Page 1,030«..1020..1,0291,0301,0311,032..1,0401,050..»

Category Archives: Transhuman News

[Full text] Current status of gene therapy for the treatment of cancer | BTT – Dove Medical Press

Posted: March 21, 2021 at 4:59 pm

Background

After DNA helical structure discovery, the world continuous staircase outburst of several advanced technologies, which are currently heading toward translation into clinical practice. Over the last decades, several molecular techniques developed that help to edit the DNA codes and modify mRNA by post-transcriptional modifications. Gene therapy is the delivery of specific genetic material to modify the encoding of a gene product or to change the biological properties of tissues for the management of various disorders.1 Gene therapy overcomes the limitations associated with the recombinant therapeutic use of peptides, such as low bioavailability, instability, severe toxicity, clearance rates, and high production cost.2 Gene therapies act by different mechanisms including, replacing malfunction genes with the therapeutic genes, gene knockdown, or deactivating problem genes, and insert a new gene to treat a disease.3 Gene therapy can be done in either somatic or germline cells. In somatic cells, gene therapy only the modified tissues will be affected, but in germline cell gene therapy, genetic changes transmit to the offspring. So, there is no clinical trial on human germline gene therapy.4 Currently, somatic gene therapy is safe for the management of several disorders in human beings. Gene therapy effectively treats several diseases due to increased understanding of disease pathogenesis and improved gene delivery technologies.5 Gene therapy uses genetic material (ie, RNA or DNA) via a vector that facilitates the delivery of foreign genetic material into the host organ. The genetic material is administered into the target organ (in vivo gene therapy) or used to modify cells taken from the host that are then re-administered (ex vivo gene therapy). Gene therapy aims to provide a functional gene copy of the damaged gene(s), increase the availability of disease-modifying genes or suppress the activity of a damaged gene.6,7 Gene therapy has a broad spectrum of applications, from gene replacement and knockdown for genetic disorders including cancer, hemophilia, hypercholesterolemia, and neurodegenerative diseases to vaccination, each with different requirements for gene administration.8 Gene delivery systems consist of three components: a gene that expresses essential therapeutic peptides, a plasmid-based gene encoding system that regulates the activity of a gene in the target organ, and a gene delivery system that regulates the administration of the encoding gene to host tissue.9

Conventional gene therapy mostly depends on viral-based delivery of genes that either randomly integrates into the host genome (eg retroviruses) or remains as extrachromosomal DNA copy (eg AAV]) and expresses a protein that is missing or mutated in human disorder. In contrast to traditional gene therapy, gene editing provides more versatile tools for gene therapy, for example, precisely correct point variants, place an extra, healthy gene at a safe genomic location or disrupt a gene. The Current gene-editing process depends on the introduction of endogenous double-strand DNA breaks (DSBs) and repair mechanisms. When DSBs occur by nucleases, cellular DNA repair mechanisms are activated. There are two main mechanisms for repairing double-strand breaks, non-homologous end joining (NHEJ) and homology-directed repair (HDR). Genome-editing nucleases can be modified to recognize and break the genome at specific DNA sequences, resulting in DSBs, which are efficiently repaired by either NHEJ or HDR.10,11

NHEJ repair damaged DNA without a homologous template. Due to this reason, NHEJ may lead to deletions or insertions of nucleotides in the damaged loci; thus, it is error-prone. HDR differs from NHEJ since it repairs DNA damages using a homologous template. Generally, having used a homologous sequence, this form of DNA repair has less chance to cause errors. From a clinical viewpoint, HDR is favorable for restoring mutations in genes or for integrating genes for therapeutic purposes.1013

Currently, there are four different gene-editing nuclease enzymes available based on their structures: meganucleases, zinc-finger nucleases, transcription activator-like effector nucleases, and CRISPR-associated nucleases.

Are sequence-specific endonucleases that recognize unique large (1440 bp) target sites. It has low cytotoxicity that makes it an attractive tool for genome editing. Existing engineering techniques include the creation of fusion protein from existing MN domains and engineering MN specificity via the direct alteration of protein residues in the DNA-binding domain. The complexity in re-engineering and low editing efficiency limits the uses of MNs.14

Artificially produced by fusing site-specific zinc finger protein with the non-specific cleavage domain of the FokI restriction endonuclease. The DNA-binding component has 36 zinc finger repeats, and each can identify between 9 and 18 base pairs. ZFN has three zinc fingers that each identifies three base pair DNA sequence to form a three-finger array that attaches to nine base pair target sites and the non-specific cleavage domain.14,15 ZFPs deliver a site-specific DSB to the genome and facilitate local homologous recombination that enhances targeted genome editing. The ZFN-encoding plasmid-based targeted administration of the required genes decreases the limitations of viral administration. If ZFNs are not specific at the target site, off-target break may occur. Such off-target breakage may cause DBS that causes cell death. An Off-target break may facilitate the random integration of donor DNA.15,16

Are artificial DNA nucleases formed by fusing a DNA-binding domain with a nonspecific nuclease domain derived from Fok I endonuclease that specifically cut the required DNA sequence.15 TALE effectors DNA-binding domain has a repeating unit of 3335 conserved amino acids. Each repeat is similar, except positions 12 and 13, which are variable and have a strong correlation with specific nucleotide recognition. DNA cleavage domain is nonspecific from FokI endonuclease. The FokI domain acts as a dimer that needs two constructs with unique DNA binding for sites in the target genome. Both the number of amino acids between the TALE DNA binding domain and the FokI cleavage domain are essential for better activity. TALEN uses to edit genomes by inducing DSB that cells respond to with repair mechanisms.17,18

CRISPR is a heritable, adaptive immune system of bacteria that provides them with the memory of previous virus infections and defends against re-infection. Contrary to the human adaptive immune system, CRISPR is passed on to the next generation of bacteria, rendering the colony immune to future virus infections. CRISPR immunity depends on the integration of the invaders DNA (virus or plasmid) into the bacterial genome.19 CRISPR helps the bacterium to identify the viral sequences and break. CRISPR stands for Clustered Regularly Interspaced Short Palindromic Repeats, which are interrupted by spacer sequences. These spacer sequences are viral sequences integrated during past viral infections when transcribed into short RNA sequences, are capable of guiding the Cas endonuclease to complementary sequences of viral DNA. Upon target identification, Cas binds to the viral DNA and cleaves it, protecting the prokaryotic cell from infection.20,21 CRISPR immune system modified to create a gene-editing tool that can target changes to the DNA. The most common is CRISPR/Cas9, which posses the Cas9 endonuclease and a short noncoding guide RNA (gRNA) that contains two components: a target-specific CRISPR RNA (crRNA) and a helper trans-activating RNA (tracrRNA). The gRNA unit guides Cas9 to a specific genomic locus via base pairing between the crRNA sequence and the target sequence.22 CRISPR-Cas-mediated gene repair, disruption, insertion, or deletion are thus finding applications in several areas of biomedical research, medicine, agriculture, and biotechnology.22,23

Since the emergence of recombinant DNA technology that helps gene-therapy, how to effectively and safely administer gene products is the major challenge. Vector is a vehicle that uses to deliver the gene of interest. An ideal vector can administer a gene to a specific tissue, accommodate enough foreign gene size, achieve the level and duration of transgenic expression enough to correct the defect gene, non-immunogenic, and safe. Delivery of the gene products done by Viral Vectors, Bactofection, and none viral Vectors (chemical and physical) method as summarized in Figure 1.24 The most important step in achieving gene therapy is choosing the vectors.

Figure 1 Overview of the delivery systems used in gene therapy.

Viruses were the first and the most widely used vectors to administer genes into the target tissue. Viral vectors ensure that almost all cells can infect, without affecting cell viability. Viruses have distinctive features that make them suitable for gene delivery in clinical practice. Surface proteins on viruses interact with their host receptors, which activate endocytosis. Once entered, viruses release their genome into the nucleus for viral gene expression.25,26 Herpes simplex virus (HSV), adenovirus (Ad), adeno-associated virus (AAV), and lentivirus (LV) are the most important viral vectors.27,28

Some bacteria specifically target tumor cells leading to RNA interference (RNAi) and gene silencing by inhibiting RNA activity, such as protein synthesis. Several in vivo and in vitro studies revealed that intracellular bacteria such as Salmonella spp., Listeria monocytogenes, Shigella flexneri, Bifidobacterium longum, E. coli, and Yersinia enterocolitica use to deliver plasmids pro-drug converting enzymes and cytotoxic agents into the target cell.29 Phase I trial is undergoing by using Listeria, Bifidobacterium, Salmonella, Shigella, and Clostridium gene therapy against cancer. Another clinical trial is ongoing on the effects of Lactococcus synthesizing interleukin 10 against colitis in Phase II.30,31

Viral-vectors-based gene transfer displays better and long-term gene encoding but has some limitations like immunogenicity, less specific to the target cell, carcinogenicity, high cost and cannot deliver large genome size. Non-viral methods display better advantages due to relatively safe, can deliver a large genome, and ease for production.3235 Chemical vectors, also known as non-viral vectors grouped as organic and inorganic vectors. The organic vectors consist of cationic lipid-based vectors: synthetic cationic polymers-based vector and peptide-based vectors. These cationic organic vectors form complexes with negatively charged DNA via an electrostatic bond. The complexes protect the genomic material and enhance cell uptake and intracellular delivery. Generally, non-viral vectors help to deliver small DNA, large DNA (plasmid DNA), and RNA (Si RNA, m RNA) into the target tissue.3638 Physical methods use different mechanical forces to facilitate the administration of gene material into the host tissues. It is an alternative to viral and chemical methods to decrease barriers that limit DNA delivery into the host tissues.39 It is feasible to deliver genes into target tissues by mechanical force. Indeed, there are several methods, and most have a similar mode of gene delivery, ie, physically formed transient pores in the cell membrane through which the genetic material enters into the host cell.40,41 Needle and jet injection, hydrodynamic gene transfer, electroporation, sonoporation, magnetofection, and gene gun bombardment are examples of physical DNA delivering methods.4244

Cancer occurs due to disrupting the normal cell proliferation and apoptosis process. Advances in cancer therapy need a novel therapeutic agent with novel mode of action, several mechanisms of cell death, and synergy with conventional management. Gene therapies possess all these profiles. Several gene therapy approaches were developed for the management of cancer, including anti-angiogenic gene therapy, suicide gene therapy, immunotherapy, siRNA therapy, pro-apoptotic gene therapy, oncolytic virotherapy, and gene directed-enzyme prodrug therapy.45 By November 2017, greater than 2597 clinical trials were conducted on gene therapy in the world. Among these trials, greater than 65% are associated with cancer, followed by monogenetic and cardiovascular diseases.8 The use of CAR T cell therapy showed promising results for the management of both myeloid and lymphoid leukemia. Until August 2019, only 22 gene products were approved for the treatment of different disorders. Most gene products used for the treatment variety types of cancers as shown in Table 1. Immuno-gene therapy is a potential treatment approach for the treatment of p53-deficient tumors (Imlygic, Gendicine, Yescarta, and Kymriah.47

Table 1 Gene Therapies Products Approved for Therapeutic Use

Oncolytic virotherapy (OV) is the most promising approach for tumor immunotherapy. OV uses replication-competent viruses that can proliferate selectively at tumor cells. Oncolytic viruses grouped as naturally occurring or genetically modified viruses. Natural occurring viruses like parvoviruses, and Newcastle disease viruses that selectively replicate in tumor cell without genetic modification. The second virus category, such as vesicular stomatitis viruses, adenoviruses, measles viruses, HSV and vaccinia viruses, genetically modified to improve the safety, tumor-specificity, and decrease virus pathogenicity. The therapeutic use of oncolytic viruses for cancer treatment is an immune-related treatment alternative. Oncolytic viruses act by directly lyses tumor cells and by introducing wild-type tumor suppressor genes into cells that lack the tumor suppressor gene.48,49 Change in p53 gene function is present in half of all malignancies, and the induction of wild-type p53 gene re-establishes the normal p53 expression. Several recombinant OVs expressing p53 were developed with the aim of producing more potent OVs that act in combination with host immunity or with other treatments modality to destroy tumor cells.49,50

Was the first approved gene product for the management of neck and head squamous cell carcinoma in 2003.50 Gendicine is a non-replicative an adenoviral vector, where the E1 gene is replaced with the tumor suppressor p53 cDNA gene. The expression of p53 in tumor cells triggers the antitumor effect by activating the apoptotic pathway, inhibit damaged DNA repair, and anti-apoptotic activity. P53 gene mutation is prevalent in several cancers. Therefore, Gendicine induces the expression of p53 restores its activity and destroys the tumor cells. Generally, Gendicine management showed 3040% complete response and 5060% partial response with a total response rate of 90%96% in different therapeutic use. Up-to-date greater than 30,000 patients managed by Gendicine.50,51

It is the first replicative, oncolytic recombinant ad5 (rAd5-H101) approved to treat refractory nasopharyngeal cancer. Loss of p53 gene linked with drug resistance and survival rate reduction in non-small cell cancer patients.50 Oncorine is an ad5 virus with a deletion in the E1B 55K gene. Host cell p53 gene inactivation is essential for wild-type to block the activation of apoptotic pathway. The removal of the E1B 55K gene inhibits viral proliferation in normal cells, allowing only proliferate in p53-deficient host cells. In tumor cells, viral proliferation causes oncolysis that is the mechanism to treat solid tumors. Following cancer cell lysis, adenoviruses release and infect another cell activating a serious of Oncorine-mediated cell death.52,53

It is a genetically modified oncolytic HSV-1 approved in Europe in 2015 for the management of non-resectable metastatic melanoma. Imlygic is the first oncolytic virus used for the management of advanced melanoma.48 The replacement of 34.5 and 47 genes with the human granulocyte-macrophage colony-stimulating factor (GM-CSF) gene modifies the HSV-1 gene. The 34.5 gene deletion causes tumor cell-selective replication and suppression of pathogenicity. The 34.5 gene blocks protein synthesis of the host cell during viral infection. Thus, suppressing 34.5 seizes the virus proliferation in normal cells. In tumor cells, the 34.5 gene deleted HSV-1 can replicate. The 47gene inhibits the host cell transporter associated with antigen presentation. The depletion of 47gene reduces MHC class I expression that increases antitumor immune activity.53 Besides, two human GM-CSF genes inserted into the virus providing high levels of GM-CSF production, and stimulate immune responses. Administration of Imlygic causes apoptosis of tumor cell enhanced antigen presentation and increased antitumor response.49,54

Is the first targeted injectable vector approved for the management of metastatic cancers. It is a replication-incompetent retroviral vector showing a SIG-binding peptide to bind to abnormal Signature (SIG) proteins in the tumor cell that increase vector concentration in tumor cells and express a dominant-negative human cyclin G1 inhibitor. After the entrance into the tumor cells, Rexin-G synthesizes cytocidal dnG1 proteins that inhibit the cell cycle in the G1 phase resulting in apoptosis of cancer cells.55,56

T cells destroy infected and tumor cells by detecting nonself antigens with the T cell receptor (TCR). CAR T is a T cell transduced with a chimeric antigen receptor specific to a tumor-associated antigen. CAR is chimeric because it contains the antigen-binding site of the B cell receptor and an intracellular TCR activation domain. CAR has three domains, an extracellular domain that has cancer-specific epitopes (scfv region) made from light (VL) and heavy (VH) chains of immunoglobin that target antigen (such as CD19), a transmembrane domain, and intracellular TCR derived stimulatory domains as showed in Figure 2. The scfv component binds to the target antigen in the MHC independent way leading to CAR clustering and stimulating T-cell via intracellular region that posses the TCR-derived CD3 chain, with or without co-stimulatory domains. Stimulated CAR T-cells give target-specific memory cells that inhibit tumor relapse.57 CD19targeted CAR T cells were the first CARs to be studied. CD19 is a promising target due to its expression limited to the B cell. Firstgeneration, CD19targeted CAR T cells were safe but ineffective. Second-generation CARs have a costimulatory domain with the CD3 activation domain show enhanced T cell activity. Two secondgeneration, CD19targeted CARs are in clinical use contain a 41BB costimulatory domain (19BBz) and a CD28 costimulatory domain and those with more than one additional co-stimulatory molecule are known as third-generation CAR.5759

Figure 2 Schematic diagram of CAR-T-cell products.

It is the first FDA approved CAR T-cell-based gene product to treat relapsed B-cell acute lymphoblastic leukemia. Kymriah has autologous T cells, modified with the lent virus to encode a CAR consist of a murine single-chain antibody fragment (scFv) selective for CD19, an intracellular domain 41BB (CD137), and CD3 zeta with CD8 transmembrane hinge. After binding to CD19 antigen-expressing cells, Kymriah initiates the antitumor effect via the CD3 domain. The intracellular 41BB co-stimulatory domains enhance the antitumor activity. The CD19 antigen is a 95-kD glycoprotein encoded as a surface antigen in diffuse large B-cell lymphoma (DLBCL) and other B-cell lymphomas.60,61 High response rates were recorded in patients with refractory DLBCL in Phase 2 clinical trials. The response rate was 50% at 3 months, 43% with a complete response at 6 months, and there were no patients with a complete response at 6 months who had a relapse by the median of 28.6 months.62

It is another CAR T-cell therapy used for the management of aggressive non-Hodgkin lymphoma. It is CD19 antigen-specific ex-vivo modified autologous T cells infected with a gamma-retroviral. It encodes a CAR comprising an extracellular murine anti-CD19 single-chain variable fragment fused to a cytoplasmic domain that possesses CD28 and CD3-zeta co-stimulatory domains.63,64

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) uses for the management of several hematopoietic malignancies. But, acute graft-versus-host-disease (aGvHD) and Graft rejection are barriers to its success. The treatment strategy for haplo-HSCT depends on T-cell depletion or administration of lymphotoxin agents like cyclophosphamide after stem cell infusion to selectively deplete activated alloreactive lymphocytes but causes prolonged immunodeficiency post-transplantation. Thus, treatment to enhance immune reconstitution after transplantation is necessary.65 Zalmoxis is a genetically modified allogeneic T cell using a retroviral vector encoding a human low-affinity nerve growth factor receptor (LNGFR) and HSV-TK Mut2 to transduce the allogeneic T immune cells. The LNGFR expression uses as a marker of the transduced T cells, and the HSV-TK Mut2 expression provides the suicide gene induction during the administration of the prodrug ganciclovir (GCV). Administration of the genetically modified donor T cells to T cell-depleted transplant patients (HSCT) reconstitutes the immunity to defend from infections. But, donor cells may specifically act as the host cells leading to Graft Versus Host Disease (GVHD). In this case, induction of suicide gene by GCV administration may kill the donor T cells encoding HSV-TK and control GVHD. Zalmoxis is a potential curative agent for HSCT patients when the matched donor does not exist. Zalmoxis provides post-transplant GvHD control, Graft versus Leukemia (GvL) improvement, relapse decrease, and immune reconstitution causes reduced infection.52,66

Gene silencing therapy is RNA interference (RNAi)-mediated knockdown of specific genes in tumor cells. RNAi is single or double-stranded noncoding RNAs (21 ribonucleotides) that induce sequence-specific degradation of complementary mRNAs via the cells internal machinery.67 siRNA is vital because most genes do not have inhibitors due to a lack of ligand binding sites and amino acid sequence homology with other proteins that limit target selectivity. RNAi consists of microRNA (miRNA), Small Interfering RNA (siRNA) and short hairpin RNA (shRNA). Two decades later after the discovery of RNAi, ONPATTRO (patisiran) approved for the first time for the management of the polyneuropathy of hereditary transthyretinmediated (hATTR) amyloidosis.68 Tumor suppressor genes, oncogenes, genes involved in cancer progression, and drug-resistance are promising targets for gene silencing by RNAi-based cancer treatment due to selective gene silencing effect and relatively fewer adverse effects than conventional chemotherapy.69 The merits of RNAi in cancer treatment are targeting several genes of different cellular pathways involved in cancer progression and develop a drug for a specific patient.70 Several studies conducted on animals revealed that targeting vital proteins in the cell cycle, such as Protein kinase N3 (PKN3), kinesin spindle protein (KSP), and polo-like kinase 1 (PLK1) by siRNA displayed a potent antitumor effect. Several liposomal siRNA dose preparations are in Phase 1 trials, such as treatments for pancreatic cancer (PKN3 siRNA), liver cancer (CEBPA siRNA), and neuroendocrine tumors (PLK1 siRNA).71

Suicide gene therapy uses viral or bacterial genes into malignant cells that metabolize non-toxic prodrug into a toxic compound. Several suicide gene systems were identified including the HSV-thymidine kinase gene (HSV-TK) with ganciclovir (GCV) and the cytosine deaminase gene (CD) with 5-fluorocytosine (5-FC).72 Gene-mediated cytotoxic immunotherapy is one strategy where an adenoviral vector possessing the herpes virus thymidine kinase gene (AdV-TK) is administered locally into the tumor site that causes local expression of the HSV-TK gene to the synthesis of viral thymidine kinase that converts GCV to GCV monophosphate. The next step is the administration of GCV that is a substrate of HSV-TK and phosphorylated to produce GCV monophosphate. Then, cellular kinases metabolize GVC-monophosphate into GVC-triphosphate. GCV triphosphate is a deoxyguanosine triphosphate analog, incorporated into the DNA chain causing chain termination and tumor cell death.73

The anti-tumor effect of the TK/GCV system showed promising results in animal models. A study on hormone-refractory prostate cancer patients treated with HSV-TK delivered by adenovirus followed by GCV. The result showed response was at the surrogate marker level and safe. Several studies are in Phase III trials.74 The cytosine deaminase (CD) enzyme exists in fungi and bacteria but not in mammalian cells, metabolizes cytosine into uracil. CD metabolizes the non-toxic prodrug 5-FC into 5-FU, which is subsequently metabolized by cellular enzymes into 5-FdUMP, 5-FdUTP, and 5-FUTP. Inhibition of thymidylate synthase and production of (5-FU) DNA and RNA are the mode of cell death induced by the CD/5-FC suicide system. 5-FU uses for cancer treatment but requires a high dose. This suicide system results in tumor-targeted chemotherapy with few side effects. The CD/5-FC system improved by the inclusion uracil phosphoribosyltransferase (UPRT) gene that phosphorylates 5-FU to 5-fluorouridine mono-phosphate, the first step of its pathway to activation.75 The anti-tumor effect of the CD/5-FC combination showed a better efficacy in animal models. A study on refractory cancer patients that involved intratumoral administration of TAPET-CD attenuated Salmonella bacterium encoding the E. coli CD gene in three patients. The study showed a significant effect and lack of side effects. An oncolytic adenovirus possessing a CD/HSV-1 TK gene was used in a phase I study in patients with prostate cancer. The result showed that the transgene encoding persistence in the prostate for 3 weeks after administration.76

Tumor-driven angiogenesis several growth factors are involved, such as vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), angiopoietins or IL-8, to secure oxygen and nutrients supply. Two major approaches are being pursued to block tumor angiogenesis. The first approach is down-regulation of pro-angiogenic factors expression, such as VEGF, and the second approach is up-regulation of expression of anti-angiogenic factors such as angiostatin, endostatin, and human soluble FMS-like tyrosine kinase receptor. Despite the successful therapeutic use of mAb like Bevacizumab for targeted therapy of cancer, the production and administration of therapeutic mAb are limited due to costly production. Therefore, gene-based studies were done to develop an angiogenesis-targeted cancer treatment.77,78

Gene therapy represents a novel alternative for the management of diseases that have no satisfactory cure. Gene therapy for cancer treatment has good progress in the last three decades, few drugs approved, while others are still in trials. Relatively gene therapy has better safety with tolerable adverse effects than chemotherapy for the treatment of cancer. In the future, tumor genomic analysis, assessment of host humoral and cellular immunity will facilitate a better selection of the most appropriate patient for gene therapy. Recent progress in developing safe and effective vectors for gene delivery, and understanding the activity of nucleases facilitate future genome editing as new treatment approaches for untreatable diseases like cancer.

The success of using autologous and allogenic chimeric antigen receptor integrated T-lymphocytes in mediating adoptive immunotherapy enhances the safety and effectiveness of gene therapy. Besides, the enhanced biological research, cheaper gene vectors will be available in the market, which increases gene therapy accessibility for most cancer patients. This will change the future of cancer treatment, from generalized cancer treatment strategies to individualized cancer treatment, based on the patients specific genome, immune status, and genetic profile of the tumor. Gene therapy is expected to be fast, effective, less toxic, and inexpensive, with higher cure rates. In November 2017, more than 2597 clinical trials are ongoing in several countries and a few of them are listed in Table 2. Until August 2019, 22 gene medicines had been approved by the drug regulatory agencies from various countries.79 Gene therapy gradually accepted by the government and the public since the 1980s and has become an important alternative to the existing treatments in the past few years. Therefore, gene therapy drugs, with safe vectors and advanced biotechnologies, would play a greater role in the prophylaxis and management of cancer in the future.

Table 2 Gene Therapies Products Candidates Under Clinical Trial

ADA, adenosine deaminase; Ad, adenovirus; AAV, adeno-associated virus; aGvHD, acute graft-versus-host-disease; allo-HSCT, allogeneic hematopoietic stem cell transplantation; CRISPR, Clustered Regularly Interspaced Short Palindromic Repeats; CAR, chimeric antigen receptor; DSBs, double-strand breaks; ERT, enzyme replacement therapy; HDR, homology-directed repair; HSV, herpes simplex virus; IRDs, inherited retinal degenerations; LV, lentivirus; NHEJ, non-homologous end joining; NMDs, neuromuscular disorders; OV, oncolytic virotherapy; tracrRNA, trans-activating RNA; TCR, T cell receptor; MNs, meganucleases.

All data are provided in the manuscript or found from published papers as cited.

I would like to acknowledge Mrs Fasika Abu for editing the manuscript for English Style.

The authors declare no competing interests in this work.

1. Wirth T, Parker N, Yl-Herttuala S. History of gene therapy. Gene. 2013;525(2):162169. doi:10.1016/j.gene.2013.03.137

2. Sharma AR, Kundu SK, Nam JS, et al. Next generation delivery system for proteins and genes of therapeutic purpose: why and how? Biomed Res Int. 2014;2014.

3. Templeton NS, editor. Gene and Cell Therapy: Therapeutic Mechanisms and Strategies. Crc Press; 2008.

4. Vermezovic J, Stergiou L, Hengartner MO, Di Fagagna FD. Differential regulation of DNA damage response activation between somatic and germline cells in Caenorhabditis elegans. Cell Death Differ. 2012;19(11):18471855. doi:10.1038/cdd.2012.69

5. Reis RL. Encyclopedia of Tissue Engineering and Regenerative Medicine. Academic Press; 2019.

6. Colella P, Ronzitti G, Mingozzi F. Emerging Issues in AAV-Mediated InVivo Gene Therapy. Mol Ther Methods Clin Dev. 2018;8:87104. doi:10.1016/j.omtm.2017.11.007

7. Merten O-W, Charrier S, Laroudie N, et al. Large-scale manufacture and characterization of a lentiviral vector produced for clinical ex vivo gene therapy application. Hum Gene Ther. 2011;22(3):343356. doi:10.1089/hum.2010.060

8. Ginn SL, Amaya AK, Alexander IE, Edelstein M, Abedi MR. Gene therapy clinical trials worldwide to 2017: an update. J Gene Med. 2018;20(5):e3015.

9. Sung YK, Kim SW. The practical application of gene vectors in cancer therapy. Integrat Cancer Sci Therap. 2018;5:15.

10. Suleiman AA, Saedi WY, Muhaidi MJ. Widely used gene editing strategies in cancer treatment a systematic review. Gene Rep. 2020;100983.

11. Zhang HX, Zhang Y, Yin H. Genome editing with mRNA encoding ZFN, TALEN, and Cas9. Mol Therapy. 2019;27(4):735746. doi:10.1016/j.ymthe.2019.01.014

12. Ernst MP, Broeders M, Herrero-Hernandez P, Oussoren E, van der Ploeg AT, Pijnappel WP. Ready for repair? Gene editing enters the clinic for the treatment of human disease. Mol Therapy Methods Clin Dev. 2020.

13. Sallmyr A, Tomkinson AE. Repair of DNA double-strand breaks by mammalian alternative end-joining pathways. J Biol Chem. 2018;293(27):1053610546. doi:10.1074/jbc.TM117.000375

14. Zaman QU, Li C, Cheng H, Hu Q. Genome editing opens a new era of genetic improvement in polyploid crops. Crop J. 2019;7(2):141150. doi:10.1016/j.cj.2018.07.004

15. Palpant NJ, Dudzinski D. Zinc finger nucleases: looking toward translation. Gene Ther. 2013;20(2):121127. doi:10.1038/gt.2012.2

16. Cassandri M, Smirnov A, Novelli F, et al. Zinc-finger proteins in health and disease. Cell Death Discovery. 2017;3(1):12. doi:10.1038/cddiscovery.2017.71

17. Sun N, Zhao H. Transcription activatorlike effector nucleases (TALENs): a highly efficient and versatile tool for genome editing. Biotechnol Bioeng. 2013;110(7):18111821. doi:10.1002/bit.24890

18. Nakano C, Kitabatake Y, Takeyari S, et al. Genetic correction of induced pluripotent stem cells mediated by transcription activator-like effector nucleases targeting ALPL recovers enzyme activity and calcification in vitro. Mol Genet Metab. 2019;127(2):158165. doi:10.1016/j.ymgme.2019.05.014

19. Hille F, Richter H, Wong SP, Bratovi M, Ressel S, Charpentier E. The biology of CRISPR-Cas: backward and forward. Cell. 2018;172(6):12391259.

20. Rath D, Amlinger L, Rath A, Lundgren M. The CRISPR-Cas immune system: biology, mechanisms and applications. Biochimie. 2015;117:119128. doi:10.1016/j.biochi.2015.03.025

21. Terns RM, Terns MP. CRISPR-based technologies: prokaryotic defense weapons repurposed. Trends Genetics. 2014;30(3):111118. doi:10.1016/j.tig.2014.01.003

22. Li Y, Glass Z, Huang M, Chen ZY, Xu Q. Ex vivo cell-based CRISPR/Cas9 genome editing for therapeutic applications. Biomaterials. 2020;234:119711. doi:10.1016/j.biomaterials.2019.119711

23. Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 2014;157(6):12621278. doi:10.1016/j.cell.2014.05.010

24. Mali S. Delivery systems for gene therapy. Indian J Hum Genet. 2013;19(1):3. doi:10.4103/0971-6866.112870

25. Sung YK, Kim SW. Recent advances in the development of gene delivery systems. Biomaterials Res. 2019;23(1):8. doi:10.1186/s40824-019-0156-z

26. Humphreys IR, Sebastian S. Novel viral vectors in infectious diseases. Immunology. 2018;153(1):19. doi:10.1111/imm.12829

27. Bouard D, AlazardDany N, Cosset FL. Viral vectors: from virology to transgene expression. Br J Pharmacol. 2009;157(2):153165. doi:10.1038/bjp.2008.349

28. Goswami R, Subramanian G, Silayeva L, et al. Gene therapy leaves a vicious cycle. Front Oncol. 2019;9:297.

29. Pilgrim S, Stritzker J, Schoen C, et al. Bactofection of mammalian cells by Listeria monocytogenes: improvement and mechanism of DNA delivery. Gene Ther. 2003;10(24):20362045. doi:10.1038/sj.gt.3302105

30. Johnson SA, Ormsby MJ, McIntosh A, Tait SW, Blyth K, Wall DM. Increasing the bactofection capacity of a mammalian expression vector by removal of the f1 ori. Cancer Gene Ther. 2019;26(7):183194. doi:10.1038/s41417-018-0039-9

31. Celec P, Gardlik R. Gene therapy using bacterial vectors. Front Biosci. 2017;22:8195. doi:10.2741/4473

32. Wang W, Li W, Ma N, Steinhoff G. Non-viral gene delivery methods. Curr Pharm Biotechnol. 2013;14(1):4660.

33. Ramamoorth M, Narvekar A. Non viral vectors in gene therapy-an overview. J Clin Diagnostic Res. 2015;9(1):GE01. doi:10.7860/JCDR/2015/10443.5394

34. Buck J, Grossen P, Cullis PR, Huwyler J, Witzigmann D. Lipid-based DNA therapeutics: hallmarks of non-viral gene delivery. ACS Nano. 2019;13(4):37543782. doi:10.1021/acsnano.8b07858

35. Yang R, Chen F, Guo J, Zhou D, Luan S. Recent advances in polymeric biomaterials-based gene delivery for cartilage repair. Bioactive Mater. 2020;5(4):9901003. doi:10.1016/j.bioactmat.2020.06.004

36. Van Bruggen C, Hexum JK, Tan Z, Dalal RJ, Reineke TM. Nonviral gene delivery with cationic glycopolymers. Acc Chem Res. 2019;52(5):13471358. doi:10.1021/acs.accounts.8b00665

37. Darr JA, Zhang J, Makwana NM, Weng X. Continuous hydrothermal synthesis of inorganic nanoparticles: applications and future directions. Chem Rev. 2017;117(17):1112511238. doi:10.1021/acs.chemrev.6b00417

38. Riley MK, Vermerris W. Recent advances in nanomaterials for gene deliverya review. Nanomaterials. 2017;7(5):94. doi:10.3390/nano7050094

39. Alsaggar M, Liu D. Physical Methods for Gene Transfer. In Advances in Genetics. Vol. 89. Academic Press; 2015:124.

40. Li X, Ruddy B, Taberner A. Characterization of needle-assisted jet injections. J Controlled Release. 2016;243:195203. doi:10.1016/j.jconrel.2016.10.010

41. Suda T, Liu D. Hydrodynamic gene delivery: its principles and applications. Mol Therapy. 2007;15(12):20632069. doi:10.1038/sj.mt.6300314

42. Herrero MJ, Sendra L, Miguel A, Alio SF. Physical Methods of Gene Delivery. In Safety and Efficacy of Gene-Based Therapeutics for Inherited Disorders. Cham: Springer; 2017:113135.

43. Du X, Wang J, Zhou Q, et al. Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv. 2018;25(1):15161525. doi:10.1080/10717544.2018.1480674

44. Smolders S, Kessels S, Smolders SM, et al. Magnetofection is superior to other chemical transfection methods in a microglial cell line. J Neurosci Methods. 2018;293:169173. doi:10.1016/j.jneumeth.2017.09.017

45. Li T, Kang G, Wang T, Huang H. Tumor angiogenesis and anti-angiogenic gene therapy for cancer. Oncol Lett. 2018;16(1):687702. doi:10.3892/ol.2018.8733

46. Mendell JR, Al-Zaidy SA, Rodino-Klapac LR, et al. Current clinical applications of in vivo gene therapy with AAVs. Mol Ther. 2021;29(2):464488. doi:10.1016/j.ymthe.2020.12.007

47. Tristn-Manzano M, Justicia-Lirio P, Maldonado-Prez N, Cortijo-Gutirrez M, Benabdellah K, Martin F. Externally-controlled systems for immunotherapy: from bench to bedside. Front Immunol. 2020;11.

48. Bommareddy PK, Patel A, Hossain S, Kaufman HL. Talimogene laherparepvec (T-VEC) and other oncolytic viruses for the treatment of melanoma. Am J Clin Dermatol. 2017;18(1):15. doi:10.1007/s40257-016-0238-9

49. Sostoa JD, Dutoit V, Migliorini D. Oncolytic viruses as a platform for the treatment of malignant brain tumors. Int J Mol Sci. 2020;21(20):7449. doi:10.3390/ijms21207449

50. Zhang -W-W, Li L, Li D, et al. The first approved gene therapy product for cancer Ad-p53 (Gendicine): 12 Years in the Clinic. Hum Gene Ther. 2018;29(2):160179. doi:10.1089/hum.2017.218

51. Xia Y, Li X, Sun W. Applications of recombinant adenovirus-p53 gene therapy for cancers in the clinic in China. Curr Gene Ther. 2020;20(2):127141. doi:10.2174/1566523220999200731003206

52. Shahryari A, Saghaeian Jazi M, Mohammadi S, Razavi Nikoo H, Nazari Z, Hosseini ES. Development and clinical translation of approved gene therapy products for genetic disorders. Front Genet. 2019;10:868. doi:10.3389/fgene.2019.00868

53. Russell L, Peng K-W. The emerging role of oncolytic virus therapy against cancer. Chine Clin Oncol. 2018;7(2):16. doi:10.21037/cco.2018.04.04

54. Osman AE, Luke JJ. The impact of the fecal microbiome on cancer immunotherapy. BioDrugs. 2019;33(1)::17. doi:10.1007/s40259-018-0328-8

55. Chawla SP, Bruckner H, Morse MA, Assudani N, Hall FL, Gordon EM. A Phase I-II study using rexin-g tumor-targeted retrovector encoding a dominant-negative cyclin g1 inhibitor for advanced pancreatic cancer. Mol Therapy Oncolytics. 2019;12:5667. doi:10.1016/j.omto.2018.12.005

56. Gordon EM, Hall FL. Rexin-G, a targeted genetic medicine for cancer. Expert Opin Biol Ther. 2010;10(5):819832. doi:10.1517/14712598.2010.481666

57. Bb Pettitt D, Arshad Z, Smith J, Stanic T, Hollnder G, Brindley D. CAR-T cells: a systematic review and mixed methods analysis of the clinical trial landscape. Mol Therapy. 2018;26(2):342353. doi:10.1016/j.ymthe.2017.10.019

58. Oluwole OO, Davila ML. At The Bedside: clinical review of chimeric antigen receptor (CAR) T cell therapy for B cell malignancies. J Leukoc Biol. 2016;100(6):12651272. doi:10.1189/jlb.5BT1115-524R

59. Ali S, Kjeken R, Niederlaender C, et al. The European Medicines Agency Review of Kymriah (Tisagenlecleucel) for the Treatment of Acute Lymphoblastic Leukemia and Diffuse Large B-Cell Lymphoma. oncologist. 2020;25(2):e321. doi:10.1634/theoncologist.2019-0233

60. Gill S, Maus MV, Porter DL. Chimeric antigen receptor T cell therapy: 25years in the making. Blood Rev. 2016;30(3):157167. doi:10.1016/j.blre.2015.10.003

61. Liu Y, Chen X, Han W, Zhang Y. Tisagenlecleucel, an approved anti-CD19 chimeric antigen receptor T-cell therapy for the treatment of leukemia. Drugs Today. 2017;53(11):597. doi:10.1358/dot.2017.53.11.2725754

62. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Eng J Med. 2019;380(1):4556. doi:10.1056/NEJMoa1804980

63. Bouchkouj N, Kasamon YL, de Claro RA, et al. FDA approval summary: axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma. Clin Cancer Res. 2019;25(6):17021708. doi:10.1158/1078-0432.CCR-18-2743

Go here to read the rest:
[Full text] Current status of gene therapy for the treatment of cancer | BTT - Dove Medical Press

Posted in Gene Medicine | Comments Off on [Full text] Current status of gene therapy for the treatment of cancer | BTT – Dove Medical Press

Sarepta Therapeutics’ Investigational Gene Therapy SRP-9003 for the Treatment of Limb-Girdle Muscular Dystrophy Type 2E Shows Sustained Expression and…

Posted: at 4:59 pm

-- Protein expression in muscle was sustained for two years following treatment in the low dose cohort, with mean beta-sarcoglycan expression of 54% at 24 months, compared to 36% at Day 60, as measured by western blot ---- Mean NSAD score improvement of 5.7 points from baseline was sustained through 24 months in low-dose cohort, and mean NSAD score improvement of 4.0 points from baseline at one year in high-dose cohort ---- Results in both cohorts continue to reinforce the safety and tolerability profile of SRP-9003 --

CAMBRIDGE, Mass., March 18, 2021 (GLOBE NEWSWIRE) -- Sarepta Therapeutics, Inc.(NASDAQ:SRPT), the leader in precision genetic medicine for rare diseases, today shared new results from the ongoing study of SRP-9003 (rAAVrh74.MHCK7.hSGCB), the Companys investigational gene therapy for limb-girdle muscular dystrophy Type 2E (LGMD2E). In the first look at expression data from biopsies taken two years after a single administration of SRP-9003, results found sustained protein expression in muscle tissue. In functional outcomes assessments taken two years following treatment in Cohort 1 (low-dose cohort) and one year after treatment in Cohort 2 (high-dose cohort), patients continued to demonstrate stability in their NSAD (North Star Assessment for Dysferlinopathies) total score and improvements on timed function tests. Results are being presented today at the 2021 Muscular Dystrophy Association (MDA) Annual Clinical and Scientific Conference.

SRP-9003 is in development for the treatment of LGMD2E (also known as beta-sarcoglycanopathy and LGMDR4), a devastating monogenic neuromuscular disease caused by a lack of beta-sarcoglycan (beta-SG) proteins. SRP-9003 is a gene therapy construct that transduces skeletal and cardiac muscle, delivering a gene that codes for the full-length beta-SG protein, the absence of which is the sole cause of the progressive degeneration and a shortened lifespan characterized by the disease.

This data is the first look at longer-term expression data with any gene therapy for muscular dystrophy. The meaningful and sustained levels of beta-sarcoglycan protein expression at two years and continued strength of the functional outcomes measured are tremendously positive and support continued advancement of this investigational treatment for patients, said Louise Rodino-Klapac, Ph.D., executive vice president and chief scientific officer, Sarepta Therapeutics. In Cohort 2, we also saw strong expression of delta-sarcoglycan and gamma-sarcoglycan proteins in addition to beta-sarcoglycan, which suggests that SRP-9003 is working to restore the dystrophin associated protein complex, or DAPC, which provides biological support for the sustained functional benefits observed in both cohorts. LGMD2E is one of the most severe forms of LGMD and causes significant disability in children while frequently leading to early mortality and the data continue to suggest this treatment could bring much needed hope to these patients.

Efficient transduction in skeletal muscle and robust beta-sarcoglycan protein expression were seen in both dose cohorts following infusion with SRP-9003, and significant creatine kinase (CK) reductions were observed.

Cohort 1 (Dosed at 1.851013 vg/kg), 24 months following treatment:

Cohort 2 (Dosed at 7.411013 vg/kg), 12 months following treatment:

In an exploratory evaluation of all SRP-9003 treated patients compared to a natural history cohort; patients treated with SRP-9003 demonstrated significant improvements in functional outcomes after 24 months. The mean decline in total NSAD score for patients in the natural history cohort was 4.6 points while SRP-9003 treated patients demonstrated a mean improvement of 4.6 points for a clinically meaningful difference of 9.2 points.

Since the last update from this study in October 2020, there have been no new drug-related safety signals observed, and no decreases in platelet counts outside of the normal range and no evidence of clinical complement activation observed in either dose cohort.

About SRP-9003 and the StudySRP-9003 uses the AAVrh74 vector, which is designed to be systemically and robustly delivered to skeletal, diaphragm and cardiac muscle, making it an ideal candidate to treat peripheral neuromuscular diseases. AAVrh74 has lower immunogenicity rates than reported with other human AAV vectors. The MHCK7 promoter has been chosen for its ability to robustly express in the heart, which is critically important for patients with limb-girdle muscular dystrophy Type 2E (LGMD2E), also known as beta-sarcoglycanopathy and LGMDR4, many of whom die from pulmonary or cardiac complications.

This open label, first-in-human study is evaluating a single intravenous infusion of SRP-9003 among children with LGMD2E between the ages of 4 and 15 years with significant symptoms of disease. The SRP-9003 study has two cohorts, each studying a different dose-per-kilogram based on the weight of the patient. Three participants in the low-dose cohort (Cohort 1) were treated with a one-time infusion of SRP-9003 dosed at 1.851013 vg/kg and an additional three participants in the high-dose cohort (Cohort 2) received a one-time infusion dosed at 7.411013 vg/kg based on linear standard qPCR titer method. The six participants were between the ages of 4 and 13. Post-treatment biopsies were taken at 60 days.

Sarepta has exclusive rights to the LGMD2E gene therapy program initially developed at the Abigail Wexner Research Institute at Nationwide Childrens Hospital.

About Limb-girdle Muscular DystrophyLimb-girdle muscular dystrophies are genetic diseases that cause progressive, debilitating weakness and wasting that begin in muscles around the hips and shoulders before progressing to muscles in the arms and legs.

Patients with limb-girdle muscular dystrophy Type 2E (LGMD2E) begin showing neuromuscular symptoms such as difficulty running, jumping and climbing stairs before age 10. The disease, which is an autosomal recessive subtype of LGMD, progresses to loss of ambulation in the teen years and often leads to early mortality. There is currently no treatment or cure for LGMD2E.

Sarepta has five LGMD gene therapy programs in development, including subtypes for LGMD2E, LGMD2D, LGMD2C, LGMD2B and LGMD2L, and holds an option for a sixth program for LGMD2A.

AboutSarepta TherapeuticsAt Sarepta, we are leading a revolution in precision genetic medicine and every day is an opportunity to change the lives of people living with rare disease. The Company has built an impressive position in Duchenne muscular dystrophy (DMD) and in gene therapies for limb-girdle muscular dystrophies (LGMDs), mucopolysaccharidosis type IIIA, Charcot-Marie-Tooth (CMT), and other CNS-related disorders, with more than 40 programs in various stages of development. The Companys programs and research focus span several therapeutic modalities, including RNA, gene therapy and gene editing. For more information, please visitwww.sarepta.comor follow us onTwitter,LinkedIn,InstagramandFacebook.

Forward-Looking StatementsThis press release contains "forward-looking statements." Any statements contained in this press release that are not statements of historical fact may be deemed to be forward-looking statements. Words such as "believes," "anticipates," "plans," "expects," "will," "intends," "potential," "possible" and similar expressions are intended to identify forward-looking statements. These forward-looking statements include statements regarding, SRP-9003 being the ideal candidate to treat peripheral neuromuscular diseases; the potential benefits of SRP-9003, including its potential to restore the dystrophin associated protein complex (DAPC); the potential benefits of MHCK7 and the AAVrh74 vector, including its potential to be systemically and robustly delivered to skeletal, diaphragm and cardiac muscle; and potential market opportunities.

These forward-looking statements involve risks and uncertainties, many of which are beyond our control. Known risk factors include, among others: success in preclinical trials and clinical trials, especially if based on a small patient sample, does not ensure that later clinical trials will be successful; the data presented in this release may not be consistent with the final data set and analysis thereof or result in a safe or effective treatment benefit; different methodologies, assumptions and applications we utilize to assess particular safety or efficacy parameters may yield different statistical results, and even if we believe the data collected from clinical trials of our product candidates are positive, these data may not be sufficient to support approval by the FDA or foreign regulatory authorities; if the actual number of patients suffering from LGMD is smaller than estimated, our revenue and ability to achieve profitability may be adversely affected; we may not be able to execute on our business plans and goals, including meeting our expected or planned regulatory milestones and timelines, clinical development plans, and bringing our product candidates to market, due to a variety of reasons, some of which may be outside of our control, including possible limitations of company financial and other resources, manufacturing limitations that may not be anticipated or resolved for in a timely manner, regulatory, court or agency decisions, such as decisions by the United States Patent and Trademark Office with respect to patents that cover our product candidates and the COVID-19 pandemic; and even if Sareptas programs result in new commercialized products, Sarepta may not achieve the expected revenues from the sale of such products; and those risks identified under the heading Risk Factors in Sareptas most recent Annual Report on Form 10-K for the year ended December 31, 2020 filed with the Securities and Exchange Commission (SEC) as well as other SEC filings made by the Company which you are encouraged to review.

Any of the foregoing risks could materially and adversely affect the Companys business, results of operations and the trading price of Sareptas common stock. For a detailed description of risks and uncertainties Sarepta faces, you are encouraged to review the SEC filings made by Sarepta. We caution investors not to place considerable reliance on the forward-looking statements contained in this press release. Sarepta does not undertake any obligation to publicly update its forward-looking statements based on events or circumstances after the date hereof.

InternetPosting of InformationWe routinely post information that may be important to investors in the 'For Investors' section of our website atwww.sarepta.com.Weencourageinvestorsandpotentialinvestorsto consult our website regularly for important information about us.

Source:Sarepta Therapeutics, Inc.

Sarepta Therapeutics, Inc.

Investors:Ian Estepan, 617-274-4052iestepan@sarepta.com

Media:Tracy Sorrentino, 617-301-8566tsorrentino@sarepta.com

Read more here:
Sarepta Therapeutics' Investigational Gene Therapy SRP-9003 for the Treatment of Limb-Girdle Muscular Dystrophy Type 2E Shows Sustained Expression and...

Posted in Gene Medicine | Comments Off on Sarepta Therapeutics’ Investigational Gene Therapy SRP-9003 for the Treatment of Limb-Girdle Muscular Dystrophy Type 2E Shows Sustained Expression and…

Record funding flowed into cell, gene therapy companies last year – BioPharma Dive

Posted: at 4:59 pm

Dive Brief:

Biotech as a whole had a strong year in 2020. The Nasdaq Biotechnology index, which tracks the industry's stock market performance, rose by nearly 25%, recovering from a spring slump as COVID-19 became a pandemic to regain ground strongly.

Cell and gene therapy companies did even better, according to ARM, which calculated in its report stock performance that surpassed the broader NBI index.

The regenerative medicine sector, which includes tissue-based treatments as well as cell- and gene-based medicines, got larger, too. ARM counted roughly 1,100 developers worldwide, up about 100 from 2019.

"The future is now," said Janet Lambert, ARM's CEO, in an interview. "It's not like we're waiting for there to be a big and meaningful cell and gene therapy sector. There is a big and meaningful cell and gene therapy sector."

Recently, however, some of the most advanced companies have run into regulatory roadblocks or revealed disappointing study results. Cancer cases reported in trials of two closely followed gene therapies have renewed safety concerns, even if it appears the experimental treatments have not played a causative role.

Setbacks are to be expected amid the sector's fast growth, said Lambert, who noted the roughly 150 late-stage studies now ongoing. Many of those programs likely won't succeed, given the usual rates of clinical trial failure in biotech.

Unlike in the past, however, the pipeline of cell and gene therapies is so broad, and the number of companies involved so high, that setbacks for any one program are less likely to slow the entire sector than in past decades. And while the Food and Drug Administration has not cleared any new gene therapies since landmark approvals for Roche's inherited blindness treatment Luxturna and Novartis's spinal muscular atrophy therapy Zolgensma, the agency recently OK'd new CAR-T cell therapies for types of lymphoma.

Across Europe, the U.S. and China, regulators are expected to decide on approvals for eight regenerative medicine therapies this year, according to ARM. In the U.S., cancer cell therapies from Bristol Myers Squibb and Johnson & Johnson could reach market, as well as a tissue-based treatment from Mallinckrodt for severe burns.

Developers and regulators are also learning quickly, particularly in areas like manufacturing and quality control.

"One of the important things we need to work on is how best to regulate the [chemistry, manufacturing and control] aspects of cell and gene therapy," said Lambert.

"It's clearly a place we've struggled," she added, noting recent disagreements between the FDA and developers over CMC issues like testing assays.

ARM members are hoping to have more conversations with the agency earlier, Lambert said, although the FDA division in charge of cell and gene therapies has been stretched thin. In comments to the agency, ARM has advocated for the division to receive more resources and staff in renegotiations for the next FDA user fee agreement that will start in 2023.

Originally posted here:
Record funding flowed into cell, gene therapy companies last year - BioPharma Dive

Posted in Gene Medicine | Comments Off on Record funding flowed into cell, gene therapy companies last year – BioPharma Dive

Sarepta Therapeutics to Present Results from its Gene Therapy and RNA Platforms at the 2021 Annual MDA Clinical and Scientific Conference -…

Posted: at 4:59 pm

-- Ten abstracts, including four podium presentations, reflect Sareptas ongoing commitment to advancing genetic medicine for rare neuromuscular disease and facilitating greater understanding of these devastating conditions --

CAMBRIDGE, Mass., March 15, 2021 (GLOBE NEWSWIRE) -- Sarepta Therapeutics, Inc. (NASDAQ:SRPT), the leader in precision genetic medicine for rare diseases, will present at the Muscular Dystrophy Association (MDA) Annual Clinical and Scientific Conference, which will take place virtually March 15-18, 2021. Among the research that will be presented:

All posters are available on-demand throughout the Congress beginning on Monday, March 15, 2021 at 6:00 a.m. ET. Podium presentations will take place on Thursday, March 18, 2021. The full MDA 2021 Virtual Congress program is available here: https://mdaconference.org.

Podium Presentations:

Poster Presentations:

Presentations will be archived on the events and presentations page in the Investor Relations section of http://www.sarepta.com for one year following their presentation at MDA.

AboutSarepta TherapeuticsAt Sarepta, we are leading a revolution in precision genetic medicine and every day is an opportunity to change the lives of people living with rare disease. The Company has built an impressive position in Duchenne muscular dystrophy (DMD) and in gene therapies for limb-girdle muscular dystrophies (LGMDs), mucopolysaccharidosis type IIIA, Charcot-Marie-Tooth (CMT), and other CNS-related disorders, with more than 40 programs in various stages of development. The Companys programs and research focus span several therapeutic modalities, including RNA, gene therapy and gene editing. For more information, please visitwww.sarepta.com or follow us on Twitter, LinkedIn, Instagram and Facebook.

Internet Posting of Information

We routinely post information that may be important to investors in the 'For Investors' section of our website atwww.sarepta.com. We encourage investors and potential investors to consult our website regularly for important information about us.

Source: Sarepta Therapeutics, Inc.

Sarepta Therapeutics, Inc.

Investors:Ian Estepan, 617-274-4052, iestepan@sarepta.com

Media:Tracy Sorrentino, 617-301-8566, tsorrentino@sarepta.com

Excerpt from:
Sarepta Therapeutics to Present Results from its Gene Therapy and RNA Platforms at the 2021 Annual MDA Clinical and Scientific Conference -...

Posted in Gene Medicine | Comments Off on Sarepta Therapeutics to Present Results from its Gene Therapy and RNA Platforms at the 2021 Annual MDA Clinical and Scientific Conference -…

Be Bio Announces Appointment of Cell and Gene Therapy Veterans Joanne Smith-Farrell, Ph.D., as Chief Executive Officer and Rick Morgan, Ph.D., as…

Posted: at 4:59 pm

CAMBRIDGE, Mass.--(BUSINESS WIRE)--Be Biopharma (Be Bio), whose mission is to pioneer the emerging new category of engineered B cells as medicines, today announced that Joanne Smith-Farrell, Ph.D., has been appointed Chief Executive Officer and Director. Dr. Smith-Farrell will be joined by Chief Scientific Officer, Richard Morgan, Ph.D., a leading expert in cell and gene therapies.

Be Bios rapidly growing team of scientists, drug developers, manufacturing experts, and business builders is leading the creation of a new category of cellular therapies, engineered B cell medicines. B cells are exquisitely designed by nature to embody a unique mix of functionalities, including prolific protein production, tissue targeting, and durable engraftment in cellular niches. Be Bio was founded by Longwood Fund in October 2020 with a $52 million Series A investment led by Atlas Ventures and RA Capital, and joined by Alta Partners and Takeda Ventures to unlock this rich biology by precisely engineering B cells as therapies to develop a broad pipeline of potent and potentially curative cellular medicines.

Prior to joining Be Bio as Chief Executive Officer, Dr. Smith-Farrell was Chief Operating Officer and Business Unit Head, Oncology, at bluebird bio, where she led the growth of bluebird Oncology from an early single-candidate effort into a leading oncology cell therapy business. Prior to this, she held executive leadership roles as Chief Business Officer of bluebird bio, Vice President of Transactions at Merck, and Vice President of Business Development at Pfizer, as well as executive positions in public and private biotechs. Prior to entering the biopharmaceutical industry, she worked in the healthcare practice at The Boston Consulting Group. Dr. Smith-Farrell did her postdoctoral research in Biomedical Engineering in Bob Langers lab at the Harvard-MIT Division for Health Science and Technology and holds a Ph.D. in Physics from The Catholic University of America and a B.S. in Physics and Mathematics from Vanderbilt University.

Be Bios mission - to unleash the power of B cells, natures protein factories, on many of humanitys most challenging diseases is an inspiring and humbling journey to be joining, said Dr. Smith-Farrell. It has been a great privilege to participate in the birth of the first generation of cell therapies to come to market, and to witness, first-hand, cell therapys power to transform the lives of patients with devastating diseases. Be Bios aspiration, fueled by the broad utility of engineered B cell medicines to offer previously impossible solutions across a wide array of therapeutic areas, takes the potential of cell therapy to an entirely new level.

Rick Morgan, Ph.D., joins Be Bio as Chief Scientific Officer, and brings decades of experience as one of the pioneers of cell and gene therapy. Most recently, Dr. Morgan was Senior Vice President of Immunogenetics at Editas Medicine, where he focused on genome engineering to produce off-the-shelf cell medicines for cancer. Prior to that, he was Vice President of Immunotherapy at bluebird bio in 2013, where he led pre-clinical activities for bluebirds first oncology medicine, the anti-B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T cell therapy idecabtagene vicleucel (ide-cel)the first CAR-T for the treatment of multiple myeloma filed in the U.S. and Europe. He started his career at the National Institutes of Health, where he conducted groundbreaking research in the development of gene therapy for genetic diseases such as hemophilia, HIV/AIDS, and cancer immunotherapy. He was a member of the team that published the first approved human gene transfer experiment in 1990, and was also the first to report the successful use of T-cell receptor gene therapy for the treatment of cancer in 2006.

By exploiting the intrinsic drug-like properties of B cells, we can make redosable medicines with superior pharmacokinetic profiles that can be administered without toxic conditioning regimens, said Dr. Morgan. Be Bios ability to engineer B cells is a true paradigm shift in gene therapy that creates major opportunities to treat diseases such as cancer, autoimmune conditions, infectious disease, and protein deficiencies. As CSO, I am excited to have the rare opportunity to shape the development of a new class of medicine from the very start.

Extraordinary science attracts extraordinary leaders, said David Steinberg, Longwood General Partner, Director and and outgoing Chief Executive Officer, Be Bio. For over 25 years, Dr. Smith-Farrell has been leading teams that are committed to conquering cancer and rare diseases, most recently at bluebird bio where she built a 400 person oncology cell therapy business unit. We are very fortunate to have her joining alongside Dr. Morgan, an internationally recognized trailblazer in cell and gene therapy and a member of an elite group of scientists who have successfully developed these groundbreaking medicines. Together, Joanne and Rick will be invaluable to Be Bio as they lead our efforts to unlock the potential of B cell medicines, bringing transformational therapies to patients in need.

About Be Biopharma

Be Biopharma is a leader in developing B cells as medicines, treating disease with the human bodys native protein factories. We precisely engineer B cells to harness their intrinsic drug-like properties remarkable protein production, selective tissue targeting, and fine control of their cellular environment to forge a new category of cell therapy. These medicines are designed to be durable, re-dosable and administered without toxic conditioning, creating new avenues to halt or reverse severe diseases like cancer, autoimmune conditions, and enzyme deficiency. Founded by Longwood Fund and B cell engineering pioneers David Rawlings, M.D., and Richard James, Ph.D., Be Biopharma is re-imagining medicine based on the power of B cell therapy. For more information, please visit Be Biopharma.

Continued here:
Be Bio Announces Appointment of Cell and Gene Therapy Veterans Joanne Smith-Farrell, Ph.D., as Chief Executive Officer and Rick Morgan, Ph.D., as...

Posted in Gene Medicine | Comments Off on Be Bio Announces Appointment of Cell and Gene Therapy Veterans Joanne Smith-Farrell, Ph.D., as Chief Executive Officer and Rick Morgan, Ph.D., as…

ElevateBio Announces Chief Scientific Officer of Regenerative Medicine, Melissa Carpenter, PhD, Elected to the International Society for Stem Cell…

Posted: at 4:59 pm

CAMBRIDGE, Mass.--(BUSINESS WIRE)--ElevateBio, a cell and gene therapy technology company focused on powering transformative cell and gene therapies, today announced that the companys Chief Scientific Officer of Regenerative Medicine, Dr. Melissa Carpenter, has been appointed to the International Society for Stem Cell Research (ISSCR) Board of Directors. In this role, Dr. Carpenter will work with the ISSCR officers and board to advance the organizations mission of bringing together researchers, clinicians, academics, and industry to promote excellence in stem cell science and applications to human health.

I am honored to have been elected to, and serve on the Board of, the ISSCR and foster the continued progress in advancing stem cell science alongside this impressive leadership and fellow board members, said Melissa Carpenter, PhD, Chief Scientific Officer of Regenerative Medicine at ElevateBio. Collaboration across the stem cell professional community is critical to our ability to translate promising stem cell research and regenerative medicine science into treatments that can have dramatic benefit for global human health globally.

Dr. Carpenter served on the ISSCR Task Force to revise the Guidelines for Stem Cell Research and Clinical Translation that will be released in May and advocated in support of the value of stem cell research as part of the Societys 2019 Advocacy Day, meeting with members of the U.S. Congress. She has also served on the Clinical Translation Committee.

We are delighted to welcome Melissa Carpenter to the ISSCR Board of Directors, said Christine Mummery, ISSCR President. Melissas dedication to supporting the translation of stem cell discoveries into therapeutics and her leadership has been crucial for advancing the clinical development of multiple therapies. Her experience will be an asset to the Board as the field of stem cell science continues to rapidly evolve.

The International Society for Stem Cell Research is the preeminent global, cross-disciplinary, science-based organization dedicated to stem cell research and its translation to the clinic. With nearly 4,000 members from more than 60 countries, the ISSCR mission is to promote excellence in stem cell science and applications to human health.

About ElevateBio:

ElevateBio is a cell and gene therapy technology company built to power the development of transformative cell and gene therapies today and for many decades to come. The company has assembled industry-leading talent, built world-class facilities, and integrated diverse technology platforms necessary for rapid innovation and commercialization of cell, gene, and regenerative therapies. The company has built an initial technology stack, including gene editing, induced pluripotent stem cells, and protein, viral, and cellular engineering. At the center of the business model is ElevateBio BaseCamp, a centralized R&D and manufacturing company that offers research and development (R&D), process development (PD), and Current Good Manufacturing Practice (CGMP) manufacturing capabilities. The company is focused on increasing long-term collaborations with industry partners while also continuing to develop its own highly innovative cell and gene therapies. ElevateBio's team of scientists, drug developers, and company builders are redefining what it means to be a technology company in the world of drug development, blurring the line between technology and healthcare.

ElevateBio is headquartered in Cambridge, Mass, with ElevateBio BaseCamp located in Waltham, Mass. For more information, visit us at http://www.elevate.bio, or follow Elevate on LinkedIn, Twitter, or Instagram.

*As of the date of this press release, SoftBank Group Corp. has made capital contributions to allow investments by SoftBank Vision Fund 2 ("SVF 2") in certain portfolio companies. The information included herein is made for informational purposes only and does not constitute an offer to sell or a solicitation of an offer to buy limited partnership interests in any fund, including SVF 2. SVF 2 has yet to have an external close, and any potential third-party investors shall receive additional information related to any SVF 2 investments prior to closing.

The rest is here:
ElevateBio Announces Chief Scientific Officer of Regenerative Medicine, Melissa Carpenter, PhD, Elected to the International Society for Stem Cell...

Posted in Gene Medicine | Comments Off on ElevateBio Announces Chief Scientific Officer of Regenerative Medicine, Melissa Carpenter, PhD, Elected to the International Society for Stem Cell…

New gene therapy innovation centre to advance scientific discoveries into life-changing treatments – University of Sheffield News

Posted: at 4:59 pm

A pioneering gene therapy innovation centre at the University of Sheffield is set to advance scientific discoveries into promising treatment options for millions of patients with life-threatening diseases.

The Gene Therapy Innovation and Manufacturing Centre (GTIMC), led by Professor Mimoun Azzouz, is one of three pioneering hubs announced today (18 March 2021) in a new 18 million network funded by LifeArc and the Medical Research Council (MRC), with support from the Biotechnology and Biological Sciences Research Council (BBSRC).

Gene therapy is a promising treatment option for more than 7,000 rare diseases that currently have no cure. It aims to treat these conditions, by engineering another gene to replace, silence or manipulate the faulty one.

The UK has a world-class genetics research base however, to date, academics have found it difficult to get access to the clinical materials, facilities and expertise required to progress gene therapy research into clinical trials.

Professor Mimoun Azzouz, Director of the GTIMC and Chair of Translational Neuroscience at the University of Sheffield, said: The Gene Therapy Innovation and Manufacturing Centre will tackle major challenges in gene therapy development for some of the most devastating diseases.

Gene therapies are pioneering medical advances that have the potential to offer much-needed, novel, effective treatments for many rare and incurable diseases that cannot be treated by conventional drug compounds.

This is a momentous milestone for revolutionary medical advances not only for Sheffield and South Yorkshire, but also for the UK.

Sheffield has emerged as one of the leading players in the cell and gene therapy and this national network of partners, facilities and training programmes will allow us to keep pace with translational discoveries for new and potentially life changing treatments

Professor Mimoun Azzouz

Director of the GTIMC and Chair of Translational Neuroscience at the University of Sheffield

The new centre, which will be the first in the north of England, builds on the University of Sheffields strong history of translational research and its reputation as an international centre of excellence for gene therapeutics.

Professor Dame Pam Shaw, Director of the NIHR Sheffield Biomedical Research Centre and co-applicant on the GTIMC application said: This exciting development and partnership will speed up the pull through of new gene therapies into early phase clinical trials and offer hope to patients with neurological and other rare diseases that can be addressed in this way.

The support given to this initiative will greatly accelerate the translational potential of genetic therapies in the UK and bring benefits in key areas of unmet medical need.

The state-of-the-art centre will bring together academic institutions, NHS trusts, non-profit and industry partners across the north of England, Midlands and Wales enabling academic-led clinical trials of novel gene therapies. The GTIMC will deliver essential translational and regulatory support alongside extensive training and skills programmes to enable upskilling and address shortage of skills in Good Manufacturing Practice (GMP) manufacturing.

Professor Koen Lamberts, President and Vice-Chancellor of the University of Sheffield, said: At the University of Sheffield we focus our research on finding real-world solutions to some of the biggest global challenges. The Gene Therapy Innovation and Manufacturing Centre will unlock development pathways for new treatments for people affected by devastating genetic disorders, many of which have no cure.

We are delighted that our University is at the forefront of research in this pioneering field of medicine and that this new centre will build on our reputation as an international centre of excellence for gene therapeutics.

This is fantastic news for the City Region and the North of England and we look forward to working collaboratively to share technical skills and resources across the coordinated network.

Alongside the national network funding from LifeArc, the MRC and BBSRC, the GTIMC was made possible thanks to a 3 million donation from The Law Family Charitable Foundation, established by Andrew Law and his wife Zo. This funding was part of a record 5 million donation from the University of Sheffield alumnus, which will also see the launch of a new student support programme.

Andrew Law, who is Chairman and CEO of Caxton Associates, said: The University of Sheffield is rapidly developing a global reputation in gene therapy. The new Gene Therapy Innovation and Manufacturing Centre will drive innovation and world-class research, while presenting a real opportunity to catalyse the creation of new start-up companies to facilitate commercialisation in the North.

This investment will enhance vital partnerships with biotechnology and pharmaceutical companies to help accelerate gene therapy programmes and clinical trials, at the same time as supporting regional economic growth and job creation.

The GTIMC is planned for a site on the University of Sheffields Innovation District close to existing translational research facilities and will contribute to an ongoing programme of regional investment and regeneration.

The centre will include a cutting-edge GMP (good manufacturing practice) facility that will support gene therapy projects emerging from universities across the UK.

The facility will utilise highly efficient processes to manufacture clinical grade adeno-associated viruses (AAV) and provide all the necessary quality assurance, regulatory certification and governance for human trials at Advanced Therapies Treatment Centres and NHS trusts within the GTIMC and the national network.

The three national hubs, located at the University of Sheffield, Kings College London, and NHS Blood and Transplant in Bristol will operate as a coordinated network, sharing technical skills and resources to enable innovative gene therapy research.

Dr Melanie Lee, CEO of LifeArc, said: Recent innovations in gene therapies hold enormous potential for treating conditions such as rare diseases, but often promising ideas particularly in academia are not making it through to patients. Through our collaboration, we aim to meet the need for researchers to have access to the essential facilities and translational advice to progress promising research.

The GTIMC will manufacture commonly used vectors including both lentivirus and adeno-associated virus that are needed for genetic therapy trials, while positioning the UK for significant bioprocessing innovation work with the potential to radically increase yields and reduce productivity barriers in future years. The hub network will also design and share commercially ready platforms, using common cell-lines, plasmids and reagents to reduce costs, facilitate simplified licensing agreements and streamline regulatory reviews. A key aim is to smooth the transition between small-scale supply for early clinical trials through to larger-scale manufacture for patient trials, and beyond.

Professor Fiona Watt, MRC's Executive Chair, said: The new network of Innovation Hubs for gene therapies will build on the UKs great strengths in this area, providing targeted investment in vital infrastructure to accelerate academic research programmes down the path to patient benefit, supporting the delivery of a new wave of genetic medicines.

Notes for editors

Sheffield GTIMC Lead Institution

The University of Sheffield

Sheffield GTIMC Co-Lead Institutions

Cell & Gene Therapy Catapult

Centre of Process Innovation (CPI)

University of Birmingham

University of Liverpool

The Midlands-Wales Advanced Therapies Treatment Centre [MW-ATTC]

Sheffield GTIMC Partners

University of Leeds

University of Leicester

University of York

Cardiff University

University of Bradford

Northern Health Science Alliance (NHSA)

NHS Hospital Trusts of Sheffield, Leeds, Birmingham Womens and Children Hospital

Leeds Teaching Hospitals NHS Foundation Trust

Genomic Laboratory Hubs

Lonza AG

Cobra Biologics

Advanced Manufacturing Research Centre (AMRC)

National Horizons Centre (NHC)

National Institute for Biological Standards

View original post here:
New gene therapy innovation centre to advance scientific discoveries into life-changing treatments - University of Sheffield News

Posted in Gene Medicine | Comments Off on New gene therapy innovation centre to advance scientific discoveries into life-changing treatments – University of Sheffield News

LogicBio Therapeutics Reports Full Year 2020 Financial Results and Provides Business Update – PRNewswire

Posted: at 4:59 pm

LEXINGTON, Mass., March 15, 2021 /PRNewswire/ --LogicBio Therapeutics, Inc. (Nasdaq:LOGC) (LogicBio or the Company), a clinical- stage genetic medicine company pioneering gene delivery and gene editing platforms to address rare and serious diseases from infancy through adulthood, today reported financial results for the year ended December 31, 2020 and provided a business update.

"The fourth quarter of 2020 marked a continuation of strong momentum for LogicBio which has also extended into 2021," said Frederic Chereau, President and CEO. "Most notably, we continue to anticipate enrolling the first patient in our SUNRISE Trial of LB-001 for the treatment of MMA in the very near future. We expect this event will be followed by additional enrollments soon after and allow us to begin to generate data regarding the safety, and clinical impact of treatment with LB-001 for these children. In addition, we also recently announced that we have extended our collaboration with the Children's Medical Research Institute of Australia to continue to develop sAAVy, our next generation capsids platform, which has already yielded novel liver-tropic capsids that we believe are superior to ones that are currently used in the clinic. We believe these capsids will allow LogicBio to continue to expand its gene editing platform, GeneRide, and potentially initiate gene therapy programs, either alone or in collaboration with strategic partners. We look forward to providing further updates on these programs throughout 2021." Mr. Chereau concluded by saying, "I am also very excited by the new members we have added to our team. Both Mariana Nacht, our new CSO, and Cecilia Jones, our new CFO, bring a wealth of biotech industry experience, and I am delighted they have chosen to join LogicBio as we move into this exciting next phase of progress."

Anticipated LogicBio Milestones for 2021:

LB-001 for MMA

Pipeline

Full Year 2020 Financial Results

Twelve Months Ended December 31, 2020 and 2019

About LogicBio Therapeutics

LogicBio Therapeutics is a clinical-stage genetic medicine company pioneering gene delivery and gene editing platforms to address rare and serious diseases from infancy through adulthood. The company's proprietary GeneRide platform is a new approach to precise gene insertion that harnesses a cell's natural DNA repair process leading to durable therapeutic protein expression levels. LogicBio's cutting-edge sAAVy capsid development platform is designed to support development of treatments in a broad range of indications and tissues. The company is based in Lexington, MA. For more information, visit http://www.logicbio.com.

Forward Looking Statements

This press release contains "forward-looking" statements within the meaning of the federal securities laws, including those related to the Company's plans to enroll patients in, advance and complete its planned Phase 1/2 SUNRISE clinical trial of LB-001 in MMA and the potential benefits to patients of LB-001; the timing, progress and results of the Company's research and development activities, including those related to the GeneRide technology platform and sAAVy, our next generation capsids program; and the sufficiency of our cash on hand to fund our operating expenses and capital expenditures. These are not statements of historical facts and are based on management's beliefs and assumptions and on information currently available. They are subject to risks and uncertainties that could cause the actual results and the implementation of the Company's plans to vary materially, including the risks associated with the initiation, cost, timing, progress and results of the Company's current and future research and development activities and preclinical studies and potential future clinical trials. In particular, the impact of the COVID-19 pandemic on the Company's ability to progress with its research, development, manufacturing and regulatory efforts, including the Company's plans to enroll patients in, advance and complete its Phase 1/2 clinical trial for LB-001 in MMA, and the value of and market for the Company's common stock, will depend on future developments that are highly uncertain and cannot be predicted with confidence at this time, such as the ultimate duration of the pandemic, travel restrictions, quarantines, social distancing and business closure requirements in the United States and in other countries, and the effectiveness of actions taken globally to contain and treat the disease. These risks are discussed in the Company's filings with the U.S. Securities and Exchange Commission (SEC), including, without limitation, the Company's Annual Report on Form 10-K filed on March 16, 2020 with the SEC, the Company's Quarterly Report on Form 10-Q filed on May 11, 2020, and the Company's subsequent Quarterly Reports on Form 10-Q and other filings with the SEC. Except as required by law, the Company assumes no obligation to update these forward-looking statements publicly, even if new information becomes available in the future.

LogicBio Therapeutics, Inc.

Condensed Consolidated Statements of Operations

(In thousands, except share and per share data)

Year Ended December 31,

2020

2019

REVENUE

Service revenue

$ 3,454

$

Total revenue

3,454

OPERATING EXPENSES

Research and development

22,753

30,656

General and administrative

12,212

10,385

Total operating expenses

34,965

41,041

LOSS FROM OPERATIONS

(31,511)

(41,041)

OTHER (EXPENSE) INCOME, NET:

Interest income

181

1,500

Interest expense

(1,098)

(546)

Other expense, net

(5)

(19)

Total other (expense) income, net

(922)

935

Loss before income taxes

(32,433)

(40,106)

Income tax provision

(188)

(22)

Net loss

$ (32,621)

$ (40,128)

Net loss per sharebasic and diluted

$ (1.29)

$ (1.78)

Weighted-average common stock outstandingbasic and diluted

25,364,453

22,602,954

LogicBio Therapeutics, Inc.

Condensed Consolidated Balance Sheets

(In thousands)

As of

December 31, 2020

December 31, 2019

Cash, cash equivalents and investments

$ 70,075

$ 50,647

Other assets

10,565

5,013

TOTAL ASSETS

$ 80,640

$ 55,660

Accounts payable, accrued expenses and other liabilities

$ 19,213

$ 13,373

Stockholders' equity

61,427

42,287

TOTAL LIABILITIES AND STOCKHOLDERS' EQUITY

$ 80,640

$ 55,660

SOURCE LogicBio Therapeutics, Inc.

http://www.logicbio.com

View post:
LogicBio Therapeutics Reports Full Year 2020 Financial Results and Provides Business Update - PRNewswire

Posted in Gene Medicine | Comments Off on LogicBio Therapeutics Reports Full Year 2020 Financial Results and Provides Business Update – PRNewswire

Generation Bio Reports Fourth Quarter and Full Year 2020 Financial Results – BioSpace

Posted: at 4:59 pm

$262.3 million cash balance at the end of 2020 bolstered by additional net proceeds of $211.3 million from follow-on offering, funding key milestones into 2024

Ron Cooper appointed to Board of Directors

Company remains on track to begin IND-enabling studies for hemophilia A in 2021

CAMBRIDGE, Mass., March 18, 2021 (GLOBE NEWSWIRE) --Generation BioCo. (Nasdaq: GBIO), an innovative genetic medicines company creating a new class of non-viral gene therapy, reported recent business highlights and fourth quarter and full year 2020 financial results.

2020 was a foundational year for Generation Bio in which we strengthened the leadership team, advanced our platform science, and added substantially to our balance sheet to fund our lead programs to human data, said Geoff McDonough, M.D., president and chief executive officer of Generation Bio. We entered 2021 with preclinical data in mice and non-human primates that we believe demonstrate our platforms potential to go beyond what is currently possible with gene therapy for hemophilia A. We expect to announce additional data throughout the remainder of this year that will support the rapid expansion of our pipeline and showcase our differentiated approach to creating a new class of genetic medicine.

Recent Business Highlights

Selected Anticipated Company Milestones in 2021

Fourth Quarter and Full Year 2020 Financial Results

About Generation Bio

Generation Bio is an innovative genetic medicines company focused on creating a new class of non-viral gene therapy to provide durable, redosable treatments for people living with rare and prevalent diseases. The companys non-viral platform incorporates a proprietary, high-capacity DNA construct called closed-ended DNA, or ceDNA; a cell-targeted lipid nanoparticle delivery system, or ctLNP; and an established, scalable capsid-free manufacturing process. The platform is designed to enable multi-year durability from a single dose of ceDNA and to allow titration and redosing if needed. The ctLNP is engineered to deliver large genetic payloads, including multiple genes, to specific tissues to address a wide range of indications. The companys efficient, scalable manufacturing process supports Generation Bios mission to extend the reach of gene therapy to more people, living with more diseases, in more places around the world.

Forward-Looking Statements

Any statements in this press release about future expectations, plans and prospects for the Company, including statements about our strategic plans or objectives, our technology platforms, our research and clinical development plans, and other statements containing the words believes, anticipates, plans, expects, and similar expressions, constitute forward-looking statements within the meaning of The Private Securities Litigation Reform Act of 1995. Actual results may differ materially from those indicated by such forward-looking statements as a result of various important factors, including: uncertainties inherent in the identification and development of product candidates, including the conduct of research activities, the initiation and completion of preclinical studies and clinical trials and clinical development of the Companys product candidates; uncertainties as to the availability and timing of results from preclinical studies and clinical trials; whether results from preclinical studies will be predictive of the results of later preclinical studies and clinical trials; expectations for regulatory approvals to conduct trials or to market products; challenges in the manufacture of genetic medicine products; whether the Companys cash resources are sufficient to fund the Companys operating expenses and capital expenditure requirements for the period anticipated; the impact of the COVID-19 pandemic on the Companys business and operations; as well as the other risks and uncertainties set forth in the Risk Factors section of our most recent quarterly report on Form 10-Q, which is on file with the Securities and Exchange Commission, and in subsequent filings the Company may make with the Securities and Exchange Commission. In addition, the forward-looking statements included in this press release represent the Companys views as of the date hereof. The Company anticipates that subsequent events and developments will cause the Companys views to change.However, while the Company may elect to update these forward-looking statements at some point in the future, the Company specifically disclaims any obligation to do so.These forward-looking statements should not be relied upon as representing the Companys views as of any date subsequent to the date on which they were made.

Contacts:

InvestorsMaren KillackeyGeneration Bio541-646-2420mkillackey@generationbio.com

MediaAlicia WebbGeneration Bio847-254-4275awebb@generationbio.com

Stephanie SimonTen Bridge Communications617-581-9333stephanie@tenbridgecommunications.com

GENERATION BIO CO.CONSOLIDATED BALANCE SHEET DATA (Unaudited)(In thousands)

GENERATION BIO CO.CONSOLIDATED STATEMENTS OF OPERATIONS (Unaudited)(in thousands, except share and per share data)

View original post here:
Generation Bio Reports Fourth Quarter and Full Year 2020 Financial Results - BioSpace

Posted in Gene Medicine | Comments Off on Generation Bio Reports Fourth Quarter and Full Year 2020 Financial Results – BioSpace

UVA Scientist Developing Gene Therapy to Help Girls With Rett Syndrome – University of Virginia

Posted: at 4:59 pm

AUniversity of Virginia School of Medicinescientist is developing an innovative gene therapy she hopes will slow disease progression and improve movement, coordination and communication in children with Rett syndrome. The approach also may be useful for battling other genetic disorders involving the X chromosome.

UVA researcher Sanchita Bhatnagar discovered that tiny bits of RNA, called microRNAs, play an important role in Rett, a rare genetic disorder that can impair childrens ability to speak, move and even breathe. Based on that finding, she is seeking to sop up those RNA bits, called microRNA, using absorbent particles called microRNA sponges.

Early work in lab models has produced promising results, and she hopes the approach could lead to a better quality of life for children with Rett.

We are seeing that lab animals treated with this gene therapy are more mobile. Theyre moving faster, theyre smarter, Bhatnagar said. And if that translates into even modest improvements for children, it could make a big difference, she said: If we can help a child to move more independently, or improve their ability to communicate, I think for a parent, thats a big win.

Rett syndrome affects approximately 1 in 10,000 children, almost exclusively girls. Thats because its caused by a mutation in theMECP2gene found on the X chromosome. Girls have two X chromosomes, while boys have an X and a Y.

Bhatnagars discovery of the role of microRNA in Rett came as a surprise, because it was the first time microRNA had been linked to X chromosome biology. That finding dovetailed perfectly with her labs expertise in creating microRNA sponges that can target microRNA, absorbing it and then breaking it down.

We are not altering the genomic DNA, Bhatnagar emphasized. We are just using them [the microRNA sponges] as inhibitors that are delivered through [adeno-associated viral] vectors. These vectors do not go and integrate into the genome, so we hope for minimal secondary effects.

Bhatnagar hopes her new approach could be useful both for treating Rett and other genetic disorders that involve the X chromosome, such as Fragile X syndrome, a condition that causes learning disabilities, developmental delays and behavioral problems. To advance her research toward patient impact, she works with the UVA Licensing & Ventures Group to navigate patent protection and industry partnerships. After disclosing her discovery to the Licensing & Ventures Group in 2019, Bhatnagar presented her research and attracted a top expert in developing new gene therapies, Kathrin Meyer of the Abigail Wexner Research Institute at Nationwide Childrens Hospital, as a research collaborator. Massachusetts-based Alcyone Therapeutics Inc. is sponsoring the research at both UVA and the Wexner Research Institute to support its transition to clinic.

The research collaborators hope to begin clinical trials in the near future. If the initial trials prove safe and successful, that would lead to larger trials to evaluate the effectiveness of the approach. The federal government would then consider the trial results to determine if the approach should be approved as a treatment.

We think what Dr. Bhatnagar has come up with is very elegant, and we are excited about its potential to improve the lives of many patients, PJ Anand, CEO of Alcyone Therapeutics, said. The beautiful part is that this gene therapy is what we call in the industry a pipeline in a product. The exact same gene therapy product can be used to potentially address diseases that are caused by other genes on the X chromosome. Once we have the initial proof of concept in the clinic for the first disease, we can use much of that same information to move toward clinical trials in other X-linked diseases as well, so it will really be an efficient path forward.

Bhatnagar, of UVAs Department of Biochemistry and Molecular Genetics, said she feels very emotional about the potential clinical implications of her work, especially knowing the dramatic impact Rett has on the affected girls and their families.

Rett syndromeleads to severe impairments, affecting nearly every aspect of a childs life.Its often a full-time job as a parent, because these girls need assistance with all their living skills, Bhatnagar said. I dont know if this therapy is going to fix all of that. But it may be able to help them, and any help will be a big step forward.

The early-stage studies for the project in Bhatnagar lab were supported by the Hartwell Foundation and the UVA Brain Center.

To keep up with the latest medical research news from UVA, subscribe to theMaking of Medicineblog.

See the original post here:
UVA Scientist Developing Gene Therapy to Help Girls With Rett Syndrome - University of Virginia

Posted in Gene Medicine | Comments Off on UVA Scientist Developing Gene Therapy to Help Girls With Rett Syndrome – University of Virginia

Page 1,030«..1020..1,0291,0301,0311,032..1,0401,050..»