are on the X chromosome, so males are affected and women are carriers.

From transfusions to gene therapy

I interviewed the first person to receive gene therapy for hemophilia A, in 2000. Born in 1949, Don Miller was raising sheep when he became the first participant in a clinical trial at the University of Pittsburgh, receiving the retrovirus-bound factor VIII gene on June 1, 1999. His story chronicles the evolution of hemophilia treatment. It began with nearly bleeding to death following his circumcision as a newborn.
Ryan White
“When I was 3, I fell out of my crib and I was black and blue from my waist to the top of my head. One time I fell at my grandmother’s house and had a 1-inch-long cut on the back of my leg. It took five weeks to stop bleeding, just leaking real slowly,” he told me. After that he limited activities and had frequent transfusions – those were the days before AIDS, which would infect 90% of the people with hemophilia before screening blood donations began. Ryan White was a teen with hemophilia A who drew attention to the vulnerability of the hemophilia community and ended use of the word “hemophiliac.”
By the time Miller had married, in 1969, he was receiving gamma globulin pooled from donated plasma and frozen, pooled factor VIII, called “cryoprecipitate.” He got into the clinical trial because he’d managed to avoid the HIV infection that kept many people with hemophilia out: “I lucked out.”
It’s just one case, but Don Miller improved – he needed boosters of factor VIII (now made more safely using recombinant DNA technology) less frequently, he had no more spontaneous bleeding, and nosebleeds stopped within minutes – as they would for anyone with normal clotting.
Alas Mr. Miller’s good response had exquisitely poor timing, for 1999 was a tragic year for gene therapy. On September 17, 1999, the death of 18-year-old Jesse Gelsinger from an overwhelming immune response to gene therapy to treat a urea cycle disorder paralyzed the field. Also that year the first boys received gene therapy for an immune deficiency that used a retrovirus, which would cause leukemia two years later, again paralyzing the field.
Clotting factor IX
But gene therapy re-emerged, with the hemophilias at the forefront. Ongoing hemophilia B clinical trials include those sponsored by UniQure Biopharma and St. Jude’s Children’s Hospital. Spark Therapeutics, in collaboration with Pfizer, recently announced preliminary data from 10 patients that showed persistent and sustained FIX production, although some patients needed steroids to quell a change in liver enzymes that could indicate an immune response. But on May 10, Dimension Therapeutics halted its trial for hemophilia B, citing “inability to achieve a minimum target product profile.” Sources tell me off-the-record that this means elevated liver enzymes that could signal an immune response.

Enter zinc fingers

Treating hemophilia B at the protein level costs $100,000 a year for “bleeds” and up to $250,000 if given two or three times a week to prevent bleeding. That’s about $20 million over a lifetime. Gene therapy seems a great alternative, a possibly one-time fix. The gene is small and only a small increase in the level of the clotting factor in plasma can make a big difference in how a patient feels.
A report from 2014 charts the “Long-Term Safety and Efficacy of Factor IX Gene Therapy in Hemophilia B,” but the vector, a type of adeno-associated virus, deposits the FIX gene in an episome, which is a bit of DNA outside of a chromosome. Like peeing in the ocean, a gene added in an episome becomes diluted as the chromosomes duplicate as cells divide, but the episomes do not. Still, gene therapy might provide enough factor IX to help adults. But young children might be a different story. For them, genome editing might be a longer-lasting approach because it inserts the healing gene at a selected site in a chromosome.
A gene therapy or genome editing approach to hemophilia would eliminate the need for frequent infusions of clotting factor.
All three genome editing technologies – CRISPR-Cas9, TALENs, and ZFNS – borrow from bacteria to create double-stranded breaks in DNA and then heal the damage. Unlike CRISPRs, which use RNA “guides,” ZFN deploys proteins, which are much more targeted and less likely to insert willy-nilly into undesired parts of the genome.
A zinc finger is a small protein that folds into an oblong shape when it binds zinc atoms, and it binds to a specific sequence of DNA found in about 3 percent of all human genes. A DNA-cutting enzyme attracted to a bound zinc finger forms a scissor-like molecular tool.
The power of zinc fingers comes from their natural affinity for transcription factor genes, which in turn encode proteins that control which suites of genes are turned on or off in a particular place and time. Zinc fingers were discovered in the African clawed frog in 1985, and first used as genetic switches in 1994.
Macrae explains how it works:
“We’ve designed zinc fingers to recognize intron 2 of the albumin gene, land on the DNA, and the restriction enzyme attaches, dimerizes, and cuts the DNA. At the same time as the ZFN cuts, we introduce a separate AAV with a good copy of the gene that encodes the missing protein.”
Some of the targeted liver cells take up the cargo and then release factor IX, which makes its way to various tissues.
He calls use of the albumin gene a “’safe harbor’ because you don’t need all of the albumin you produce. It has nice introns where we can land it and we will never remove but a fraction of the albumin genes from producing albumin.”
Meanwhile, he and his colleagues at The company is also launching clinical trials to tackle two mucopolysaccharidoses (Hunter and Hurler syndromes), and has treated 100 people with HIV with zinc fingers targeted to the CCR5 gene. ZFNs can hit any target in the genome efficiently. “We think zinc fingers is the leading genome editing technology. But eventually there will be others, and CRISPR will sort out its problems,” Macrae said.Sangamo are looking forward to the day when zinc fingers can help the youngest patients with hemophilia B:

“Parents whose children have hemophilia B came to see us, and they talked about holding their babies down to find a vein in the scalp to give an infusion. Some patients need clotting factor on a daily basis, and getting it dominates their lives. Patients die. We see what we do as part of the journey to eventually offer patients a chance to address the disease in a fundamental way.”