Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where James C. Kaczmarek is active.

Publication


Featured researches published by James C. Kaczmarek.


Genome Medicine | 2017

Advances in the delivery of RNA therapeutics: from concept to clinical reality

James C. Kaczmarek; Piotr S. Kowalski; Daniel G. Anderson

The rapid expansion of the available genomic data continues to greatly impact biomedical science and medicine. Fulfilling the clinical potential of genetic discoveries requires the development of therapeutics that can specifically modulate the expression of disease-relevant genes. RNA-based drugs, including short interfering RNAs and antisense oligonucleotides, are particularly promising examples of this newer class of biologics. For over two decades, researchers have been trying to overcome major challenges for utilizing such RNAs in a therapeutic context, including intracellular delivery, stability, and immune response activation. This research is finally beginning to bear fruit as the first RNA drugs gain FDA approval and more advance to the final phases of clinical trials. Furthermore, the recent advent of CRISPR, an RNA-guided gene-editing technology, as well as new strides in the delivery of messenger RNA transcribed in vitro, have triggered a major expansion of the RNA-therapeutics field. In this review, we discuss the challenges for clinical translation of RNA-based therapeutics, with an emphasis on recent advances in delivery technologies, and present an overview of the applications of RNA-based drugs for modulation of gene/protein expression and genome editing that are currently being investigated both in the laboratory as well as in the clinic.


Angewandte Chemie | 2016

Polymer–Lipid Nanoparticles for Systemic Delivery of mRNA to the Lungs

James C. Kaczmarek; Asha K. Patel; Kevin J. Kauffman; Owen S. Fenton; Matthew J. Webber; Michael Heartlein; Frank Derosa; Daniel G. Anderson

Therapeutic nucleic acids hold great promise for the treatment of disease but require vectors for safe and effective delivery. Synthetic nanoparticle vectors composed of poly(β-amino esters) (PBAEs) and nucleic acids have previously demonstrated potential utility for local delivery applications. To expand this potential utility to include systemic delivery of mRNA, hybrid polymer-lipid nanoformulations for systemic delivery to the lungs were developed. Through coformulation of PBAEs with lipid-polyethylene glycol (PEG), mRNA formulations were developed with increased serum stability and increased in vitro potency. The formulations were capable of functional delivery of mRNA to the lungs after intravenous administration in mice. To our knowledge, this is the first report of the systemic administration of mRNA for delivery to the lungs using degradable polymer-lipid nanoparticles.


Cancer treatment and research | 2015

Cancer Nanotherapeutics in Clinical Trials

Abigail K. R. Lytton-Jean; Kevin J. Kauffman; James C. Kaczmarek; Robert Langer

To be legally sold in the United States, all drugs must go through the FDA approval process. This chapter introduces the FDA approval process and describes the clinical trials required for a drug to gain approval. We then look at the different cancer nanotherapeutics and in vivo diagnostics that are currently in clinical trials or have already received approval. These nanotechnologies are catagorized and described based on the delivery vehicle: liposomes, polymer micelles, albumin-bound chemotherapeutics, polymer-bound chemotherapeutics, and inorganic particles.


Advanced Materials | 2017

Synthesis and Biological Evaluation of Ionizable Lipid Materials for the In Vivo Delivery of Messenger RNA to B Lymphocytes

Owen S. Fenton; Kevin J. Kauffman; James C. Kaczmarek; Rebecca L. Mcclellan; Siddharth Jhunjhunwala; Mark W. Tibbitt; Manhao D. Zeng; Eric A. Appel; Joseph R. Dorkin; Faryal F. Mir; Jung H. Yang; Matthias A. Oberli; Michael Heartlein; Frank Derosa; Robert Langer; Daniel G. Anderson

B lymphocytes regulate several aspects of immunity including antibody production, cytokine secretion, and T-cell activation; moreover, B cell misregulation is implicated in autoimmune disorders and cancers such as multiple sclerosis and non-Hodgkins lymphomas. The delivery of messenger RNA (mRNA) into B cells can be used to modulate and study these biological functions by means of inducing functional protein expression in a dose-dependent and time-controlled manner. However, current in vivo mRNA delivery systems fail to transfect B lymphocytes and instead primarily target hepatocytes and dendritic cells. Here, the design, synthesis, and biological evaluation of a lipid nanoparticle (LNP) system that can encapsulate mRNA, navigate to the spleen, transfect B lymphocytes, and induce more than 60 pg of protein expression per million B cells within the spleen is described. Importantly, this LNP induces more than 85% of total protein production in the spleen, despite LNPs being observed transiently in the liver and other organs. These results demonstrate that LNP composition alone can be used to modulate the site of protein induction in vivo, highlighting the critical importance of designing and synthesizing new nanomaterials for nucleic acid delivery.


Molecular therapy. Nucleic acids | 2018

Rapid, Single-Cell Analysis and Discovery of Vectored mRNA Transfection In Vivo with a loxP-Flanked tdTomato Reporter Mouse

Kevin J. Kauffman; Matthias A. Oberli; J. Robert Dorkin; Juan E. Hurtado; James C. Kaczmarek; Shivani Bhadani; Jeff Wyckoff; Robert Langer; Ana Jaklenec; Daniel G. Anderson

mRNA therapeutics hold promise for the treatment of diseases requiring intracellular protein expression and for use in genome editing systems, but mRNA must transfect the desired tissue and cell type to be efficacious. Nanoparticle vectors that deliver the mRNA are often evaluated using mRNA encoding for reporter genes such as firefly luciferase (FLuc); however, single-cell resolution of mRNA expression cannot generally be achieved with FLuc, and, thus, the transfected cell populations cannot be determined without additional steps or experiments. To more rapidly identify which types of cells an mRNA formulation transfects in vivo, we describe a Cre recombinase (Cre)-based system that permanently expresses fluorescent tdTomato protein in transfected cells of genetically modified mice. Following in vivo application of vectored Cre mRNA, it is possible to visualize successfully transfected cells via Cre-mediated tdTomato expression in bulk tissues and with single-cell resolution. Using this system, we identify previously unknown transfected cell types of an existing mRNA delivery vehicle in vivo and also develop a new mRNA formulation capable of transfecting lung endothelial cells. Importantly, the same formulations with mRNA encoding for fluorescent protein delivered to wild-type mice did not produce sufficient signal for any visualization in vivo, demonstrating the significantly improved sensitivity of our Cre-based system. We believe that the system described here may facilitate the identification and characterization of mRNA delivery vectors to new tissues and cell types.


Advanced Healthcare Materials | 2018

Poly(β‐amino ester)‐co‐poly(caprolactone) Terpolymers as Nonviral Vectors for mRNA Delivery In Vitro and In Vivo

Umberto Capasso Palmiero; James C. Kaczmarek; Owen S. Fenton; Daniel G. Anderson

The production of new proteins with messenger RNA (mRNA) has gained a broad interest due to its potential for addressing a wide range of diseases. Here, the design and characterization of novel ionizable poly(β-amino ester)-co-poly(caprolactone) terpolymers, synthesized via the combination of the ring opening polymerization and the Michael step-growth polymerization, are reported. The versatility of this method is demonstrated by varying the number of caprolactone units attached to each poly(β-amino ester) (PBAE) terpolymer. The ability of the novel poly-caprolactone (PCL)-based PBAE materials to deliver mRNA is shown to depend on the physiochemical characteristics of the material, such as lipophilicity, as well as the formulation method used to complex the polymer with the oligonucleotide. This latter variable represents a previously unstudied aspect of PBAE library screens that can play an important role in identifying true top candidates for nucleic acid delivery. The most stable terpolymer is injected intravenously (IV) in mice and shows a transfection efficacy several times higher than the polyethylenimine (PEI) which is focused in the spleen, opening the possibility to use these biodegradable carriers in the intravenous delivery of antigen-encoding mRNA for cancer immunotherapy and vaccination.


Nano Letters | 2018

Optimization of a Degradable Polymer–Lipid Nanoparticle for Potent Systemic Delivery of mRNA to the Lung Endothelium and Immune Cells

James C. Kaczmarek; Kevin J. Kauffman; Owen S. Fenton; Kaitlyn Sadtler; Asha K. Patel; Michael Heartlein; Frank Derosa; Daniel G. Anderson

mRNA therapeutics hold great potential for treating a variety of diseases through protein-replacement, immunomodulation, and gene editing. However, much like siRNA therapy the majority of progress in mRNA delivery has been confined to the liver. Previously, we demonstrated that poly(β-amino esters), a class of degradable polymers, are capable of systemic mRNA delivery to the lungs in mice when formulated into nanoparticles with poly(ethylene glycol)-lipid conjugates. Using experimental design, a statistical approach to optimization that reduces experimental burden, we demonstrate herein that these degradable polymer-lipid nanoparticles can be optimized in terms of polymer synthesis and nanoparticle formulation to achieve a multiple order-of-magnitude increase in potency. Furthermore, using genetically engineered Cre reporter mice, we demonstrate that mRNA is functionally delivered to both the lung endothelium and pulmonary immune cells, expanding the potential utility of these nanoparticles.


Biomaterials | 2016

Efficacy and immunogenicity of unmodified and pseudouridine-modified mRNA delivered systemically with lipid nanoparticles in vivo

Kevin J. Kauffman; Faryal F. Mir; Siddharth Jhunjhunwala; James C. Kaczmarek; Juan E. Hurtado; Jung H. Yang; Matthew J. Webber; Piotr S. Kowalski; Michael Heartlein; Frank Derosa; Daniel G. Anderson


PMC | 2016

Polymer-Lipid Nanoparticles for Systemic Delivery of mRNA to the Lungs

Michael Heartlein; Frank Derosa; James C. Kaczmarek; Asha K. Patel; Kevin J. Kauffman; Owen S. Fenton; Matthew J. Webber; Daniel G. Anderson


Angewandte Chemie | 2018

Customizable Lipid Nanoparticle Materials for the Delivery of siRNAs and mRNAs

Owen S. Fenton; Kevin J. Kauffman; Rebecca L. Mcclellan; James C. Kaczmarek; Manhao D. Zeng; Jason L. Andresen; Luke Hyunsik Rhym; Michael Heartlein; Frank Derosa; Daniel G. Anderson

Collaboration


Dive into the James C. Kaczmarek's collaboration.

Top Co-Authors

Avatar

Daniel G. Anderson

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Kevin J. Kauffman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Owen S. Fenton

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Webber

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Robert Langer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Asha K. Patel

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Faryal F. Mir

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Juan E. Hurtado

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jung H. Yang

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Manhao D. Zeng

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge