CRISPR基因编辑

Search documents
首次人体实验成功,基因编辑胰岛细胞“隐身”植入,可正常分泌胰岛素
3 6 Ke· 2025-09-16 07:48
Core Insights - A breakthrough in diabetes treatment has been achieved with the first successful transplantation of CRISPR-edited pancreatic cells into a type 1 diabetes patient, allowing for insulin secretion without the need for immunosuppressants [1][2]. Group 1: Research and Methodology - The study published in the New England Journal of Medicine and reported on Nature's website highlights the potential of CRISPR gene editing in treating type 1 diabetes, which affects approximately 9.5 million patients globally [2]. - Researchers extracted pancreatic cells from a 60-year-old deceased donor and utilized CRISPR-Cas12b technology to edit these cells by knocking out two key genes, B2M and CIITA, which typically signal T cells to attack foreign invaders [2][3]. - To further protect the edited cells from immune system attacks, a gene encoding the CD47 protein was introduced, which sends a "do not eat me" signal to the immune system [2]. Group 2: Clinical Application - The final cell preparation, named UP421, consisted of three types of cells: fully edited cells lacking HLA and expressing high levels of CD47, partially edited cells with some HLA and maintaining endogenous CD47 levels, and wild-type cells with varying CD47 levels [6]. - The edited pancreatic cells were implanted into a 42-year-old patient with 37 years of type 1 diabetes through 17 injections, totaling 79.6 million engineered cells [8][11]. - Remarkably, the entire procedure did not involve any glucocorticoids, anti-inflammatory drugs, or immunosuppressants, and after 12 weeks, the cells showed no signs of rejection while effectively regulating the patient's blood sugar levels [12]. Group 3: Results and Future Plans - C-peptide levels, a direct marker of endogenous insulin secretion, were undetectable at baseline but showed significant increases at weeks 4, 8, and 12 post-intervention, indicating successful insulin production [13]. - Even six months post-transplant, the edited cells continued to evade immune detection and attack [13]. - However, the study involved only one participant, and the treatment duration was insufficient to eliminate the need for insulin injections, prompting the company to plan further clinical trials starting next year for more comprehensive research [14].
首次人体实验成功!基因编辑胰岛细胞“隐身”植入,可正常分泌胰岛素
量子位· 2025-09-16 05:58
Core Viewpoint - The article highlights a significant breakthrough in diabetes treatment, where CRISPR-edited pancreatic cells were successfully transplanted into a type 1 diabetes patient, showing promising results in insulin secretion and immune evasion [1][2][3]. Group 1: Research Background - Type 1 diabetes is an autoimmune disease where the immune system attacks insulin-secreting pancreatic cells, leading to uncontrolled blood sugar levels [4][5]. - The research conducted by Sana Biotechnology aims to provide a potential cure for approximately 9.5 million type 1 diabetes patients globally [8]. Group 2: Methodology - Researchers extracted pancreatic cells from a 60-year-old deceased donor and utilized CRISPR-Cas12b technology to edit these cells by knocking out two key genes, B2M and CIITA, which typically mark foreign invaders for the immune system [9][10]. - To further protect the cells from immune surveillance, a gene encoding the CD47 protein was introduced, which sends a "don't eat me" signal to the immune system [12]. Group 3: Clinical Application - The edited pancreatic cells, totaling 79.6 million, were implanted into a 42-year-old patient with 37 years of type 1 diabetes through 17 injections into muscle tissue [20][24]. - Notably, the entire procedure did not involve any glucocorticoids, anti-inflammatory drugs, or immunosuppressants [25]. Group 4: Results and Future Plans - After 12 weeks post-transplant, the edited cells showed no signs of rejection and continued to secrete insulin, effectively regulating the patient's blood sugar levels [26]. - C-peptide levels, a direct marker of endogenous insulin secretion, were significantly elevated at 4, 8, and 12 weeks post-intervention [28]. - Sana Biotechnology plans to conduct more comprehensive clinical trials starting next year to further investigate the treatment's efficacy [30].
Nature:首批CRISPR基因编辑马诞生,肌肉更强,跑得更快!
生物世界· 2025-09-06 04:05
Core Viewpoint - The article discusses the birth of the world's first CRISPR gene-edited horses, highlighting the implications of this technology in animal breeding and the controversies surrounding it [3][5][7]. Group 1: CRISPR Gene-Edited Horses - The first CRISPR gene-edited horses were born in Argentina, created by Kheiron Biotech, using CRISPR-Cas9 technology to enhance muscle growth by knocking out the myostatin gene [7]. - These horses are clones of the award-winning racehorse Polo Pureza and exhibit stronger muscles and faster speeds compared to ordinary horses [5][7]. - The introduction of these gene-edited horses has sparked controversy, particularly in the equestrian community, with concerns about the impact on traditional breeding practices and livelihoods [8]. Group 2: Broader Applications of CRISPR Technology - Prior to the CRISPR horses, gene editing had been widely applied in agriculture and disease treatment, such as the development of PRLR-SLICK cattle, which are more heat-resistant due to a gene edit [9]. - The FDA approved PRLR-SLICK for meat production in 2022, showcasing the regulatory acceptance of gene-edited animals [9]. - Other examples include CRISPR sheep for increased meat yield and CRISPR pigs that are resistant to diseases like PRRS, with FDA approval for market sale expected by 2026 [10]. Group 3: Ethical and Health Concerns - The rise of CRISPR gene-edited animals raises ethical questions, particularly regarding animal health and the potential unforeseen consequences of genetic modifications [11]. - Concerns include the long-term health effects on the animals and the possibility of genetic changes being passed to future generations or affecting wild populations [12]. - There is a call for further research to monitor any adverse health impacts resulting from gene editing in animals [12].
Editas Medicine(EDIT) - 2025 FY - Earnings Call Transcript
2025-09-03 13:00
Financial Data and Key Metrics Changes - The company is advancing its lead development candidate, Edit 401, which has shown a 90% mean reduction in LDL cholesterol in preclinical studies, significantly outperforming current standard treatments that achieve 40% to 60% reductions [4][18][19] - Edit 401 is expected to provide a one-time treatment option with a durable lifetime reduction in LDL levels, which could transform the treatment landscape for hyperlipidemia [5][18] Business Line Data and Key Metrics Changes - Edit 401 has been selected as the lead program due to its compelling preclinical data, which supports rapid progression to human proof of concept studies expected by the end of 2026 [5][19] - The company is also focusing on optimizing candidates for its hematopoietic stem cell (HSC) program while advancing Edit 401 [20] Market Data and Key Metrics Changes - The market potential for Edit 401 is substantial, with the U.S. healthcare system projected to incur over $300 billion in expenditures related to atherosclerotic cardiovascular disease by 2035 [5] - Hyperlipidemia affects over 70 million patients in the U.S., indicating a significant patient population that could benefit from Edit 401 [6] Company Strategy and Development Direction - The company aims to be a leader in in vivo gene editing by developing CRISPR-based medicines that are best in class or first in class therapeutics [3] - The strategy involves a differentiated approach focusing on functional upregulation of gene expression rather than knockdown strategies used by competitors, allowing the company to target areas others cannot [10] Management's Comments on Operating Environment and Future Outlook - Management expressed confidence in Edit 401's potential to deliver meaningful benefits beyond current therapies, particularly for patients with refractory hyperlipidemia [7][18] - The company remains committed to advancing its pipeline and optimizing resources for its lead program while exploring other therapeutic areas [19][20] Other Important Information - Edit 401 utilizes a proprietary CRISPR-Cas9 enzyme and dual guide RNAs to target the LDLR gene, with a delivery strategy involving lipid nanoparticles [13] - Preclinical studies have shown no adverse effects, with transient increases in liver enzymes resolving within a week [13][110] Q&A Session Summary Question: What patient populations will Edit 401 target? - The company plans to consider segments such as heterozygous familial hypercholesterolemia and other refractory patient populations, with final selections depending on regulatory discussions [24] Question: Is there evidence that 90% LDL reduction leads to lower cardiovascular risk? - Clinical trials indicate that every 40 mg/dL reduction in LDL correlates with a 20% risk reduction for cardiovascular events, supporting the potential for greater risk reduction with Edit 401 [33] Question: How will LDL reduction in mice translate to humans? - The company is optimistic about the translatability of LDL reduction observed in non-human primates to humans, projecting that a human dose will be below 1 mg/kg [36] Question: How does Edit 401 fit into the competitive landscape? - Edit 401's significant LDL reduction effect size positions it as a potential treatment for severe patients, addressing the unmet needs of the 75% of patients who do not achieve their LDL targets with current therapies [42][43] Question: What are the safety profiles and off-target editing concerns? - The company has designed its gene editing tools to be highly specific, with no observed off-target effects or significant adverse events in preclinical studies [109] Question: Will there be room for redosing if needed? - The company believes there is potential for redosing if necessary, based on their analysis from non-human primate studies [120]
Editas Medicine (EDIT) Update / Briefing Transcript
2025-09-02 13:02
Summary of Editas Medicine (EDIT) Webinar on Edit 401 Company Overview - **Company**: Editas Medicine (EDIT) - **Focus**: Development of CRISPR-based gene editing medicines, specifically targeting hyperlipidemia through Edit 401 Key Industry Insights - **Industry**: Gene editing and biopharmaceuticals - **Market Potential**: Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death globally, with U.S. healthcare expenditures projected to exceed $300 billion by 2035 [5][6] Core Product Insights - **Product**: Edit 401 - **Mechanism**: A one-time, in vivo CRISPR gene editing medicine aimed at significantly reducing LDL cholesterol levels - **Efficacy**: Demonstrated a mean reduction of 90% in LDL cholesterol in preclinical studies, compared to 40%-60% reductions achieved by current standard treatments like statins and PCSK9 inhibitors [4][17] - **Treatment Paradigm**: Potential to transform hyperlipidemia treatment by providing a durable, lifetime reduction in LDL levels, thus reducing cardiovascular event risks [5][17] Clinical Development Plans - **Next Steps**: Progressing Edit 401 towards human proof of concept studies, with expected data by the end of 2026 [5][18] - **Patient Segments**: Initial focus may include patients with heterozygous familial hypercholesterolemia (HeFH) and other refractory segments [23][132] Competitive Advantages - **Unique Approach**: Edit 401 utilizes a proprietary CRISPR-Cas9 enzyme and dual-guide RNAs to target the LDLR 3' untranslated region (UTR), leading to increased LDLR protein expression and enhanced LDL clearance [12][11] - **Safety Profile**: Preclinical studies showed no significant adverse effects, with transient increases in liver enzymes resolving within a week [108][109] Market Dynamics - **Patient Population**: Over 70 million patients in the U.S. suffer from elevated LDL levels, with 75% of patients failing to achieve LDL targets with current therapies [6][42] - **Cost Efficiency**: Edit 401 is expected to reduce overall healthcare costs by minimizing the need for multiple therapies and improving patient compliance [18][17] Additional Considerations - **Long-term Safety**: Ongoing studies will assess the durability of LDL reduction and the potential need for redosing [117][119] - **Regulatory Engagement**: Future patient segment selection for clinical trials will be determined in consultation with regulatory bodies [132] Conclusion - Editas Medicine is positioned to potentially revolutionize the treatment of hyperlipidemia with Edit 401, leveraging advanced gene editing technology to achieve significant LDL reductions and improve patient outcomes while addressing a substantial market need.
Cell子刊:带来衰老和炎症!揭示造血干细胞CRISPR基因编辑的意外不良后果
生物世界· 2025-06-08 23:55
Core Viewpoint - The study highlights the unintended adverse effects of CRISPR-Cas9/AAV6-mediated gene editing in hematopoietic stem cells (HSPC), specifically focusing on senescence and inflammation, which can negatively impact the efficacy and safety of gene therapy [4][5][15]. Group 1: Gene Editing Techniques and Challenges - Gene editing in HSPC represents a transformative approach for treating immune, blood, and metabolic genetic diseases, utilizing platforms like CRISPR-Cas9 and AAV6 for targeted gene modifications [8]. - A major limitation of efficient homologous recombination (HDR) gene editing is that primitive HSCs are in a quiescent state, which hinders long-term proliferation of HDR-edited human HSPC [9][10]. - The activation of DNA damage response (DDR) due to double-strand breaks (DSB) can adversely affect the adaptability of HSPC, leading to increased sensitivity and reduced clonal diversity in xenotransplantation [10]. Group 2: Adverse Effects and Mitigation Strategies - The study found that CRISPR-Cas9/AAV6-mediated gene editing induces senescence-like phenotypes and inflammatory responses in HSPC, which impair the functionality of HDR-edited cells [12][15]. - Short-term suppression of p53 can alleviate the negative impacts of gene editing-induced DDR, but complete inactivation of p53 raises concerns about genetic toxicity [11]. - The use of Anakinra, an IL-1 signaling antagonist, has been identified as a promising strategy to enhance the clonal output of HDR-edited cells while minimizing genetic toxicity risks associated with the editing process [5][14][15]. Group 3: Research Findings and Implications - The core findings of the research indicate that CRISPR-Cas9/AAV6 gene editing leads to significant accumulation of senescence markers in HDR-edited HSPC, which can be mitigated to enhance long-term hematopoietic reconstitution [15]. - The study proposes strategies to overcome key obstacles in HDR-based gene therapy for HSPC, aiming to improve clinical efficacy and safety [6][17].
Cell子刊:孙洁/钱鹏旭/裴善赡/黄河团队开发CRISPR基因编辑增强的CAR-T细胞疗法,治疗急性髓系白血病
生物世界· 2025-05-28 03:58
Core Viewpoint - Acute Myeloid Leukemia (AML) presents significant treatment challenges due to the inherent heterogeneity of leukemic cells and the resistance of leukemia stem cells (LSC) to chemotherapy, leading to poor prognosis and high relapse rates [2][3]. Group 1: AML Characteristics and Challenges - AML is characterized by the obstruction of differentiation and abnormal proliferation of immature myeloid cells, resulting in hematopoietic dysfunction and life-threatening cytopenias [2]. - The incidence of AML increases with age, with over two-thirds of diagnoses occurring in individuals over 55 years old. The overall survival rate is approximately 40%-45% for patients under 65, while it drops to 10%-15% for those over 65 [2]. - More than half of AML patients experience disease relapse within one year, and less than one-third achieve durable remission [2]. Group 2: CAR-T Cell Therapy and CD97 Targeting - CAR-T cell therapy has shown success in treating refractory or relapsed B-cell malignancies but has not met expectations in AML due to the lack of specific targets like CD19 found in B-cells [3]. - Most antigens expressed on AML cells and LSCs are also present on normal cells, particularly hematopoietic stem cells (HSC), leading to off-target toxicity and complications such as neutropenia, anemia, and thrombocytopenia [3]. - A study published on May 26, 2025, identified CD97 as an ideal target for CAR-T cell therapy in AML, demonstrating that CD97-directed CAR-T cells could effectively eradicate AML in various xenograft models [3][9]. Group 3: CD97 Characteristics and Implications - CD97, encoded by the ADGRE5 gene, is a member of the adhesion G protein-coupled receptor family and is associated with promoting the invasion of various cancer cells, including those in AML [5]. - CD97 expression is significantly elevated in primary AML cells and LSCs compared to normal bone marrow cells, and its high expression correlates with poor prognosis in AML patients [5]. - CD97 plays a critical role in the proliferation and survival of AML cells, maintaining their undifferentiated state, making it a promising therapeutic target [5]. Group 4: Research Findings on CD97-targeted CAR-T Cells - The research team utilized CRISPR-Cas9 technology to knock out CD97 in T cells, creating CD97 KO CAR-T cells that effectively eliminate AML cells while showing tolerable toxicity to HSCs [6][9]. - Optimized CD97 KO CAR-T cells demonstrated sustained anti-tumor activity in vitro and in various xenograft mouse models, indicating their potential for durable therapeutic effects [6][9]. - The study supports CD97 as a promising target for CAR-T cell therapy in AML, highlighting the importance of gene editing strategies to mitigate self-targeting issues in T cells [9].