Network


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

Hotspot


Dive into the research topics where Jonathan E. Zuckerman is active.

Publication


Featured researches published by Jonathan E. Zuckerman.


Nature | 2010

Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles

Mark E. Davis; Jonathan E. Zuckerman; Chung Hang J. Choi; David Seligson; Anthony William Tolcher; Christopher A. Alabi; Yun Yen; Jeremy D. Heidel; Antoni Ribas

Therapeutics that are designed to engage RNA interference (RNAi) pathways have the potential to provide new, major ways of imparting therapy to patients. Long, double-stranded RNAs were first shown to mediate RNAi in Caenorhabditis elegans, and the potential use of RNAi for human therapy has been demonstrated by the finding that small interfering RNAs (siRNAs; approximately 21-base-pair double-stranded RNA) can elicit RNAi in mammalian cells without producing an interferon response. We are at present conducting the first in-human phase I clinical trial involving the systemic administration of siRNA to patients with solid cancers using a targeted, nanoparticle delivery system. Here we provide evidence of inducing an RNAi mechanism of action in a human from the delivered siRNA. Tumour biopsies from melanoma patients obtained after treatment show the presence of intracellularly localized nanoparticles in amounts that correlate with dose levels of the nanoparticles administered (this is, to our knowledge, a first for systemically delivered nanoparticles of any kind). Furthermore, a reduction was found in both the specific messenger RNA (M2 subunit of ribonucleotide reductase (RRM2)) and the protein (RRM2) levels when compared to pre-dosing tissue. Most notably, we detect the presence of an mRNA fragment that demonstrates that siRNA-mediated mRNA cleavage occurs specifically at the site predicted for an RNAi mechanism from a patient who received the highest dose of the nanoparticles. Together, these data demonstrate that siRNA administered systemically to a human can produce a specific gene inhibition (reduction in mRNA and protein) by an RNAi mechanism of action.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Targeting kidney mesangium by nanoparticles of defined size

Chung Hang J. Choi; Jonathan E. Zuckerman; Paul Webster; Mark E. Davis

Nanoparticles are being investigated for numerous medical applications and are showing potential as an emerging class of carriers for drug delivery. Investigations on how the physicochemical properties (e.g., size, surface charge, shape, and density of targeting ligands) of nanoparticles enable their ability to overcome biological barriers and reach designated cellular destinations in sufficient amounts to elicit biological efficacy are of interest. Despite proven success in nanoparticle accumulation at cellular locations and occurrence of downstream therapeutic effects (e.g., target gene inhibition) in a selected few organs such as tumor and liver, reports on effective delivery of engineered nanoparticles to other organs still remain scarce. Here, we show that nanoparticles of ~75 ± 25-nm diameters target the mesangium of the kidney. These data show the effects of particle diameter on targeting the mesangium of the kidney. Because many diseases originate from this area of the kidney, our findings establish design criteria for constructing nanoparticle-based therapeutics for targeting diseases that involve the mesangium of the kidney.


Journal of Translational Medicine | 2010

Differential sensitivity of melanoma cell lines with BRAFV600E mutation to the specific Raf inhibitor PLX4032

Jonas Sondergaard; Ramin Nazarian; Qi Wang; Deliang Guo; Teli Hsueh; Stephen Mok; Hooman Sazegar; Laura E. MacConaill; Jordi Barretina; Sarah M. Kehoe; Narsis Attar; Erika von Euw; Jonathan E. Zuckerman; Bartosz Chmielowski; Begoña Comin-Anduix; Richard C. Koya; Paul S. Mischel; Roger S. Lo; Antoni Ribas

