Applications of CRISPR-Cas Systems in Gene Therapy

Gene therapy is a revolutionary approach to treating diseases by introducing foreign genes into target cells, aiming to correct conditions caused by mutated or defective genes. While gene therapy initially considered both somatic and germline cells, ethical and technical complexities have currently limited its application primarily to somatic cell gene therapy. Traditional methods like homologous recombination and lentiviral delivery, although used, present challenges such as low efficiency and potential risks from random genome insertion. The emergence of CRISPR-Cas systems has dramatically changed the landscape, offering a more efficient and precise tool for gene therapy across a spectrum of human diseases, including monogenic disorders, infectious diseases, and cancer. CRISPR-mediated genome editing therapies are rapidly advancing, with several already in clinical trials, signaling a promising future for this technology.

The power of CRISPR-Cas systems lies in their precision and versatility in gene editing, making them particularly attractive for therapeutic applications. Unlike earlier gene therapy techniques, CRISPR allows for targeted modifications to the genome, reducing off-target effects and enhancing therapeutic efficacy. This precision is crucial in addressing the root cause of many diseases at the genetic level. The following sections will delve into the applications of CRISPR-Cas systems in treating various diseases, highlighting ongoing clinical trials and the potential impact on patient care.

Clinical Trials of Gene Therapy Using Genome-Editing Technology

Genome-editing technologies, including CRISPR-Cas9, TALENs, and ZFNs, are at the forefront of gene therapy advancements. The table below summarizes ongoing clinical trials utilizing these technologies, showcasing the breadth of diseases being targeted and the institutions leading this innovative research. These trials represent crucial steps in translating the promise of gene editing into tangible treatments for patients.

Number Disease Intervention/treatment Nuclease Company/institute Country Year Clinicaltrials.gov ID
1 HIV/HIV Infections Biological: ZFN modified T cells ZFN Sangamo Therapeutics USA 2009 NCT00842634
2 HIV Genetic: SB-728mR-HSPC Infusion 3 days following busulfan conditioning ZFN Sangamo Therapeutics USA 2015 NCT02500849
3 HIV Drug: ZFN Modified CD4 + T Cells ZFN National Institute of Allergy and Infectious Diseases (NIAID) USA 2015 NCT02388594
4 Human Papillomavirus-Related Malignant Neoplasm Biological: ZFN-603 and ZFN-758 ZFN Huazhong University of Science and Technology China 2016 NCT02800369
5 Hemophilia B Biological: SB-FIX ZFN Sangamo Therapeutics USA 2016 NCT02695160
6 Mucopolysaccharidosis I Biological: SB-318 ZFN Sangamo Therapeutics USA 2016 NCT02702115
7 Mucopolysaccharidosis II Biological: SB-913 ZFN Sangamo Therapeutics USA 2017 NCT03041324
8 HIV Biological: CD4 CAR+CCR5 ZFN T-cells ZFN University of Pennsylvania USA 2018 NCT03617198
9 Transfusion Dependent Beta-thalassemia Genetic: ST-400 Investigational product ZFN Sangamo Therapeutics/ USA 2018 NCT03432364
10 Acute Myeloid Leukemia Biological: UCART123 TALEN Cellectis S.A. USA 2017 NCT03190278
11 Human Papillomavirus-Related Malignant Neoplasm Biological: TALENBiological: CRISPR/Cas TALEN First Affiliated Hospital, Sun Yat-Sen University China 2017 NCT03057912
12 Multiple Myeloma Biological: UCARTCS1A TALEN Cellectis S.A. USA 2019 NCT04142619
13 B-cell Acute Lymphoblastic Leukemia Biological: UCART22 TALEN Cellectis S.A. USA 2019 NCT04150497
14 Acute Myeloid Leukaemia Biological: UCART123 TALEN Cellectis S.A UK 2019 NCT04106076
15 Metastatic Non-small Cell Lung Cancer Other: PD-1 Knockout T Cells CRISPR-Cas9 Chengdu MedGenCell, Co., Ltd. China 2016 NCT02793856
16 HIV-1-infection Genetic: CCR5 gene modification CRISPR-Cas9 Affiliated Hospital of Academy to Military Medical Sciences China 2017 NCT03164135
17 B Cell Leukemia/B Cell Lymphoma Biological: UCART019 CRISPR-Cas9 Chinese PLA General Hospital China 2017 NCT03166878
18 EBV positive advanced stage malignancies PD-1 knockout-T cells from autologous origin CRISPR-Cas9 The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School China 2017 NCT03044743
19 Esophageal Cancer Other: PD-1 Knockout T Cells CRISPR-Cas9 Anhui Kedgene Biotechnology Co.,Ltd China 2017 NCT03081715
20 T cell malignancy Genetic: CD7.CAR/28zeta CAR T cells CRISPR-Cas9 Baylor College of Medicine USA 2018 NCT03690011
21 Sickle Cell Disease Biological: CTX001 CRISPR-Cas9 CRISPR Therapeutics USA 2018 NCT03745287
22 Thalassemia Biological: iHSCs treatment CRISPR-Cas9 Allife Medical Science and Technology USA 2018 NCT03728322
23 β-Thalassemia Biological: CTX001 CRISPR-Cas9 CRISPR Therapeutics USA 2018 NCT03655678
24 Solid Tumor Biological: Mesothelin-directed CAR-T cells CRISPR-Cas9 Chinese PLA General Hospital China 2018 NCT03747965
25 B Cell Leukemia/B Cell Lymphoma Biological: Universal Dual Specificity CD19 and CD20 or CD22 CAR-T Cells CRISPR-Cas9 Chinese PLA General Hospital China 2018 NCT03398967
26 Multiple Myeloma/Melanoma/Synovial Sarcoma/Liposarcoma Biological: NY-ESO-1 redirected autologous T cells with CRISPR edited endogenous TCR and PD-1 CRISPR-Cas9 Parker Institute for Cancer Immunotherapy USA 2018 NCT03399448
27 Solid Tumor Biological: anti-mesothelin CAR-T cells CRISPR-Cas9 Chinese PLA General Hospital China 2018 NCT03545815
28 Thalassemia Major Biological: γ-globin reactivated autologous hematopoietic stem cells CRISPR-Cas9 Shanghai Bioray Laboratory Inc. China 2019 NCT04211480
29 B-cell malignancies Biological: CTX110 CRISPR-Cas9 CRISPR Therapeutics AG USA 2019 NCT04035434
30 β-thalassemia Major Biological: β-globin restored autologous HSC CRISPR-Cas9 Shanghai Bioray Laboratory Inc. China 2019 NCT04205435
31 Leber Congenital Amaurosis 10 (LAC10) Drug: AGN-151587 CRISPR-Cas9 Editas Medicine, Inc. USA 2019 NCT03872479
32 CD19+ leukemia or lymphoma Genetic: XYF19 CAR-T cell CRISPR-Cas9 Xi’An Yufan Biotechnology Co.,Ltd China 2019 NCT04037566
33 Gastro-Intestinal (GI) Cancer Biological: Tumor-Infiltrating Lymphocytes (TIL) CRISPR-Cas9 Intima Bioscience, Inc. USA 2020 NCT04426669
34 Multiple Myeloma Biological: CTX120 CRISPR-Cas9 CRISPR Therapeutics AG USA 2020 NCT04244656
35 Renal Cell Carcinoma Biological: CTX130 CRISPR-Cas9 CRISPR Therapeutics AG Australia 2020 NCT04438083
36 Advanced Hepatocellular Carcinoma Biological: PD-1 knockout engineered T cells CRISPR-Cas9 Central South University China 2020 NCT04417764

β-Thalassemia, a serious inherited blood disorder, stands as one of the most prevalent and health-threatening monogenic diseases globally. This condition arises from mutations in the β-globin (HBB) gene, which leads to a significant reduction in hemoglobin (Hb) levels and consequent severe anemia. Currently, hematopoietic stem cell transplantation (HSCT) is the only curative treatment for β-thalassemia. However, its widespread application is hindered by high costs and the scarcity of suitable donors. Alternative treatments, such as blood transfusions, can manage symptoms and sustain life but do not offer a cure. CRISPR-Cas9 technology is being explored to address this therapeutic gap by repairing the defective β-globin gene in induced pluripotent stem cells (iPSCs) derived from β-thalassemia patients. The promise is that corrected red blood cells can then be produced, effectively curing the disease. Another innovative approach involves reactivating fetal hemoglobin (HbF) expression by knocking out the BCL11A gene, a known suppressor of fetal hemoglobin expression.

Sickle cell disease (SCD), another hematologic disorder, and hemophilia B (HB) are also being targeted by CRISPR-Cas systems. SCD is caused by a single-nucleotide mutation in the β-globin gene, resulting in abnormal hemoglobin S (HbS). CRISPR-Cas9 is employed to either repair this mutation or reactivate HbF expression as therapeutic strategies. Hemophilia B, an X-linked bleeding disorder due to coagulation factor IX deficiency, is traditionally managed by factor supplementation. However, gene correction using CRISPR-Cas9 offers a potential long-term solution. Studies in HB mouse models involving in vivo and ex vivo gene editing have demonstrated promising results in mitigating coagulation deficiency. Furthermore, in situ genome editing using CRISPR-Cas9 has shown to improve hemostatic efficiency and survival rates in HB mice.

Duchenne muscular dystrophy (DMD), a devastating X-chromosome recessive disease, leads to progressive muscle weakness and atrophy due to mutations in the DMD gene, which encodes dystrophin protein. Current treatments for DMD are limited to symptom management, with no definitive cure available. CRISPR-Cas9 is being investigated for its ability to remove mutated transcripts, potentially leading to the production of a functional, albeit truncated, dystrophin protein. Base editing systems also hold promise for DMD treatment by correcting single base mutations or inducing exon skipping.

Retinitis pigmentosa (RP), a group of hereditary retinal degenerative diseases, leads to progressive vision loss. Its genetic complexity and varied inheritance patterns pose significant therapeutic challenges. CRISPR-Cas9 mediated gene correction in mouse models of RP has shown potential in preventing retinal degeneration and improving visual function by targeting genes like RHO, PRPF31, and RP1.

Leber Congenital Amaurosis type 10 (LCA10), a cause of severe early-onset vision loss, is often due to the IVS26 mutation in the CEP290 gene. CRISPR-Cas9 systems are being used to knock out the intronic region containing this mutation to restore normal CEP290 expression. Clinical trials involving subretinal injections of EDIT-101 in animal models and humans are evaluating the safety and efficacy of this approach.

Hutchinson-Gilford Progeria Syndrome (HGPS), a rare genetic disorder characterized by accelerated aging, results from a mutation in the lamin A gene. CRISPR-Cas based gene therapy offers a potential avenue for treatment. Studies in HGPS mice have shown that CRISPR-Cas9 can reduce progerin expression, improving health and prolonging lifespan. Another approach, SATI, has also shown promise in repairing the HGPS-causing mutation in mouse models.

Hereditary tyrosinemia (HT) and cystic fibrosis (CF) are additional diseases where CRISPR-Cas systems are showing therapeutic potential. HT, a metabolic disorder, has been successfully treated in mouse models using CRISPR-Cas9 to correct the Fah mutation in the liver. Cystic fibrosis, a severe respiratory disease caused by CFTR gene mutations, is being addressed in cell models using CRISPR-Cas9 to correct the Δ508 mutation, leading to restored CFTR function in airway epithelial cells.

Human immunodeficiency virus (HIV) poses a persistent global health challenge. While antiretroviral therapy can manage HIV-1 replication, it does not eliminate the virus integrated into the host genome. CRISPR-Cas9 systems offer a potential strategy for complete HIV-1 eradication by targeting and destroying viral proviruses. Furthermore, disrupting the HIV co-receptor CCR5 gene in CD4+ T cells using CRISPR-Cas9 can induce resistance to HIV-1 infection.

Cervical cancer, often linked to human papillomavirus (HPV) infection, is another area of CRISPR-Cas application. By targeting the HPV E6 and E7 oncogenes with CRISPR-Cas9, researchers aim to block their expression, restore normal tumor suppressor gene function, and induce tumor cell apoptosis. This approach has shown promise in in vivo experiments, demonstrating suppressed tumor growth and viral elimination.

Immunotherapy, particularly CAR-T cell therapy, represents a significant advancement in cancer treatment. CAR-T cell therapy involves genetically modifying a patient’s T cells to target cancer cells. CRISPR-Cas9 is being utilized to enhance CAR-T cell therapy by developing universal CAR-T cells that can be used across multiple patients and by improving CAR-T cell function through gene editing of signaling molecules and inhibitory receptors like PD-1 and CTLA-4.

Conclusion

CRISPR-Cas systems have emerged as a transformative tool in gene therapy, offering unprecedented precision and efficiency in genome editing. From monogenic blood disorders like β-thalassemia and sickle cell disease to complex conditions like cancer and HIV, the applications are vast and rapidly expanding. The ongoing clinical trials underscore the accelerating translation of CRISPR-Cas technology from bench to bedside. While challenges remain, the progress in CRISPR-Cas-mediated gene therapy holds immense promise for revolutionizing the treatment of a wide range of diseases, offering hope for more effective and potentially curative therapies. The continuous advancements in delivery methods, specificity, and safety protocols will further solidify the role of CRISPR-Cas systems in shaping the future of medicine.

Comments

No comments yet. Why don’t you start the discussion?

Leave a Reply

Your email address will not be published. Required fields are marked *