ASH Clinical News FINAL_ACN_3.14_FULL_ISSUE_DIGITAL | Page 105

TRAINING and EDUCATION FIGURE 1. Therapeutic Genome Editing FIGURE 2. Delivery strategies and target cells for in vivo and ex vivo genome editing. Genome-Editing Systems HSCs = hematopoietic stem cells; iPSCs = induced pluripotent stem cells Source: Hoban MD, Bauer DE. A genome editing primer for the hematologist. Blood. 2016;127:2525-35. Somatic gene therapy products are classified as “biological products” and are regulated by the U.S. Food and Drug Administration’s (FDA) Center for Biologics Evaluation and Research. Proposals for gene therapy clinical trials funded by the National Institutes of Health (NIH) are reviewed by its Recombi- nant DNA Advisory Committee (RAC), which collaborates with the FDA. 6 The FDA is prohibited from reviewing pro- posals for studies that use germline editing to alter human embryos. Should this restriction expire, the National Academies of Sciences, Engineering, and Medicine proposed strin- gent criteria for adopting germline editing, recommending that germline editing research trials be permitted “only for compelling pur- poses of treating or preventing serious disease or disabilities, and only if there is a stringent oversight system.” 7 However, many scientists believe that any germline editing using cur- rent technologies would be dangerous and unethical. 8 As far as CRISPR/Cas9 is concerned, Dr. Orkin noted, “There are no ethical concerns unless one talks about germline editing, which is not generally thought of at this time for hematologic disease.” The Role of CRISPR/Cas9 in Hematology To date, no commercialized CRISPR/Cas9 products have begun clinical trials in the U.S. “That being said, there are multiple programs in development – both in academia and industry – heading in that direction,” said Matthew Porteus, MD, PhD, associate profes- sor of pediatrics at Stanford University in ASHClinicalNews.org California, and founder of CRISPR Therapeu- tics. “We are likely to see clinical trials in 2018 and 2019.” At Stanford, Dr. Porteus’s group is moving toward a clinical trial for CRISPR/Cas9 gene editing in sickle cell disease (SCD). 9 “It is rather remarkable that the system was first described as useful in mammalian cells in 2013, and we learned how to use the system efficiently in clinically relevant human cells in 2015,” he added. “The pace to the clin- ic is amazingly fast given the careful scientific and regulatory work that needs to be done before one can give genetically engineered cells to patients.” Enhancing Cellular Immunotherapy To date, the FDA has approved two gene therapies, both autologous chimeric antigen receptor (CAR) T-cell immunotherapies for treating types of leukemia and lymphoma. 10,11 Therapy involves collection of the patient’s own T cells, genetic modification to express a CAR that targets tumor cells, and infusion of modified T cells back into the patient’s system. 12 These CAR-modified T cells are made using viral delivery systems, which randomly insert the CAR gene into the T-cell genome and may result in unwanted genetic effects. Scientists are now using CRISPR/Cas9 to generate CAR T cells and have found that controlling where CAR integrates can enhance their potency. 13 CRISPR/Cas9 gene editing also is being investigated as a tool to enhance CAR T-cell function by disabling genes that encode inhib- itory receptors or signaling molecules, such as programmed cell death protein 1 (PD1). 12 The first CRISPR/Cas9 clinical trial, initiated in China in 2016, is doing just that. 14 In the U.S., a proposed CRISPR/Cas9 clinical trial has already been approved by RAC, but still needs approval from the FDA. This clinical trial will focus on the safety of CRISPR/Cas9-modified T cells for the treat- ment of myeloma, sarcoma, and melanoma. CRISPR/Cas9 will be used to knock out PD1 as well as the endogenous T-cell receptor. 15 “Such gene editing promises to improve the potency and specificity of lymphocyte products and may even enable production of cell therapies from universal donors,” Daniel Bauer, MD, PhD, from the department of pediatric he- matology/oncology at Boston Children’s Hospital and assistant professor of pediatrics at Harvard Medical School in Boston, Massachusetts. CRISPR/Cas9 also could help maximize efficacy and minimize unwanted toxicity of cellular immunotherapy in acute myeloid leukemia (AML). Researchers have proposed that, by knocking out the AML antigen CD33 in normal hematopoietic stem cells (HSCs), CD33-directed immunotherapy can be used against AML without disrupting normal my- eloid function. 16 Correcting Genetic Defects While cellular immunotherapy is the first application of CRISPR/Cas9 being evaluated in the clinic, Dr. Bauer pointed out that “not far behind may be applications for CRISPR in HSCs, where a variety of nonmalignant blood disorders could be ameliorated by permanent genetic modification. In addition, genome ASH Clinical News 103