Page 26«..1020..25262728..4050..»

Archive for the ‘Crispr’ Category

CRISPR-engineered T cells in patients with refractory cancer – Science Magazine

CRISPR takes first steps in humans

CRISPR-Cas9 is a revolutionary gene-editing technology that offers the potential to treat diseases such as cancer, but the effects of CRISPR in patients are currently unknown. Stadtmauer et al. report a phase 1 clinical trial to assess the safety and feasibility of CRISPR-Cas9 gene editing in three patients with advanced cancer (see the Perspective by Hamilton and Doudna). They removed immune cells called T lymphocytes from patients and used CRISPR-Cas9 to disrupt three genes (TRAC, TRBC, and PDCD1) with the goal of improving antitumor immunity. A cancer-targeting transgene, NY-ESO-1, was also introduced to recognize tumors. The engineered cells were administered to patients and were well tolerated, with durable engraftment observed for the study duration. These encouraging observations pave the way for future trials to study CRISPR-engineered cancer immunotherapies.

Science, this issue p. eaba7365; see also p. 976

Most cancers are recognized and attacked by the immune system but can progress owing to tumor-mediated immunosuppression and immune evasion mechanisms. The infusion of ex vivo engineered T cells, termed adoptive T cell therapy, can increase the natural antitumor immune response of the patient. Gene therapy to redirect immune specificity combined with genome editing has the potential to improve the efficacy and increase the safety of engineered T cells. CRISPR coupled with CRISPR-associated protein 9 (Cas9) endonuclease is a powerful gene-editing technology that potentially allows the ability to target multiple genes in T cells to improve cancer immunotherapy.

Our first-in-human, phase 1 clinical trial (clinicaltrials.gov; trial NCT03399448) was designed to test the safety and feasibility of multiplex CRISPR-Cas9 gene editing of T cells from patients with advanced, refractory cancer. A limitation of adoptively transferred T cell efficacy has been the induction of T cell dysfunction or exhaustion. We hypothesized that removing the endogenous T cell receptor (TCR) and the immune checkpoint molecule programmed cell death protein 1 (PD-1) would improve the function and persistence of engineered T cells. In addition, the removal of PD-1 has the potential to improve safety and reduce toxicity that can be caused by autoimmunity. A synthetic, cancer-specific TCR transgene (NY-ESO-1) was also introduced to recognize tumor cells. In vivo tracking and persistence of the engineered T cells were monitored to determine if the cells could persist after CRISPR-Cas9 modifications.

Four cell products were manufactured at clinical scale, and three patients (two with advanced refractory myeloma and one with metastatic sarcoma) were infused. The editing efficiency was consistent in all four products and varied as a function of the single guide RNA (sgRNA), with highest efficiency observed for the TCR chain gene (TRAC) and lowest efficiency for the TCR chain gene (TRBC). The mutations induced by CRISPR-Cas9 were highly specific for the targeted loci; however, rare off-target edits were observed. Single-cell RNA sequencing of the infused CRISPR-engineered T cells revealed that ~30% of cells had no detectable mutations, whereas ~40% had a single mutation and ~20 and ~10% of the engineered T cells were double mutated and triple mutated, respectively, at the target sequences. The edited T cells engrafted in all three patients at stable levels for at least 9 months. The persistence of the T cells expressing the engineered TCR was much more durable than in three previous clinical trials during which T cells were infused that retained expression of the endogenous TCR and endogenous PD-1. There were no clinical toxicities associated with the engineered T cells. Chromosomal translocations were observed in vitro during cell manufacturing, and these decreased over time after infusion into patients. Biopsies of bone marrow and tumor showed trafficking of T cells to the sites of tumor in all three patients. Although tumor biopsies revealed residual tumor, in both patients with myeloma, there was a reduction in the target antigens NY-ESO-1 and/or LAGE-1. This result is consistent with an on-target effect of the engineered T cells, resulting in tumor evasion.

Preliminary results from this pilot trial demonstrate that multiplex human genome engineering is safe and feasible using CRISPR-Cas9. The extended persistence of the engineered T cells indicates that preexisting immune responses to Cas9 do not appear to present a barrier to the implementation of this promising technology.

T cells (center) were isolated from the blood of a patient with cancer. CRISPR-Cas9 ribonuclear protein complexes loaded with three sgRNAs were electroporated into the normal T cells, resulting in gene editing of the TRAC, TRBC1, TRBC2, and PDCD1 (encoding PD-1) loci. The cells were then transduced with a lentiviral vector to express a TCR specific for the cancer-testis antigens NY-ESO-1 and LAGE-1 (right). The engineered T cells were then returned to the patient by intravenous infusion, and patients were monitored to determine safety and feasibility. PAM, protospacer adjacent motif.

CRISPR-Cas9 gene editing provides a powerful tool to enhance the natural ability of human T cells to fight cancer. We report a first-in-human phase 1 clinical trial to test the safety and feasibility of multiplex CRISPR-Cas9 editing to engineer T cells in three patients with refractory cancer. Two genes encoding the endogenous T cell receptor (TCR) chains, TCR (TRAC) and TCR (TRBC), were deleted in T cells to reduce TCR mispairing and to enhance the expression of a synthetic, cancer-specific TCR transgene (NY-ESO-1). Removal of a third gene encoding programmed cell death protein 1 (PD-1; PDCD1), was performed to improve antitumor immunity. Adoptive transfer of engineered T cells into patients resulted in durable engraftment with edits at all three genomic loci. Although chromosomal translocations were detected, the frequency decreased over time. Modified T cells persisted for up to 9 months, suggesting that immunogenicity is minimal under these conditions and demonstrating the feasibility of CRISPR gene editing for cancer immunotherapy.

Gene editing offers the potential to correct DNA mutations and may offer promise to treat or eliminate countless human genetic diseases. The goal of gene editing is to change the DNA of cells with singlebase pair precision. The principle was first demonstrated in mammalian cells when it was shown that expression of a rare cutting endonuclease to create double-strand DNA breaks resulted in repair by homologous and nonhomologous recombination (1). A variety of engineered nucleases were then developed to increase efficiency and enable potential therapeutic applications, including zinc finger nucleases, homing endonucleases, transcription activatorlike effector nucleases, and CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats associated with Cas9 endonuclease) (2). The first pilot human trials using genome editing were conducted in patients with HIV/AIDS and targeted the white blood cell protein CCR5, with the goal of mutating the CCR5 gene by nonhomologous recombination and thereby inducing resistance to HIV infection (3, 4). The incorporation of multiple guide sequences in CRISPR-Cas9 permits, in principle, multiplex genome engineering at several sites within a mammalian genome (59). The ability of CRISPR to facilitate efficient multiplex genome editing has greatly expanded the scope of possible targeted genetic manipulations, enabling new possibilities such as simultaneous deletion or insertion of multiple DNA sequences in a single round of mutagenesis. The prospect of using CRISPR engineering to treat a host of diseases, such as inherited blood disorders and blindness, is moving closer to reality.

Recent advances in CRISPR-Cas9 technology have also permitted efficient DNA modifications in human T cells, which holds great promise for enhancing the efficacy of cancer therapy. T lymphocytes are specialized immune cells that are largely at the core of the modern-day cancer immunotherapy revolution. The T cell receptor (TCR) complex is located on the surface of T cells and is central for initiating successful antitumor responses by recognizing foreign antigens and peptides bound to major histocompatibility complex molecules. One of the most promising areas of cancer immunotherapy involves adoptive cell therapy, whereby the patients own T cells are genetically engineered to express a synthetic (transgenic) TCR that can specifically detect and kill tumor cells. Recent studies have shown safety and promising efficacy of such adoptive T cell transfer approaches using transgenic TCRs specific for the immunogenic NY-ESO-1 tumor antigen in patients with myeloma, melanoma, and sarcoma (1012). One limitation of this approach is that the transgenic TCR has been shown to mispair and/or compete for expression with the and chains of the endogenous TCR (1315). Mispairing of the therapeutic TCR and chains with endogenous and chains reduces therapeutic TCR cell surface expression and potentially generates self-reactive TCRs.

A further shortcoming of adoptively transferred T cells has been the induction of T cell dysfunction or exhaustion leading to reduced efficacy (16). Programmed cell death protein 1 (PD-1)deficient allogeneic mouse T cells with transgenic TCRs showed enhanced responses to alloantigens, indicating that the PD-1 protein on T cells plays a negative regulatory role in antigen responses that are likely to be cell intrinsic (17). The adoptive transfer of PD-1deficient T cells in mice with chronic lymphocytic choriomeningitis virus infection initially leads to enhanced cytotoxicity and later to enhanced accumulation of terminally differentiated T cells (18). Antibody blockade of PD-1, or disruption or knockdown of the gene encoding PD-1 (i.e., PDCD1), improved chimeric antigen receptor (CAR) or TCR T cellmediated killing of tumor cells in vitro and enhanced clearance of PD-1 ligandpositive (PD-L1+) tumor xenografts in vivo (1923). In preclinical studies, we and others found that CRISPR-Cas9mediated disruption of PDCD1 in human T cells transduced with a CAR increased antitumor efficacy in tumor xenografts (2426). Adoptive transfer of transgenic TCR T cells specific for the cancer antigen NY-ESO-1, in combination with a monoclonal antibody targeting PD-1, enhanced antitumor efficacy in mice (27). We therefore designed a first-in-human, phase 1 human clinical trial to test the safety and feasibility of multiplex CRISPR-Cas9 genome editing for a synthetic biology cancer immunotherapy application. We chose to target endogenous TRAC, TRBC, and PDCD1 on T cells to increase the safety and efficacy profile of NY-ESO-1 TCRexpressing engineered cells. In principle, this strategy allowed us to increase exogenous TCR expression and reduce the potential for mixed heterodimer formation (i.e., by deleting the and TCR domain genes TRAC and TRBC, respectively) and to limit the development of T cell exhaustion, which can be triggered by the checkpoint ligands PD-L1 and PD-L2 (i.e., by deleting PDCD1).

The phase 1 human trial (clinicaltrials.gov; trial NCT03399448) was designed to assess the safety and feasibility of infusing autologous NY-ESO-1 TCRengineered T cells in patients after CRISPR-Cas9 editing of the TRAC, TRBC, and PDCD1 loci. During the manufacturing process, cells were taken out of the cancer patient, engineered, and then infused back into the individual. The genetically engineered T cell product was termed NYCE (NY-ESO-1transduced CRISPR 3X edited cells) and is referred to as NYCE hereafter. During clinical development of the protocol, we elected to use a TCR rather than a CAR because the incidence of cytokine release syndrome is generally less prevalent using TCRs (11). In principle, this allowed a more discriminating assessment of whether gene editing with Cas9 was potentially immunogenic or toxic when compared with the baseline low level of adverse events observed in our previous clinical trial targeting NY-ESO-1 with transgenic TCRs (11). The autologous T cells were engineered by lentiviral transduction to express an HLA-A2*0201restricted TCR specific for the SLLMWITQC peptide in NY-ESO-1 and LAGE-1. The manufacturing process, vector design, and clinical protocol for NYCE T cells are described in the materials and methods and are depicted schematically (figs. S1 and S2). Of the six patients who were initially enrolled, four patients had successfully engineered T cells that were subjected to detailed release criteria testing as specified in the U.S. Food and Drug Administration (FDA)accepted Investigational New Drug application (table S1) (see fig. S3 for the consort diagram). Of the four patients with cell products available, one patient assigned unique patient number (UPN) 27 experienced rapid clinical progression and was no longer eligible for infusion owing to the inability to meet protocol-mandated safety criteria (see supplementary materials). Of the three patients who were infused with CRISPR-Cas9engineered T cells, two patients had refractory advanced myeloma and one patient had a refractory metastatic sarcoma not responding to multiple prior therapies (Table 1). The patients were given lymphodepleting chemotherapy with cyclophosphamide and fludarabine on days 5 to 3 (i.e., before administration with CRISPR-Cas9engineered T cells) and a single infusion of 1 108 manufactured CRISPR-Cas9engineered T cells per kilogram on day 0 of the protocol (fig. S2). No cytokines were administered to the patients.

MM, multiple myeloma; BM, bone marrow; XRT, radiation therapy; ASCT, autologous hematopoietic stem cell transplant; ND, not done.

The T cell product was manufactured by electroporation of ribonucleoprotein complexes (RNPs) comprising recombinant Cas9 loaded with equimolar mixtures of single guide RNA (sgRNA) for TRAC, TRBC, and PDCD1 followed by lentiviral transduction of the transgenic TCR (Fig. 1A). All products were expanded to >1 1010 T cells by the time of harvest (Fig. 1B). The transgenic TCR could be detected by flow cytometric staining for V8.1 or dextramer staining, ranging from 2 to 7% of T cells in the final product (Fig. 1C). The frequency of editing, as determined by digital polymerase chain reaction (PCR), varied according to the sgRNA and was about 45% for TRAC, 15% for TRBC, and 20% for PDCD1 (Fig. 1D). Final product transduction efficiency, CD4:CD8 ratio, and dosing are shown in table S2.

(A) Schematic representation of CRISPR-Cas9 NYCE T cells. (B) Large-scale expansion of NYCE T cells. Autologous T cells were transfected with Cas9 protein complexed with sgRNAs (RNP complex) against TRAC, TRBC (i.e., endogenous TCR deletion), and PDCD1 (i.e., PD-1 deletion) and subsequently transduced with a lentiviral vector to express a transgenic NY-ESO-1 cancer-specific TCR. Cells were expanded in dynamic culture for 8 to 12 days. On the final day of culture, NYCE T cells were harvested and cryopreserved in infusible medium. The total number of enriched T cells during culture is plotted for all four subjects (UPN07, UPN27, UPN35, and UPN39). (C) NY-ESO-1 TCR transduction efficiency was determined in harvested infusion products by flow cytometry. Data are gated on live CD3-expressing and V8.1- or dextramer-positive lymphocytes and further gated on CD4-positive and/or CD8-positive cells. (D) The frequencies of TRAC, TRBC, and PDCD1 gene-disrupted total cells in NYCE infusion products were measured using chip-based digital PCR. All data are representative of at least two independent experiments. Error bars represent mean SEM.

The potency of the final engineered T cells was assessed by coculture with HLA-A2+ tumor cells engineered to express NY-ESO-1 (Fig. 2A). The engineered T cells had potent antigen-specific cytotoxicity over a wide range of effector-totarget cell ratios. Interestingly, the cells treated with CRISPR-Cas9 were more cytotoxic than control cells transduced with the TCR but electroporated without CRISPR-Cas9 (i.e., cells that retained endogenous TCR). This is consistent with previous findings in mouse T cells, when a transgenic TCR was inserted into the endogenous locus, ablating expression of the endogenous TCR (15). Further studies will be required to determine if PD-1 knockout contributes to the increased potency afforded by knockout of the endogenous TCR.

(A) Cytotoxicity of NYCE T cells cocultured with HLA-A*0201positive Nalm-6 tumor cells engineered to express NY-ESO-1 and luciferase. Patient T cells transduced with the NY-ESO-1 TCR without CRISPR-Cas9 editing (NY-ESO-1 TCR) and untransduced T cells with CRISPR-Cas9 editing of TRAC, TRBC, and PDCD1 (labeled CRISPR) were included as controls (n = 4 patient T cell infusion products). Asterisks indicate statistical significance determined by paired Students t tests between groups (*P < 0.05). Error bars represent SEM. (B) Levels of soluble interferon- produced by patient NYCE T cell infusion products (labeled NYCE) after a 24-hour coculture with anti-CD3 and anti-CD28 antibody-coated beads or NY-ESO-1expressing Nalm-6 target cells. Patient NY-ESO-1 TCRtransduced T cells (NY-ESO-1 TCR) and untransduced, CRISPR-Cas9edited T cells (labeled CRISPR) served as controls. Error bars represent SEM. (C) Quantification of residual Cas9 protein in NYCE T cell infusion products in clinical-scale manufacturing is shown over time. Asterisks indicate statistical significance determined by paired Students t tests between time points (*P < 0.05). (D) Results from the fluorescence-based indirect ELISA screen performed to detect antibodies against Cas9 protein in the sera of three patients treated with NYCE T cells. Each dot represents the amount of anti-Cas9 signals detected in patient serum before T cell infusion (indicated by a vertical black arrow) and at various time points after NYCE T cell transfer. RFU, relative fluorescent units. (E) Immunoreactive Cas9-specific T cells in baseline patient leukapheresis samples were detected. Representative flow cytometry plots (left) from two patients whose T cells were positive for interferon- in response to Cas9 peptide stimulation. Unstimulated T cells treated with vehicle alone (dimethyl sulfoxide, DMSO) served as a negative control, whereas matched T cells stimulated with phorbol myristate acetate (PMA) and ionomycin served as a positive control. Bar graphs (right) show the frequency of ex vivo CD4+ and CD8+ T cells from patients or healthy donor controls (n = 6) that secrete interferon- in response to stimulation with three different Cas9 peptide pools. The background frequency of interferon-expressing T cells (unstimulated control group, DMSO alone) is subtracted from the values shown in the bar graph. Error bars represent SD.

We developed a sensitive immunoassay for detection of Streptococcus pyogenes Cas9 protein and quantified Cas9 early in the manufacturing process, showing declining levels that were <0.75 fg per cell in the harvested final product (Fig. 2C). Using a competitive fluorescence enzyme-linked immunosorbent assay (ELISA) screen, we found that healthy donors have humoral reactivity to Cas9 in serum (data not shown) and T cells (Fig. 2E), confirming previous reports (2830). Interestingly, we found that the three patients tested at a variety of time points after infusion of the engineered T cells did not develop humoral responses to Cas9. The lack of immunization to Cas9 is consistent with the extended persistence of the infused cells (Fig. 3) and could be a consequence of the low content of Cas9 in the infused product and/or to the immunodeficiency in the patients as a result of their extensive previous treatment histories (Table 1).

(A) The total number of vector copies per microgram of genomic DNA of the NY-ESO-1 TCR transgene in the peripheral blood (UPN07, UPN35, and UPN39), bone marrow (UPN07 and UPN35; multiple myeloma), and tumor (UPN39; sarcoma) is shown pre and postNYCE T cell infusion. (B) Calculated absolute numbers of NY-ESO-1 TCRexpressing T cells per microliter of whole blood from the time of infusion to various postinfusion time points in the study are shown. The limit of detection is about 2.5 cells per microliter of whole blood. (C) Frequencies of CRISPR-Cas9edited T cells (TRAC, TRBC, and PDCD1 knockout) before and after adoptive cell transfer are depicted. Error bars represent SD.

Three patients with advanced, refractory cancer were given infusions of the CRISPR-Cas9engineered T cells. The infusions were well tolerated, with no serious adverse events (Table 2); importantly, there were no cases of cytokine release syndrome, which is a potentially life-threatening systemic inflammatory response that has been associated with cancer immunotherapies (31). All three patients were infused with 1 108 cells/kg, and, owing to the considerable variation in TCR transduction efficiencies (table S2), the absolute number of infused engineered T cells ranged from 6.0 107 to 7.1 108 cells. Despite the variation in engineered cells, there were high peak levels and sustained persistence of the engineered cells in the blood of all three patients (Fig. 3A). The peak and steady-state levels of engineered cells were lowest in patient UPN35, who also had the lowest transduction efficiency (table S2). The persistence of the transduced cells is notably stable from 3 to 9 months after infusion, varying from 5 to 50 cells per microliter of blood (Fig. 3B). Using a subject-specific piecewise linear model, the decay half-lives of the transduced cells were 20.3, 121.8, and 293.5 days for UPN07, UPN35, and UPN39, respectively. The average decay half-life was 83.9 days (15 to 153 days, 95% confidence interval) for the three subjects, as estimated by a piecewise linear mixed-effects model that assumes cells decay linearly from day 14 postexpansion and random effects to allow varying level of expansion (or peak values) across subjects. The stable engraftment of our engineered T cells is notably different from previously reported trials with NY-ESO-1 TCRengineered T cells, in which the half-life of the cells in blood was ~1 week (11, 32, 33). Biopsy specimens of bone marrow in the myeloma patients and tumor in the sarcoma patient demonstrated trafficking of the engineered T cells to the tumor in all three patients at levels approaching those in the blood compartment (Fig. 3A).

indicates no adverse event.

To determine the engraftment frequency of the CRISPR-Cas9 gene-edited cells, we initially used chip-based digital PCR. With this assay, engraftment of cells with editing at the TRAC and PDCD1 loci was evident in all three patients (Fig. 3C). There was sustained persistence of TRAC and PDCD1 edits in patients UPN39 and UPN07 at frequencies of 5 to 10% of circulating peripheral blood mononuclear cells (PBMCs), whereas TRBC-edited cells were lowest in frequency and only transiently detected. The low-level engraftment of TRBC-edited cells is likely related to the observation that this locus had the lowest level of editing efficiency in our preclinical studies (25) and in the harvested products (Fig. 1D).

On- and off-target editing efficiency was assessed in the NYCE cells at the end of product manufacturing. Details of the analysis for UPN07 are shown as an example in Fig. 4, with detailed analysis of the other three manufactured products shown in table S3. The average on-target CRISPR-Cas9 editing efficiency for all engineered T cell products for each target is shown in Table 3. We used iGUIDE (34), a modification of the GUIDE sequencing (GUIDE-seq) method (35), to analyze the Cas9-mediated cleavage specificity. A complication of assays to assess repair by nonhomologous end joining (NHEJ) is that DNA double-strand breaks are formed spontaneously during cell division at high rates in the absence of added nucleases (36), which can increase the background in assays of off-target cleavage. The distribution of on- and off-target cleavage is expected to vary for the three sgRNAs that were used in the manufacturing process (fig. S1A). Of the three sgRNAs, there were more off-target mutations identified for TRBC than for the other loci (Fig. 4C and figs. S4 and S5). The sgRNA for PDCD1 was the most specific, because very few off-target edits were identified in more than 7000 sites of cleavage and there were very few off-target reads identified at the TRAC1 and TRAC2 loci (Fig. 4C).

(A) Genomic distribution of oligonucleotide (dsODN) incorporation sites, which mark locations of double-strand breaks. The ring indicates the human chromosomes aligned end to end, plus the mitochondrial chromosome (labeled M). The targeted cleavage sites are on chromosomes 2, 7, and 14. The frequency of cleavage and subsequent dsODN incorporation is shown on a log scale on each ring (pooled over 10-Mb windows). The purple innermost ring plots all alignments identified. The green ring shows pileups of three or more overlapping sequences, the blue ring shows alignments extending along either strand from a common dsODN incorporation site (flanking pairs), and the red ring shows reads with matches to the gRNA (allowing <6 mismatches) within 100 bp (target matched). (B) Distribution of inferred positions of cleavage and dsODN incorporation at an on-target locus. Incorporations in different strand orientations are shown on the positive (red) and negative (blue) y axis. The percentage in the bottom right corner is an estimate of the number of incorporations associated with the on-target site (based on pileups) captured within the allowed window of 100 bp. (C) Sequences of sites of cleavage and dsODN incorporation are shown, annotated by whether they are on target or off target (Target); the total number of unique alignments associated with the site (Abund.); and an identifier indicating the nearest gene (Gene ID). An asterisk after the gene name indicates that the site is within the transcription unit of the specific gene, whereas ~ indicates that the gene appears on the allOnco cancer-associated gene list.

The genomic localization of identified DNA cleavage sites was as expected, given the chromosomal location of the three targeted genes on chromosomes 2, 7, and 14 (Fig. 4A). The distribution of the incorporation of the double-stranded oligodeoxynucleotide (dsODN) label around on-target sites, based on pileups within a window of 100 base pairs (bp), is shown in Fig. 4B and fig. S4. Although most mutations were on target, there were off-target mutations identified (Fig. 4C and fig. S5). For the TRAC sgRNA, there were low-abundance mutations within the transcriptional unit of CLIC2 (chloride intracellular channel 2); however, disruption of CLIC2 in T cells is not expected to have negative consequences because it is not reported to be expressed in T cells. For the TRBC sgRNA, off-target edits were identified in genes encoding a transcriptional regulator (ZNF609) and a long intergenic nonprotein coding RNA (LINC00377) (table S3). In addition to the above post hoc investigations of multiplex editing specificity, all products were shown not to have cellular transformation by virtue of the absence of long-term growth before infusion (table S1).

In addition to the above detection of repair of double-strand DNA breaks by NHEJ, on-target mutagenesis by engineered nucleases can result in deletions, duplications, inversions, and translocations and can also lead to complex chromosomal rearrangements under some conditions (37). CRISPR-Cas9 has been used to intentionally create oncogenic chromosomal rearrangements (38). In preclinical studies with human T cells, simultaneous gene editing of TRAC and CD52 using TALENs led to translocations that were detected at frequencies of 104 to 102 (39). In a subsequent clinical report using dual-gene editing with TALENs, chromosomal rearrangements were observed in 4% of infused cells (40). To study the safety and genotoxicity of multiplex CRISPR-Cas9 genome editing on three chromosomes, we used stringent release criteria of the manufactured cells and assays to detect translocations (fig. S6). We developed and qualified quantitative PCR (qPCR) assays to quantify the 12 potential translocations that could occur with the simultaneous editing of four loci: TRAC, TRBC1, TRBC2, and PDCD1 (see materials and methods). We observed translocations in all manufactured products; however, the translocations were at the limit of detection for the assay in patient UPN39 (Fig. 5A). TRBC1:TRBC2 was the most abundant rearrangement (Fig. 5A), resulting in a 9.3-kb deletion (supplementary materials). The deletion and translocations peaked on days 5 to 7 of manufacturing and then declined in frequency until cell harvest. The translocations and the TRBC1:TRBC2 deletion were evident in the three patients between 10 days after infusion and 30 to 170 days after infusion (Fig. 5B). However, the rearrangements declined in frequency in vivo, suggesting that they conferred no evidence of a growth advantage over many generations of expansion in the patients on this trial (Fig. 3, A and B). At days 30, 150, and 170 in patients UPN07, UPN35, and UPN39, respectively, chromosomal translocations were at the limits of detection or not detected for all rearrangements except for the 9.3-kb deletion for TRBC1:TRBC2.

(A) Evaluation of chromosomal translocations in NYCE T cell infusion products during the course of large-scale culture is shown. For the 12 monocentromeric translocation assays conducted, a positive reference sample that contains 1 103 copies of the synthetic template plasmid was evaluated as a control, and the percent difference between expected and observed marking was calculated. The absence of amplification from the 12 reactions that correspond to the different chromosomal translocations indicates assay specificity (see methods). (B) Longitudinal analysis of chromosomal translocations in vivo in three patients pre and postNYCE T cell product infusion is displayed. In (A) and (B), error bars represent SD. For graphical purposes, the proportions of affected cells were plotted on a log scale; a value of 0.001% indicates that translocations were not detected.

We used single-cell RNA sequencing (scRNA-seq) to comprehensively characterize the transcriptomic phenotype of the NYCE T cells and their evolution over time in patient UPN39 (fig. S7). UPN39 was chosen because they had the highest level of cell engraftment and because this patient had evidence of tumor regression. CRISPR-Cas9engineered T cells were infused to patient UPN39 and recovered after infusion from the blood on day 10 and at ~4 months (day 113) and were analyzed by scRNA-seq, as described in the materials and methods. For each sample (infusion product, day 10 and day 113), T cells were sorted on the basis of expression of CD4 or CD8 and processed using droplet-based 5 scRNA-seq. From the gene expression libraries, PCR was used to further amplify cellular cDNA corresponding to the NY-ESO-1 TCR transgene, as well as TRAC, TRBC, and PDCD1 target sequences, allowing us to genotype single cells as wild type or mutant. In the infusion product, cells were identified that contained mutations in all three target sequences (Fig. 6, A and B). The most commonly mutated gene was TRAC. About 30% of cells had no mutations identified, whereas ~40% had one mutation, and ~20 and ~10% of the T cells in the manufactured product were double mutated and triple mutated, respectively, at the target sequences. Of the transgenic TCR+ cells in the infusion product, monogenic mutations were less frequent than digenic and trigenic mutations (Fig. 6A). Single-cell genotyping of UPN39 cells at 10 days and 4 months after infusion showed a decline in the frequency of gene-edited T cells from the levels in the infusion product, and this decline occurred regardless of whether the cells were transduced with the NY-ESO-1 TCR (Fig. 6C). The frequency of gene-edited cells was quite stable between day 10 and 4 months postinfusion, and notably, about 40% of the peripheral bloodcirculating T cells in this patient 4 months after infusion were mutated at any one of the targeted genes (Fig. 6, B and C, and table S4).

(A) Venn diagram showing relative numbers of NY-ESO-1 TCRpositive cells with TRAC, TRBC, and/or PDCD1 mutations in the NYCE T cell infusion product (IP) (day 0). (B) Proportions of preinfusion (IP, day 0) and postinfusion (days 10 and 113) wild-type T cells with TRAC, TRBC, or PDCD1 mutations or expressing the NY-ESO-1 TCR transgene. Numbers of cells belonging to each of these categories are listed below the graph. (C) Analysis of NY-ESO-1 TCRpositive (right) and NY-ESO-1 TCRnegative (left) cells without mutations (wild type) or with single, double, or triple mutations at day 0 (NYCE T cell infusion product) and day 113 postNYCE T cell infusion. Numbers of analyzed cells for each time point are listed above the bars. (D) Uniform manifold approximation and projection (UMAP) plots of gene expression data. Analysis was performed on all T cells integrated across time points, but only NY-ESO-1 TCRexpressing cells, split by time point, are shown (top). The increase in TCF7 expression is indicative of an acquired central memory phenotype (bottom, same cells). (E) Heatmap showing scaled expression of differentially expressed genes in NY-ESO-1 TCRpositive T cells across time points. Color scheme is based on scaled gene expression from 2 (purple) to 2 (yellow).

Of particular interest is the frequency and evolution of PD-1deficient T cells owing to the previous mention that genetic disruption of PDCD1 in CAR and TCR T cells enhances antitumor efficacy in preclinical models (19, 2124). We found that ~25% of the T cells expressing the NY-ESO-1 TCR in the infusion product had mutations in the PDCD1 locus (fig. S8). It is interesting that the frequency of cells with edits in the PDCD1 locus decreased to ~5% of the cells expressing the transgenic TCR at 4 months postinfusion. This would be consistent with mouse studies of chronic infection in which PD-1deficient T cells are less able to establish memory (18).

Figure 6D shows the distribution of engineered T cells expressing the NY-ESO-1 TCR transgene in the infusion product of patient UPN39, and again at 4 months in vivo as they evolve from the infused cells. In the heatmap (Fig. 6E), the most differentially expressed genes in the cells expressing the NY-ESO-1 transcript at the various time points are shown in table S5. Notably, UPN39 had increases in expression of genes associated with central memory (IL7R and TCF7) over time (Fig. 6, D and E, and table S4). This is in marked contrast to the recently published results with NY-ESO-1 T cells in the absence of genome editing, in which the infused transgenic T cells evolved to a terminally differentiated phenotype and displayed characteristics of T cell exhaustion in cancer patients (12).

The clinical course of the three infused cancer patients is shown in Fig. 7 (and described in the materials and methods). No patient experienced cytokine release syndrome or overt side effects attributed to the cell infusion (table S5). The best clinical responses were stable disease in two patients. UPN39 had a mixed response, with a ~50% decrease in a large abdominal mass that was sustained for 4 months (Fig. 7D), although other lesions progressed. As of December 2019, all patients have progressed: Two are receiving other therapies, and UPN07 died from progressive myeloma.

(A) Swimmers plot describing time on study for each patient, duration of follow-up off study (defined as survival beyond progression or initiation of other cancer therapy), and present status (differentially colored) is shown. Arrows indicate ongoing survival. SD, stable disease; PD, progressive disease. (B) Changes in kappa light chain levels (mg/liter 103) in patient UPN07 after NYCE T cell product infusion are depicted. Vertical black arrow indicates initiation of a D-ACE salvage chemotherapy regimen (defined as intravenous infusion of cisplatin, etoposide, cytarabine, and dexamethasone). (C) Longitudinal M-spike levels (g/dl) in patient UPN35 postNYCE T cell product administration are shown. Vertical black arrows indicate administration of combination therapy with elotuzumab, pomalidomide, and dexamethasone. (D) Computed tomography scans demonstrating tumor regression in patient UPN39 after administration of an autologous NYCE T cell infusion product. Radiologic studies were obtained before therapy and after adoptive transfer of NYCE T cells. Tumor is indicated by red X. (E) Cytolytic capacity of NY-ESO-1specific CD8+ T cells recovered at the indicated month after infusion and expanded from patients is shown. PBMC samples collected after NYCE T cell product infusion were expanded in vitro in the presence of NY-ESO-1 peptide and interleukin-2. The ability of expanded effector cells to recognize antigen and elicit cytotoxicity was tested in a 4-hour 51Cr release assay incorporating Nalm-6 NY-ESO-1+, parental Nalm-6 (NY-ESO-1), and A375 melanoma cells (NY-ESO-1+). All target cell lines were HLA-A*02 positive. Assays were performed in triplicate, and error bars represent SD.

Biopsies of bone marrow and tumor showed trafficking of the NYCE-engineered T cells to the sites of tumor in all three patients (Fig. 3A). It is interesting to note that even though the tumor biopsies revealed residual tumor, in both patients with myeloma, there was a reduction in the target antigens NY-ESO-1 and/or LAGE-1 (fig. S9). The reduction of target antigen was transient in patient UPN07 and persistent in patient UPN35. This result is consistent with an on-target effect of the infused cells, likely resulting in tumor editing (41).

To determine whether the NYCE cells retained antitumor activity after infusion, samples of blood obtained from patients 3 to 9 months after infusion were expanded in culture in the presence of NY-ESO-1 peptide and assessed for cytotoxicity against tumor cells (Fig. 7E and fig. S10). Antigen-specific cytotoxicity was observed in all three patients. It is interesting to note that the most potent antitumor cytotoxicity was observed in UPN39, because UPN39 was the only patient to have tumor regression after infusion of the CRISPR-Cas9engineered T cells (Fig. 7D).

Our phase 1 first-in-human pilot study demonstrates the initial safety and feasibility of multiplex CRISPR-Cas9 T cell human genome engineering in patients with advanced, refractory cancer. In one patient analyzed at depth, a frequency of 30% of digenic and trigenic editing was achieved in the infused cell population, and 20% of the TCR transgenic T cells in circulation 4 months later had persisting digenic and trigenic edits. We chose to redirect specificity of the T cells with a T cell receptor, rather than a CAR, to avoid the CAR-associated potential toxicities such as cytokine release syndrome (31). This provided a lower baseline toxicity profile, thus enhancing the ability to detect toxicity specifically associated with the CRISPR-Cas9engineering process. We observed mild toxicity, and most of the adverse events were attributed to the lymphodepleting chemotherapy. We note that although the initial clinical results have acceptable safety, experience with more patients given infusions of CRISPR-engineered T cells with higher editing efficiencies, and longer observation after infusion, will be required to fully assess the safety of this approach.

Our large-scale product manufacturing process resulted in gene-editing efficiencies similar to those in our preclinical studies (24). A surprising finding was the high-level engraftment and long-term persistence of the infused CRISPR-Cas9engineered T cells. In previous clinical studies testing adoptively transferred NY-ESO-1 transgenic T cells, the engrafted cells had an initial decay half-life of about 1 week (1012). The explanation for the extended survival that we observed remains to be determined and could include the editing of the endogenous TCR, PD-1, and/or the choice of the TCR and vector design.

The use of scRNA-seq technology permitted the analysis of the transcriptome of the infused NY-ESO-1specific T cells (i.e., CRISPR-Cas9engineered T cells) at baseline and for up to 4 months in vivo. The results shown for UPN39 revealed that the infused cells evolved to a state consistent with central memory. These results are in contrast to a recent study in which the infused NY-ESO-1 T cells evolved to a state consistent with T cell exhaustion (12). A limitation of our in vivo single-cell analysis is that for purposes of feasibility, it is limited to the one patient who had the highest level of engraftment. Another limitation is that we were not able to compare the transcriptional state of the modified cells in the tumor microenvironment with circulating NYCE T cells.

Analysis of the manufacturing process in vitro demonstrated monochromosomal translocations and rearrangements, and some of these persisted in vivo. The translocations were not random in occurrence and occurred most frequently between PDCD1:TRAC and TRBC1:TRBC2. The frequency of translocations that we observed with trigenic editing is similar to that reported for digenic editing using TALEN-mediated gene editing in preclinical and clinical studies, in which rearrangements were detected in about 4% of cells (39, 40). It is important to note that healthy individuals often harbor oncogenic translocations in B and T cells (4244). T cells bearing translocations can persist for months to years without evidence of pathogenicity (4547).

Antagonism of the PD-1:PD-L1 costimulatory pathway can result in organ-specific and systemic autoimmunity (17, 48). PD-1 has been reported to function as a haploinsufficient tumor suppressor in mouse T cells (49). Our patients have had engraftment with PD-1deficient T cells, and to date, there is no evidence of autoimmunity or T cell genotoxicity.

In conclusion, our phase 1 human pilot study has confirmed that multiplex CRISPR-Cas9 editing of the human genome is possible at clinical scale. We note that although the initial clinical results suggest that this treatment is safe, experience with more patients given infusions with higher editing efficiencies and longer observation after infusion will be required to fully assess the safety of this approach. The potential rejection of infused cells due to preexisting immune responses to Cas9 (28, 29) does not appear to be a barrier to the application of this promising technology. Finally, it is important to note that our manufacturing was based on the reagents available in 2016, when our protocol had been reviewed by the National Institutes of Health (NIH) Recombinant DNA Advisory Committee and received approval. Our Investigational New Drug application was subsequently reviewed and accepted by the FDA. There has been rapid progress in the field since that time, with the development of reagents that should increase efficiencies and decrease off-target editing using CRISPR-based technology (50).

The clinical protocol is listed at clinicaltrials.gov, trial NCT03399448. Protocol no. 1604-1524 Phase 1 trial of autologous T cells engineered to express NY-ESO-1 TCR and CRISPR gene edited to eliminate endogenous TCR and PD-1 (NYCE T Cells) was reviewed and approved by the U.S. National Institutes of Health Recombinant DNA Advisory Committee on 21 June 2016. See fig. S1B for clinical trial design. Patient demographics are shown in Table 1. A list of adverse events is depicted in Table 2.

The genomic gRNA target sequences with protospacer adjacent motif (PAM) underlined were: TRAC1 and TRAC2: 5-TGTGCTAGACATGAGGTCTATGG-3, TRBC: 5-GGAGAATGACGAGTGGACCCAGG-3, and PDCD1: 5-GGCGCCCTGGCCAGTCGTCTGGG-3. In vitro transcribed gRNA was prepared from linearized DNA (Aldevron) using Bulk T7 Megascript 5X (Ambion) and purified using RNeasy Maxi Kit (Qiagen).

Cas9 recombinant protein derived from S. pyogenes was TrueCut Cas9 v2 (catalogue no. A36499, ThermoFisher). Cas9 RNP was made by incubating protein with gRNA at a molar ratio of 1:1 at 25C for 10 min immediately before electroporation.

The 8F TCR recognizes the HLA-A*0201 SLLMWITQC epitope on NY-ESO-1 and LAGE-1. The 8F TCR was isolated from a T cell clone obtained from patient after vaccination with NY-ESO-1 peptide. The TCR sequences were cloned into a transfer plasmid that contains the EF-1 promoter, a cPPT sequence, a Rev response element and a woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), as shown in fig. S1B. Plasmid DNA was manufactured at Puresyn, Inc. Lentiviral vector was produced at the University of Pennsylvania Center for Advanced Retinal and Ocular Therapeutics using transient transfection with four plasmids expressing the transfer vector, Rev, VSV-G, and gag-pol, in human embryonic kidney 293T cells.

Go here to read the rest:
CRISPR-engineered T cells in patients with refractory cancer - Science Magazine

Can CRISPR Therapeutics Double Your Money in 2020? – The Motley Fool

CRISPR Therapeutics (NASDAQ:CRSP) has done very well for itself, with the company's stock almost doubling in 2019 as optimism surrounding its gene-editing technology continues to grow. Considering how much of a game-changer gene-editing technology can be for patients with incurable conditions, it makes sense that people are excited.

While shares have tumbled over the past couple of months, this might be a good thing for investors looking to buy this promising stock at a cheaper price. If you're on the fence about CRISPR or are looking for a stock with great upside potential, here are a few reasons why CRISPR looks like a good pick in 2020.

Image source: Getty Images.

CRISPR is arguably the top name in the relatively young gene-editing market right now. The company currently has nine drug candidates, with four either having begun clinical testing or close to starting.

CRISPR's flagship candidate is CTX001, a drug that targets sickle cell disease and transfusion-dependent beta-thalassemia (TBT). Patients with either of these conditions have malformed red blood cells that struggle to deliver oxygen throughout the body. Approximately 300,000 infants are born with sickle cell disease each year, with another 60,000 born annually with TBT.At present, there are no treatments for either condition.

The other three noteworthy candidates in CRISPR's pipeline are CTX110, CTX120, and CTX130. These drug candidates are a type of new cancer-immunology treatment known as a chimeric antigen receptor T cell (CAR-T) therapy. These types of treatments involve modifying a patient's immune cells in a lab to make them better at killing cancer cells. While it's traditionally quite expensive, CRISPR's technology could possibly make these new CAR-T therapies cheaper than the competition.

While the drug is still in early clinical testing, CRISPR reported some success with CTX001 back in November when it announced that two patients had been treated successfully with the drug. The two patients, one diagnosed with sickle cell disease and the other with TBT, managed to eliminate all of their symptoms following a single CTX001 infusion.

In the case of TBT, the number of required blood transfusions dropped to zero, while the sickle cell disease patient experienced zero occlusive crises (blood vessel blockages that occur due to the abnormal shape of the patient's blood vessels).

CRISPR has confirmed that it will be providing more data for both CTX001 and its cancer immunotherapies sometime this year. That means that investors can look forward to further potential catalysts in 2020, likely toward the latter half of the year.

In general, investors shouldn't take too much stock in the financial figures of early-stage biotechstocks unless there's something really alarming going on (like not having enough cash). Revenue figures change dramatically once a drug receives approval, and companies tend to report significant losses until drug candidates reach late clinical stages.

However, CRISPR's situation is different. In its recently released fourth-quarter financial results, the company reported an impressive $77 million in revenue, a substantial improvement from the mere $100,000 seen last year. Annual revenue for 2019 came in at $289.6 million in comparison to 2018's $3.1 million.

While this virtually all comes from CRISPR's collaboration agreement with Vertex Pharmaceuticals, the important point is that CRISPR is now reporting a profit. Net income for the fourth quarter came in at $30.5 million, whereas last year the company saw a net loss of $47.6 million.

Data source: YCharts, CRISPR Therapeutics.

No other notable gene-editing stock out there is reporting a profit right now. Even if CRISPR ends up dipping into a net loss again in subsequent quarters, the fact that the company managed to report a positive net income this early on in its drug development program is impressive.

Given how young the gene-editing industry is and how experimental this technology can be, positive clinical results in this field can have a positive effect on all stocks in the sector. When Intellia Therapeuticspresented new data regarding two of its drug-editing programs earlier in February, shares of all gene-editing stocks -- including CRISPR -- shot up, despite the fact that they are all competitors.

While this might seem strange at first, it makes sense given how young this industry is. Further clinical proof that gene-editing drugs work, no matter where it comes from, is good for the entire sector. A rising tide lifts all ships, and CRISPR investors also should look out for potential catalysts from other gene-editing companies, which could act as an indirect catalyst for CRISPR's stock.

Intellia, Editas Medicine, and Sangamo Therapeuticsare all working on sickle cell disease and transfusion-dependent beta-thalassemia treatments of their own. Positive developments from their treatments could have a spillover effect on CRISPR's stock. Editas stated recently that it expects to file an Investigational New Drug (IND) application for EDIT-301, its sickle cell drug, by the end of 2020.

The answer is yes, it definitely can. CRISPR Therapeutics has plenty of good things going for it, and there is a lot of long-term enthusiasm surrounding both the company and the industry. While shares of CRISPR have fallen a fair bit over the past couple of months -- down 14% since the start of 2020-- so have other gene-editing stocks. As such, it doesn't seem to be as much of a problem with CRISPR in particular as it is a sector-wide phenomenon. Since there's no real news that appears to be behind this decline, I wouldn't worry about it too much.

Instead, now looks like a good time to buy gene-editing stocks, because they're trading at a bit of a discount. Back in November, Oppenheimer analyst Silvan Turkcan issued a price target of $80 for CRISPR Therapeutics, suggesting at least a 57.1% upside to the stock based on current prices. That seems very reasonable, and I wouldn't be surprised if CRISPR does much better than that in 2020.

However, CRISPR still remains a high-risk investment given the fact it's an early-stage biotech stock. If you want to buy shares right now, keep your position on the smaller side. Never risk too much of your portfolio on a single stock, no matter how promising it might seem.

More:
Can CRISPR Therapeutics Double Your Money in 2020? - The Motley Fool

CRISPR Therapeutics Proposes Changes to the Board of Directors – Yahoo Finance

ZUG, Switzerland and CAMBRIDGE, Mass., Feb. 26, 2020 (GLOBE NEWSWIRE) -- CRISPR Therapeutics (CRSP), a biopharmaceutical company focused on creating transformative gene-based medicines for serious diseases, today announced it proposes to elect Doug Treco, Ph.D. to its Board of Directors at the Companys upcoming annual general meeting to be held later this year. The Company also announced that Pablo Cagnoni, M.D., Chief Executive Officer of Rubius Therapeutics, will resign from the Board of Directors to focus on other commitments, effective immediately.

On behalf of our Board of Directors and management team, I would like to thank Pablo for his years of service and his many contributions to CRISPR Therapeutics, and I wish him the best in his future endeavors, said Rodger Novak, M.D., President and Chairman of the Board of CRISPR Therapeutics. We are grateful for his thoughtful guidance and support over the years.

Dr. Novak added: We are excited to invite Doug to our Board during an important time in CRISPR Therapeutics continued evolution. He has an impressive track record of success in advancing the development of numerous drug candidates, with a unique focus on rare disease, gene targeting, and gene therapy. His deep expertise and leadership experience will make him an outstanding addition to our Board, and we look forward to the valuable insights he will bring.

Doug co-founded Ra Pharmaceuticals, Inc. (RARX) in 2008 and has been Chief Executive Officer and a member of the Board of Directors since its inception. Ra Pharma is a leader in macrocyclic peptide and small molecule therapeutics targeting the complement pathway and has advanced its lead molecule, zilucoplan, into the clinic for multiple neuromuscular indications, including an ongoing pivotal Phase 3 study in myasthenia gravis. In October 2019, Ra Pharma entered into a merger agreement with UCB pursuant to which UCB will acquire Ra Pharma. He was an Entrepreneur-in-Residence at Morgenthaler Ventures from January 2008 to May 2014. In 1988, Doug co-founded Transkaryotic Therapies Inc. (TKT), a multi-platform biopharmaceutical company developing protein and gene therapy products, where he led the discovery of a number of approved biopharmaceuticals, including Dynepo, Replagal, Elaprase, and Vpriv. TKT (formerly Nasdaq: TKTX) was acquired by Shire Pharmaceuticals Group plc in 2005. He was a Visiting Scientist in the Department of Molecular Biology at Massachusetts General Hospital and a Lecturer in Genetics at Harvard Medical School from 2004 to 2007. Doug received his Ph.D. in Biochemistry and Molecular Biology from the State University of New York at Stony Brook and performed postdoctoral studies at the Salk Institute for Biological Studies and Massachusetts General Hospital.

About CRISPR TherapeuticsCRISPR Therapeutics is a leading gene editing company focused on developing transformative gene-based medicines for serious diseases using its proprietary CRISPR/Cas9 platform. CRISPR/Cas9 is a revolutionary gene editing technology that allows for precise, directed changes to genomic DNA. CRISPR Therapeutics has established a portfolio of therapeutic programs across a broad range of disease areas including hemoglobinopathies, oncology, regenerative medicine and rare diseases. To accelerate and expand its efforts, CRISPR Therapeutics has established strategic partnerships with leading companies including Bayer, Vertex Pharmaceuticals and ViaCyte, Inc. CRISPR Therapeutics AG is headquartered in Zug, Switzerland, with its wholly-owned U.S. subsidiary, CRISPR Therapeutics, Inc., and R&D operations based in Cambridge, Massachusetts, and business offices in San Francisco, California and London, United Kingdom. For more information, please visit http://www.crisprtx.com.

Important Additional Information and Where to Find ItCRISPR Therapeuticswill file a proxy statement with theUnited States Securities and Exchange Commission(SEC) in connection with the solicitation of proxies for its 2020 annual general meeting (2020 Annual Meeting). SHAREHOLDERS ARE STRONGLY ADVISED TO READ THE PROXY STATEMENT WHEN IT BECOMES AVAILABLE BECAUSE IT WILL CONTAIN IMPORTANT INFORMATION. Shareholders may obtain a free copy of the proxy statement, any amendments or supplements to the proxy statement and other documents thatCRISPR Therapeutics files with theSECfrom the SECs website atwww.sec.govor CRISPR Therapeutics website atwww.crisprtx.comas soon as reasonably practicable after such materials are electronically filed with, or furnished to, theSEC.

Story continues

Certain Information Regarding ParticipantsCRISPR Therapeutics, its directors, nominees for election as director, executive officers and other persons related toCRISPR Therapeutics may be deemed to be participants in the solicitation of proxies from CRISPR Therapeutics shareholders in connection with the matters to be considered at the 2020 Annual Meeting. Information concerning the interests of CRISPR Therapeutics participants in the solicitation is set forth in the materials filed byCRISPR Therapeutics with theSEC, including in its definitive proxy statement filed with theSEConApril 30, 2019, and will be set forth in the proxy statement relating to the 2020 Annual Meeting when it becomes available.

Investor Contact:Susan Kimsusan.kim@crisprtx.com

Media Contact:Rachel EidesWCG on behalf of CRISPR617-337-4167 reides@wcgworld.com

Read more:
CRISPR Therapeutics Proposes Changes to the Board of Directors - Yahoo Finance

Global CRISPR And CRISPR-Associated (Cas) Genes Market Is Set to Boom in 2020,Coming Years – News Times

The Global CRISPR And CRISPR-Associated (Cas) Genes Market report by Globalmarketers.Biz sets out the production, consumption, revenue, gross margin, cost, gross, market share, CAGR, and global market influencing factors of the market for 2020-2025. The segmentation of regional market included the historical and forecast mandates for North America, Europe, Asia-Pacific, Latin America, the Middle East and Africa. The CRISPR And CRISPR-Associated (Cas) Genes Market report provides a far-reaching industry analysis by types, applications, players and regions.

Request a Sample PDF Copy of CRISPR And CRISPR-Associated (Cas) Genes Report Here: https://www.globalmarketers.biz/report/life-sciences/global-crispr-and-crispr-associated-(cas)-genes-market-2019-by-manufacturers,-regions,-type-and-application,-forecast-to-2024/131472 #request_sample

The Top Key Players Are Covered In This Report Are As Follows:

Caribou BiosciencesAddgeneCRISPR THERAPEUTICSMerck KGaAMirus Bio LLCEditas MedicineTakara Bio USAThermo Fisher ScientificHorizon Discovery GroupIntellia TherapeuticsGE Healthcare Dharmacon

Above are the leading companies and brands that are driving the CRISPR And CRISPR-Associated (Cas) Genes Market. The CAGR numbers are looking quite impressive for the forecast period of 2020-2025 in the CRISPR And CRISPR-Associated (Cas) Genes Market. The sales, import, export and revenue figures are also skyrocketing in the forecast period. The key players and brands are making their moves by product launches, their researches, their joint ventures, merges, and accusations and are getting successful results. Complete study compiled with over 100+ pages, list of tables & figures, profiling 10+ companies.

Market Segment by Type, covers

Genome EditingGenetic engineeringgRNA Database/Gene LibrarCRISPR PlasmidHuman Stem CellsGenetically Modified Organisms/CropsCell Line Engineering

Market Segment by Applications, can be divided into

Biotechnology CompaniesPharmaceutical CompaniesAcademic InstitutesResearch and Development Institutes

Market Segment by Regions, regional analysis covers

Enquiry before Buying At https://www.globalmarketers.biz/report/life-sciences/global-crispr-and-crispr-associated-(cas)-genes-market-2019-by-manufacturers,-regions,-type-and-application,-forecast-to-2024/131472 #inquiry_before_buying

CRISPR And CRISPR-Associated (Cas) Genes Market Report Structure at a Glance:

Reasons to Purchase this Report :

Table of Content:

Get Detailed Table Of Cotents : https://www.globalmarketers.biz/report/life-sciences/global-crispr-and-crispr-associated-(cas)-genes-market-2019-by-manufacturers,-regions,-type-and-application,-forecast-to-2024/131472 #table_of_contents

Do You Have Any Question Or Specific Requirement? Ask to Our Industry Expert @ [emailprotected]

Note : You Can Request for the Customisation of Particular Report as per Your Needs. We Ensure You That You Will Get Report As You Want. Thanking You!

Excerpt from:
Global CRISPR And CRISPR-Associated (Cas) Genes Market Is Set to Boom in 2020,Coming Years - News Times

Viewpoint: We can sustainably feed 10 billion people. Here’s how CRISPR and GMO crops can help – Genetic Literacy Project

Agriculture is responsible for the production of a quarter of the total human-generated greenhouse gases. Growing food also uses about 70 percent of the water available to us. Moreover, agriculture (especially meat production) is the single most significant driver of deforestation and biodiversity loss. Food production is detrimental to the health of the planetbut it doesnt end there. Once the food reaches plates, poor-quality diets cause malnutrition, claiming more lives than tobacco, drug and alcohol combined.

Search for malnutrition online and you will see pictures of frail and sick children. But along with stunting, wasting, vitamin and mineral deficiency, malnutrition also includes overweight, obesity and other diet-related illnesses. Yes, 1 in 9 people around the world go to sleep hungry, but nearly 2 billion adults are also overweight or obese. As such, more than one-third of the world population suffers from at least one form of malnutrition.

With the climate and biodiversity crises, and the global public-health crisis in the form of malnutrition, we must find a healthy and environmentally sustainable diet to feed the growing population. In 2019, the EAT-Lancet Commission brought together leading experts in nutrition, health, sustainability and policy to recommend ways to transform the global food system to achieve a healthy and sustainable diet.

The EAT-Lancet report recommends that planetary health diets to feed 10 billion people by 2050 requires cutting down meat consumption by half and eating twice as much as fruits, vegetables, beans and nuts. Despite recognizing the need to make healthy food affordable for the poor, the EAT-Lancet Commission didnt review the cost and affordability of the ideal diet. Therefore, in a recent global study, scientists reviewed prices for nearly 750 food items to calculate the value of healthy and sustainable diets in 159 countries.

The research, published in Lancet Global Health, shows that many people in low and lower-middle-income countries are too poor to afford EAT-Lancets ideal diet. EAT-Lancet says that we would need to eat twice as much as many fruits and vegetables, and get more protein and fats from plant-source foods. However, the new study found that fruits, vegetables, beans and nuts are the most expensive items of the ideal diet accounting for half of its total price.

Shifting foodsystems

A key challenge of the 21st-century is to change our food system to produce a healthy diet that is both economically and environmentally sustainable. As EAT-Lancets ideal diet isnt affordable for much of the worlds low-income population, authorities must make several parallel interventions to tackle global food inequality.

Lower food prices and higher earnings would give poor people more purchasing power. We must also find cheaper, nutritious food alternatives that are affordable and accessible to people living in low-income areas. I believe that biotechnology has the power to lower the cost of locally and globally grown food, making the ideal diet economically viable to those that need it the most.

One problem is the lack of available, affordable options, which partly stems from decreasing agrobiodiversity. Just three crops (rice, wheat and corn) provide over half of the plant-derived calories worldwide. Shifting calories away from the starchy staple foods towards more nutritious fruits, vegetables and other protein-sourced food remains a significant challenge in meeting EAT-Lancet targets. Grand challenges require great technological solutions, and genetic engineering technology is among the most powerful tools at our disposal.

Power of biotechnology

Biotechnology can improve agrobiodiversity and provide more locally-grown food options for people in low-income areas. One way to do this would be to make inedible plants into a good source of nutrition and calories. Take cottonseed, for example, which has the potential to be a cheaper alternative to nuts. Cottonseeds are highly nutritious, containing oils and proteins in abundance, but many low-income cotton farmers cant eat cottonseeds because they produce toxins called gossypol.

Now, scientists have engineered cotton plants to remove the toxin, making cottonseeds safe for us to eat. And recently, the U.S. Food and Drug Administration approved genetically modified (GM) cottonseed for human consumption. Biotech cottonseed can act as an excellent alternative dietary source in low-income regions, where people struggle to meet the costs of the ideal diet recommended by EAT-Lancet.

Genetic engineering can also enable widespread cultivation of local plants. The groundcherry plant in its native form has a wild, sprawling growth habit which causes its fruits to drop to the ground while still small. Difficulties in cultivating the wildcherry mean its an orphan plant. However, scientists used genetic engineering to improve wildcherrys undesirable traits, including the plants weedy shape, flower production and fruit size. Now there are hopes for large-scale cultivation of genetically engineered groundcherry, which is native to Central and South America.

Millions of children and adults around the world suffer from micronutrient deficiencies, and biotechnology can also help fortify current crops to improve their vitamin and micronutrient contents. For example, scientists have recently developed biofortified cassava, which has higher zinc and iron contents than regular cassava. The biofortified cassava may one day prevent illnesses related to iron and zinc deficiencies.

Golden Rice is perhaps the prime example of a biofortified cropconventional rice that is genetically engineered to produce the vitamin A precursor beta-carotene. Golden Rice, acting as a source of vitamin A, can address vitamin A deficiency that blinds and kills hundreds of thousands of children every year. After a rigorous biosafety assessment in the Philippines, the Department of Agriculture-Bureau of Plant Industry found Golden Rice to be safe as conventional rice. Golden Rice regulation application is under review in Bangladesh, as well. This biofortified crop can provide much-needed micronutrients, taking the everyday staple food further to meet peoples dietary requirements in the poorest regions of the world.

Economic benefits

Improved agrobiodiversity and availability of local food varieties, enabled by biotechnology, will bring down the cost of the ideal diet, reducing food inequality. But GM technology also has the power to lift people out of poverty and increase the spending power of the low-income communities in developing regions.

Higher farm productivity, especially in low-income areas, can lower food prices. A meta-analysis of studies published after 1995 found that adopting GM technology has widespread benefits, including economic gains for farmers that grow GM crops. The meta-analysis found that GM technology increases crop yields by 21 percent. Some GM crops are engineered to be more resistant to pest damage, which helps achieve higher yields, for example.

The meta-study also found that GM crops require 37 percent less pesticide, which reduces pesticide costs by 39 percent and helps spare the environment. Even though GM seeds are more expensive than non-GM seeds, savings in pest control and pesticide use mean that farmers adopting GM crops enjoy 68 percent more profit. Therefore, GM crops can increase farmers spending power, which is excellent news for the quarter of the worlds working population employed in agriculture . More importantly, the yield and profit from GM crops are higher in developing countries than in developed countries.

If adopted widely, genetic engineering technology will bring us closer to meeting the EAT-Lancet dietary targets, which will help us protect the environment, public health, and reduce inequality.

Rupesh Paudyal holds a PhD in plant science and covers agriculture and the environment as a freelance writer. Visit his website and follow him on Twitter @TalkPlant

More here:
Viewpoint: We can sustainably feed 10 billion people. Here's how CRISPR and GMO crops can help - Genetic Literacy Project

Gene editing ‘rice revolution’: CRISPR could be used to grow one of the world’s most important crops in salt water – Genetic Literacy Project

Traditional agriculture requires many inputs; fertilizer, specific chemicals, manual labor and water. Most of the water used in agriculture is for irrigation, and some crops require more water to grow than others. Rice is one of the most water-intensive crops, and also one of the most widely consumed worldwide.

Manipulating the rice genome is not entirely new. The Golden Rice Project emerged in 1999 to address the rampant vitamin A deficiency, and resulting blindness in manycountries where rice is a staple food. Other research into increasing photosynthetic efficiency, drought resistance, and methane reduction of rice is in the works as well, and all requires genetic modification.

Opposition to genetically modified organisms (GMOs) in food has halted progress on a project that the founders believe could save billions of people who eat rice every day. GMO use is a divisive topic, and many scientists and companies are choosing to stay away from them to avoid public disdain and regulatory challenges.

Agrisea is taking a different approach to food science. They want to grow rice in the ocean by using gene-editing, which would amplify the expression of genes already found in rice that control salt-tolerance. Salt-tolerant rice could be grown in salty ocean water without the use of soil, fertilizer or fresh water. Rather than inserting genes from other species, they have identified the genes that control for salt expulsion, cellular insulation and DNA protection, and are enhancing the expression of those genes.

Together these genes act in a network, just like they do in nature, Luke Young, CEO and co-founder of Agrisea said. We just encourage them along the pathways that nature has formed in plants that can thrive in a salty environment. The co-founders explained that they could use repeated selective breeding in rice to get the same result, but gene-editing just speeds up the process.

Read the original post

Read more:
Gene editing 'rice revolution': CRISPR could be used to grow one of the world's most important crops in salt water - Genetic Literacy Project

CRISPR and CAS Gene Market Demand Analysis by 2026 | Caribou Biosciences Inc., CRISPR Therapeutics, Mirus Bio LLC, Editas Medicine, Takara Bio Inc.,…

Global CRISPR and CAS Gene Marketreport includes the worlds crucial region market share, size (volume), recent trends including the product profit, value (revenue), price, production, supply/demand, capability utilization, and industry growth rate.

The Global CRISPR and CAS Gene Market2020-2026 Research Report offers extended insights on requisite forecasts of the CRISPR and CAS Gene market trends and macro and micro factors. Also, this report serves to understand the measures that are operating and restraining the requirement and application in the CRISPR and CAS Gene market. However, the research explores the main highlights of the current market trends and gives a prediction for the CRISPR and CAS Gene industry future.

The competitive evaluation of the application market brings monitoring into the product usage types of the present top players. Also, the study highlights characteristic features & CRISPR and CAS Gene price, beneficial reviews on the crucial products in the worldwide market. The report offered key facts and figures on the CRISPR and CAS Gene market statistics, key competitors and is an important source of guidance and business direction and an individuals interests in the CRISPR and CAS Gene industry.

Get Free Sample Copy of Research Report @ https://www.coherentmarketinsights.com/insight/request-sample/2598

This report provides an overview of the CRISPR and CAS Gene industry, including its basic introduction, applications, and advanced manufacturing techniques. So as to get a more extreme view of the market size, the competitive landscape is served. This includes CRISPR and CAS Gene market revenue share (%) by key players (2013-2018) and revenue (in Million USD) by top leading companies (2013-2018).

Competitive Analysis:

The major companies are exceedingly focused on innovation in CRISPR and CAS Gene production technology to enhance ledge life and efficiency. The best long-term development path for CRISPR and CAS Gene market can be caught by guaranteeing financial pliancy to invest in the optimal strategies and current process improvement.

The CRISPR and CAS Gene industry company profile section ofCaribou Biosciences Inc., CRISPR Therapeutics, Mirus Bio LLC, Editas Medicine, Takara Bio Inc., Synthego, Thermo Fisher Scientific, Inc., GenScript, Addgene, Merck KGaA (Sigma-Aldrich), Integrated DNA Technologies, Inc., Transposagen Biopharmaceuticals, Inc., OriGene Technologies, Inc., New England Biolabs, Dharmacon, Cellecta, Inc., Agilent Technologies, and Applied StemCell, Inc.

Each manufacturer or CRISPR and CAS Gene market players growth rate, revenue figures, and gross profit margin is provided in a tabular, simple format for few years and an individual section on CRISPR and CAS Gene market recent development such as collaboration, acquisition, mergers, and any new service or new product launching in the market is offered.

Topographical Study: Europe, US, Japan, Southeast Asia, and Central & South America, China and India.

A detailed profile for more than 10 leading manufacturers is included, along with the financial history, to analyze the latest performance of the CRISPR and CAS Gene market. Latest and revised discussion of major CRISPR and CAS Gene market and influences the market is considered with a thought-provoking qualitative state on CRISPR and CAS Gene market future threats, challenges, and opportunities. This report integrates the best of statistically applicable quantitative data from the CRISPR and CAS Gene industry, along with detailed and relevant qualitative study and comment.

Purchase Copy of This Business Report: https://www.coherentmarketinsights.com/insight/buy-now/2598

Customized country-level and region-wise reports for the following regions:

North America: US, Canada, and Mexico.

South & Central America: Chile, Argentina, and Brazil.

Middle East & Africa: UAE, Turkey, Saudi Arabia, Egypt, and South Africa.

Europe: United Kingdom, France, Spain, Italy, Germany, and Russia.

Asia-Pacific: Japan, India, China, Singapore, South Korea, Indonesia, and Australia.

The following years taken into consideration in this research to forecast the global CRISPR and CAS Gene market size are as follows:

History Year: 2013-2018

Base Year: 2018

Estimated Year: 2019

Forecast Year: 2020 to 2026

Download The Free PDF Brochure: https://www.coherentmarketinsights.com/insight/request-pdf/2598

About Coherent Market Insights:

Coherent Market Insights is a prominent market research and consulting firm offering action-ready syndicated research reports, custom market analysis, consulting services, and competitive analysis through various recommendations related to emerging market trends, technologies, and potential absolute dollar opportunity.

Contact Us:Name: Mr.ShahPhone: US +12067016702 / UK +4402081334027Email:[emailprotected] Visit our Blog: https://hospitalhealthcareblog.wordpress.com/

See the rest here:
CRISPR and CAS Gene Market Demand Analysis by 2026 | Caribou Biosciences Inc., CRISPR Therapeutics, Mirus Bio LLC, Editas Medicine, Takara Bio Inc.,...

Why So Many Biotechs Are Scrambling to Develop a Drug for the Same Rare Disease – The Motley Fool

Nearly 1.8 million Americans were diagnosed with cancer last year. Around the world, the total was close to 17 million. It's not surprising, then, that more than 700 biopharmaceutical companies have experimental cancer drugs in late-stage development.

Beta-thalassemia, on the other hand, is a rare disease that affects around 1,000 or so people in the United States. It's more prevalent in some countries but still impacts only one in 100,000 individuals.

You might expect one or maybe two biotechs could be developing therapies to treat beta-thalassemia. However, by my count, at least half a dozen companies have programs targeting the blood disorder. Why are a disproportionate number of biotechs scrambling to develop drugs for the same rare disease?

Image source: Getty Images.

Probably the main reason why a relatively large group of drugmakers are targeting beta-thalassemia is that the cause of the disease is straightforward. Understanding the why behind a disease is a critical prerequisite to treating it.

Beta-thalassemia is usually caused by a mutation in the HBB gene, which provides instructions on how to build beta-globin proteins. These proteins are part of hemoglobin, the protein in red blood cells that carries oxygen throughout the body. The HBB mutations that cause beta-thalassemia result in dysfunctional red blood cells that can't carry enough oxygen, which leads to patients experiencing anemia.

Another potential reason why biotechs are attracted to beta-thalassemia, though, is that it's not the only disease that is caused by mutations in the HBB gene. Sickle cell disease (SCD) is a related disease where HBB mutations cause red blood cells to form a sickle (or crescent) shape. These misshaped red blood cells can get stuck in blood vessels and cause multiple health complications, including anemia, infections, frequent pain, and heart problems.

While beta-thalassemia is rare, SCD is the most common genetic blood disorder in the U.S. It affects up to 100,000 Americans. SCD is even more prevalent in Africa, impacting up to 3% of newborns in some parts of the continent.

Drugmakers that identify a way to treat beta-thalassemia can be on the right track to target sickle cell disease as well. And with a much larger patient population, the market potential for successful therapies is greater.

One product has already been approved by the FDA for treating beta-thalassemia. Acceleron Pharma (NASDAQ:XLRN) developed luspatercept in collaboration with Celgene. In November 2019, Celgene won FDA approval for luspatercept in treating transfusion-dependent beta-thalassemia. Bristol-Myers Squibb (NYSE:BMY) closed its acquisition of Celgene a few weeks later and is marketing the drug under the brand name Reblozyl. Luspatercept is also in a mid-stage clinical study for treating non-transfusion-dependent beta-thalassemia.

Bluebird bio (NASDAQ:BLUE) won European approval for Lentiglobin in June 2019 for treating transfusion-dependent beta-thalassemia. Lentiglobin is a gene therapy that transplants cells with healthy HBB genes into patients. The biotech launched the therapy in Germany in January with the brand name Zynteglo. Bluebird plans to roll out Zynteglo in other key European markets later this year and should file for U.S. approval within the next few months.

Several biotechs are developing gene-editing approaches to treat beta-thalassemia. The company with the most advanced gene-editing program is Sangamo Therapeutics (NASDAQ:SGMO). However, there are some worries about ST-400, the experimental gene therapy that Sangamo is developing with Sanofi. In December 2019, Sangamo announced preliminary results from an early stage clinical study that, while showing promise, raised safety concerns.

CRISPR Therapeutics (NASDAQ:CRSP) and its big partner, Vertex Pharmaceuticals (NASDAQ:VRTX), are evaluating CTX001 in early stage clinical studies for treating beta-thalassemia and SCD. CTX-001 uses CRISPR gene editing, a different method than the zinc-finger nuclease (ZFN) gene-editing approach that Sangamo uses. CRISPR Therapeutics and Vertex reported promising preliminary results in December 2019 from both of its clinical studies.

Editas Medicine (NASDAQ:EDIT) is also using CRISPR gene editing to target both beta-thalassemia and SCD. The biotech hasn't advanced its experimental therapy to a clinical study in humans yet but plans to file for FDA approval later in 2020 to begin clinical testing. Editas thinks that its gene-editing approach is superior to the ones being taken by CRISPR Therapeutics and Sangamo.

Trailing the pack is Syros Pharmaceuticals (NASDAQ:SYRS). In December, Syros and Global Blood Therapeuticssigned a deal to work together to develop drugs targeting beta-thalassemia and SCD based on Syros' gene control platform. Instead of trying to directly edit the gene mutations, Syros' gene control therapies attempt to control the expression of genes through genomic switches in other parts of DNA. The biotech hasn't said how soon it will be able to advance to clinical testing with its experimental drug.

There are a couple of big problems for investors with so many companies chasing after the same rare disease. First, it's impossible to know which experimental therapies will be successful. Second, if multiple drugs win regulatory approvals, the competition could be so fierce that no product is a huge moneymaker.

It's also important to know that several of the products being developed hold the potential to cure beta-thalassemia. These therapies could wipe out the opportunities for drugs that aren't curative.

One solution to this investor's dilemma is to avoid all of the biotech stocks that are focused on beta-thalassemia. However, that's like throwing the baby out with the bathwater. I think that a better alternative is to invest in the big drugmakers with beta-thalassemia programs.

Bristol-Myers Squibb already has one FDA approval under its belt for Reblozyl. BMS also owns 5.3% of CRISPR Therapeutics and is partnering with Editas on developing gene-editing therapies targeting cancer. Vertex is partnering with CRISPR Therapeutics and owns 10.2% of the small biotech. Both BMS and Vertex stand to win with their beta-thalassemia drugs but also have plenty of other growth drivers.

Excerpt from:
Why So Many Biotechs Are Scrambling to Develop a Drug for the Same Rare Disease - The Motley Fool

UC Professor On How The Monkeyflower Gets Its Spots – Sierra Sun Times

The yellow monkeyflowers distinctive red spots serve as landing pads for bees and other pollinators, helping them access the sweet nectar inside. A new study reveals the genetic programming that creates these attractive patterns. (Image byPollyDotviaPixaBay)

February 24, 2020 - ByKara Manke- The intricate spotted patterns dappling the bright blooms of the monkeyflower plant may be a delight to humans, but they also serve a key function for the plant. These patterns act as bee landing pads, attracting nearby pollinators to the flower and signaling the best approach to access the sweet nectar inside.

They are like runway landing lights, helping the bees orient so they come in right side up instead of upside down, said Benjamin Blackman, assistant professor of plant and molecular biology at the University of California, Berkeley.

In a new paper, Blackman and his group at UC Berkeley, in collaboration with Yaowu Yuan and his group at the University of Connecticut, reveal for the first time the genetic programming that helps the monkeyflower and likely other patterned flowers achieve their spotted glory. Thestudywas published online today (Thursday, Feb. 20) in the journalCurrent Biology.

While we know a good deal about how hue is specified in flower petals whether it is red or orange or blue, for instance we dont know a lot about how those pigments are then painted into patterns on petals during development to give rise to these spots and stripes that are often critical for interacting with pollinators, Blackman said. Our lab, in collaboration with others, has developed the genetic tools to be able to identify the genes related to these patterns and perturb them so that we can confirm whats actually going on.

In the study, the research team used CRISPR-Cas9 gene editing to recreate the yellow monkeyflower patterns found in nature. On the left, a wild-type monkeyflower exhibits the typical spotted pattern. In the middle, a heterozygote with one normal RTO gene and one damaged RTO gene exhibits blotchier spots. And on the right, homozygote with two copies of the damaged RTO gene is all red, with no spots. (UC Berkeley photo by Srinidhi Holalu)

The positions of petals spots arent mapped out ahead of time, like submarines in a game of battleship, Blackman said. Instead, scientists have long theorized that they could come about through the workings of an activator-repressor system, following what is known as a reaction-diffusion model, in which an activator molecule stimulates a cell to produce the red-colored pigment that produces a spot. At the same time, a repressor molecule is expressed and sent to neighboring cells to instruct themnotto produce the red pigment.

The results are small, dispersed bunches of red cells surrounded by cells that keep the background yellow color.

By tweaking the parameters how strongly a cell turns on an inhibitor, how strongly the inhibitor can inhibit the activator, how quickly it moves between cells it can lead to big spots, small spots, striped patterns, really interesting periodic patterns, Blackman said.

In the study, UC Berkeley postdoctoral researcher Srinidhi Holalu and research associate Erin Patterson identified two natural varieties of the yellow monkeyflower one type with the typical red spots in the throat of the flower and a second type with an all-red throat appearing in multiple natural populations in California and Oregon, including at theUC Davis McLaughlin Reserve. In parallel, UConn postdoctoral researcher Baoqing Ding worked with a very similar plant with fully red-throated flowers found when surveying a population of Lewiss monkeyflower that had induced DNA mutations.

When the scientists presented bees in the lab with the two types of monkeyflowers, they preferred the red tongue variety to the spotted variety, though the red tongue variety is less common in nature. (UC Berkeley video by Erin Patterson and Anna Greenlee)

In a previous study, the Yuan lab had found that a gene called NEGAN (nectar guide anthocyanin) acts as an activator in the monkeyflower petals, signaling the cells to produce the red pigment. Through detailed genomic analysis in both monkeyflower species, the two groups were able to pinpoint that a gene called RTO, short for red tongue, acts as the inhibitor.

The red-throated forms of the monkeyflower have defective RTO inhibitor genes, resulting in a characteristic all-red throat, rather than red spots. To confirm their findings, Holalu used the CRISPR-Cas9 gene editing system to knock out the RTO gene in spotted variants of the flower. The result was flowers with a flashy red throat. Further experiments revealed how the functional form of the RTO protein moves to neighboring cells and represses NEGAN to prevent the spread of pigmentation beyond the local spots. This study is the first reported use of CRISPR-Cas9 editing to research the biology of monkeyflowers.

The team also collaborated with Michael Blinov at the UConn School of Medicine to develop a mathematical model to explain how different self-organized patterns might arise from this genetic system.

This work is the simplest demonstration of the reaction-diffusion theory of how patterns arise in biological systems, said Yaowu Yuan, associate professor of ecology and evolutionary biology at UConn. We are closer to understanding how these patterns arise throughout nature.

Monkeyflower plants with the RTO gene knocked out by CRISPR-Cas9 gene editing produce one big patch where all flowers exhibit a fully red throat, in contrast to wild fields where red-tongued flowers appear in small dispersed spots (UC Berkeley photo by Srinidhi Holalu)Source: UC Berkeley

Read more from the original source:
UC Professor On How The Monkeyflower Gets Its Spots - Sierra Sun Times

New CRISPR base-editing technology slows ALS progression in mice – University of Illinois News

CHAMPAIGN, Ill. With a new CRISPR gene-editing methodology, scientists from the University of Illinois at Urbana-Champaign inactivated one of the genes responsible for an inherited form of amyotrophic lateral sclerosis a debilitating and fatal neurological disease for which there is no cure. The novel treatment slowed disease progression, improved muscle function and extended lifespan in mice with an aggressive form of ALS.

ALS unfortunately has few treatment options. This is an important first step in showing that this new form of gene editing could be used to potentially treat the disease, said bioengineering professor Thomas Gaj, who co-led the study with bioengineering professor Pablo Perez-Pinera.

The method relied on an emerging gene-editing technology known as CRISPR base editors.

Traditional CRISPR gene-editing technologies cut both strands of a DNA molecule, which can introduce a variety of errors in the DNA sequence, limiting its efficiency and potentially leading to a number of unintended mutations in the genome. The Illinois group instead used base editing to change one letter of the DNA sequence to another without cutting through both DNA strands, Perez-Pinera said.

Base editors are too large to be delivered into cells with one of the most promising and successful gene therapy vectors, known as adeno-associated virus, Gaj said. However, in 2019, Perez-Pineras group developed a method of splitting the base editor proteins into halves that can be delivered by two separate AAV particles. Once inside the cell, the halves reassemble into the full-length base editor protein.

By combining the power of AAV gene delivery and split-base editors, Gaj and Perez-Pinera targeted and permanently disabled a mutant SOD1 gene, which is responsible for roughly 20% of inherited forms of ALS. They published their results in the journal Molecular Therapy.

Many ALS studies are focused on preventing or delaying the onset of the disease. However, in the real world, most patients are not diagnosed until symptoms are advanced, said graduate student Colin Lim. Slowing progression, rather than preventing it, may have a greater impact on patients. Lim is the co-first author of the study along with graduate students Michael Gapinske and Alexandra Brooks.

CRISPR base editing decreased the amount of a mutant protein (blue) that contributes to ALS in the spinal cord. Left, a spinal cord section from an untreated mouse. Right, a spinal cord section from an animal treated by base editing.

Image courtesy of Thomas Gaj

Edit embedded media in the Files Tab and re-insert as needed.

The researchers first tested the SOD1 base editor in human cells to verify reassembly of the split CRISPR base editor and inactivation of the SOD1 gene. Then they injected AAV particles encoding the base editors into the spinal columns of mice carrying a mutant SOD1 gene that causes a particularly severe form of ALS that paralyzes the mice within a few months after birth.

The disease progressed more slowly in treated mice, which had improved motor function, greater muscle strength and less weight loss. The researchers observed an 85% increase in time between the onset of the late stage of the disease and the end stage, as well as increased overall survival.

We were excited to find that many of the improvements happened well after the onset of the disease. This told us that we were slowing the progression of the disorder, Gapinske said.

The base editor introduces a stop signal near the start of the SOD1 gene, so it has the advantage of stopping the cell from making the malfunctioning protein no matter which genetic mutation a patient has. However, it potentially disrupts the healthy version of the gene, so the researchers are exploring ways to target the genes mutant copy.

Moving forward, we are thinking about how we can bring this and other gene-editing technologies to the clinic so that we can someday treat ALS in patients, Gaj said. For that, we have to develop new strategies capable of targeting all of the cells involved in the disease. We also have to further evaluate the efficiency and safety of this approach in other clinically relevant models.

The split base editor approach has potential for treating other diseases with a genetic basis as well, Perez-Pinera said. Though ALS was the first demonstration of the tool, his group has studies underway applying it to Duchenne muscular dystrophy and spinal muscular atrophy.

The Muscular Dystrophy Association, the Judith and Jean Pape Adams Foundation, the American Heart Association and the National Institutes of Health supported this work. Gaj and Perez-Pinera are affiliated with the Carl R. Woese Institute for Genomic Biology at Illinois. Perez-Pinera also is affiliated with the Carle Illinois College of Medicine and the Cancer Center at Illinois.

View post:
New CRISPR base-editing technology slows ALS progression in mice - University of Illinois News

Some organic farming advocates poised to embrace CRISPR and other New Breeding Techniques because of their sustainability benefits – Genetic Literacy…

There is a new ongoing debate as to what plant breeding technologies constitute genetic modification. The transgenic GM crops introduced more than 20 years ago remain verboten for organic food production. If the pro-organic Cornucopia Institute and other organic food industry proponents have their way, all forms of gene editing and [New Breeding Techniques] would be classified as GM and join the list of practices prohibited for the production of food products eligible to be certified USDA Organic.

We strongly oppose any efforts to revisit the issue of any type of genetic engineering in organic certification, and we will work to ensure that all genetic engineering remains an excluded method, says Organic Farmers Association President David Colson. Any suggestion that we should explore gene-editing or any other type of genetic engineering, would distract from the core issues the organic market is facing right now.

On the other hand, some organic growers do see NBTs as a potential boon to their industry and are calling for revised rules that would allow growers to benefit from crop improvements created using gene editing. Klaas Martens, a prominent voice in the organic movement and a third-generation grain and livestock farmer, operates a 1,600-acre farm in New Yorks Finger Lakes region. He also owns a feed and seed business. Martens says he would be receptive to using CRISPR gene editing technology to grow versions of naturally occurring crops that restore soil health.

If it could be used in a way that enhances the natural system, and mimicked it, then I would want to use it, Martens says. But it would definitely have to be case by case.

The farmers who are opposed to an absolute ban of biotechnology for organic production underscore the belief held by many that USDA Certified Organic crops can help farming become more sustainable as a rising global population demands more food.

In my view, the use of genetic engineering technologies is the most powerful and honest organic tool we have, says Oliver Peoples, president and CEO of Yield10 Bioscience, an agricultural bioscience company focusing on the development of disruptive technologies to produce step-change improvements in crop yield for food and feed crops.

Read the original post

Read this article:
Some organic farming advocates poised to embrace CRISPR and other New Breeding Techniques because of their sustainability benefits - Genetic Literacy...

CRISPR Technology: Global Industry Share, Size, Trends, Growth, Investment Analysis, Development Factors, Future Scope, Challenges and Forecast to…

The research study on Modest recovery in Global CRISPR Technology Market is inclusive of a detailed summary of this industry. A highly focused approach to subjective research has been undertaken, with the description of product scope and elaborate industry insights and outlook until 2025. Introduced by Research Reports Inc., this report delivers information about the product pertaining to the parameters of cost, demand and supply graph, market trends, and the nature of the transaction.

Also, the report is liable to help shareholders and prominent investors understand the demands of customers for efficiently marketing the products and services.

A detailed analysis of the CRISPR Technology market has been provided in the report. The analysis is undertaken on the basis of the overall historical data, valid projections on the market size, qualitative insights, and more. The predictions of this report have been inferred based on conclusive analysis techniques and assumptions. In essence, this research report works like a repository of analysis as well as information for all the aspects of the industry including and not limited to:

A detailed evaluation of the popular trends prevalent in the CRISPR Technology market has been given in the report, in tandem with the microeconmic pointers and regulatory mandates. With this analysis, the report projects the lucrativeness of every market segment over the forecast period, 2020-2025.

Important factors analyzed in worldwide CRISPR Technology market report

Revenue and Sales Estimation: Historical remuneration, as well as sales volume, have been specified in the report this helps in preparing an accurate budget. The data is segmented with the help of bottom-up and top-down approaches to predict the overall market share as well as to calculate forecast numbers for the major geographies in the report in tandem with the key Types and Applications.

Manufacturing Analysis: The report is presently evaluated in terms of the numerous product types and applications. The global CRISPR Technology market study delivers essential highlights of the manufacturing process analysis that has been verified through primaries. These primaries have been collected via industry professionals and also major representatives of all the firms profiled in the report, in order to prepare courses of action to support the industry growth effectively.

Competition: Major contenders have been studied on the basis of their company profile, product/service price, sales, capacity, product portfolio, and cost to find out the present competitors strengths as well as weaknesses.

Demand & Supply and Effectiveness: CRISPR Technology report also delivers information about the production, distribution, consumption & export/import, and break-even point & marginal revenue). ** If applicable

Ask For Customized Report as per Your Business Requirement

Thermo Fisher Scientific, Merck KGaA, GenScript, Integrated DNA Technologies (IDT), Horizon Discovery Group, Agilent Technologies, Cellecta, GeneCopoeia, New England Biolabs, Origene Technologies, Synthego Corporation, Toolgen

Graphically, this report is split into numerous regions, with details on production, consumption, supply, and demand, growth rate, and market share of CRISPR Technology Market in these regions, between 2020 to 2025 (forecast), covering:- North America, Europe, Asia Pacific, Latin America, and Middle East & Africa

Brief introduction about CRISPR Technology Market:

Chapter 1. Global CRISPR TechnologyMarket Size (Sales) Market Share by Type (Product Category) [1,2,3,] in 2020

Chapter 2. CRISPR TechnologyMarket by Application/End Users [1,2,3]

Chapter 3. Global CRISPR TechnologySales (Volume) and Market Share Comparison by Applications

Chapter 4. Global CRISPR TechnologySales and Growth Rate (2020-2025)

Chapter 5. CRISPR TechnologyMarket Competition by Players/Suppliers, Region, Type, and Application

Chapter 6. CRISPR Technology(Volume, Value and Sales Price) structure specified for each geographic region included.

Chapter 7. Global CRISPR TechnologyPlayers/Suppliers Profiles and Sales Data

Chapter 8. Company primary Information and Top Competitors list are being provided for each vendor listed in the report.

Chapter 9. Market Sales, Revenue, Price and Gross Margin (2020-2025) table for each product type which includes Cost Structure Analysis, Key Raw Materials Analysis & Price Trends

Chapter 10. Supply Chain, Sourcing approach and Downstream Buyers, Industrialized Chain Analysis

Directly Buy This Report

Closure: A detailed point-by-point analysis, that contains information on the estimation of the parent market-relevant diversity in market segmentation and market dynamics until the second or third level. Historical, present, and projected market scope from the perspective of cost and capacity. The report also provides details on the reporting as well as interpretation of the latest industry progress, in tandem with market shares and strategies of major players, emerging niche segments as well as regional markets. An objective analysis of the growth curve of the market has been provided, that would guide stakeholders to increase their foothold in the market.

About Research Reports Inc:

Research Reports Inc. is one of the leading destinations for market research reports across all industries, companies, and technologies. Our repository features an exhaustive list of market research reports from thousands of publishers worldwide. We take pride in curating a database covering virtually every market category and an even more comprehensive collection of market research reports under these categories and sub-categories. We are one of the premier sources for such reports & report customization services.

Contact Us:

David ( Sales Manager )

US: +1-855-419-2424

UK : +440330807757

Email: ([emailprotected])

View original post here:
CRISPR Technology: Global Industry Share, Size, Trends, Growth, Investment Analysis, Development Factors, Future Scope, Challenges and Forecast to...

Intellia Therapeutics to Hold Conference Call to Discuss Fourth Quarter and Full-Year 2019 Earnings and Company Update – Yahoo Finance

CAMBRIDGE, Mass., Feb. 20, 2020 (GLOBE NEWSWIRE) -- Intellia Therapeutics, Inc. (NTLA), a leading genome editing company focused on developing curative therapeutics using CRISPR/Cas9 technology both in vivo and ex vivo, will present fourth quarter and full-year 2019 financial results and operational highlights in a conference call on February 27, 2020 at 8 a.m. ET.

To join the call:

U.S. callers should dial 1-877-317-6789 and use conference ID# 10138773, approximately five minutes before the call.

International callers should dial 1-412-317-6789 and use conference ID# 10138773, approximately five minutes before the call.

A replay of the call will be available through the Events and Presentations page of the Investor Relations section of the companys website at http://www.intelliatx.com, beginning on February 27, 2020 at 12 p.m. ET.

About Intellia Therapeutics

Intellia Therapeutics is a leading genome editing company focused on developing proprietary, curative therapeutics using the CRISPR/Cas9 system. Intellia believes the CRISPR/Cas9 technology has the potential to transform medicine by permanently editing disease-associated genes in the human body with a single treatment course, and through improved cell therapies that can treat cancer and immunological diseases, or can replace patients diseased cells. The combination of deep scientific, technical and clinical development experience, along with its leading intellectual property portfolio, puts Intellia in a unique position to unlock broad therapeutic applications of the CRISPR/Cas9 technology and create a new class of therapeutic products. Learn more about Intellia Therapeutics and CRISPR/Cas9 at intelliatx.com and follow us on Twitter @intelliatweets.

Intellia Contacts:

Investor Contact: Lina LiAssociate Director, Investor Relations+1 857-706-1612lina.li@intelliatx.com

Story continues

Media Contact:Jennifer Mound SmoterSenior Vice President, External Affairs & Communications+1 857-706-1071jenn.smoter@intelliatx.com

Visit link:
Intellia Therapeutics to Hold Conference Call to Discuss Fourth Quarter and Full-Year 2019 Earnings and Company Update - Yahoo Finance

Stocks to Follow: Mastercard Incorporated (MA) and CRISPR Therapeutics AG (CRSP) – BOV News

MORGAN STANLEY ASIA LTD. bought a fresh place in Mastercard Incorporated (NYSE:MA). The institutional investor bought 2.3 million shares of the stock in a transaction took place on 12/31/2019. In another most recent transaction, which held on 12/31/2019, SWEDBANK ROBUR FONDER AB bought approximately 1.6 million shares of Mastercard Incorporated. In a separate transaction which took place on 12/31/2019, the institutional investor, GOLDMAN SACHS & CO. LLC (PRIVATE bought 1.3 million shares of the companys stock. The total Institutional investors and hedge funds own 77.30% of the companys stock.

In the most recent purchasing and selling session, Mastercard Incorporated (MA)s share price increased by 0.97 percent to ratify at $344.56. A sum of 3010854 shares traded at recent session and its average exchanging volume remained at 3.31M shares. The 52-week price high and low points are important variables to concentrate on when assessing the current and prospective worth of a stock. Mastercard Incorporated (MA) shares are taking a pay cut of 0.43% from the high point of 52 weeks and flying high of 59.57% from the low figure of 52 weeks.

Mastercard Incorporated (MA) shares reached a high of $347.24 and dropped to a low of $342.6349 until finishing in the latest session at $343.99. Traders and investors may also choose to study the ATR or Average True Range when concentrating on technical inventory assessment. Currently at 5.68 is the 14-day ATR for Mastercard Incorporated (MA). The highest level of 52-weeks price has $343.10 and $215.93 for 52 weeks lowest level. After the recent changes in the price, the firm captured the enterprise value of $344.4B, with the price to earnings ratio of 43.37 and price to earnings growth ratio of 2.44. The liquidity ratios which the firm has won as a quick ratio of 1.40, a current ratio of 1.40 and a debt-to-equity ratio of 1.56.

Having a look at past record, were going to look at various forwards or backwards shifting developments regarding MA. The firms shares rose 4.12 percent in the past five business days and grew 6.46 percent in the past thirty business days. In the previous quarter, the stock rose 22.72 percent at some point. The output of the stock increased 23.91 percent within the six-month closing period, while general annual output gained 56.20 percent. The companys performance is now positive at 15.40% from the beginning of the calendar year.

According to WSJ, Mastercard Incorporated (MA) obtained an estimated Buy proposal from the 36 brokerage firms currently keeping a deep eye on the stock performance as compares to its rivals. 0 equity research analysts rated the shares with a selling strategy, 2 gave a hold approach, 29 gave a purchase tip, 4 gave the firm a overweight advice and 1 put the stock under the underweight category. The average price goal of one year between several banks and credit unions that last year discussed the stock is $359.44.

CRISPR Therapeutics AG (CRSP) shares on Wednesdays trading session, jumped 4.35 percent to see the stock exchange hands at $58.11 per unit. Lets a quick look at companys past reported and future predictions of growth using the EPS Growth. EPS growth is a percentage change in standardized earnings per share over the trailing-twelve-month period to the current year-end. The company posted a value of $0.97 as earning-per-share over the last full year, while a chance, will post -$5.03 for the coming year. The current EPS Growth rate for the company during the year is 134.10% and predicted to reach at -13.40% for the coming year. In-depth, if we analyze for the long-term EPS Growth, the out-come was 54.00% for the past five years.

The last trading period has seen CRISPR Therapeutics AG (CRSP) move -21.47% and 88.98% from the stocks 52-week high and 52-week low prices respectively. The daily trading volume for CRISPR Therapeutics AG (NASDAQ:CRSP) over the last session is 1.35 million shares. CRSP has attracted considerable attention from traders and investors, a scenario that has seen its volume jump 4.33% compared to the previous one.

Investors focus on the profitability proportions of the company that how the company performs at profitability side. Return on equity ratio or ROE is a significant indicator for prospective investors as they would like to see just how effectively a business is using their cash to produce net earnings. As a return on equity, CRISPR Therapeutics AG (NASDAQ:CRSP) produces 11.70%. Because it would be easy and highly flexible, ROI measurement is among the most popular investment ratios. Executives could use it to evaluate the levels of performance on acquisitions of capital equipment whereas investors can determine that how the stock investment is better. The ROI entry for CRSPs scenario is at 4.90%. Another main metric of a profitability ratio is the return on assets ratio or ROA that analyses how effectively a business can handle its assets to generate earnings over a duration of time. CRISPR Therapeutics AG (CRSP) generated 9.60% ROA for the trading twelve-month.

Volatility is just a proportion of the anticipated day by day value extendthe range where an informal investor works. Greater instability implies more noteworthy benefit or misfortune. After an ongoing check, CRISPR Therapeutics AG (CRSP) stock is found to be 5.65% volatile for the week, while 4.66% volatility is recorded for the month. The outstanding shares have been calculated 63.38M. Based on a recent bid, its distance from 20 days simple moving average is 5.27%, and its distance from 50 days simple moving average is -3.80% while it has a distance of 16.08% from the 200 days simple moving average.

The Williams Percent Range or Williams %R is a well-known specialized pointer made by Larry Williams to help recognize overbought and oversold circumstances. CRISPR Therapeutics AG (NASDAQ:CRSP)s Williams Percent Range or Williams %R at the time of writing to be seated at 32.64% for 9-Day. It is also calculated for different time spans. Currently for this organization, Williams %R is stood at 28.28% for 14-Day, 28.28% for 20-Day, 66.19% for 50-Day and to be seated 41.23% for 100-Day. Relative Strength Index, or RSI(14), which is a technical analysis gauge, also used to measure momentum on a scale of zero to 100 for overbought and oversold. In the case of CRISPR Therapeutics AG, the RSI reading has hit 52.87 for 14-Day.

See the rest here:
Stocks to Follow: Mastercard Incorporated (MA) and CRISPR Therapeutics AG (CRSP) - BOV News

Gene Editing Tools Market- increasing demand with Industry Professionals: Thermofisher Scientific, CRISPR Therapeutics – Instant Tech News

A new Profession Intelligence Report released by Stats and Reports with the title Global Gene Editing Tools Market can grow into the most important market in the world that has played an important role in making progressive impacts on the global economy. Global Gene Editing Tools Market Report presents a dynamic vision to conclude and research market size, market hope and competitive environment. The study is derived from primary and secondary statistical data and consists of qualitative and numerical analysis. The main company in this survey is Thermofisher Scientific, CRISPR Therapeutics, Editas Medicine, NHGRI, Intellia Therapeutics, Merck KGaA, Horizon.

Gene editing or Genome editingis a way of making specific changes to the DNA of a cell or organism. An enzyme cuts the DNA at a specific sequence, and when this is repaired by the cell a change or edit is made to the sequence.

Free Sample Report @:www.statsandreports.com/request-sample/340551-global-gene-editing-tools-market-size-status-and-forecast-2019-2025

This report clearly shows that the Gene Editing Tools industry has achieved significant growth since 2018. It is based on an in-depth assessment of the industry. The analysis provided in this report shows the leading segments to gain a strong presence in the industry and the insights that help determine new strategies. In conclusion, analysts who value unbiased information about stakeholders, investors, product managers, marketing executives, and supply, demand, and future predictions value the report.

Preliminary Data:Get raw market data and contrast from wide front. Data is constantly filtered so that only validated and authenticated sources are considered. The data is also collected from many reputable paid databases and many reports in our repository. A comprehensive understanding of the market is essential to understanding and facilitating the complete value chain. We collect data from raw material suppliers, distributors, and buyers.

Furthermore, the years considered for the study are as follows:Historical year 2014-2018Base year 2019Forecast period** 2019 to 2025[** unless otherwise stated]

Research Methodology:The market engineering process uses a top-down and bottom-up approach and several data triangulation methods to evaluate and validate the size of the entire market and other dependent sub-markets listed in this report. Numerous qualitative and quantitative analyzes have been conducted in the market engineering process to list key information / insights. The major players in the market were identified through the second survey and the market rankings were determined through the first and second surveys.

Exclusive Discount Offer on Quick Purchase@www.statsandreports.com/check-discount/340551-global-gene-editing-tools-market-size-status-and-forecast-2019-2025

Crucial Research:During the first survey, we interviewed various key sources of supply and demand to obtain qualitative and quantitative information related to this report. Key supply sources include key industry participants, subject matter specialists from key companies, and consultants from several major companies and organizations active in the digital signage market.

Minor Research:The second study was conducted to obtain key information on the supply chain of the industry, the markets currency chain, pools of major companies, and market segmentation, with the lowest level, geographical market, and technology-oriented perspectives. Secondary data was collected and analyzed to reach the total market size, which was verified by the first survey.

This research many focuses on future market segments or regions or countries to channel efforts and investments to maximize growth and profitability. The report presents an in-depth analysis of key vendors or key players in the market competitive landscape and market.The research provides answers to the following key questions:

What are the Major applications of the Gene Editing Tools Market?Applications cover in these Reports Is:Sickle Cell Disease, Heart Disease, Diabetes, Alzheimers Disease, Obesity and Others

what are the Types of the Gene Editing Tools Market?Types Cover in this Research :Zinc finger nucleases (ZFNs), Transcription Activator-Like Effector-based Nucleases (TALENs), CRISPR-Cas system

Who are the main competitors in the market and what are their priorities, strategies, and developments?Lists of Competitors in Research Is:Thermofisher Scientific, CRISPR Therapeutics, Editas Medicine, NHGRI, Intellia Therapeutics, Merck KGaA, Horizo n

Read Full TOC of Gene Editing Tools Research Study at @www.statsandreports.com/report/340551-global-gene-editing-tools-market-size-status-and-forecast-2019-2025

All percent shares, breaks, and classifications were determined using the secondary sources and confirmed through the primary sources. All parameters that may affect the market covered in this study have been extensively reviewed, researched through basic investigations, and analyzed to obtain final quantitative and qualitative data. This has been the study of key quantitative and qualitative insights through interviews with industry experts, including CEOs, vice presidents, directors and marketing executives, as well as annual and financial reports from top market participants.

Years considered for the study are:Historical year 2014-2018Disreputable year 2019Estimate period** 2019 to 2025 [** unless otherwise stated]

Essentials of Table of Content:

1 Report Overview1.1 Research Scope1.2 Key Market Segments1.3 Target Player1.4 Market Analysis by Type1.5 Market by Application1.6 Learning Objectives1.7 years considered

Buy the Up-to-date Full Report @www.statsandreports.com/placeorder?report=340551-global-gene-editing-tools-market-size-status-and-forecast-2019-2025

2 Global Growth Trends2.1 Global Gene Editing Tools Market Size2.2 Trends of Gene Editing Tools Growth by Region2.3 Corporate trends

3 Gene Editing Tools Market shares by key players3.1 Global Gene Editing Tools Market Size by Manufacturer3.2 Global Gene Editing Tools Key players Provide headquarters and local3.3 Major Players Products / Solutions / Services3.4 Enter the Barriers in the Gene Editing Tools Market3.5 Mergers, acquisitions and expansion plans

4 Market By-products4.1 Global Gene Editing Tools Sales by Product4.2 Global Gene Editing Tools by Product Revenue4.3 Global Gene Editing Tools

Note: Regional Breakdown & Sectional purchase Available We provide Pie chats Best Customize Reports As per Requirements.

About Us

Stats and Reports is a global market research and consulting service provider specialized in offering wide range of business solutions to their clients including market research reports, primary and secondary research, demand forecasting services, focus group analysis and other services. We understand that how data is important in todays competitive environment and thus, we have collaborated with industrys leading research providers who works continuously to meet the ever-growing demand for market research reports throughout the year.

Contact:Stats and ReportsSatish K. (Global Sales Manager)Mangalam Chamber, Office No 16, Paud RoadSankalp Society, Kothrud, Pune, Maharashtra 411038Phone: +1 650-646-3808Email: [emailprotected]Web: https://www.statsandreports.comFollow Us on: LinkedIN| Twitter|

See the rest here:
Gene Editing Tools Market- increasing demand with Industry Professionals: Thermofisher Scientific, CRISPR Therapeutics - Instant Tech News

How The Monkeyflower Gets Its Spots | Berkeley – Patch.com

From the University of California, Berkeley:

The yellow monkeyflower's distinctive red spots serve as 'landing pads' for bees and other pollinators, helping them access the sweet nectar inside. A new study reveals the genetic programming that creates these attractive patterns.

The intricate spotted patterns dappling the bright blooms of the monkeyflower plant may be a delight to humans, but they also serve a key function for the plant. These patterns act as "bee landing pads," attracting nearby pollinators to the flower and signaling the best approach to access the sweet nectar inside.

"They are like runway landing lights, helping the bees orient so they come in right side up instead of upside down," said Benjamin Blackman, assistant professor of plant and molecular biology at the University of California, Berkeley.

In a new paper, Blackman and his group at UC Berkeley, in collaboration with Yaowu Yuan and his group at the University of Connecticut, reveal for the first time the genetic programming that helps the monkeyflower and likely other patterned flowers achieve their spotted glory. The study was published online today (Thursday, Feb. 20) in the journal Current Biology.

"While we know a good deal about how hue is specified in flower petals whether it is red or orange or blue, for instance we don't know a lot about how those pigments are then painted into patterns on petals during development to give rise to these spots and stripes that are often critical for interacting with pollinators," Blackman said. "Our lab, in collaboration with others, has developed the genetic tools to be able to identify the genes related to these patterns and perturb them so that we can confirm what's actually going on."

In the study, the research team used CRISPR-Cas9 gene editing to recreate the yellow monkeyflower patterns found in nature. On the left, a wild-type monkeyflower exhibits the typical spotted pattern. In the middle, a heterozygote with one normal RTO gene and one damaged RTO gene exhibits blotchier spots. And on the right, homozygote with two copies of the damaged RTO gene is all red, with no spots.

The positions of petals' spots aren't mapped out ahead of time, like submarines in a game of battleship, Blackman said. Instead, scientists have long theorized that they could come about through the workings of an activator-repressor system, following what is known as a reaction-diffusion model, in which an activator molecule stimulates a cell to produce the red-colored pigment that produces a spot. At the same time, a repressor molecule is expressed and sent to neighboring cells to instruct them not to produce the red pigment.

The results are small, dispersed bunches of red cells surrounded by cells that keep the background yellow color.

"By tweaking the parameters how strongly a cell turns on an inhibitor, how strongly the inhibitor can inhibit the activator, how quickly it moves between cells it can lead to big spots, small spots, striped patterns, really interesting periodic patterns," Blackman said.

In the study, UC Berkeley postdoctoral researcher Srinidhi Holalu and research associate Erin Patterson identified two natural varieties of the yellow monkeyflower one type with the typical red spots in the throat of the flower and a second type with an all-red throat appearing in multiple natural populations in California and Oregon, including at the UC Davis McLaughlin Reserve. In parallel, UConn postdoctoral researcher Baoqing Ding worked with a very similar plant with fully red-throated flowers found when surveying a population of Lewis's monkeyflower that had induced DNA mutations.

Monkeyflower plants with the RTO gene knocked out by CRISPR-Cas9 gene editing produce one big patch where all flowers exhibit a fully red throat, in contrast to wild fields where red-tongued flowers appear in small dispersed spots.

In a previous study, the Yuan lab had found that a gene called NEGAN (nectar guide anthocyanin) acts as an activator in the monkeyflower petals, signaling the cells to produce the red pigment. Through detailed genomic analysis in both monkeyflower species, the two groups were able to pinpoint that a gene called RTO, short for red tongue, acts as the inhibitor.

The red-throated forms of the monkeyflower have defective RTO inhibitor genes, resulting in a characteristic all-red throat, rather than red spots. To confirm their findings, Holalu used the CRISPR-Cas9 gene editing system to knock out the RTO gene in spotted variants of the flower. The result was flowers with a flashy red throat. Further experiments revealed how the functional form of the RTO protein moves to neighboring cells and represses NEGAN to prevent the spread of pigmentation beyond the local spots. This study is the first reported use of CRISPR-Cas9 editing to research the biology of monkeyflowers.

The team also collaborated with Michael Blinov at the UConn School of Medicine to develop a mathematical model to explain how different self-organized patterns might arise from this genetic system.

"This work is the simplest demonstration of the reaction-diffusion theory of how patterns arise in biological systems," said Yaowu Yuan, associate professor of ecology and evolutionary biology at UConn. "We are closer to understanding how these patterns arise throughout nature."

This press release was produced by the University of California, Berkeley. The views expressed here are the author's own.

Link:
How The Monkeyflower Gets Its Spots | Berkeley - Patch.com

Crispr gene-editing technology: what is it and why you need to know about it – The Irish Times

If youve never heard of Crispr then get ready, because this is a technology youre likely to be hearing a lot more about in future. From genetically modified Chinese babies to foods that deliver man-made health benefits, the potential of Crispr is enormous but its implications have some deep thinkers concerned.

Announced in 2012, Crispr stand for Clustered Regularly Interspaced Short Palindromic Repeats a scientific name that doesnt do much to tell the average person what it does. However the technology represents an astonishing breakthrough, allowing scientists to essentially edit genes and change aspects of DNA in ways previously thought impossible.

Late last year, gene editing was voted to be the innovation of the last decade by readers of The Irish Times, beating out social media, the cloud and even smartphones.

So just what is it and how can something so low-key be so significant?

Crispr (pronounced crisper) allows users to edit genomes and alter DNA sequences to modify gene function. It can be used to correct genetic defects, treating and preventing the spread of diseases and improving crops. While gene editing was possible before Crispr, it cost an enormous amount of money and was relatively imprecise.

The comparison has been made that old-style gene editing could be likened to a blunt mallet while Crispr is more like a laser beam, capable of surgical accuracy at the level of DNA. The science behind this is predictably complicated, but, in essence, Crispr allows scientists to find a specific bit of DNA inside a cell and then alter that piece of DNA.

It can be used to turn genes on or off without altering their sequence and it means that scientists can alter the DNA of plants, animals and potentially human beings to do any number of things. The most significant include altering the DNA of living people to turn off genes that have resulted in them suffering from genetic disorders, or making changes to an individual persons genome so that they dont pass on any genetic defect.

In Ireland in 2018, scientists from Trinity College discovered a therapy for one of the most common soft tissue cancers using Crispr. Synovial sarcoma affects teenagers and young adults and has survival rates of less than 50 per cent.

Pre-clinical trials in mice have showed that drugs developed using information gained through Crispr were able to target cancerous cells, crucially leaving normal cells alone.

Crispr is a two or three component system that allows you to use specific targeting RNA molecules to direct an enzyme that will cut and edit DNA and in theory you can change the DNA any which way you might want to, said Dr Gerard Brien, senior research fellow researching childhood cancers in the genetics department at Trinity College.

In theory, if a disease is caused by a specific mutation, then you could fix that mutation, he said.

But its still early days. In theory, all sorts of things are possible but the practicalities of how to do these things in a way that is effective and safe were not anywhere near a point of understanding how to do that.

The problem, according to Dr Brien, is that of unwanted off target effects.

Were not yet at the point where we can make one clear and specific change and not unintentionally create other changes at the same time. Its these other unwanted changes that are the problem and that will be the stumbling block in using Crispr ethically in humans for the next twenty years, if not more, he said.

In 2020 though, Crispr has applications outside of medicine. Its already being used to alter the DNA of plants and animals to create new kinds of super-foods, genetically modified (GM) to be healthier and offer advantages to the consumer.

To date, some GM foods have been treated with suspicion by the public because their modifications have mostly been for the benefit of the farmers or retailers tomatoes that last longer on the shelf, for example but instead of this, think of coeliac-friendly GM wheat, rapeseed oil high in beneficial omega-3s and even GM potatoes that dont produce harmful cancer-causing acrylamides when fried.

Crispr is a complicated beast, however.

The problem with making changes to the DNA of a plant, animal or person is that its often quite hard to predict the full range of consequences. Tweak something here, and something over there can be affected without you realising.

Its for this reason that experiments on human beings are considered hugely unethical. But that hasnt stopped everyone from tinkering with the technology. In late 2019 Chinese biophysicist He Jiankui was jailed for three years and fined 3 million yuan (about 393,000) when he was convicted of violating a government ban on experimenting on human embryos.

He claimed to have edited the genes of a set of human twins, known by the pseudonyms Lulu and Nana, to give them protection against HIV, but was globally condemned when news of his actions broke. The Chinese court accused the man of having essentially gone on a glory run, saying the people involved in the experiment had acted in the pursuit of personal fame and gain. Theyve crossed the bottom line of ethics in scientific research and medical ethics.

The consequences of Hes actions are still unknown, but the effects will be permanent. If the twins he experimented on grow up and have children of their own, they will inherit his genetic modifications and potentially introduce a permanent change to the human genome.

One of the issues with the Chinese-born twins is that the germline was edited so they will pass their edits on to future generations and we dont know what the long-term effects of that will be, it could be that these changes to their genes could initiate cancer down the line. We just dont know, said Dr Oliver Feeney, a bioethicist and lecturer in University College Cork.

There is this thing called the precautionary principle which states that you shouldnt do something unless you have all the down-side risks guarded against and you know exactly whats going to happen. The problem is we dont do that in any other context. We generally just bulldoze through life and see what happens.

The question here is should we shape humans and the way they develop in future? Is that ethical? Most people would agree that Crispr-enabled treatments for diseases would be a good thing if theyre effective and safe, but what about enhancements? Were only at the start of the regulatory landscape that will be required to manage this.

Dr Feeney suggests that the concept of gene editing is inherently scary to some people and that can colour the way in which it is viewed.

There is a possibility that we could be too concerned about Crispr and any issues that might arise. Thats not to minimise the risks or anything, but there is a certain level of hype around this technology and perhaps also excessive fear about what it can do, he said.

Some people have gone as far as to suggest that engaging in gene editing opens the door to eugenics and a world of genetic haves and have-nots. Its ironic to consider that this is the same scenario presented in the movie Gattaca and here we are having the conversation for real 20 years later.

Read the original post:
Crispr gene-editing technology: what is it and why you need to know about it - The Irish Times

CRISPR technology opens door to vertical farming of dozens of crops, from strawberries and cucumbers to mango and almond trees – Genetic Literacy…

Do you want it locally grown, water-saving and pesticide-free? Urban agriculture might suit you, with a little help from gene editing. Zachary Lippmans team has already succeeded with Solanaceae fruit crops, optimizing tomatoes and ground-cherries for indoor production (see their paper in Nature Biotechnology).

By targeting three genes (SlER, SPG5, and SP), they made the plants display compact growth habit and early yield. The tomatoes produced were slightly smaller than the wild type, but each plant bore more fruit, and they tasted good.

Commenting the paper in the news and views section, Cathryn O Sullivan and colleagues foresee a whole CRISPR menu coming from urban agriculture in the future. It is unlikely that wheat or rice will ever be grown indoors, but urban farms will be interested in producing any plant that has high value and is eaten fresh.

First of all fruits and vegetables that grow on bushes or vines, such as tomato, strawberry, raspberry, blueberry, cucumber, capsicum, grapes, kiwifruit. Specialist crops such as hops, vanilla, saffron, coffee, and also medicinal or cosmetic crops may come next.

They think that one day even small trees (chocolate, mango, almonds) may be grown indoors. However, for indoor farming to be broadly adopted, the capital and operating costs of climate-controlled farms must be reduced, or they will benefit only the wealthiest communities.

Read the original article

Go here to read the rest:
CRISPR technology opens door to vertical farming of dozens of crops, from strawberries and cucumbers to mango and almond trees - Genetic Literacy...

Caribou Biosciences and ProMab Biotechnologies Announce Sale and Assignment Agreement for Humanized scFv Targeting BCMA | More News | News Channels -…

DetailsCategory: More NewsPublished on Thursday, 20 February 2020 13:06Hits: 157

BERKELEY, CA & RICHMOND, CA, USA I February 19, 2020 I Caribou Biosciences, Inc., a leading CRISPR genome editing company, and ProMab Biotechnologies, Inc., a biotechnology CRO/CDMO specializing in antibody engineering and CAR-T development, today announced a sale and assignment agreement under which Caribou gains access to a ProMab humanized single-chain variable fragment (scFv) targeting the B Cell Maturation Antigen (BCMA) for use in allogeneic engineered cell therapies. Caribou intends to utilize this scFv in the development of its CB-011 program, an allogeneic CAR-T therapy targeting BCMA-positive tumors including multiple myeloma.

We are excited for the opportunity to have access to this highly advanced, humanized molecule and believe it will significantly advance our promising CB-011 CAR-T program, said Steven Kanner, PhD, Chief Scientific Officer of Caribou.

We anticipate that our humanized BCMA scFv will aid greatly in Caribous efforts to further its allogeneic CAR-T program, and hope our technology continues to improve the field of preclinical and clinical stage immunotherapy research by providing broad choices of validated antibodies, said John Wu, MD, Chief Executive Officer of ProMab.

Under the terms of the agreement, ProMab received an upfront payment and is eligible for royalties on net sales of licensed products containing the BCMA scFv.

About Caribou Biosciences, Inc. Caribou is a leading company in CRISPR genome editing founded by pioneers of CRISPR-Cas9 biology. The company is developing an internal pipeline of off-the-shelf CAR-T cell therapies, other gene-edited cell therapies, and engineered gut microbes. Additionally, Caribou offers licenses to its CRISPR-Cas9 foundational IP in multiple fields including research tools, internal research use, diagnostics, and industrial biotechnology. Interested companies may contact Caribou at This email address is being protected from spambots. You need JavaScript enabled to view it.. For more information about Caribou, visit http://www.cariboubio.com and follow the Company @CaribouBio. Caribou Biosciences and the Caribou logo are registered trademarks of Caribou Biosciences, Inc.

About ProMab Biotechnologies, Inc. ProMab Biotechnologies focuses on developing and commercializing mouse, rabbit, and human monoclonal antibodies as well as chimeric antigen receptor-T Cell (CAR-T) products. ProMabs CAR-T platform covers both hematological and solid cancers with intensive in vitro and in vivo pre-clinical validation designed for safer and better treatment. As a CRO in the immunology field for 19 years, ProMab offers standard laboratory procedures and animal studies for antibody discovery through the integration of the newest techniques in antibody library construction, next generation sequencing, unique humanization modeling, high-throughput screening, and artificial intelligence analysis systems. ProMab aims to out-license antibodies validated in CAR-T therapy in the preclinical stage or to bring CAR-T technologies to the early stage market of clinical study. ProMab has partnered with top biotechnology startups, medical institutions, and pharmaceutical companies to advance the development of cell therapies as well as bispecific antibodies targeting multiple cancers. For more information, visit http://www.promab.com.

SOURCE: Caribou Biosciences

Read the original:
Caribou Biosciences and ProMab Biotechnologies Announce Sale and Assignment Agreement for Humanized scFv Targeting BCMA | More News | News Channels -...

A Huge Discovery in the World of Viruses – The Atlantic

Though common, these big phages would have been completely missed by traditional lab techniques. It used to be that scientists could only discover viruses by first growing themand they often filtered out anything above a certain size. In science, you tend to find what you look for. The huge phages dont fit the standard conception of what a virus should be, so no one went looking for them. But Banfield used a different method, which she pioneered in the 1990s: Her team took environmental samplesscoops of soil or drops of waterand simply analyzed all the DNA within to see what popped out. And once Banfield realized that the huge phages existed, it wasnt hard to find more.

Read: Beware the Medusavirus

Her team, including researchers Basem Al-Shayeb and Rohan Sachdeva, identified huge phages in French lakes, in Tibetan springs, and on the Japanese seafloor. They found the viruses in geysers in Utah, salt from Chiles Atacama Desert, stomach samples from Alaskan moose, a neonatal intensive-care unit in Pittsburgh, and spit samples from Californian women. All of these phages have at least 200,000 DNA letters in their genome, and the largest of them has 735,000.

The team included researchers from nine countries, and so named the new viruses using words for huge in their respective languages. Hence: Mahaphage (Sanskrit), Kaempephage (Danish), Kyodaiphage (Japanese), and Jabbarphage (Arabic), but also Whopperphage (American English).

These huge phages have other strange characteristics. With so much DNA, the viruses are probably physically bigger than typical phages, which means that they likely reproduce in unusual ways. When phages infect bacteria, they normally make hundreds of copies of themselves before exploding outwards. But Banfield says that an average bacterium doesnt have enough room to host hundreds of huge phages. The giant viruses can probably only make a few copies of themselves at a timea strategy more akin to that of humans or elephants, which only raise a few young at a time, than to the reproduction of rodents or most insects, which produce large numbers of offspring.

Giant phages also seem to exert more control over their bacterial hosts than a typical virus. All viruses co-opt their hosts resources to build more copies of themselves, but the huge phages seem to carry out a much more thorough and directed takeover, Banfield says. Their target is the ribosomea manufacturing plant found in all living cells, which reads the information encoded in genes and uses that to build proteins. The huge phages seem equipped to fully commandeer the ribosome so that it ignores the hosts genes, and instead devotes itself to building viral proteins.

This takeover involves an unorthodox use of CRISPR. Long before humans discovered CRISPR and used it to edit DNA, bacteria invented it as a way of defending themselves against viruses. The bacteria store genetic snippets of phages that have previously attacked them, and use these to send destructive scissorlike enzymes after new waves of assailants. But Banfields team found that some huge phages have their own versions of CRISPR, which they use in two ways. First, they direct their own scissors at bacterial genes, which partly explains why they can so thoroughly take over the ribosomes of their hosts. Second, they seem to redirect the bacterial scissors into attacking other phages. They actually boost their hosts immune system to take out the competition.

See the rest here:
A Huge Discovery in the World of Viruses - The Atlantic

What is CRISPR? – NBC News

CRISPR is a powerful gene-editing technology that scientists use to change the genetic blueprint of plants and animals and even humans.

Since its development about 10 years ago, its been used to change the DNA of living things in beneficial ways creating pink tomatoes and mushrooms that dont go brown, for example, and crops that resist insect attacks.

This technology operates efficiently in virtually all cell types of organisms in which its been tested, CRISPR co-inventor Jennifer Doudna, a biochemist at the University of California, Berkeley, said in an interview last May. It was really quite amazing how quickly it was possible to harness this technology once it was clear how it operated.

CRISPR (also known as CRISPR/Cas9) could also be used to create human designer babies with specific traits for example, a specific eye color or possibly enhanced intelligence. Most scientists have scrupulously avoided pursuing this controversial line of research, although a Chinese scientist stoked controversy in 2018 when he claimed to have used CRISPR to edit the genes of twin girls before their birth in order to make them immune to HIV, the virus that causes AIDS.

The name CRISPR is an acronym for clustered regularly interspaced short palindromic repeats, but you dont need to understand that brain-boggling term in order to understand how CRISPR works.

In short, it works by identifying a specific strand of DNA for example, the genetic instructions that determine eye color and replacing it by cutting" the original DNA and pasting in replacement DNA.

There are other gene-editing techniques, but they are slow and expensive in comparison to CRISPR. What used to take weeks or months can now be done in days with CRISPR.

Some modern CRISPR gene-editing kits, consisting of a few petri dishes, pipettes and bottles of special proteins, are small enough to keep on a shelf in the fridge and it can take as little as two days to see results.

Beyond creating better crops and hardier farm animals, CRISPR offers the tantalizing prospect of revolutionizing human health by bringing cures for genetic diseases: We are really on the threshold of a technology that is going to enable that to treat it at its source, by correcting the code in the DNA, Doudna said in a recent video.

In a series of experiments conducted a few years ago at the Broad Institute, a biomedical institute of MIT and Harvard in Cambridge, Massachusetts, scientists used CRISPR to improve hearing in mice with a certain form of hereditary deafness.

And in experiments at several University of California campuses published in 2016, researchers fixed defective bone marrow cells in a way that could offer a cure for sickle-cell anemia, a potentially deadly condition that affects an estimated 250 million people around the world.

Scientists are also using CRISPR in an effort to wipe out malaria by creating malaria-resistant mosquitoes, which would replace the wild populations of mosquitoes that spread the disease.

Though scientists see huge potential in CRISPR technology for treating human genetic diseases, theyve generally avoided using CRISPR to edit the genes of human embryos, citing the potential dangers of the technology and the ethical issues that surround its use for that purpose.

The actions of the Chinese researcher, He Jiankui, have drawn stern criticism from scientists and bioethicists, who called the work dangerous, unethical and even amateur. They point out that scientific knowledge of CRISPR and human genetics is far from perfect and that the twin girls could suffer as they grow up from genetic problems created by the CRISPR editing of their genes.

FOLLOW NBC NEWS MACH ON TWITTER, FACEBOOK, AND INSTAGRAM.

Link:
What is CRISPR? - NBC News

Dreaming of hypoallergenic cats and how CRISPR could ‘come to the rescue’ – Genetic Literacy Project

Given that just two genes are responsible for making cats a problem for many people, it seemed like a no-brainer to engineer cats that lacked those genes, or to simply breed cats with versions of the genes that made the animals less allergenic.

But so far, itchy-eyed cat lovers have been left disappointed.

But for all those who havent given up hope, there may be new options around the corner. An allergic owner might pop open a can of allergy-fighting food for the cat. Or maybe vaccinate the cat to produce fewer allergens. And allergy shots for owners might shift from burdensome weekly or monthly injections to a shot that offers immediate relief.

The new gene-editing technology called CRISPR/Cas9 might even come to the rescue, delivering the ultimate dream to those who can afford it: a cat that doesnt produce allergens at all. One company has made some progress applying CRISPR/Cas9 to cats.

Success in taming cat allergies could bring good news for people whose allergies have nothingto do with cats. If any of the cat allergyfighting measures prove safe and effective, they could be deployed against other allergens, especially airborne ones like pollen, dog dander or dust mites.

Read the original post

Visit link:
Dreaming of hypoallergenic cats and how CRISPR could 'come to the rescue' - Genetic Literacy Project

Carl June on CRISPR, CART and how the Vietnam War dropped him into medicine – Endpoints News

In August of 2011, Carl June and his team published a landmark paper showing their CART treatment had cleared a patient of cancer. A year-to-the-month later, Jennifer Doudna made an even bigger splash when she published the first major CRISPR paper, setting off a decade of intense research and sometimes even more intense public debate over the ethics of what the gene-editing tool could do.

Last week, June, whose CART work was eventually developed by Novartis into Kymriah, published in Sciencethe first US paper showing how the two could be brought together. It was not only one of the first time scientists have combined the groundbreaking tools, but the first peer-reviewed American paper showing how CRISPR could be used in patients.

June used CRISPR to edit the cells of three patients with advanced blood cancer, deleting the traditional T cell receptor and then erasing the PD1 gene, a move designed to unleash the immune cells. The therapy didnt cure the patients, but the cells remained in the body for a median of 9 months, a major hurdle for the therapy.

Endpoints caught up with June about the long road both he and the field took to get here, if the treatment will ever scale up, and where CRISPR and other advancements can lead it.

The interview has been condensed and edited.

Youve spoken in the past about howyou started working in this field in the mid-90s after your wife passed away from cancer. What were some of those early efforts? How did you start?

Well, I graduated from high school and had a low draft number [for the Vietnam War] and was going to go to study engineering at Stanford, but I was drafted and went into the Naval Academy in 1971, and I did that so I wouldnt have to go to the rice fields.

The war ended in 73, 74, so when I graduated in 1975, I was allowed to go to medical school, and then I had a long term commitment to the Navy because they paid for the Acadamy and Medical school. And I was interested in research and at the time, what the Navy cared about was a small scale nuclear disaster like in a submarine, and like what happened at Chernobyl and Fukushima. So they sent me to the Fred Hutchinson Cancer Center where I got trained in cancer, as a medical oncologist. I was going to open a bone marrow transplant center in Bethesda because the Navy wanted one in the event of a nuclear catastrophe.

And then in 1989, the Berlin Wall came down and there was no more Cold War. I had gone back to the Navy in 86 for the transplant center, which never happened, so then I had to work in the lab full time. But in the Navy, all the research has to be about combat and casualty. They care about HIV, so my first papers were on malaria and infectious disease. And the first CAR-T trials were on HIV in the mid-90s.

In 96, my wife got diagnosed with ovarian cancer and she was in remission for 3-4 years. I moved to the University of Pennsylvania in 1999 and started working on cancer because I wasnt allowed to do that with the Navy. My wife was obviously a lot of motivation to do that. She passed away in 2001. Then I started working with David Porter on adoptive transfer T cells.

I got my first grant to do CAR-T cells on HIV in 2004, and I learned a whole lot. I was lucky to have worked on HIV because we did the first trials using lentiviruses, which is an engineered HIV virus.

I was trained in oncology, and then because of the Navy forced to work on HIV. It was actually a blessing in disguise.

So if you hadnt been drafted, you wouldve become an engineer?

Yes. Thats what I was fully intending. My dad was a chemical engineer, my brother is an engineer. Thats what I thought I was going to do. No one in my family was ever a physician. Its one of those many quirks of fate.

Back then, we didnt have these aptitude tests. It was just haphazard. I applied to three schools Berkeley, Stanford and Caltech and I got into all three. It was just luck, fate.

And it turned out when I went to the Naval Academy, they had added a pre-med thing onto the curriculum the year before, so thats what I did when I started, I did chemistry.

I wouldve [otherwise] been in nuclear submarines. The most interesting thing in the Navy then was the nuclear sub technology.

You talked about doing the first CAR-T trials on HIV patients because thats where the funding was. Was it always in your head that this was eventually going to be something for cancer?

So I got out of the Navy in 99 and moved to Penn. I started in 98 working on treating leukemia, and then once I got to Penn, I continued working one day a week on HIV.

Its kind of a Back-to-the-Future thing because now cancer has paved out a path to show that CART cells can work and put down the manufacturing and its going to be a lot cheaper making it for HIV. I still think thats going to happen.

Jim Riley, who used to be a postdoc in my lab, has some spectacular results in monkeys with HIV models. They have a large NIH and NIAID research program.

So were going to see more and more of that. The CAR technology is going to move outside of cancer, and into autoimmune and chronic infections.

I want to jump over to cytotoxic release syndrome (CRS)because a big part of the CRISPR study was that it didnt provoke this potentially deadly adverse effect. When did you first become aware that CRS was going to be a problem?

I mean we saw it in the very first patient we treated but in all honesty, we missed it. Im an MD, but I dont see the patient and David Porter tookcare of the first three patients and our first pediatric patient,Emily Whitehead.

In our first patients, 2 out of 3, had complete remission and there were fevers and it was CRS but we thought it was just an infection, and we treated with antibiotics for 3 weeks and[eventually] it went away. And sort of miraculously he was in remission and is still in remission, 9 years later.

And then when we treated Emily. She was at a 106-degree fever over three days, and there was no infection.

Ive told this story before. My daughter has rheumatoid arthritis, and I had been president of the Clinical Immunologists Society from 2009 to 2010, and the first good drug for juvenile rheumatoid arthritisthat came out. I was invited to give the Japanese scientist Tadamitsu Kishimoto the presidential award for inventing the drug.

Then in 2012, Emily Whitehead was literally dying from CRS, she had multiple organ failures. And her labs came back and IL-6 levels were 1000x normal. It turns out the drug I was looking at for my daughter, it blocks IL-6 levels. I called the physician and I said, listen theres something actionable here, since its in your formulary to give it to her off-label.

And she gave her the appropriate dose for rheumatoid arthritis. It was miraculous. She woke up very rapidly.

Now its co-labeled. When the FDA approvedKymriah, it was co-labeled. It kind of saved the field.

How were you feeling during this time? Did you have any idea what was happening to her?

No, not until we got the cytokine levels, and then it was really clear. The cytokine levels go up and it exactly coincided. Then we retroactively checked out adults and they had adverse reactions and it easy to see. We hadnt been on the lookout because it wasnt in our mouse models.

And it appeared with those who got cured. Its one of the first on-target toxicities seen in cancer, a toxicity that happens when you get better. All the toxicities from chemotherapy are off-target: like leukopenia or hair loss.

I had a physician who had a fever of 106, I saw him on a fever when he was starting to get CRS. When the nurse came in and it said 106, they thought the thermometer must be broken. On Monday, I saw him, and said how are you feeling and he said fine. And I looked at the thermometer and histemperature was still 102.

People will willingly tolerate on-target toxicity thats very different from chemotherapy if they know it helps get them better. Thats a new principle in cancer therapy.

You had these early CART results almost at the same time that Doudna publishes the first CRISPR papers, then still in bacteria. When did you first start thinking about combining the two?

Yeah, it was published inSciencein 2012 and thats when Emily Whitehead got treated. Its an amazing thing.

Thats something so orthogonal. You think how in the heck can that ever benefit CART cells? but my lab had done the first edited cells in patients, published in 2012. And we used zinc-fingered nucleases, which were the predecessors to CRISPR. It knocked out one gene at a time, but we showed it was safe.

I was already into gene editing because it could make T cells resistant to HIV. So it was pretty obvious that there were candidates in T cells that you can knock out. And almost every lab started working on some with CRISPR, cause it was much easier.

We were the first to get full approval by the FDA, so we worked on it from 2012, had all the preclinical data by 2016, and then it takes a while to develop a lot of new assays for this as we were very cautious to optimize safety and it took longer than we wanted, but in the end, we learned a tremendous amount.

So what did we learn?

First of all our patients had advanced metastatic cancer and had had a lot of chemotherapy. The first patient had had 3 bone marrow transplants.

One thing is feasibility: could you really do all the complex engineering? So we found out we could. feasibility was passed.

Another was the fact that cas9 came out of bacteria, forms of strep and staph. Everyone has pre-existing immunity to Cas9 and we had experience from the first trial with Sangamo[with zinc-finger nucleases] where some patients had a very high fever. In that case, we had used adenoviruses, and it turned out our patients had very high levels of baseline immune response to adenoviruses, so we were worried that would happen with CRISPR, and it did not happen.

It did not have any toxicity. If it had, it would have really set the field back. If there was animmune response to cas9 and CRISPR, there couldve been a real barrier to the field.

And then, the cells survived in the patients. The furthest on, it was 9 months. The cells had a very high level of survival. In the previous trials, the cells survived less than 7 days. In our case, the half-life was 85 days. We dont know the mechanism yet.

And we found very big precision in the molecular scissors, and that was a good thing for the field. You could cut 3 different genes on 3 different chromosomes and have such high fidelity.

It [CRISPR] is living up to the hype. Its going to fix all these diseases.

Whats the potential in CAR-T, specifically?

Well theres many many genes that you can add. There are many genes that knocking outwill make the cells work better. We started with the cell receptor. There are many, I think, academics and biotechs doing this now and it should make the cells more potent and less toxic.

And more broadly, what else are you looking at for the future of CART? The week before your paper, there were the results from MD Anderson on natural killer cells.

Different cell types, natural killer cells, stem cells putting CAR molecules into stem cells, macrophages. One of my graduate students started a company to do CAR macrophages and macrophages actually eat tumor cells, as opposed to T cells that punch holes in them.

There will be different cell types and there will be many more ways to edit cells. The prime editing and base editing. All different new variations.

Youve talked about how people used to think the immuno-oncology, if it ever worked, would nevertheless be a boutique treatment. Despite all the advancements, Novartis and Gilead still have not met the sales they once hoped to grab from their CART treatments. Are you confident CART will ever be widely accessible?

Oh yeah, Novartis sales are going up. They had a hiccup launching.

Back in 96 or 97, when Genentech launched Herceptin, their commercial antibody, they couldnt meet the demand either and then they scaled up and learned how to do better cultures. So right now Novartis is using tech invented in my lab in the 1990s culture tech thats complex and requires a lot of labor, so the most expensive part is human labor. A lot can be made robotic. The scale problem will be much easier.

Thats an engineering problem that will become a thing of the past. The manufacturing problem will get a lot cheaper. Here in the US, we have a huge problem with how drugs are priced. We have a problem with pricing. Thats a political issue.

But in cell therapy, its just kind of the growth things you see in a new industry. Itll get worked out.

This article has been updated to reflect that Jim Riley conducted work on CAR in HIV.

Originally posted here:
Carl June on CRISPR, CART and how the Vietnam War dropped him into medicine - Endpoints News

Viewpoint: If Europe wants to be ‘carbon neutral,’ it needs to embrace biotechnologyGMO and CRISPR crops included – Genetic Literacy Project

A revolution is taking place in the knowledge base for life sciences and biotechnology, opening up new applications in healthcare, agriculture, and environmental protection. Political awareness of this potential dates back to 2001, when the European Commission recognized life sciences and biotech through the adoption of its life science and biotechnology strategy.

With the European Green Deal, the new European Commission has set out an ambitious roadmap towards a climate neutral continent in 2050. With that, Europe strives to become a global frontrunner and lead the way in tackling the climate crisis. Taking the potential of biotechnology and life sciences in benefiting people and planet, a renewed focus and impetus on life sciences and biotechnology are all the more necessary. Regaining leadership in the sector should be a fundamental priority for the EU.

In agriculture, biotechnology offers sustainable food solutions through applying newest technologies. Biotechnology, (including genetic modification of crops), has increased farmers yields and incomes while reducing CO2 emissions, and the need for farmer inputs. Meanwhile, a science-based, risk-proportionate and non-discriminatory regulatory framework that allows for gene editing in crops could pave the way for products which offer health and consumer benefits ..

Read the original post

Read the rest here:
Viewpoint: If Europe wants to be 'carbon neutral,' it needs to embrace biotechnologyGMO and CRISPR crops included - Genetic Literacy Project

Gene Editing Might Let China Create The Perfect Human Being – The National Interest Online

Key point: Humanity isn't ready for gene editing.

More than a year ago, the world was shocked by Chinese biophysicist He Jiankuis attempt to use CRISPR technology to modify human embryos and make them resistant to HIV, which led to the birth of twins Lulu and Nana.

Now, crucial details have been revealed in a recent release of excerpts from the study, which have triggered a series of concerns about how Lulu and Nanas genome was modified.

How CRISPR works

CRISPR is a technique that allows scientists to make precise edits to any DNA by altering its sequence.

When using CRISPR, you may be trying to knock out a gene by rendering it inactive, or trying to achieve specific modifications, such as introducing or removing a desired piece of DNA.

Read more: What is CRISPR gene editing, and how does it work?

Gene editing with the CRISPR system relies on an association of two molecules. One is a protein, called Cas9, that is responsible for cutting the DNA. The other molecule is a short RNA (ribonucleic acid) molecule which works as a guide that brings Cas9 to the position where it is supposed to cut.

The system also needs help from the cells being edited. DNA damage is frequent, so cells regularly have to repair the DNA lesions. The associated repair mechanisms are what introduce the deletions, insertions or modifications when performing gene editing.

How the genomes of Lulu and Nana were modified

He Jiankui and his colleagues were targeting a gene called CCR5, which is necessary for the HIV virus to enter into white blood cells (lymphocytes) and infect our body.

One variant of CCR5, called CCR5 32, is missing a particular string of 32 letters of DNA code. This variant naturally occurs in the human population, and results in a high level of resistance to the most common type of HIV virus.

The team wanted to recreate this mutation using CRISPR on human embryos, in a bid to render them resistant to HIV infection. But this did not go as planned, and there are several ways they may have failed.

First, despite claiming in the abstract of their unpublished article that they reproduced the human CCR5 mutation, in reality the team tried to modify CCR5 close to the 32 mutation.

As a result, they generated different mutations, of which the effects are unknown. It may or may not confer HIV resistance, and may or may not have other consequences.

Worryingly, they did not test any of this, and went ahead with implanting the embryos. This is unjustifiable.

A second source of errors could have been that the editing was not perfectly efficient. This means that not all cells in the embryos were necessarily edited.

When an organism has a mixture of edited and unedited cells, it is called a mosaic. While the available data are still limited, it seems that both Lulu and Nana are mosaic.

This makes it even less likely that the gene-edited babies would be resistant to HIV infection. The risk of mosaicism should have been another reason not to implant the embryos.

Read more: 'Designer' babies won't be common anytime soon despite recent CRISPR twins

Moreover, editing can have unintended impacts elsewhere in the genome.

When designing a CRISPR experiment, you choose the guide RNA so that its sequence is unique to the gene you are targeting. However, off-target cuts can still happen elsewhere in the genome, at places that have a similar sequence.

He Jiankui and his team tested cells from the edited embryos, and reported only one off-target modification. However, that testing required sampling the cells, which were therefore no longer part of the embryos - which continued developing.

Thus, the remaining cells in the embryos had not been tested, and may have had different off-target modifications.

This is not the teams fault, as there will always be limitations in detecting off-target and mosaicism, and we can only get a partial picture.

However, that partial picture should have made them pause.

A bad idea to begin

Above, we have described several risks associated with the modifications made on the embryos, which could be passed on to future generations.

Embryo editing is only ethically justifiable in cases where the benefits clearly outweigh the risks.

Technical issues aside, the researchers did not even address an unmet medical need.

While the twins father was HIV-positive, there is already a well-established way to prevent an HIV-positive father from infecting embryos. This sperm washing method was actually used by the team.

The only benefit of the attempted gene modification, if proven, would have been a reduced risk of HIV infection for the twins later in life.

But there are safer existing ways to control the risk of infection, such as condoms and mandatory testing of blood donations.

Implications for gene editing as a field

Gene editing has endless applications. It can be used to make plants such as the Cavendish banana more resistant to devastating diseases. It can play an important role in the adaptation to climate change.

In health, we are already seeing promising results with the editing of somatic cells (that is, non-heritable modifications of the patients own cells) in beta thalassemia and sickle cell disease.

However, we are just not ready for human embryo editing. Our techniques are not mature enough, and no case has been made for a widespread need that other techniques, such as preimplantation genetic testing, could not address.

Read more: Experts call for halt to CRISPR editing that allows gene changes to pass on to children

There is also much work still needed on governance. There have been individual calls for a moratorium on embryo editing, and expert panels from the World Health Organisation to UNESCO.

Yet, no consensus has emerged.

It is important these discussions move in unison to a second phase, where other stakeholders, such as patient groups, are more broadly consulted (and informed). Engagement with the public is also crucial.

Correction: this article originally described RNA (ribonucleic acid) as a protein, rather than a molecule.

Dimitri Perrin, Senior Lecturer, Queensland University of Technology and Gaetan Burgio, Geneticist and Group Leader, The John Curtin School of Medical Research, Australian National University

This article is republished from The Conversation under a Creative Commons license. Read the original article.

Image: Reuters

Link:
Gene Editing Might Let China Create The Perfect Human Being - The National Interest Online

Archives