Blocking oncogenic signaling induced by the BRAFV600E mutation is a promising approach for melanoma treatment. We tested the anti-tumor effects of a specific inhibitor of Raf protein kinases, PLX4032/RG7204, in melanoma cell lines. PLX4032 decreased signaling through the MAPK pathway only in cell lines with the BRAFV600E mutation. Seven out of 10 BRAFV600E mutant cell lines displayed sensitivity based on cell viability assays and three were resistant at concentrations up to 10 μM. Among the sensitive cell lines, four were highly sensitive with IC50 values below 1 μM, and three were moderately sensitive with IC50 values between 1 and 10 μM. There was evidence of MAPK pathway inhibition and cell cycle arrest in both sensitive and resistant cell lines. Genomic analysis by sequencing, genotyping of close to 400 oncogeninc mutations by mass spectrometry, and SNP arrays demonstrated no major differences in BRAF locus amplification or in other oncogenic events between sensitive and resistant cell lines. However, metabolic tracer uptake studies demonstrated that sensitive cell lines had a more profound inhibition of FDG uptake upon exposure to PLX4032 than resistant cell lines. In conclusion, BRAFV600E mutant melanoma cell lines displayed a range of sensitivities to PLX4032 and metabolic imaging using PET probes can be used to assess sensitivity.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Polycation-siRNA nanoparticles can disassemble at the kidney glomerular basement membrane

Jonathan E. Zuckerman; Chung Hang J. Choi; Han Han; Mark E. Davis

Despite being engineered to avoid renal clearance, many cationic polymer (polycation)-based siRNA nanoparticles that are used for systemic delivery are rapidly eliminated from the circulation. Here, we show that a component of the renal filtration barrier—the glomerular basement membrane (GBM)—can disassemble cationic cyclodextrin-containing polymer (CDP)-based siRNA nanoparticles and, thereby, facilitate their rapid elimination from circulation. Using confocal and electron microscopies, positron emission tomography, and compartment modeling, we demonstrate that siRNA nanoparticles, but not free siRNA, accumulate and disassemble in the GBM. We also confirm that the siRNA nanoparticles do not disassemble in blood plasma in vitro and in vivo. This clearance mechanism may affect any nanoparticles that assemble primarily by electrostatic interactions between cationic delivery components and anionic nucleic acids (or other therapeutic entities).


Nature Reviews Drug Discovery | 2015

Clinical experiences with systemically administered siRNA-based therapeutics in cancer

Jonathan E. Zuckerman; Mark E. Davis

Small interfering RNA (siRNA)-based therapies are emerging as a promising new anticancer approach, and a small number of Phase I clinical trials involving patients with solid tumours have now been completed. Encouraging results from these pioneering clinical studies show that these new therapeutics can successfully and safely inhibit targeted gene products in patients with cancer, and have taught us important lessons regarding appropriate dosages and schedules. In this Review, we critically assess these Phase I studies and discuss their implications for future clinical trial design. Key challenges and future directions in the development of siRNA-containing anticancer therapeutics are also considered.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA

Jonathan E. Zuckerman; Ismael Gritli; Anthony W. Tolcher; Jeremy D. Heidel; Dean Lim; Robert J. Morgan; Bartosz Chmielowski; Antoni Ribas; Mark E. Davis; Yun Yen

Significance CALAA-01 is a targeted nanoparticle containing siRNA that is a first-in-class experimental therapeutic for cancer. To our knowledge, it is the first targeted, polymer-based nanoparticle-carrying siRNA to be systemically administered to humans. Results from a human phase Ia/Ib clinical trial are presented and correlated to preclinical animal data to provide an initial assessment of how this class of experimental therapeutics is translated from animals to humans. Nanoparticle-based experimental therapeutics are currently being investigated in numerous human clinical trials. CALAA-01 is a targeted, polymer-based nanoparticle containing small interfering RNA (siRNA) and, to our knowledge, was the first RNA interference (RNAi)–based, experimental therapeutic to be administered to cancer patients. Here, we report the results from the initial phase I clinical trial where 24 patients with different cancers were treated with CALAA-01 and compare those results to data obtained from multispecies animal studies to provide a detailed example of translating this class of nanoparticles from animals to humans. The pharmacokinetics of CALAA-01 in mice, rats, monkeys, and humans show fast elimination and reveal that the maximum concentration obtained in the blood after i.v. administration correlates with body weight across all species. The safety profile of CALAA-01 in animals is similarly obtained in humans except that animal kidney toxicities are not observed in humans; this could be due to the use of a predosing hydration protocol used in the clinic. Taken in total, the animal models do appear to predict the behavior of CALAA-01 in humans.


Journal of Biological Chemistry | 2008

Anti-apoptotic Bcl-2 family proteins disassemble ceramide channels

Leah J. Siskind; Laurence Feinstein; Tingxi Yu; Joseph Samuel Davis; David H. Jones; Jinna Choi; Jonathan E. Zuckerman; Wenzhi Tan; R. Blake Hill; J. Marie Hardwick; Marco Colombini

Early in mitochondria-mediated apoptosis, the mitochondrial outer membrane becomes permeable to proteins that, when released into the cytosol, initiate the execution phase of apoptosis. Proteins in the Bcl-2 family regulate this permeabilization, but the molecular composition of the mitochondrial outer membrane pore is under debate. We reported previously that at physiologically relevant levels, ceramides form stable channels in mitochondrial outer membranes capable of passing the largest proteins known to exit mitochondria during apoptosis (Siskind, L. J., Kolesnick, R. N., and Colombini, M. (2006) Mitochondrion 6, 118–125). Here we show that Bcl-2 proteins are not required for ceramide to form protein-permeable channels in mitochondrial outer membranes. However, both recombinant human Bcl-xL and CED-9, the Caenorhabditis elegans Bcl-2 homologue, disassemble ceramide channels in the mitochondrial outer membranes of isolated mitochondria from rat liver and yeast. Importantly, Bcl-xL and CED-9 disassemble ceramide channels in the defined system of solvent-free planar phospholipid membranes. Thus, ceramide channel disassembly likely results from direct interaction with these anti-apoptotic proteins. Mutants of Bcl-xL act on ceramide channels as expected from their ability to be anti-apoptotic. Thus, ceramide channels may be one mechanism for releasing pro-apoptotic proteins from mitochondria during the induction phase of apoptosis.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Correlating preclinical animal studies and human clinical trials of a multifunctional, polymeric nanoparticle.

Scott Eliasof; Douglas Lazarus; Christian G. Peters; Roy I. Case; Roderic O. Cole; Jungyeon Hwang; Thomas Schluep; Joseph Chao; Jimmy Lin; Yun Yen; Han Han; Devin T. Wiley; Jonathan E. Zuckerman; Mark E. Davis

Nanoparticles are currently being investigated in a number of human clinical trials. As information on how nanoparticles function in humans is difficult to obtain, animal studies that can be correlative to human behavior are needed to provide guidance for human clinical trials. Here, we report correlative studies on animals and humans for CRLX101, a 20- to 30-nm-diameter, multifunctional, polymeric nanoparticle containing camptothecin (CPT). CRLX101 is currently in phase 2 clinical trials, and human data from several of the clinical investigations are compared with results from multispecies animal studies. The pharmacokinetics of polymer-conjugated CPT (indicative of the CRLX101 nanoparticles) in mice, rats, dogs, and humans reveal that the area under the curve scales linearly with milligrams of CPT per square meter for all species. Plasma concentrations of unconjugated CPT released from CRLX101 in animals and humans are consistent with each other after accounting for differences in serum albumin binding of CPT. Urinary excretion of polymer-conjugated CPT occurs primarily within the initial 24 h after dosing in animals and humans. The urinary excretion dynamics of polymer-conjugated and unconjugated CPT appear similar between animals and humans. CRLX101 accumulates into solid tumors and releases CPT over a period of several days to give inhibition of its target in animal xenograft models of cancer and in the tumors of humans. Taken in total, the evidence provided from animal models on the CRLX101 mechanism of action suggests that the behavior of CRLX101 in animals is translatable to humans.


Journal of Controlled Release | 2012

Systemic delivery of siRNA nanoparticles targeting RRM2 suppresses head and neck tumor growth

Mohammad Aminur Rahman; A.R.M. Ruhul Amin; Xu Wang; Jonathan E. Zuckerman; Chung Hang J. Choi; Bingsen Zhou; Dongsheng Wang; Sreenivas Nannapaneni; Lydia Koenig; Zhengjia Chen; Zhuo Georgia Chen; Yun Yen; Mark E. Davis; Dong M. Shin

Systemic delivery of siRNA to solid tumors remains challenging. In this study, we investigated the systemic delivery of a siRNA nanoparticle targeting ribonucleotide reductase subunit M2 (RRM2), and evaluated its intratumoral kinetics, efficacy and mechanism of action. Knockdown of RRM2 by an RNAi mechanism strongly inhibited cell growth in head and neck squamous cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC) cell lines. In a mouse xenograft model of HNSCC, a single intravenous injection led to the accumulation of intact nanoparticles in the tumor that disassembled over a period of at least 3days, leading to target gene knockdown lasting at least 10days. A four-dose schedule of siRNA nanoparticle delivering RRM2 siRNA targeted to HNSCC tumors significantly reduced tumor progression by suppressing cell proliferation and inducing apoptosis. These results show promise for the use of RRM2 siRNA-based therapy for HNSCC and possibly NSCLC.


Proceedings of the National Academy of Sciences of the United States of America | 2016

CRLX101 nanoparticles localize in human tumors and not in adjacent, nonneoplastic tissue after intravenous dosing

Andrew J. Clark; Devin T. Wiley; Jonathan E. Zuckerman; Paul Webster; Joseph Chao; James Lin; Yun Yen; Mark E. Davis

Significance Nanoparticle-based therapeutics rely on the enhanced permeability and retention effect to localize in solid tumors and not healthy tissue. These phenomena are rationalized from animal models of human disease. Since these models poorly represent human tumors, there is a need to obtain information from humans to better understand how nanoparticle-based therapeutics perform in humans. Here, we collected tumor and nonneoplastic tissue biopsies from cancer patients who have been administered CRLX101 and show that the intact nanoparticles localize in human tumors and not in adjacent tissues. Sufficient concentrations reach the tumors to cause down-regulation of topoisomerase I and carbonic anhydrase IX. These results will aid in better understanding how nanoparticle therapeutics function in humans and how to better design future therapeutics. Nanoparticle-based therapeutics are being used to treat patients with solid tumors. Whereas nanoparticles have been shown to preferentially accumulate in solid tumors of animal models, there is little evidence to prove that intact nanoparticles localize to solid tumors of humans when systemically administered. Here, tumor and adjacent, nonneoplastic tissue biopsies are obtained through endoscopic capture from patients with gastric, gastroesophageal, or esophageal cancer who are administered the nanoparticle CRLX101. Both the pre- and postdosing tissue samples adjacent to tumors show no definitive evidence of either the nanoparticle or its drug payload (camptothecin, CPT) contained within the nanoparticle. Similar results are obtained from the predosing tumor samples. However, in nine of nine patients that were evaluated, CPT is detected in the tumor tissue collected 24–48 h after CRLX101 administration. For five of these patients, evidence of the intact deposition of CRLX101 nanoparticles in the tumor tissue is obtained. Indications of CPT pharmacodynamics from tumor biomarkers such as carbonic anhydrase IX and topoisomerase I by immunohistochemistry show clear evidence of biological activity from the delivered CPT in the posttreatment tumors.

Collaboration


Dive into the Jonathan E. Zuckerman's collaboration.

Top Co-Authors

Avatar

Mark E. Davis

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Chung Hang J. Choi

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Rong Ma

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Antoni Ribas

University of California

View shared research outputs
Top Co-Authors

Avatar

Yanxia Wang

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Sarika Chaudhari

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Yun Yen

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Devin T. Wiley

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Han Han

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Joseph Chao

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge