13 results on '"Katalin Karikó"'
Search Results
2. Development of GPC2-directed chimeric antigen receptors using mRNA for pediatric brain tumors
- Author
-
David M Barrett, Daniel Martinez, Tiffany Smith, Jessica B Foster, Crystal Griffin, Jo Lynne Rokita, Allison Stern, Cameron Brimley, Komal Rathi, Maria V Lane, Samantha N Buongervino, Peter J Madsen, Alberto Delaidelli, Poul H Sorensen, Robert J Wechsler-Reya, Katalin Karikó, Phillip B Storm, Adam C Resnick, John M Maris, and Kristopher R Bosse
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Pediatric brain tumors are the leading cause of cancer death in children with an urgent need for innovative therapies. Glypican 2 (GPC2) is a cell surface oncoprotein expressed in neuroblastoma for which targeted immunotherapies have been developed. This work aimed to characterize GPC2 expression in pediatric brain tumors and develop an mRNA CAR T cell approach against this target.Methods We investigated GPC2 expression across a cohort of primary pediatric brain tumor samples and cell lines using RNA sequencing, immunohistochemistry, and flow cytometry. To target GPC2 in the brain with adoptive cellular therapies and mitigate potential inflammatory neurotoxicity, we used optimized mRNA to create transient chimeric antigen receptor (CAR) T cells. We developed four mRNA CAR T cell constructs using the highly GPC2-specific fully human D3 single chain variable fragment for preclinical testing.Results We identified high GPC2 expression across multiple pediatric brain tumor types including medulloblastomas, embryonal tumors with multilayered rosettes, other central nervous system embryonal tumors, as well as definable subsets of highly malignant gliomas. We next validated and prioritized CAR configurations using in vitro cytotoxicity assays with GPC2-expressing neuroblastoma cells, where the light-to-heavy single chain variable fragment configurations proved to be superior. We expanded the testing of the two most potent GPC2-directed CAR constructs to GPC2-expressing medulloblastoma and high-grade glioma cell lines, showing significant GPC2-specific cell death in multiple models. Finally, biweekly locoregional delivery of 2–4 million GPC2-directed mRNA CAR T cells induced significant tumor regression in an orthotopic medulloblastoma model and significantly prolonged survival in an aggressive orthotopic thalamic diffuse midline glioma xenograft model. No GPC2-directed CAR T cell related neurologic or systemic toxicity was observed.Conclusion Taken together, these data show that GPC2 is a highly differentially expressed cell surface protein on multiple malignant pediatric brain tumors that can be targeted safely with local delivery of mRNA CAR T cells, laying the framework for the clinical translation of GPC2-directed immunotherapies for pediatric brain tumors.
- Published
- 2022
- Full Text
- View/download PDF
3. ASL mRNA-LNP Therapeutic for the Treatment of Argininosuccinic Aciduria Enables Survival Benefit in a Mouse Model
- Author
-
Owen Daly, Azita Josefine Mahiny, Sara Majeski, Kevin McClintock, Julia Reichert, Gábor Boros, Gábor Tamás Szabó, Jonas Reinholz, Petra Schreiner, Steve Reid, Kieu Lam, Marlen Lepper, Melanie Adler, Tracy Meffen, James Heyes, Katalin Karikó, Pete Lutwyche, and Irena Vlatkovic
- Subjects
lipid nanoparticle-mRNA (LNP-mRNA) ,mRNA optimization ,mRNA therapeutic ,rare disease ,argininosuccinic aciduria (ASA) ,argininosuccinate lyase deficiency (ASLD) ,Biology (General) ,QH301-705.5 - Abstract
Argininosuccinic aciduria (ASA) is a metabolic disorder caused by a deficiency in argininosuccinate lyase (ASL), which cleaves argininosuccinic acid to arginine and fumarate in the urea cycle. ASL deficiency (ASLD) leads to hepatocyte dysfunction, hyperammonemia, encephalopathy, and respiratory alkalosis. Here we describe a novel therapeutic approach for treating ASA, based on nucleoside-modified messenger RNA (modRNA) formulated in lipid nanoparticles (LNP). To optimize ASL-encoding mRNA, we modified its cap, 5′ and 3′ untranslated regions, coding sequence, and the poly(A) tail. We tested multiple optimizations of the formulated mRNA in human cells and wild-type C57BL/6 mice. The ASL protein showed robust expression in vitro and in vivo and a favorable safety profile, with low cytokine and chemokine secretion even upon administration of increasing doses of ASL mRNA-LNP. In the ASLNeo/Neo mouse model of ASLD, intravenous administration of the lead therapeutic candidate LNP-ASL CDS2 drastically improved the survival of the mice. When administered twice a week lower doses partially protected and 3 mg/kg LNP-ASL CDS2 fully protected the mice. These results demonstrate the considerable potential of LNP-formulated, modified ASL-encoding mRNA as an effective alternative to AAV-based approaches for the treatment of ASA.
- Published
- 2023
- Full Text
- View/download PDF
4. Ribozyme Assays to Quantify the Capping Efficiency of In Vitro-Transcribed mRNA
- Author
-
Irena Vlatkovic, János Ludwig, Gábor Boros, Gábor Tamás Szabó, Julia Reichert, Maximilian Buff, Markus Baiersdörfer, Jonas Reinholz, Azita Josefine Mahiny, Uğur Şahin, and Katalin Karikó
- Subjects
mRNA capping efficiency ,ribozyme ,in vitro-transcribed (IVT) mRNA ,cap ,quality control ,Pharmacy and materia medica ,RS1-441 - Abstract
The presence of the cap structure on the 5′-end of in vitro-transcribed (IVT) mRNA determines its translation and stability, underpinning its use in therapeutics. Both enzymatic and co-transcriptional capping may lead to incomplete positioning of the cap on newly synthesized RNA molecules. IVT mRNAs are rapidly emerging as novel biologics, including recent vaccines against COVID-19 and vaccine candidates against other infectious diseases, as well as for cancer immunotherapies and protein replacement therapies. Quality control methods necessary for the preclinical and clinical stages of development of these therapeutics are under ongoing development. Here, we described a method to assess the presence of the cap structure of IVT mRNAs. We designed a set of ribozyme assays to specifically cleave IVT mRNAs at a unique position and release 5′-end capped or uncapped cleavage products up to 30 nt long. We purified these products using silica-based columns and visualized/quantified them using denaturing polyacrylamide gel electrophoresis (PAGE) or liquid chromatography and mass spectrometry (LC–MS). Using this technology, we determined the capping efficiencies of IVT mRNAs with different features, which include: Different cap structures, diverse 5′ untranslated regions, different nucleoside modifications, and diverse lengths. Taken together, the ribozyme cleavage assays we developed are fast and reliable for the analysis of capping efficiency for research and development purposes, as well as a general quality control for mRNA-based therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
5. ASL mRNA-LNP Therapeutic for the Treatment of Argininosuccinic Aciduria Enables Survival Benefit in a Mouse Model
- Author
-
Vlatkovic, Owen Daly, Azita Josefine Mahiny, Sara Majeski, Kevin McClintock, Julia Reichert, Gábor Boros, Gábor Tamás Szabó, Jonas Reinholz, Petra Schreiner, Steve Reid, Kieu Lam, Marlen Lepper, Melanie Adler, Tracy Meffen, James Heyes, Katalin Karikó, Pete Lutwyche, and Irena
- Subjects
lipid nanoparticle-mRNA (LNP-mRNA) ,mRNA optimization ,mRNA therapeutic ,rare disease ,argininosuccinic aciduria (ASA) ,argininosuccinate lyase deficiency (ASLD) - Abstract
Argininosuccinic aciduria (ASA) is a metabolic disorder caused by a deficiency in argininosuccinate lyase (ASL), which cleaves argininosuccinic acid to arginine and fumarate in the urea cycle. ASL deficiency (ASLD) leads to hepatocyte dysfunction, hyperammonemia, encephalopathy, and respiratory alkalosis. Here we describe a novel therapeutic approach for treating ASA, based on nucleoside-modified messenger RNA (modRNA) formulated in lipid nanoparticles (LNP). To optimize ASL-encoding mRNA, we modified its cap, 5′ and 3′ untranslated regions, coding sequence, and the poly(A) tail. We tested multiple optimizations of the formulated mRNA in human cells and wild-type C57BL/6 mice. The ASL protein showed robust expression in vitro and in vivo and a favorable safety profile, with low cytokine and chemokine secretion even upon administration of increasing doses of ASL mRNA-LNP. In the ASLNeo/Neo mouse model of ASLD, intravenous administration of the lead therapeutic candidate LNP-ASL CDS2 drastically improved the survival of the mice. When administered twice a week lower doses partially protected and 3 mg/kg LNP-ASL CDS2 fully protected the mice. These results demonstrate the considerable potential of LNP-formulated, modified ASL-encoding mRNA as an effective alternative to AAV-based approaches for the treatment of ASA.
- Published
- 2023
- Full Text
- View/download PDF
6. Progress in vaccine development for infectious diseases—a Keystone Symposia report
- Author
-
Jennifer Cable, Barney S. Graham, Richard A. Koup, Robert A. Seder, Katalin Karikó, Norbert Pardi, Dan H. Barouch, Bhawna Sharma, Susanne Rauch, Raffael Nachbagauer, Mattias N. E. Forsell, Michael Schotsaert, Ali H. Ellebedy, Karin Loré, Darrell J. Irvine, Emily Pilkington, Siri Tahtinen, Elizabeth A. Thompson, Yanis Feraoun, Neil P. King, Kevin Saunders, Galit Alter, Syed M. Moin, Kwinten Sliepen, Gunilla B. Karlsson Hedestam, Hedda Wardemann, Bali Pulendran, Nicole A. Doria‐Rose, Wan‐Ting He, Jennifer A. Juno, Sila Ataca, Adam K. Wheatley, Jason S. McLellan, Laura M. Walker, Julia Lederhofer, Lisa C. Lindesmith, Holger Wille, Peter J. Hotez, and Linda‐Gail Bekker
- Subjects
History and Philosophy of Science ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
7. Single immunizations of self-amplifying or non-replicating mRNA-LNP vaccines control HPV-associated tumors in mice
- Author
-
Jamile Ramos da Silva, Karine Bitencourt Rodrigues, Guilherme Formoso Pelegrin, Natiely Silva Sales, Hiromi Muramatsu, Mariângela de Oliveira Silva, Bruna F. M. M. Porchia, Ana Carolina Ramos Moreno, Luana Raposo M. M. Aps, Aléxia Adrianne Venceslau-Carvalho, István Tombácz, Wesley Luzetti Fotoran, Katalin Karikó, Paulo J. C. Lin, Ying K. Tam, Mariana de Oliveira Diniz, Norbert Pardi, and Luís Carlos de Souza Ferreira
- Subjects
General Medicine - Abstract
As mRNA vaccines have proved to be very successful in battling the coronavirus disease 2019 (COVID-19) pandemic, this new modality has attracted widespread interest for the development of potent vaccines against other infectious diseases and cancer. Cervical cancer caused by persistent human papillomavirus (HPV) infection is a major cause of cancer-related deaths in women, and the development of safe and effective therapeutic strategies is urgently needed. In the present study, we compared the performance of three different mRNA vaccine modalities to target tumors associated with HPV-16 infection in mice. We generated lipid nanoparticle (LNP)–encapsulated self-amplifying mRNA as well as unmodified and nucleoside-modified non-replicating mRNA vaccines encoding a chimeric protein derived from the fusion of the HPV-16 E7 oncoprotein and the herpes simplex virus type 1 glycoprotein D (gDE7). We demonstrated that single low-dose immunizations with any of the three gDE7 mRNA vaccines induced activation of E7-specific CD8 + T cells, generated memory T cell responses capable of preventing tumor relapses, and eradicated subcutaneous tumors at different growth stages. In addition, the gDE7 mRNA-LNP vaccines induced potent tumor protection in two different orthotopic mouse tumor models after administration of a single vaccine dose. Last, comparative studies demonstrated that all three gDE7 mRNA-LNP vaccines proved to be superior to gDE7 DNA and gDE7 recombinant protein vaccines. Collectively, we demonstrated the immunogenicity and therapeutic efficacy of three different mRNA vaccines in extensive comparative experiments. Our data support further evaluation of these mRNA vaccines in clinical trials.
- Published
- 2023
- Full Text
- View/download PDF
8. Development of GPC2-directed chimeric antigen receptors using mRNA for pediatric brain tumors
- Author
-
Jessica B Foster, Crystal Griffin, Jo Lynne Rokita, Allison Stern, Cameron Brimley, Komal Rathi, Maria V Lane, Samantha N Buongervino, Tiffany Smith, Peter J Madsen, Daniel Martinez, Alberto Delaidelli, Poul H Sorensen, Robert J Wechsler-Reya, Katalin Karikó, Phillip B Storm, David M Barrett, Adam C Resnick, John M Maris, and Kristopher R Bosse
- Subjects
Pharmacology ,Oncogene Proteins ,Cancer Research ,Receptors, Chimeric Antigen ,Brain Neoplasms ,Immunology ,Glioma ,Xenograft Model Antitumor Assays ,Neuroblastoma ,Oncology ,Glypicans ,Cell Line, Tumor ,Molecular Medicine ,Immunology and Allergy ,Humans ,RNA, Messenger ,Cerebellar Neoplasms ,Child ,Medulloblastoma ,Single-Chain Antibodies - Abstract
BackgroundPediatric brain tumors are the leading cause of cancer death in children with an urgent need for innovative therapies. Glypican 2 (GPC2) is a cell surface oncoprotein expressed in neuroblastoma for which targeted immunotherapies have been developed. This work aimed to characterize GPC2 expression in pediatric brain tumors and develop an mRNA CAR T cell approach against this target.MethodsWe investigated GPC2 expression across a cohort of primary pediatric brain tumor samples and cell lines using RNA sequencing, immunohistochemistry, and flow cytometry. To target GPC2 in the brain with adoptive cellular therapies and mitigate potential inflammatory neurotoxicity, we used optimized mRNA to create transient chimeric antigen receptor (CAR) T cells. We developed four mRNA CAR T cell constructs using the highly GPC2-specific fully human D3 single chain variable fragment for preclinical testing.ResultsWe identified high GPC2 expression across multiple pediatric brain tumor types including medulloblastomas, embryonal tumors with multilayered rosettes, other central nervous system embryonal tumors, as well as definable subsets of highly malignant gliomas. We next validated and prioritized CAR configurations using in vitro cytotoxicity assays with GPC2-expressing neuroblastoma cells, where the light-to-heavy single chain variable fragment configurations proved to be superior. We expanded the testing of the two most potent GPC2-directed CAR constructs to GPC2-expressing medulloblastoma and high-grade glioma cell lines, showing significant GPC2-specific cell death in multiple models. Finally, biweekly locoregional delivery of 2–4 million GPC2-directed mRNA CAR T cells induced significant tumor regression in an orthotopic medulloblastoma model and significantly prolonged survival in an aggressive orthotopic thalamic diffuse midline glioma xenograft model. No GPC2-directed CAR T cell related neurologic or systemic toxicity was observed.ConclusionTaken together, these data show that GPC2 is a highly differentially expressed cell surface protein on multiple malignant pediatric brain tumors that can be targeted safely with local delivery of mRNA CAR T cells, laying the framework for the clinical translation of GPC2-directed immunotherapies for pediatric brain tumors.
- Published
- 2022
9. A systematic dissection of determinants and consequences of snoRNA-guided pseudouridylation of human mRNA
- Author
-
Ronit Nir, Thomas Philipp Hoernes, Hiromi Muramatsu, Klaus Faserl, Katalin Karikó, Matthias David Erlacher, Aldema Sas-Chen, and Schraga Schwartz
- Subjects
RNA, Ribosomal ,Protein Biosynthesis ,Genetics ,Humans ,RNA, Small Nucleolar ,RNA, Messenger ,RNA Processing, Post-Transcriptional ,Pseudouridine - Abstract
RNA can be extensively modified post-transcriptionally with >170 covalent modifications, expanding its functional and structural repertoire. Pseudouridine (Ψ), the most abundant modified nucleoside in rRNA and tRNA, has recently been found within mRNA molecules. It remains unclear whether pseudouridylation of mRNA can be snoRNA-guided, bearing important implications for understanding the physiological target spectrum of snoRNAs and for their potential therapeutic exploitation in genetic diseases. Here, using a massively parallel reporter based strategy we simultaneously interrogate Ψ levels across hundreds of synthetic constructs with predesigned complementarity against endogenous snoRNAs. Our results demonstrate that snoRNA-mediated pseudouridylation can occur on mRNA targets. However, this is typically achieved at relatively low efficiencies, and is constrained by mRNA localization, snoRNA expression levels and the length of the snoRNA:mRNA complementarity stretches. We exploited these insights for the design of snoRNAs targeting pseudouridylation at premature termination codons, which was previously shown to suppress translational termination. However, in this and follow-up experiments in human cells we observe no evidence for significant levels of readthrough of pseudouridylated stop codons. Our study enhances our understanding of the scope, ‘design rules’, constraints and consequences of snoRNA-mediated pseudouridylation.
- Published
- 2022
10. Developing mRNA for Therapy
- Author
-
Katalin Karikó
- Subjects
Mammals ,Vaccines, Synthetic ,COVID-19 Vaccines ,Liposomes ,Animals ,COVID-19 ,Humans ,Nanoparticles ,General Medicine ,RNA, Messenger ,mRNA Vaccines - Abstract
Messenger RNA was discovered in 1961 and it took 60 years until the first mRNA became FDA-approved product in the form of COVID-19 mRNA vaccine. During those years a lot of progress has been made by hundreds of scientists. It was 1978 when the first-time isolated mRNA delivered into mammalian cells produced the encoded protein. In vitro transcription introduced in 1984 made it possible to generate any desired mRNA from the encoding plasmid using phage RNA polymerases. In the early 90s mRNA was used for therapy as well as vaccine against infectious diseases and cancer. Inflammatory nature of the mRNAs limited their in vivo use. Replacing uridine with pseudouridine made the mRNA non-immunogenic, more stable and highly translatable. Delivery of the lipid nanoparticle-formulated nucleoside-modified mRNA encoding viral antigens became a platform for effective vaccine. Labile nature of the mRNA is ideal for transient production of the viral antigen, to generate effective antibody and cellular immune response. The mRNA platform is revolutionizing the delivery of effective and safe vaccines, therapeutics and gene therapies.
- Published
- 2022
11. Breaking Through : My Life in Science
- Author
-
Katalin Karikó and Katalin Karikó
- Subjects
- COVID-19 vaccines--Development, Women immigrants--United States--Biography, Biochemists--United States--Biography, Women biochemists--United States--Biography, Immigrants--United States--Biography, mRNA vaccines--Development, Hungarians--United States--Biography, Hungarian Americans--Biography
- Abstract
A powerful memoir from Katalin Karikó, winner of the 2023 Nobel Prize in Physiology or Medicine, whose decades-long research led to the COVID-19 vaccines “Katalin Karikó's story is an inspiration.”—Bill Gates“Riveting... a true story of a brilliant biochemist who never gave up or gave in.”—Bonnie Garmus, author of Lessons in ChemistryA KIRKUS REVIEWS BEST BOOK OF THE YEARKatalin Karikó has had an unlikely journey. The daughter of a butcher in postwar communist Hungary, Karikó grew up in an adobe home that lacked running water, and her family grew their own vegetables. She saw the wonders of nature all around her and was determined to become a scientist. That determination eventually brought her to the United States, where she arrived as a postdoctoral fellow in 1985 with $1,200 sewn into her toddler's teddy bear and a dream to remake medicine. Karikó worked in obscurity, battled cockroaches in a windowless lab, and faced outright derision and even deportation threats from her bosses and colleagues. She balked as prestigious research institutions increasingly conflated science and money. Despite setbacks, she never wavered in her belief that an ephemeral and underappreciated molecule called messenger RNA could change the world. Karikó believed that someday mRNA would transform ordinary cells into tiny factories capable of producing their own medicines on demand. She sacrificed nearly everything for this dream, but the obstacles she faced only motivated her, and eventually she succeeded.Karikó's three-decade-long investigation into mRNA would lead to a staggering achievement: vaccines that protected millions of people from the most dire consequences of COVID-19. These vaccines are just the beginning of mRNA's potential. Today, the medical community eagerly awaits more mRNA vaccines—for the flu, HIV, and other emerging infectious diseases.Breaking Through isn't just the story of an extraordinary woman. It's an indictment of closed-minded thinking and a testament to one woman's commitment to laboring intensely in obscurity—knowing she might never be recognized in a culture that is driven by prestige, power, and privilege—because she believed her work would save lives.
- Published
- 2023
12. Honorary Doctorate Dr. Katalin Karikó
- Author
-
Katalin Karikó, Floris Rutjes, Katalin Karikó, and Floris Rutjes
- Subjects
- Messenger RNA, mRNA vaccines
- Abstract
On 20 October 2022, the day that Radboud University celebrated its 99th anniversary, Dr. Katalin Karikó received a Radboud honorary doctorate in recognition of her scientific contributions to developing mRNA-based vaccines. This edition includes the laudatio of the honorary supervisor and the speech of the honorary doctor. Karikó spent years researching medical applications of mRNA. Her dream was to develop synthetic mRNA and use this to cure cancer, strokes, and influenza. Eventually, after years of toil, rejection, and criticism from colleagues, she and fellow researcher Drew Weissman demonstrated that it is possible to trigger an immune response in the body with mRNA without the body turning against the mRNA itself. With this breakthrough, a new revolutionary technique was born. Thanks to Karikó's scientific work, BioNTech/Pfizer and Moderna were able to develop the current mRNA vaccines against COVID-19. A prime example of the beneficial impact that fundamental research can eventually have on society. Honorary supervisor Floris Rutjes, Professor in Organic Synthesis: “With courage and determination, she pursued her scientific vision for a very long time, and by doing so, she has ultimately made a significant contribution to the fight against viral diseases.”
- Published
- 2023
13. Reducing cell intrinsic immunity to mRNA vaccine alters adaptive immune responses in mice
- Author
-
Ziyin Wang, Egon J. Jacobus, David C. Stirling, Stefanie Krumm, Katie E. Flight, Robert F. Cunliffe, Jonathan Mottl, Charanjit Singh, Lucy G. Mosscrop, Leticia Aragão Santiago, Annette B. Vogel, Katalin Kariko, Ugur Sahin, Stephanie Erbar, and John S. Tregoning
- Subjects
MT: Bioinformatics ,RNA vaccine ,inflammation ,influenza ,innate ,sensing ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The response to mRNA vaccines needs to be sufficient for immune cell activation and recruitment, but moderate enough to ensure efficacious antigen expression. The choice of the cap structure and use of N1-methylpseudouridine (m1Ψ) instead of uridine, which have been shown to reduce RNA sensing by the cellular innate immune system, has led to improved efficacy of mRNA vaccine platforms. Understanding how RNA modifications influence the cell intrinsic immune response may help in the development of more effective mRNA vaccines. In the current study, we compared mRNA vaccines in mice against influenza virus using three different mRNA formats: uridine-containing mRNA (D1-uRNA), m1Ψ-modified mRNA (D1-modRNA), and D1-modRNA with a cap1 structure (cC1-modRNA). D1-uRNA vaccine induced a significantly different gene expression profile to the modified mRNA vaccines, with an up-regulation of Stat1 and RnaseL, and increased systemic inflammation. This result correlated with significantly reduced antigen-specific antibody responses and reduced protection against influenza virus infection compared with D1-modRNA and cC1-modRNA. Incorporation of m1Ψ alone without cap1 improved antibodies, but both modifications were required for the optimum response. Therefore, the incorporation of m1Ψ and cap1 alters protective immunity from mRNA vaccines by altering the innate immune response to the vaccine material.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.