Publications
Search Google Scholar | Search PubMed
2023
Baldwin, Paige; Sridhar, Srinivas; Singh, Bijay
Nanoencapsulated combination drug formulations Miscellaneous
2023, (US Patent 11,648,211).
BibTeX | Tags: Nanomedicine
@misc{baldwin2023nanoencapsulated,
title = {Nanoencapsulated combination drug formulations},
author = {Paige Baldwin and Srinivas Sridhar and Bijay Singh},
year = {2023},
date = {2023-05-01},
urldate = {2023-05-01},
note = {US Patent 11,648,211},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Sridhar, Srinivas
True superconductivity at near ambient temperature has not been confirmed by Dasenbrock-Gammon et al. Nature, volume 615, pages 244–250 (2023) Journal Article
In: Journal of Physics and Chemistry of Solids, pp. 111381, 2023.
BibTeX | Tags: Superconductivity
@article{sridhar2023trueb,
title = {True superconductivity at near ambient temperature has not been confirmed by Dasenbrock-Gammon et al. Nature, volume 615, pages 244–250 (2023)},
author = {Srinivas Sridhar},
year = {2023},
date = {2023-01-01},
urldate = {2023-01-01},
journal = {Journal of Physics and Chemistry of Solids},
pages = {111381},
publisher = {Pergamon},
keywords = {Superconductivity},
pubstate = {published},
tppubtype = {article}
}
Yang, Shicheng; Green, Allen; Brown, Needa; Robinson, Alexis; Senat, Merline; Testino, Bryanna; Dinulescu, Daniela M; Sridhar, Srinivas
Sustained delivery of PARP inhibitor Talazoparib for the treatment of BRCA-deficient ovarian cancer Journal Article
In: Frontiers in Oncology, vol. 13, pp. 1175617, 2023.
BibTeX | Tags: Nanomedicine
@article{yang2023sustained,
title = {Sustained delivery of PARP inhibitor Talazoparib for the treatment of BRCA-deficient ovarian cancer},
author = {Shicheng Yang and Allen Green and Needa Brown and Alexis Robinson and Merline Senat and Bryanna Testino and Daniela M Dinulescu and Srinivas Sridhar},
year = {2023},
date = {2023-01-01},
journal = {Frontiers in Oncology},
volume = {13},
pages = {1175617},
publisher = {Frontiers},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Sridhar, Srinivas
True superconductivity at near ambient temperature has not been confirmed by Dasenbrock-Gammon, et. al. Nature (2023) Journal Article
In: arXiv preprint arXiv:2303.05987, 2023.
BibTeX | Tags:
@article{sridhar2023true,
title = {True superconductivity at near ambient temperature has not been confirmed by Dasenbrock-Gammon, et. al. Nature (2023)},
author = {Srinivas Sridhar},
year = {2023},
date = {2023-01-01},
journal = {arXiv preprint arXiv:2303.05987},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2022
Thong, Phan Quoc; Huong, Le Thi Thu; Tu, Nguyen Dac; Nhung, Hoang Thi My; Khanh, Lam; Manh, Do Hung; Nam, Pham Hong; Phuc, Nguyen Xuan; Alonso, Javier; Qiao, Ju; others,
Multifunctional nanocarriers of Fe3O4@ PLA-PEG/curcumin for MRI, magnetic hyperthermia and drug delivery Journal Article
In: Nanomedicine, no. 0, 2022.
BibTeX | Tags: Nanomedicine
@article{thong2022multifunctional,
title = {Multifunctional nanocarriers of Fe3O4@ PLA-PEG/curcumin for MRI, magnetic hyperthermia and drug delivery},
author = {Phan Quoc Thong and Le Thi Thu Huong and Nguyen Dac Tu and Hoang Thi My Nhung and Lam Khanh and Do Hung Manh and Pham Hong Nam and Nguyen Xuan Phuc and Javier Alonso and Ju Qiao and others},
year = {2022},
date = {2022-01-01},
journal = {Nanomedicine},
number = {0},
publisher = {Future Medicine Ltd London, UK},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Kunjachan, Sijumon; Detappe, Alexandre; Kumar, Rajiv; Ireland, Thomas; Cameron, Lisa; Biancur, Douglas E; Motto-Ros, Vincent; Sancey, Lucie; Sridhar, Srinivas; Makrigiorgos, G Mike; others,
Nanoparticle Mediated Tumor Vascular Disruption: A Novel Strategy in Radiation Therapy (Retraction of Vol 15, Pg 7488, 2015) Miscellaneous
2022.
BibTeX | Tags: Nanomedicine
@misc{kunjachan2022nanoparticle,
title = {Nanoparticle Mediated Tumor Vascular Disruption: A Novel Strategy in Radiation Therapy (Retraction of Vol 15, Pg 7488, 2015)},
author = {Sijumon Kunjachan and Alexandre Detappe and Rajiv Kumar and Thomas Ireland and Lisa Cameron and Douglas E Biancur and Vincent Motto-Ros and Lucie Sancey and Srinivas Sridhar and G Mike Makrigiorgos and others},
year = {2022},
date = {2022-01-01},
publisher = {AMER CHEMICAL SOC 1155 16TH ST, NW, WASHINGTON, DC 20036 USA},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
2021
Cohen, Susan E; Hashmi, Sara M; III, A-Andrew D Jones; Lykourinou, Vasiliki; Ondrechen, Mary Jo; Sridhar, Srinivas; Ven, Anne L; Waters, Lauren S; Beuning, Penny J
Adapting undergraduate research to remote work to increase engagement Journal Article
In: The Biophysicist, vol. 2, no. 2, pp. 28–32, 2021.
@article{cohen2021adapting,
title = {Adapting undergraduate research to remote work to increase engagement},
author = {Susan E Cohen and Sara M Hashmi and A-Andrew D Jones III and Vasiliki Lykourinou and Mary Jo Ondrechen and Srinivas Sridhar and Anne L Ven and Lauren S Waters and Penny J Beuning},
year = {2021},
date = {2021-01-01},
urldate = {2021-01-01},
journal = {The Biophysicist},
volume = {2},
number = {2},
pages = {28–32},
publisher = {Biophysical Society},
keywords = {Education},
pubstate = {published},
tppubtype = {article}
}
Versek, Craig; Banijamali, S Mohammad Ali; Bex, Peter; Lashkari, Kameran; Kamarthi, Sagar; Sridhar, Srinivas
Portable diagnostic system for age-related macular degeneration screening using visual evoked potentials Journal Article
In: Eye and brain, pp. 111–127, 2021.
BibTeX | Tags: Neurotechnology
@article{versek2021portable,
title = {Portable diagnostic system for age-related macular degeneration screening using visual evoked potentials},
author = {Craig Versek and S Mohammad Ali Banijamali and Peter Bex and Kameran Lashkari and Sagar Kamarthi and Srinivas Sridhar},
year = {2021},
date = {2021-01-01},
urldate = {2021-01-01},
journal = {Eye and brain},
pages = {111–127},
publisher = {Taylor & Francis},
keywords = {Neurotechnology},
pubstate = {published},
tppubtype = {article}
}
Timms, Liam; Zhou, Tianyi; Lyu, Yue; Qiao, Ju; Mishra, Vishala; Lahoud, Rita Maria; Jayaraman, Gayatri Veeramani; Allegretti, Andrew S; Drew, David; Seethamraju, Ravi T; others,
Ferumoxytol-enhanced ultrashort TE MRA and quantitative morphometry of the human kidney vasculature Journal Article
In: Abdominal Radiology, vol. 46, pp. 3288–3300, 2021.
@article{timms2021ferumoxytol,
title = {Ferumoxytol-enhanced ultrashort TE MRA and quantitative morphometry of the human kidney vasculature},
author = {Liam Timms and Tianyi Zhou and Yue Lyu and Ju Qiao and Vishala Mishra and Rita Maria Lahoud and Gayatri Veeramani Jayaraman and Andrew S Allegretti and David Drew and Ravi T Seethamraju and others},
year = {2021},
date = {2021-01-01},
urldate = {2021-01-01},
journal = {Abdominal Radiology},
volume = {46},
pages = {3288–3300},
publisher = {Springer US},
keywords = {MRI},
pubstate = {published},
tppubtype = {article}
}
Yang, Shicheng; Wallach, Mia; Krishna, Apurva; Kurmasheva, Raushan; Sridhar, Srinivas
Recent developments in nanomedicine for pediatric cancer Journal Article
In: Journal of Clinical Medicine, vol. 10, no. 7, pp. 1437, 2021.
BibTeX | Tags: Nanomedicine
@article{yang2021recent,
title = {Recent developments in nanomedicine for pediatric cancer},
author = {Shicheng Yang and Mia Wallach and Apurva Krishna and Raushan Kurmasheva and Srinivas Sridhar},
year = {2021},
date = {2021-01-01},
journal = {Journal of Clinical Medicine},
volume = {10},
number = {7},
pages = {1437},
publisher = {MDPI},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Zhang, Di; Singh, Bijay; Moerland, Jessica; Mitchell, Owen; Lockwood, Lizbeth; Carapellucci, Sarah; Sridhar, Srinivas; Liby, Karen T
Sustained, local delivery of the PARP inhibitor talazoparib prevents the development of mammary gland hyperplasia in BRCA1-deficient mice Journal Article
In: Scientific Reports, vol. 11, no. 1, pp. 1234, 2021.
BibTeX | Tags: Nanomedicine
@article{zhang2021sustained,
title = {Sustained, local delivery of the PARP inhibitor talazoparib prevents the development of mammary gland hyperplasia in BRCA1-deficient mice},
author = {Di Zhang and Bijay Singh and Jessica Moerland and Owen Mitchell and Lizbeth Lockwood and Sarah Carapellucci and Srinivas Sridhar and Karen T Liby},
year = {2021},
date = {2021-01-01},
journal = {Scientific Reports},
volume = {11},
number = {1},
pages = {1234},
publisher = {Nature Publishing Group UK London},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Rodell, Christopher B; Baldwin, Paige; Fernandez, Bianca; Weissleder, Ralph; Sridhar, Srinivas; Dubach, John Matthew
Quantification of cellular drug biodistribution addresses challenges in evaluating in vitro and in vivo encapsulated drug delivery Journal Article
In: Advanced therapeutics, vol. 4, no. 3, pp. 2000125, 2021.
BibTeX | Tags: Nanomedicine
@article{rodell2021quantification,
title = {Quantification of cellular drug biodistribution addresses challenges in evaluating in vitro and in vivo encapsulated drug delivery},
author = {Christopher B Rodell and Paige Baldwin and Bianca Fernandez and Ralph Weissleder and Srinivas Sridhar and John Matthew Dubach},
year = {2021},
date = {2021-01-01},
journal = {Advanced therapeutics},
volume = {4},
number = {3},
pages = {2000125},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
2020
Wilfred, NGWA; Kumar, Rajiv; Makrigiorgos, Gerassimos; Sridhar, Srinivas; Dougan, Stephanie
Biomaterials for combined radiotherapy and immunotherapy of cancer Miscellaneous
2020, (US Patent 10,835,604).
BibTeX | Tags: Nanomedicine
@misc{wilfred2020biomaterials,
title = {Biomaterials for combined radiotherapy and immunotherapy of cancer},
author = {NGWA Wilfred and Rajiv Kumar and Gerassimos Makrigiorgos and Srinivas Sridhar and Stephanie Dougan},
year = {2020},
date = {2020-11-01},
urldate = {2020-11-01},
note = {US Patent 10,835,604},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Mitra, Ronodeep; Cheng, Ming; O'Neil, Gerard; Kulkarni, Praveen; Kumar, Rajiv; Sridhar, Srinivas; Ferris, Craig; Hamilton, James; Jo, Hanjoong; Ebong, Eno E
In Vivo Flow-Regulated Endothelial Glycocalyx Integrity Leveraged for Targeted Intravenous Nanoparticle Delivery Proceedings Article
In: 2020 Virtual AIChE Annual Meeting, AIChE 2020.
BibTeX | Tags: Nanomedicine
@inproceedings{mitra2020vivo,
title = {In Vivo Flow-Regulated Endothelial Glycocalyx Integrity Leveraged for Targeted Intravenous Nanoparticle Delivery},
author = {Ronodeep Mitra and Ming Cheng and Gerard O'Neil and Praveen Kulkarni and Rajiv Kumar and Srinivas Sridhar and Craig Ferris and James Hamilton and Hanjoong Jo and Eno E Ebong},
year = {2020},
date = {2020-01-01},
booktitle = {2020 Virtual AIChE Annual Meeting},
organization = {AIChE},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {inproceedings}
}
TANGUTOORI, Shifalika; Sridhar, Srinivas
Nanoparticle drug delivery system and method of treating cancer and neurotrauma Miscellaneous
2020.
BibTeX | Tags: Nanomedicine
@misc{tangutoori2020nanoparticle,
title = {Nanoparticle drug delivery system and method of treating cancer and neurotrauma},
author = {Shifalika TANGUTOORI and Srinivas Sridhar},
year = {2020},
date = {2020-01-01},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Singh, Bijay; Yang, Shicheng; Krishna, Apurva; Sridhar, Srinivas
Nanoparticle formulations of poly (ADP-ribose) polymerase inhibitors for cancer therapy Journal Article
In: Frontiers in Chemistry, vol. 8, pp. 594619, 2020.
BibTeX | Tags: Nanomedicine
@article{singh2020nanoparticle,
title = {Nanoparticle formulations of poly (ADP-ribose) polymerase inhibitors for cancer therapy},
author = {Bijay Singh and Shicheng Yang and Apurva Krishna and Srinivas Sridhar},
year = {2020},
date = {2020-01-01},
journal = {Frontiers in Chemistry},
volume = {8},
pages = {594619},
publisher = {Frontiers Media SA},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Kunjachan, Sijumon; Kotb, Shady; Pola, Robert; Pechar, Michal; Kumar, Rajiv; Singh, Bijay; Gremse, Felix; Taleeli, Reza; Trichard, Florian; Motto-Ros, Vincent; others,
Author correction: Selective priming of tumor Blood Vessels by Radiation therapy enhances nanodrug Delivery Journal Article
In: Scientific Reports, vol. 10, no. 1, pp. 15344, 2020.
BibTeX | Tags: Nanomedicine
@article{kunjachan2020author,
title = {Author correction: Selective priming of tumor Blood Vessels by Radiation therapy enhances nanodrug Delivery},
author = {Sijumon Kunjachan and Shady Kotb and Robert Pola and Michal Pechar and Rajiv Kumar and Bijay Singh and Felix Gremse and Reza Taleeli and Florian Trichard and Vincent Motto-Ros and others},
year = {2020},
date = {2020-01-01},
journal = {Scientific Reports},
volume = {10},
number = {1},
pages = {15344},
publisher = {Nature Publishing Group UK London},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Zhang, Di; Singh, Bijay; Moerland, Jessica; Mitchell, Owen; Lockwood, Lizbeth; Carapellucci, Sarah; Sridhar, Srinivas; Liby, Karen
Localized delivery of the PARP inhibitor Talazoparib for chemoprevention of breast cancer Journal Article
In: Cancer Research, vol. 80, no. 16_Supplement, pp. 12–12, 2020.
BibTeX | Tags: Nanomedicine
@article{zhang2020localized,
title = {Localized delivery of the PARP inhibitor Talazoparib for chemoprevention of breast cancer},
author = {Di Zhang and Bijay Singh and Jessica Moerland and Owen Mitchell and Lizbeth Lockwood and Sarah Carapellucci and Srinivas Sridhar and Karen Liby},
year = {2020},
date = {2020-01-01},
journal = {Cancer Research},
volume = {80},
number = {16_Supplement},
pages = {12–12},
publisher = {The American Association for Cancer Research},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Mueller, Romy; Yasmin-Karim, Sayeda; DeCosmo, Kaylie; Vazquez-Pagan, Ana; Sridhar, Srinivas; Kozono, David; Hesser, Juergen; Ngwa, Wilfred
Increased carcinoembryonic antigen expression on the surface of lung cancer cells using gold nanoparticles during radiotherapy Journal Article
In: Physica Medica, vol. 76, pp. 236–242, 2020.
BibTeX | Tags: Nanomedicine
@article{mueller2020increased,
title = {Increased carcinoembryonic antigen expression on the surface of lung cancer cells using gold nanoparticles during radiotherapy},
author = {Romy Mueller and Sayeda Yasmin-Karim and Kaylie DeCosmo and Ana Vazquez-Pagan and Srinivas Sridhar and David Kozono and Juergen Hesser and Wilfred Ngwa},
year = {2020},
date = {2020-01-01},
journal = {Physica Medica},
volume = {76},
pages = {236–242},
publisher = {Elsevier},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Virani, Needa A; Kelada, Olivia J; Kunjachan, Sijumon; Detappe, Alexandre; Kwon, Jihun; Hayashi, Jennifer; Vazquez-Pagan, Ana; Biancur, Douglas E; Ireland, Thomas; Kumar, Rajiv; others,
Noninvasive imaging of tumor hypoxia after nanoparticle-mediated tumor vascular disruption Journal Article
In: Plos one, vol. 15, no. 7, pp. e0236245, 2020.
BibTeX | Tags: Nanomedicine
@article{virani2020noninvasive,
title = {Noninvasive imaging of tumor hypoxia after nanoparticle-mediated tumor vascular disruption},
author = {Needa A Virani and Olivia J Kelada and Sijumon Kunjachan and Alexandre Detappe and Jihun Kwon and Jennifer Hayashi and Ana Vazquez-Pagan and Douglas E Biancur and Thomas Ireland and Rajiv Kumar and others},
year = {2020},
date = {2020-01-01},
journal = {Plos one},
volume = {15},
number = {7},
pages = {e0236245},
publisher = {Public Library of Science San Francisco, CA USA},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Schuemann, Jan; Bagley, Alexander F; Berbeco, Ross; Bromma, Kyle; Butterworth, Karl T; Byrne, Hilary L; Chithrani, B Devika; Cho, Sang Hyun; Cook, Jason R; Favaudon, Vincent; others,
Roadmap for metal nanoparticles in radiation therapy: Current status, translational challenges, and future directions Journal Article
In: Physics in Medicine & Biology, vol. 65, no. 21, pp. 21RM02, 2020.
BibTeX | Tags: Nanomedicine
@article{schuemann2020roadmap,
title = {Roadmap for metal nanoparticles in radiation therapy: Current status, translational challenges, and future directions},
author = {Jan Schuemann and Alexander F Bagley and Ross Berbeco and Kyle Bromma and Karl T Butterworth and Hilary L Byrne and B Devika Chithrani and Sang Hyun Cho and Jason R Cook and Vincent Favaudon and others},
year = {2020},
date = {2020-01-01},
journal = {Physics in Medicine & Biology},
volume = {65},
number = {21},
pages = {21RM02},
publisher = {IOP Publishing},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Mitra, Ronodeep; Qiao, Ju; Madhavan, Sudharsan; O’Neil, Gerard; Ritchie, Bailey; Kulkarni, Praveen; Sridhar, Srinivas; Ven, Anne; Cherry, Erica; Ferris, Craig; others,
High fat diet versus disturbed blood flow conditions: Implications for endothelial glycocalyx integrity and pre-atherosclerotic inflammation Journal Article
In: The FASEB Journal, vol. 34, no. S1, pp. 1–1, 2020.
BibTeX | Tags: Nanomedicine
@article{mitra2020high,
title = {High fat diet versus disturbed blood flow conditions: Implications for endothelial glycocalyx integrity and pre-atherosclerotic inflammation},
author = {Ronodeep Mitra and Ju Qiao and Sudharsan Madhavan and Gerard O’Neil and Bailey Ritchie and Praveen Kulkarni and Srinivas Sridhar and Anne Ven and Erica Cherry and Craig Ferris and others},
year = {2020},
date = {2020-01-01},
journal = {The FASEB Journal},
volume = {34},
number = {S1},
pages = {1–1},
publisher = {The Federation of American Societies for Experimental Biology},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Cheng, Ming J; Mitra, Ronodeep; Okorafor, Chinedu C; Nersesyan, Alina A; Harding, Ian C; Bal, Nandita N; Kumar, Rajiv; Jo, Hanjoong; Sridhar, Srinivas; Ebong, Eno E
Targeted intravenous nanoparticle delivery: role of flow and endothelial glycocalyx integrity Journal Article
In: Annals of biomedical engineering, vol. 48, pp. 1941–1954, 2020.
BibTeX | Tags: Nanomedicine
@article{cheng2020targetedb,
title = {Targeted intravenous nanoparticle delivery: role of flow and endothelial glycocalyx integrity},
author = {Ming J Cheng and Ronodeep Mitra and Chinedu C Okorafor and Alina A Nersesyan and Ian C Harding and Nandita N Bal and Rajiv Kumar and Hanjoong Jo and Srinivas Sridhar and Eno E Ebong},
year = {2020},
date = {2020-01-01},
urldate = {2020-01-01},
journal = {Annals of biomedical engineering},
volume = {48},
pages = {1941–1954},
publisher = {Springer International Publishing},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Versek, Craig; Banijamali, S Mohammad Ali; Bex, Peter J; Lashkari, Kameran; Kamarthi, Sagar V; Sridhar, Srinivas
Portable Objective Diagnostics using Visual Evoked Potentials for Age-related Macular Degeneration Journal Article
In: medRxiv, pp. 2020–01, 2020.
BibTeX | Tags: Neurotechnology
@article{versek2020portableb,
title = {Portable Objective Diagnostics using Visual Evoked Potentials for Age-related Macular Degeneration},
author = {Craig Versek and S Mohammad Ali Banijamali and Peter J Bex and Kameran Lashkari and Sagar V Kamarthi and Srinivas Sridhar},
year = {2020},
date = {2020-01-01},
urldate = {2020-01-01},
journal = {medRxiv},
pages = {2020–01},
publisher = {Cold Spring Harbor Laboratory Press},
keywords = {Neurotechnology},
pubstate = {published},
tppubtype = {article}
}
Cheng, Ming J; Mitra, Ronodeep; Okorafor, Chinedu C; Nersesyan, Alina A; Harding, Ian C; Bal, Nandita N; Kumar, Rajiv; Jo, Hanjoong; Sridhar, Srinivas; Ebong, Eno E
Targeted Intravenous Nanoparticle Delivery: Role of Flow and Endothelial Glycocalyx Integrity Journal Article
In: Annals of Biomedical Engineering, pp. 1–14, 2020.
Abstract | BibTeX | Tags: Nanomedicine
@article{cheng2020targeted,
title = {Targeted Intravenous Nanoparticle Delivery: Role of Flow and Endothelial Glycocalyx Integrity},
author = {Ming J Cheng and Ronodeep Mitra and Chinedu C Okorafor and Alina A Nersesyan and Ian C Harding and Nandita N Bal and Rajiv Kumar and Hanjoong Jo and Srinivas Sridhar and Eno E Ebong},
year = {2020},
date = {2020-01-01},
journal = {Annals of Biomedical Engineering},
pages = {1--14},
abstract = {Therapies for atherosclerotic cardiovascular disease should target early disease stages and specific vascular sites where disease occurs. Endothelial glycocalyx (GCX) degradation compromises endothelial barrier function and increases vascular permeability. This initiates pro-atherosclerotic lipids and inflammatory cells to penetrate vessel walls, and at the same time this can be leveraged for targeted drug delivery. In prior cell culture studies, GCX degradation significantly increased endothelial cell uptake of nanoparticle vehicles that are designed for drug delivery, compared to the effects of intact GCX. The present study assessed if the cell culture findings translate to selective nanoparticle uptake in animal vessels. In mice, the left carotid artery (LCA) was partially ligated to disturb blood flow, which induces GCX degradation, endothelial dysfunction, and atherosclerosis. After ligation, the LCA vessel wall exhibited.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Versek, Craig William; Banijamali, Mohammad Ali S; Bex, Peter J; Lashkari, Kameran; Kamarthi, Sagar V; Sridhar, Srinivas
Portable Objective Diagnostics using Visual Evoked Potentials for Age-related Macular Degeneration Journal Article
In: medRxiv, 2020.
Abstract | BibTeX | Tags: Neurotechnology, Opthalmology
@article{versek2020portable,
title = {Portable Objective Diagnostics using Visual Evoked Potentials for Age-related Macular Degeneration},
author = {Craig William Versek and Mohammad Ali S Banijamali and Peter J Bex and Kameran Lashkari and Sagar V Kamarthi and Srinivas Sridhar},
year = {2020},
date = {2020-01-01},
journal = {medRxiv},
publisher = {Cold Spring Harbor Laboratory Press},
abstract = {Delayed Dark Adapted vision Recovery (DAR) is a biomarker for Age-related Macular Degeneration (AMD); however, its measurement is burdensome for patients and examiners. We developed a portable, wireless, quick-setup system that employs a headset with a smartphone to deliver and analyze controlled dichoptic photobleach and pattern reversal stimuli, and with custom electroencephalography (EEG) electrodes, to measure objective Dark Adapted Visual Evoked Potentials (DAVEP) at multiple locations of the visual field in one comfortable 20-minute session, without requiring subject reporting. DAVEP responses post photobleach (up to 15 minutes), were measured concurrently in both eyes of 13 patients with AMD and 8 others not diagnosed with AMD. New unexpected features were observed in the DAVEP responses at high latencies to scotopic stimulus intensities. The amplitude recovery of the DAVEP response was significantly delayed in AMD patients compared with controls. We developed DAVEP1 scores, a simple metric for DAR, using it to successfully identify all 100% of AMD subjects and correctly classify 90% of subject eyes. Deficits in DAR in patients with AMD can be identified with this objective VEP based system using the DAVEP1 metric, a promising new objective biomarker for this disease that can be easily tested in a clinic.},
keywords = {Neurotechnology, Opthalmology},
pubstate = {published},
tppubtype = {article}
}
2019
Sridhar, Srinivas; Versek, Craig; Bex, Peter
Portable brain and vision diagnostic and therapeutic system Miscellaneous
2019, (US Patent App. 16/347,049).
Abstract | BibTeX | Tags: Neurotechnology, Opthalmology
@misc{sridhar2019portableb,
title = {Portable brain and vision diagnostic and therapeutic system},
author = {Srinivas Sridhar and Craig Versek and Peter Bex},
year = {2019},
date = {2019-10-01},
abstract = {A portable wireless neuromonitoring device can be used to diagnose and/or treat conditions of the brain and vision system. The device includes a sensor unit mountable on the head of a human subject and capable of recording signals from the brain in EEG and/or EFEG (electric field encephalography) mode, and the device can be used for simultaneous stimulus display and recording with latency of less than 1 millisecond. The device also includes electrodes that allow rapid set-up and measurement with low impedance contact with the scalp. The device can also be used in conjunction with virtual reality or alternate reality environments.},
note = {US Patent App. 16/347,049},
keywords = {Neurotechnology, Opthalmology},
pubstate = {published},
tppubtype = {misc}
}
Baldwin, Paige; Ohman, Anders; Medina, Jamie; Dinulescu, Daniela; Sridhar, Srinivas
NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL Proceedings Article
In: CLINICAL CANCER RESEARCH, pp. 198–198, AMER ASSOC CANCER RESEARCH 615 CHESTNUT ST, 17TH FLOOR, PHILADELPHIA, PA~… 2019.
Abstract | BibTeX | Tags: Nanomedicine
@inproceedings{baldwin2019nanoformulationc,
title = {NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL},
author = {Paige Baldwin and Anders Ohman and Jamie Medina and Daniela Dinulescu and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
booktitle = {CLINICAL CANCER RESEARCH},
volume = {25},
number = {22},
pages = {198--198},
organization = {AMER ASSOC CANCER RESEARCH 615 CHESTNUT ST, 17TH FLOOR, PHILADELPHIA, PA~…},
abstract = {Talazoparib, a potent PARP inhibitor (PARPi), induces synthetic lethality in BRCA-deficient cancers making it an attractive candidate for ovarian cancer treatment. However, its potency lends itself to side effects associated more closely with traditional chemotherapeutics than other clinically approved PARPi's. We sought to formulate Talazoparib in a nanoparticle delivery system such that the drug could be administered intraperitoneally, localizing the entire dose at the disease site, to increase therapeutic efficacy and minimize toxicity. NanoTalazoparib was formulated and characterized and found to have a mean diameter of 70 nm and a neutral surface charge. Talazoparib and NanoTalazoparib were tested on a panel of murine tubal and human HGSOC cell lines and dose response compared to the first clinically approved PARPi, Olaparib. Dose response data indicated all cell lines were more sensitive to Talazoparib and NanoTalazoparib than Olaparib and all lines showed the same sensitivity to nanoformulations as free drugs. The human cell lines had various BRCA mutations and deletions, as well as a homologous recombination proficient (HRP) line, however, the HRP line was more sensitive to treatment than some HRD lines. Therapeutic efficacy was tested in vivo in a murine cancer model that mimics disseminated peritoneal disease. NanoTalazoparib 3X weekly for 8 weeks did not shrink tumors but resulted in tumor growth inhibition of 64% while an equivalent dose of oral Talazoparib only resulted in 34% growth inhibition. NanoTalazoparib suppressed the average volume of ascites at the study endpoint by 3.45 times more than oral Talazoparib. H&E staining of the tissues indicated no significant toxicity to the organs of the mononuclear phagocyte system. These results indicate that NanoTalazoparib can be used to localize PARPi therapy to the peritoneal cavity for disseminated late stage ovarian cancer treatment. Our data suggests that NanoTalazoparib could be utilized to delay the formation of tumor ascites for women with HR-deficient disease. While NanoTalazoparib did not effectively treat the disseminated disease at this dose, it may have clinical utility, either in combination with other therapies or as a maintenance therapy. Preclinical data indicates PARP inhibitors potentiate damage when combined with other cytotoxic treatments, however, in the clinic this has resulted in enhanced toxicity, forcing dose reduction and delay. The IP administration of NanoTalazoparib may provide a route to bypass some of the toxicities that have plagued combination treatments.
Supported in part by Rivkin Foundation and CDMRP Ovarian Cancer Research Program
Citation Format: Paige Baldwin, Anders Ohman, Jamie Medina, Daniela Dinulescu, Srinivas Sridhar. NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL [abstract]. In: Proceedings of the 12th Biennial Ovarian Cancer Research Symposium; Sep 13-15, 2018; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2019;25(22 Suppl):Abstract nr NT-087.
©2019 American Association for Cancer Research.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {inproceedings}
}
Supported in part by Rivkin Foundation and CDMRP Ovarian Cancer Research Program
Citation Format: Paige Baldwin, Anders Ohman, Jamie Medina, Daniela Dinulescu, Srinivas Sridhar. NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL [abstract]. In: Proceedings of the 12th Biennial Ovarian Cancer Research Symposium; Sep 13-15, 2018; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2019;25(22 Suppl):Abstract nr NT-087.
©2019 American Association for Cancer Research.
Singh, Bijay; Yang, Shicheng; Baldwin, Paige; van de Ven, Anne; Sridhar, Srinivas
Abstract A104: Nanoformulations of PARP and CDK inhibitors for cancer therapy Miscellaneous
2019.
Abstract | BibTeX | Tags: Nanomedicine
@misc{singh2019abstract,
title = {Abstract A104: Nanoformulations of PARP and CDK inhibitors for cancer therapy},
author = {Bijay Singh and Shicheng Yang and Paige Baldwin and Anne van de Ven and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
publisher = {American Association for Cancer Research},
abstract = {Poly(ADP-ribose) polymerase (PARP) inhibitors have been majorly utilized in cancers with BRCA1/2 mutations that have deficiencies in the homologous recombination (HR) DNA repair pathway. Traditionally, PARP inhibitors have been administered through oral route in the clinic but they have resulted in poor bioavailability, low tumor accumulation and high systemic toxicity. Moreover, tumors develop resistance to PARP inhibitors necessitating the development of strategies to re-sensitize these resistant tumors. To overcome these limitations, nanoparticle drug formulation has demonstrated a great potential to increase the drug concentration and accumulation at tumor sites with low systemic toxicities. Here we report the development of several nanoformulations of PARP inhibitors and observe their efficacies by systemic administration to treat various cancer diseases. We used two FDA approved PARP inhibitors (Olaparib or Talazoparib) for nanoformulations of NanoOlaparib and NanoTalazoparib, decorated with EpCAM or EGFR antibody, and tested on breast, ovarian, prostate and lung cancer with or without irradiation. In HR deficient models, the nanoformulations showed significant inhibition of tumor cell growth in vitro and in vivo. Above all, combination of PARP inhibitor with irradiation resulted in greater efficacy in lung and prostate cancer models. To overcome PARP inhibitor resistance, we formulated nanoparticles, NanoDinaciclib of cyclin-dependent kinase 12 (CDK12) inhibitor (dinaciclib) and used in combination with nanoformulation of PARP inhibitor to arrest the tumor growth in animal model. Our current research reveals that the use of CDK12 inhibitor in PARP resistant tumors further arrests tumor growth compared with monotherapy alone. Further studies will help clarify to achieve clear understanding of the mechanism of action CDK12 inhibitor to determine the best way to use PARP inhibitors beyond HR deficiency.
Citation Format: Bijay Singh, Shicheng Yang, Paige Baldwin, Anne van de Ven, Srinivas Sridhar. Nanoformulations of PARP and CDK inhibitors for cancer therapy [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A104. doi:10.1158/1535-7163.TARG-19-A104
©2019 American Association for Cancer Research.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Citation Format: Bijay Singh, Shicheng Yang, Paige Baldwin, Anne van de Ven, Srinivas Sridhar. Nanoformulations of PARP and CDK inhibitors for cancer therapy [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A104. doi:10.1158/1535-7163.TARG-19-A104
©2019 American Association for Cancer Research.
Baldwin, Paige; Likhotvorik, Rostislav; Baig, Nabeela; Cropper, Jodie; Carlson, Ruth; Kurmasheva, Raushan; Sridhar, Srinivas
Nanoformulation of talazoparib increases maximum tolerated doses in combination with temozolomide for treatment of Ewing sarcoma Journal Article
In: Frontiers in Oncology, vol. 9, pp. 1416, 2019.
Abstract | BibTeX | Tags: Nanomedicine
@article{baldwin2019nanoformulationb,
title = {Nanoformulation of talazoparib increases maximum tolerated doses in combination with temozolomide for treatment of Ewing sarcoma},
author = {Paige Baldwin and Rostislav Likhotvorik and Nabeela Baig and Jodie Cropper and Ruth Carlson and Raushan Kurmasheva and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
journal = {Frontiers in Oncology},
volume = {9},
pages = {1416},
publisher = {Frontiers},
abstract = {The Pediatric Preclinical Testing Program previously identified the PARP inhibitor talazoparib (TLZ) as a means to potentiate temozolomide (TMZ) activity for the treatment of Ewing sarcoma. However, the combination of TLZ and TMZ has been toxic in both preclinical and clinical testing, necessitating TMZ dose reduction to ~15% of the single agent maximum tolerated dose. We have synthesized a nanoparticle formulation of talazoparib (NanoTLZ) to be administered intravenously in an effort to modulate the toxicity profile of this combination treatment. Results in Ewing sarcoma xenograft models are presented to demonstrate the utility of this delivery method both alone and in combination with TMZ. NanoTLZ reduced gross toxicity and had a higher maximum tolerated dose than oral TLZ. The dose of TMZ did not have to be reduced when combined with NanoTLZ as was required when combined with oral TLZ. This indicated the NanoTLZ delivery system may be advantageous in decreasing the systemic toxicity associated with the combination of oral TLZ and TMZ.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Baldwin, Paige; Ohman, Anders; Medina, Jamie; Dinulescu, Daniela; Sridhar, Srinivas
Abstract NT-087: NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL Miscellaneous
2019.
Abstract | BibTeX | Tags: Nanomedicine
@misc{baldwin2019abstract,
title = {Abstract NT-087: NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL},
author = {Paige Baldwin and Anders Ohman and Jamie Medina and Daniela Dinulescu and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
publisher = {American Association for Cancer Research},
abstract = {Talazoparib, a potent PARP inhibitor (PARPi), induces synthetic lethality in BRCA-deficient cancers making it an attractive candidate for ovarian cancer treatment. However, its potency lends itself to side effects associated more closely with traditional chemotherapeutics than other clinically approved PARPi's. We sought to formulate Talazoparib in a nanoparticle delivery system such that the drug could be administered intraperitoneally, localizing the entire dose at the disease site, to increase therapeutic efficacy and minimize toxicity. NanoTalazoparib was formulated and characterized and found to have a mean diameter of 70 nm and a neutral surface charge. Talazoparib and NanoTalazoparib were tested on a panel of murine tubal and human HGSOC cell lines and dose response compared to the first clinically approved PARPi, Olaparib. Dose response data indicated all cell lines were more sensitive to Talazoparib and NanoTalazoparib than Olaparib and all lines showed the same sensitivity to nanoformulations as free drugs. The human cell lines had various BRCA mutations and deletions, as well as a homologous recombination proficient (HRP) line, however, the HRP line was more sensitive to treatment than some HRD lines. Therapeutic efficacy was tested in vivo in a murine cancer model that mimics disseminated peritoneal disease. NanoTalazoparib 3X weekly for 8 weeks did not shrink tumors but resulted in tumor growth inhibition of 64% while an equivalent dose of oral Talazoparib only resulted in 34% growth inhibition. NanoTalazoparib suppressed the average volume of ascites at the study endpoint by 3.45 times more than oral Talazoparib. H&E staining of the tissues indicated no significant toxicity to the organs of the mononuclear phagocyte system. These results indicate that NanoTalazoparib can be used to localize PARPi therapy to the peritoneal cavity for disseminated late stage ovarian cancer treatment. Our data suggests that NanoTalazoparib could be utilized to delay the formation of tumor ascites for women with HR-deficient disease. While NanoTalazoparib did not effectively treat the disseminated disease at this dose, it may have clinical utility, either in combination with other therapies or as a maintenance therapy. Preclinical data indicates PARP inhibitors potentiate damage when combined with other cytotoxic treatments, however, in the clinic this has resulted in enhanced toxicity, forcing dose reduction and delay. The IP administration of NanoTalazoparib may provide a route to bypass some of the toxicities that have plagued combination treatments.
Supported in part by Rivkin Foundation and CDMRP Ovarian Cancer Research Program
Citation Format: Paige Baldwin, Anders Ohman, Jamie Medina, Daniela Dinulescu, Srinivas Sridhar. NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL [abstract]. In: Proceedings of the 12th Biennial Ovarian Cancer Research Symposium; Sep 13-15, 2018; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2019;25(22 Suppl):Abstract nr NT-087.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Supported in part by Rivkin Foundation and CDMRP Ovarian Cancer Research Program
Citation Format: Paige Baldwin, Anders Ohman, Jamie Medina, Daniela Dinulescu, Srinivas Sridhar. NANOFORMULATION OF TALAZOPARIB SUPPRESSES TUMOR GROWTH AND ASCITES IN A DISSEMINATED CANCER MODEL [abstract]. In: Proceedings of the 12th Biennial Ovarian Cancer Research Symposium; Sep 13-15, 2018; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2019;25(22 Suppl):Abstract nr NT-087.
Kunjachan, Sijumon; Kotb, Shady; Pola, Robert; Pechar, Michal; Kumar, Rajiv; Singh, Bijay; Gremse, Felix; Taleeli, Reza; Trichard, Florian; Motto-Ros, Vincent; others,
Selective priming of tumor Blood Vessels by Radiation therapy enhances nanodrug Delivery Journal Article
In: Scientific reports, vol. 9, no. 1, pp. 1–14, 2019.
Abstract | BibTeX | Tags: Nanomedicine
@article{kunjachan2019selective,
title = {Selective priming of tumor Blood Vessels by Radiation therapy enhances nanodrug Delivery},
author = {Sijumon Kunjachan and Shady Kotb and Robert Pola and Michal Pechar and Rajiv Kumar and Bijay Singh and Felix Gremse and Reza Taleeli and Florian Trichard and Vincent Motto-Ros and others},
year = {2019},
date = {2019-01-01},
journal = {Scientific reports},
volume = {9},
number = {1},
pages = {1--14},
publisher = {Nature Publishing Group},
abstract = {Effective drug delivery is restricted by pathophysiological barriers in solid tumors. In human pancreatic adenocarcinoma, poorly-permeable blood vessels limit the intratumoral permeation and penetration of chemo or nanotherapeutic drugs. New and clinically viable strategies are urgently sought to breach the neoplastic barriers that prevent effective drug delivery. Here, we present an original idea to boost drug delivery by selectively knocking down the tumor vascular barrier in a human pancreatic cancer model. Clinical radiation activates the tumor endothelial-targeted gold nanoparticles to induce a physical vascular damage due to the high photoelectric interactions. Active modulation of these tumor neovessels lead to distinct changes in tumor vascular permeability. Noninvasive MRI and fluorescence studies, using a short-circulating nanocarrier with MR-sensitive gadolinium and a long-circulating nanocarrier with fluorescence-sensitive nearinfrared dye, demonstrate more than two-fold increase in nanodrug delivery, post tumor vascular modulation. Functional changes in altered tumor blood vessels and its downstream parameters, particularly, changes in Ktrans (permeability), Kep (flux rate), and Ve (extracellular interstitial volume), reflect changes that relate to augmented drug delivery. The proposed dual-targeted therapy effectively invades the tumor vascular barrier and improve nanodrug delivery in a human pancreatic tumor model and it may also be applied to other nonresectable, intransigent tumors that barely respond to standard drug therapies.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Bal, Nandita; Cheng, Ming; Kumar, Rajiv; Sridhar, Srinivas; Ebong, Eno E
In Vivo Endothelial Uptake of Nanoparticles: Impact of Disturbed Flow and Degraded Glycocalyx Proceedings Article
In: 2019 AIChE Annual Meeting, AIChE 2019.
Abstract | BibTeX | Tags: Nanomedicine
@inproceedings{bal2019vivo,
title = {In Vivo Endothelial Uptake of Nanoparticles: Impact of Disturbed Flow and Degraded Glycocalyx},
author = {Nandita Bal and Ming Cheng and Rajiv Kumar and Srinivas Sridhar and Eno E Ebong},
year = {2019},
date = {2019-01-01},
booktitle = {2019 AIChE Annual Meeting},
organization = {AIChE},
abstract = {Statement of Purpose: New and more effective therapies for atherosclerosis-based cardiovascular disease should target early stages of the diseases and specific vascular sites where disease occurs.1 The initiation and localization of atherosclerotic plaques has been linked to the dysfunction of the endothelial glycocalyx (GCX). The GCX breaks down, resulting in compromised endothelial barrier function and an increase in vascular permeability.2 This allows lipids and inflammatory cells to penetrate vessel walls, and at the same time can be leveraged for targeted therapeutic delivery. In previous cell culture studies, enzyme-induced GCX degradation resulted in significantly increased endothelial cell uptake of nanoparticle vehicles that are designed for drug delivery, compared to the effects of intact GCX.3,4 The study presented in this abstract was designed to assess if the cell culture findings translate to selective nanoparticle uptake in animals. Specifically, it was determined if increased nanoparticle uptake occurs in animal endothelial cells that have degraded GCX. The first objective of this animal study was to identify vessel regions with intact versus degraded GCX, and confirm that circulating nanoparticles localize selectively at regions of GCX dysfunction. The second objective was to promote endothelial cell surface-specific localization of these nanoparticles by targeting the intracellular adhesion molecule (ICAM) which is upregulated in pre-atherosclerotic disturbed flow conditions.5
Methods: Mouse models of endothelial dysfunction apply partial ligation of the left carotid artery (LCA) to model acute disturbed flow in the mice, which has been reported to induce endothelial dysfunction.6,7 Seven C57BL/6 mice at 4 weeks of age underwent this partial LCA ligation surgery. Polymer- and biotin-coated 10 nm gold nanospheres (GNS) were administered to the mice at day 26 after LCA ligation. In the case of targeted delivery to the endothelial cell surface, an anti-ICAM peptide was conjugated to the GNS surface before it was administered to the mice. The animals were euthanized 2 days after receiving the GNS without or conjugated with anti-ICAM. The LCA and right carotid artery (RCA) were separately cryopreserved and sectioned onto slides. GCX was imaged after incubating LCA and RCA sections with antibody to block GNS, followed by biotinylated wheat germ agglutinin and horseradish peroxidase conjugated to streptavidin. GNS were imaged after incubation with horseradish peroxidase conjugated to streptavidin. We then applied a fluorescent reagent that reacts with horseradish peroxidase. The blood vessels were imaged by fluorescence microscopy and analyzed using ImageJ for GCX coverage and GNS uptake.
Results: The partial LCA ligation model achieved the goal of creating a vessel with disturbed flow. The model also provided a convenient comparison with a healthy RCA experiencing streamlined flow. The vessel walls of the LCA exhibited a more discontinuous GCX layer on the intima as compared to the RCA, decreasing from 76.3 ± 10.2 % in the RCA to 21.2 ± 5.9 % in the LCA. This observable dysfunction correlated to increased nanoparticle uptake, as the LCA took in approximately 2.5-fold more GNS than the RCA did, based on the fluorescence signal detected in the histology images. The studies to improve endothelial cell surface-specific localization of these nanoparticles by targeting ICAM are still ongoing.
Conclusions: A partial LCA ligation was performed to acutely disturb blood flow in a mouse vessel and observe resultant endothelial GCX dysfunction as well as passive targeting of GNS to affected areas. The affected LCA exhibited lack of continuous GCX layer, as well as increased localization of plolymer-coated GNS that are designed to deliver drug therapies. These results indicate that vessel and GCX dysfunction, both precursors of atherosclerosis and cardiovascular disease, can be induced in a mouse model to study targeted drug delivery. Passive nanoparticle uptake differences between the healthy RCA and disturbed LCA indicate a role of GCX infiltration of nanoparticles to the endothelial cells. The ongoing investigation will determine the effectiveness of active targeting with ICAM to localize these nanoparticles to the endothelial cell surface. This strategy of targeting dysfunctional vessels based on the GCX offers a new approach in cardiovascular disease therapy and prevention.
Acknowledgements: We appreciate funding from National Institutes of Health (K01 HL125499) and the Northeastern University Provost’s Tier 1 Grant.
References:
Weinbaum, S.; et. al, Annu Rev Biomed Eng 2007, 9,121-167.
Becker, B. et. al, Br J Clin Pharmacol 2015, 80, 389-402.
Cheng, et. al, Int. J. Nanomed 2016, 21, 3305-3315.
Cheng, et. al, Int. J. Nanomed 2019, 14, 319-333.
Nakashima et. al, Arteriosclerosis, Thrombosis, and Vascular Biology 1998, 18, 842-851.
Kumar, S.; et. al, J. Lab. Invest. 2017, 97, 935-945
Mitra, et. al, Trans. Med. Comm. 2018, 3-10.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {inproceedings}
}
Methods: Mouse models of endothelial dysfunction apply partial ligation of the left carotid artery (LCA) to model acute disturbed flow in the mice, which has been reported to induce endothelial dysfunction.6,7 Seven C57BL/6 mice at 4 weeks of age underwent this partial LCA ligation surgery. Polymer- and biotin-coated 10 nm gold nanospheres (GNS) were administered to the mice at day 26 after LCA ligation. In the case of targeted delivery to the endothelial cell surface, an anti-ICAM peptide was conjugated to the GNS surface before it was administered to the mice. The animals were euthanized 2 days after receiving the GNS without or conjugated with anti-ICAM. The LCA and right carotid artery (RCA) were separately cryopreserved and sectioned onto slides. GCX was imaged after incubating LCA and RCA sections with antibody to block GNS, followed by biotinylated wheat germ agglutinin and horseradish peroxidase conjugated to streptavidin. GNS were imaged after incubation with horseradish peroxidase conjugated to streptavidin. We then applied a fluorescent reagent that reacts with horseradish peroxidase. The blood vessels were imaged by fluorescence microscopy and analyzed using ImageJ for GCX coverage and GNS uptake.
Results: The partial LCA ligation model achieved the goal of creating a vessel with disturbed flow. The model also provided a convenient comparison with a healthy RCA experiencing streamlined flow. The vessel walls of the LCA exhibited a more discontinuous GCX layer on the intima as compared to the RCA, decreasing from 76.3 ± 10.2 % in the RCA to 21.2 ± 5.9 % in the LCA. This observable dysfunction correlated to increased nanoparticle uptake, as the LCA took in approximately 2.5-fold more GNS than the RCA did, based on the fluorescence signal detected in the histology images. The studies to improve endothelial cell surface-specific localization of these nanoparticles by targeting ICAM are still ongoing.
Conclusions: A partial LCA ligation was performed to acutely disturb blood flow in a mouse vessel and observe resultant endothelial GCX dysfunction as well as passive targeting of GNS to affected areas. The affected LCA exhibited lack of continuous GCX layer, as well as increased localization of plolymer-coated GNS that are designed to deliver drug therapies. These results indicate that vessel and GCX dysfunction, both precursors of atherosclerosis and cardiovascular disease, can be induced in a mouse model to study targeted drug delivery. Passive nanoparticle uptake differences between the healthy RCA and disturbed LCA indicate a role of GCX infiltration of nanoparticles to the endothelial cells. The ongoing investigation will determine the effectiveness of active targeting with ICAM to localize these nanoparticles to the endothelial cell surface. This strategy of targeting dysfunctional vessels based on the GCX offers a new approach in cardiovascular disease therapy and prevention.
Acknowledgements: We appreciate funding from National Institutes of Health (K01 HL125499) and the Northeastern University Provost’s Tier 1 Grant.
References:
Weinbaum, S.; et. al, Annu Rev Biomed Eng 2007, 9,121-167.
Becker, B. et. al, Br J Clin Pharmacol 2015, 80, 389-402.
Cheng, et. al, Int. J. Nanomed 2016, 21, 3305-3315.
Cheng, et. al, Int. J. Nanomed 2019, 14, 319-333.
Nakashima et. al, Arteriosclerosis, Thrombosis, and Vascular Biology 1998, 18, 842-851.
Kumar, S.; et. al, J. Lab. Invest. 2017, 97, 935-945
Mitra, et. al, Trans. Med. Comm. 2018, 3-10.
Qiao, Ju; Cai, Xuezhu; Xiao, Qian; Chen, Zhengxi; Kulkarni, Praveen; Ferris, Craig; Kamarthi, Sagar; Sridhar, Srinivas
Data on MRI brain lesion segmentation using K-means and Gaussian Mixture Model-Expectation Maximization Journal Article
In: Data in brief, vol. 27, pp. 104628, 2019.
@article{qiao2019data,
title = {Data on MRI brain lesion segmentation using K-means and Gaussian Mixture Model-Expectation Maximization},
author = {Ju Qiao and Xuezhu Cai and Qian Xiao and Zhengxi Chen and Praveen Kulkarni and Craig Ferris and Sagar Kamarthi and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
journal = {Data in brief},
volume = {27},
pages = {104628},
publisher = {Elsevier},
abstract = {The data in this article provide details about MRI lesion segmentation using K-means and Gaussian Mixture Model-Expectation Maximization (GMM-EM) algorithms. Both K-means and GMM-EM algorithms can segment lesion area from the rest of brain MRI automatically. The performance metrics (accuracy, sensitivity, specificity, false positive rate, misclassification rate) were estimated for the algorithms and there was no significant difference between K-means and GMM-EM. In addition, lesion size does not affect the accuracy and sensitivity for either method.},
keywords = {MRI},
pubstate = {published},
tppubtype = {article}
}
Zhang, Paige Baldwin Di; Leal, Ana S; Carapellucci, Sarah; Sridhar, Srinivas; Liby, Karen T
A nano-liposome formulation of the PARP inhibitor Talazoparib enhances treatment efficacy and modulates immune cell populations in mammary tumors of BRCA-deficient mice Journal Article
In: Theranostics, vol. 9, no. 21, pp. 6224, 2019.
Abstract | BibTeX | Tags: Nanomedicine
@article{di2019nano,
title = {A nano-liposome formulation of the PARP inhibitor Talazoparib enhances treatment efficacy and modulates immune cell populations in mammary tumors of BRCA-deficient mice},
author = {Paige Baldwin Di Zhang and Ana S Leal and Sarah Carapellucci and Srinivas Sridhar and Karen T Liby},
year = {2019},
date = {2019-01-01},
journal = {Theranostics},
volume = {9},
number = {21},
pages = {6224},
publisher = {Ivyspring International Publisher},
abstract = {Two recently approved PARP inhibitors provide an important new therapeutic option for patients with BRCA-mutated metastatic breast cancer. PARP inhibitors significantly prolong progression-free survival in patients, but conventional oral delivery of PARP inhibitors is hindered by limited bioavailability and off-target toxicities, thus compromising the therapeutic benefits and quality of life for patients. Here, we developed a new delivery system, in which the PARP inhibitor Talazoparib is encapsulated in the bilayer of a nano-liposome, to overcome these limitations.
Methods: Nano-Talazoparib (NanoTLZ) was characterized both in vitro and in vivo. The therapeutic efficacy and toxicity of Nano-Talazoparib (NanoTLZ) were evaluated in BRCA-deficient mice. The regulation of NanoTLZ on gene transcription and immunomodulation were further investigated in spontaneous BRCA-deficient tumors.
Results: NanoTLZ significantly (p<0.05) prolonged the overall survival of BRCA-deficient mice compared to all of the other experimental groups, including saline control, empty nanoparticles, and free Talazoparib groups (oral and i.v.). Moreover, NanoTLZ was better tolerated than treatment with free Talazoparib, with no significant weight lost or alopecia as was observed with the free drug. After 5 doses, NanoTLZ altered the expression of over 140 genes and induced DNA damage, cell cycle arrest and inhibition of cell proliferation in the tumor. In addition, NanoTLZ favorably modulated immune cell populations in vivo and significantly (p<0.05) decreased the percentage of myeloid derived suppressor cells in both the tumor and spleen compared to control groups.
Conclusions: Our results demonstrate that delivering nanoformulated Talazoparib not only enhances treatment efficacy but also reduces off-target toxicities in BRCA-deficient mice; the same potential is predicted for patients with BRCA-deficient breast cancer.
Keywords: PARP inhibitor, Talazoparib, Nanoparticle, BRCA-deficient breast cancer, immunomodulation},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Methods: Nano-Talazoparib (NanoTLZ) was characterized both in vitro and in vivo. The therapeutic efficacy and toxicity of Nano-Talazoparib (NanoTLZ) were evaluated in BRCA-deficient mice. The regulation of NanoTLZ on gene transcription and immunomodulation were further investigated in spontaneous BRCA-deficient tumors.
Results: NanoTLZ significantly (p<0.05) prolonged the overall survival of BRCA-deficient mice compared to all of the other experimental groups, including saline control, empty nanoparticles, and free Talazoparib groups (oral and i.v.). Moreover, NanoTLZ was better tolerated than treatment with free Talazoparib, with no significant weight lost or alopecia as was observed with the free drug. After 5 doses, NanoTLZ altered the expression of over 140 genes and induced DNA damage, cell cycle arrest and inhibition of cell proliferation in the tumor. In addition, NanoTLZ favorably modulated immune cell populations in vivo and significantly (p<0.05) decreased the percentage of myeloid derived suppressor cells in both the tumor and spleen compared to control groups.
Conclusions: Our results demonstrate that delivering nanoformulated Talazoparib not only enhances treatment efficacy but also reduces off-target toxicities in BRCA-deficient mice; the same potential is predicted for patients with BRCA-deficient breast cancer.
Keywords: PARP inhibitor, Talazoparib, Nanoparticle, BRCA-deficient breast cancer, immunomodulation
Sridhar, Srinivas; Versek, Craig; Banijamali, Ali; Tran, Anthony; Cardozo, Armando; Lashkari, Kameran; Bex, Peter
Portable VEP Diagnostics for NeuroVisual Disorders Journal Article
In: Investigative Ophthalmology & Visual Science, vol. 60, no. 9, pp. 3591–3591, 2019.
Abstract | BibTeX | Tags: Neurotechnology
@article{sridhar2019portable,
title = {Portable VEP Diagnostics for NeuroVisual Disorders},
author = {Srinivas Sridhar and Craig Versek and Ali Banijamali and Anthony Tran and Armando Cardozo and Kameran Lashkari and Peter Bex},
year = {2019},
date = {2019-01-01},
urldate = {2019-01-01},
journal = {Investigative Ophthalmology & Visual Science},
volume = {60},
number = {9},
pages = {3591--3591},
publisher = {The Association for Research in Vision and Ophthalmology},
abstract = {Purpose: Visual Evoked Potentials (VEPs) provide objective neuro-opthalmologic assessments that avoid patient task performance but utilizes invasive and cumbersome apparatus. We have developed a system that combines a scalp neuroelectric potential and field sensor with a smartphone in a portable wireless display headset called the NeuroDotVR (Figure 1). The system records VEPs and Fields (VEPF) in response to dichoptic stimuli presented on the smartphone display for a range of neuro-oplthalmologic disorders. We evaluate the NeuroDotVR for Dark Adaptation Recovers (DAR), a key biomarker for age-related macular degeneration (AMD).
Methods: DAR was measured simultaneously in both eyes of# patients with AMD and# age-matched controls. Following a 60s photobleach (400 cd/m 2 white cellphone screen), recovery of visual sensitivity was recorded with VEPFs to pattern reversal checkerboard …},
keywords = {Neurotechnology},
pubstate = {published},
tppubtype = {article}
}
Methods: DAR was measured simultaneously in both eyes of# patients with AMD and# age-matched controls. Following a 60s photobleach (400 cd/m 2 white cellphone screen), recovery of visual sensitivity was recorded with VEPFs to pattern reversal checkerboard …
Singh, Bijay; Abdelhalim, Mostafa; Warrington, Stephanee; Sridhar, Srinivas
Development of targeted nanoformulation of talazoparib for combined chemoradiation therapy in lung cancer Miscellaneous
2019.
Abstract | BibTeX | Tags: Nanomedicine
@misc{singh2019development,
title = {Development of targeted nanoformulation of talazoparib for combined chemoradiation therapy in lung cancer},
author = {Bijay Singh and Mostafa Abdelhalim and Stephanee Warrington and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
publisher = {American Association for Cancer Research},
abstract = {Poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) therapy exploits a synthetic lethality strategy in cancers with inherent damage in DNA repair or transcription pathways. Talazoparib is a potent PARPi that is currently indicated for oral inhibitor therapy in several cancer clinical trials. Oral administration of these inhibitors typically results in poor bioavailability and tumor accumulation. In contrast, nanoparticle formulation provides a safe vehicle for parenteral administration of therapeutic drugs with sustainable release reducing systemic toxicity and also protect from surveillance of immune cells, thereby increasing the bioavailability of the drugs in vivo. Moreover, targeting strategy for the nanoparticles will improve the accumulation of drugs in the tumors. Here, we developed a targeted formulation of Talazoparib (NanoTLZ) which was decorated with anti-EGFR antibody as a ligand to ameliorate the cellular uptake of NanoTLZ via EGFR-mediated endocytosis. In vitro tests showed that NanoTLZ is more effective in cell growth inhibition than free Talazoparib. When combined with radiation with different doses from 2-10Gy, NanoTLZ showed a strong radiosensitization effect as evidenced with almost no colonies formation at 6 Gy of radiation dose. The high therapeutic efficacy of combined chemoradiation therapy in Calu 6 cell line can be attributed to the higher accumulation of NanoTLZ in the cancer cells as compared to the free Talazoparib which is also marred with efflux of the drug from the cells. The slow and sustained release of Talazoparib from nanoparticle formulation inside the cells lead enhanced inhibition of DNA repair pathways. These studies provide very encouraging results to evaluate the efficacy of these nanoparticles in lung cancer animal models.
Citation Format: Bijay Singh, Mostafa Abdelhalim, Stephanee Warrington, Srinivas Sridhar. Development of targeted nanoformulation of talazoparib for combined chemoradiation therapy in lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3631.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Citation Format: Bijay Singh, Mostafa Abdelhalim, Stephanee Warrington, Srinivas Sridhar. Development of targeted nanoformulation of talazoparib for combined chemoradiation therapy in lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3631.
Baldwin, Paige; Orriols, Adrienne; Sridhar, Srinivas
Combination nanotherapy using the PARP inhibitor talazoparib and cyclin dependent kinase inhibitor dinaciclib Miscellaneous
2019.
Abstract | BibTeX | Tags: Nanomedicine
@misc{baldwin2019combination,
title = {Combination nanotherapy using the PARP inhibitor talazoparib and cyclin dependent kinase inhibitor dinaciclib},
author = {Paige Baldwin and Adrienne Orriols and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
publisher = {American Association for Cancer Research},
abstract = {Introduction: PARP inhibitors exploit defects in DNA repair pathways to selectively target cancerous cells. As such, Talazoparib (TLZ), a potent PARP inhibitor, offers a way to target the biology of a number of cancers with DNA repair defects until these tumors develop resistance. PARP inhibitors must be used in combination with other inhibitors or chemotherapeutics to reverse resistance and sensitize non-responsive tumors. Dinaciclib, a potent cyclin dependent kinase (CDK) inhibitor, has been shown to sensitize both BRCA wild-type tumors and PARP inhibitor resistant tumors to PARP inhibition through disruption of homologous recombination. In clinical trials, Talazoparib and Dinaciclib have both demonstrated hematologic toxicities, suggesting a combination of these drugs would result in compounded toxicity, leading to dose reduction and an ineffective combination. Nanoparticle delivery systems offer a means to modify the toxicity profiles of these drugs and enhance the therapeutic window, therefore allowing for effective combination treatment.
Methods: Separate nanoformulations of Talazoparib (NanoTLZ) and Dinaciclib (NanoDCB) were optimized, and pharmacokinetics and pharmacodynamics assessed. Nanoformulations were tested alone and in combination in vitro to ensure NanoDCB could sensitize a model with no known DNA repair defects to NanoTLZ. The combination of the two nanoformulations was then assessed for efficacy and toxicity in orthotopic MDA-MB-231 xenografts.
Results: Robust formulations of NanoTLZ and NanoDCB were developed. Each nanoformulation extended the half-life of the drug it encapsulates. A constant low dose of Dinaciclib sensitized MDA-MB-231 cells to Talazoparib, significantly lowering the IC50 value. As a single agent NanoDCB was more effective in vitro than free Dinaciclib. In vivo, the combination of the two nanoformulations was more effective than either single nanoformulation or the combination of the two free drugs. Assessments of hematologic toxicities are underway, but thus far, there were no signs of gross toxicity in the combination therapy group.
Conclusions: The combination of NanoDCB and NanoTLZ has provided an effective method for sensitizing tumors to PARP inhibition that are otherwise nonresponsive to this therapy. The development of two separate nanoformulations has allowed for tailored dosing. These long-circulating nanoformulations have proven more effective than the free drugs in stabilizing tumor growth and were well tolerated. This work was supported by ARMY/W81XWH-16-1-0731.
Citation Format: Paige Baldwin, Adrienne Orriols, Srinivas Sridhar. Combination nanotherapy using the PARP inhibitor talazoparib and cyclin dependent kinase inhibitor dinaciclib [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3642.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Methods: Separate nanoformulations of Talazoparib (NanoTLZ) and Dinaciclib (NanoDCB) were optimized, and pharmacokinetics and pharmacodynamics assessed. Nanoformulations were tested alone and in combination in vitro to ensure NanoDCB could sensitize a model with no known DNA repair defects to NanoTLZ. The combination of the two nanoformulations was then assessed for efficacy and toxicity in orthotopic MDA-MB-231 xenografts.
Results: Robust formulations of NanoTLZ and NanoDCB were developed. Each nanoformulation extended the half-life of the drug it encapsulates. A constant low dose of Dinaciclib sensitized MDA-MB-231 cells to Talazoparib, significantly lowering the IC50 value. As a single agent NanoDCB was more effective in vitro than free Dinaciclib. In vivo, the combination of the two nanoformulations was more effective than either single nanoformulation or the combination of the two free drugs. Assessments of hematologic toxicities are underway, but thus far, there were no signs of gross toxicity in the combination therapy group.
Conclusions: The combination of NanoDCB and NanoTLZ has provided an effective method for sensitizing tumors to PARP inhibition that are otherwise nonresponsive to this therapy. The development of two separate nanoformulations has allowed for tailored dosing. These long-circulating nanoformulations have proven more effective than the free drugs in stabilizing tumor growth and were well tolerated. This work was supported by ARMY/W81XWH-16-1-0731.
Citation Format: Paige Baldwin, Adrienne Orriols, Srinivas Sridhar. Combination nanotherapy using the PARP inhibitor talazoparib and cyclin dependent kinase inhibitor dinaciclib [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3642.
Kelada, Olivia J; Kunjachan, Sijumon; Virani, Needa A; Detappe, Alexandre; Hayashi, Jennifer; Ireland, Thomas; Biancur, Douglas E; Kumar, Rajiv; Sridhar, Srinivas; Makrigiorgos, Mike; others,
Noninvasive imaging of tumor hypoxia during radiation-induced tumor vascular disruption Miscellaneous
2019.
Abstract | BibTeX | Tags: Nanomedicine
@misc{kelada2019noninvasive,
title = {Noninvasive imaging of tumor hypoxia during radiation-induced tumor vascular disruption},
author = {Olivia J Kelada and Sijumon Kunjachan and Needa A Virani and Alexandre Detappe and Jennifer Hayashi and Thomas Ireland and Douglas E Biancur and Rajiv Kumar and Srinivas Sridhar and Mike Makrigiorgos and others},
year = {2019},
date = {2019-01-01},
publisher = {American Association for Cancer Research},
abstract = {Purpose Tumor vascular targeted gold nanoparticles induce tumor vascular disruption when combined with external beam radiation therapy. Although effective in suppressing tumor growth and improving progression-free survival, tumor hypoxia may be a potential challenge in this anti-vascular therapy. Here, we investigate, the dynamic changes in tumor hypoxia pre- and post-radiation therapy using a gold nanoparticle-based tumor vascular disrupting agent.
Materials and methods 4-6 weeks old female nude-FOXn1 mice were subcutaneously inoculated with human A549 cells (~3×106) into the left flank. Functionalized gold nanoparticles (AuNP) were used to target the tumor blood vessels and tumor vascular disruption was induced via radiation. 10 Gy radiation treatment was delivered using a clinical radiation beam (6 MV) and HypoxiSense680 based in vivo fluorescence imaging was performed to visualize changes in tumor hypoxia at 24 h, 48 h and an extended period of 10 days. Tumor hypoxia was confirmed via immunohistochemistry. Mice were divided into four treatment groups: control, AuNP only, IR only, and AuNP+IR. Following treatment, tumor progression and overall survival was measured. Further analysis of nanoparticle biodistribution and toxicity were accessed using TEM Imaging, IC-PMS, and immunohistochemistry.
Results Longitudinal changes in tumor hypoxia were observed in all radiation-based treatment conditions. Combining gold nanoparticle and radiation resulted in an increase in tumor hypoxia at 48 h (p < 0.05) and a return to baseline in 10 days. In contrast, the ‘radiation only’ group showed an increase in tumor hypoxia by a factor of 0.5 at 48 h post-IR compared to baseline while 10 days later the tumor hypoxia remained stable. Quantitative variation in the hypoxia blood factor, CA9 increased 24 h post IR in the AuNP+IR, followed by a decrease in hypoxic by day 10 in accordance with in vivo imaging data. No change was observed with the IR only group. These findings were confirmed with representative pimonidazole staining that showed an increase in tumor hypoxia a few hours after AuNP+IR treatment. The mean relative reduction in tumor size post-treatment was a factor of 5.2 (p < 0.05) in the AuNP+IR group compared to the control and 3.5 compared to the IR only group, nearly 80 days post-treatment. Overall survival showed an average gain of up to 24 days in the AuNP+IR group compared to all other treatments. Almost 100 days post-treatment, 50% survival was observed in the AuNP+IR group compared to 20% in the IR-only groups (p < 0.05).
Conclusions Noninvasive imaging showed that AuNP+IR results in a transient increase and subsequent decline in mean tumor hypoxia, leading to substantial tumor regression and an overall increase in tumor survival. High radiation-induced vascular damage may lead to better tumor reduction and prolonged survival in the human non-small cell lung cancer model.
Citation Format: Olivia J. Kelada, Sijumon Kunjachan, Needa A. Virani, Alexandre Detappe, Jennifer Hayashi, Thomas Ireland, Douglas E. Biancur, Rajiv Kumar, Srinivas Sridhar, Mike Makrigiorgos, Ross I. Berbeco. Noninvasive imaging of tumor hypoxia during radiation-induced tumor vascular disruption [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 85.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Materials and methods 4-6 weeks old female nude-FOXn1 mice were subcutaneously inoculated with human A549 cells (~3×106) into the left flank. Functionalized gold nanoparticles (AuNP) were used to target the tumor blood vessels and tumor vascular disruption was induced via radiation. 10 Gy radiation treatment was delivered using a clinical radiation beam (6 MV) and HypoxiSense680 based in vivo fluorescence imaging was performed to visualize changes in tumor hypoxia at 24 h, 48 h and an extended period of 10 days. Tumor hypoxia was confirmed via immunohistochemistry. Mice were divided into four treatment groups: control, AuNP only, IR only, and AuNP+IR. Following treatment, tumor progression and overall survival was measured. Further analysis of nanoparticle biodistribution and toxicity were accessed using TEM Imaging, IC-PMS, and immunohistochemistry.
Results Longitudinal changes in tumor hypoxia were observed in all radiation-based treatment conditions. Combining gold nanoparticle and radiation resulted in an increase in tumor hypoxia at 48 h (p < 0.05) and a return to baseline in 10 days. In contrast, the ‘radiation only’ group showed an increase in tumor hypoxia by a factor of 0.5 at 48 h post-IR compared to baseline while 10 days later the tumor hypoxia remained stable. Quantitative variation in the hypoxia blood factor, CA9 increased 24 h post IR in the AuNP+IR, followed by a decrease in hypoxic by day 10 in accordance with in vivo imaging data. No change was observed with the IR only group. These findings were confirmed with representative pimonidazole staining that showed an increase in tumor hypoxia a few hours after AuNP+IR treatment. The mean relative reduction in tumor size post-treatment was a factor of 5.2 (p < 0.05) in the AuNP+IR group compared to the control and 3.5 compared to the IR only group, nearly 80 days post-treatment. Overall survival showed an average gain of up to 24 days in the AuNP+IR group compared to all other treatments. Almost 100 days post-treatment, 50% survival was observed in the AuNP+IR group compared to 20% in the IR-only groups (p < 0.05).
Conclusions Noninvasive imaging showed that AuNP+IR results in a transient increase and subsequent decline in mean tumor hypoxia, leading to substantial tumor regression and an overall increase in tumor survival. High radiation-induced vascular damage may lead to better tumor reduction and prolonged survival in the human non-small cell lung cancer model.
Citation Format: Olivia J. Kelada, Sijumon Kunjachan, Needa A. Virani, Alexandre Detappe, Jennifer Hayashi, Thomas Ireland, Douglas E. Biancur, Rajiv Kumar, Srinivas Sridhar, Mike Makrigiorgos, Ross I. Berbeco. Noninvasive imaging of tumor hypoxia during radiation-induced tumor vascular disruption [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 85.
Versek, Craig; Rissmiller, Armen; Tran, Anthony; Taya, Munish; Chowdhury, Kaushik; Bex, Peter; Sridhar, Srinivas
Portable system for neuro-optical diagnostics using virtual reality display Journal Article
In: Military medicine, vol. 184, no. Supplement_1, pp. 584–592, 2019.
Abstract | BibTeX | Tags: Nanomedicine
@article{versek2019portable,
title = {Portable system for neuro-optical diagnostics using virtual reality display},
author = {Craig Versek and Armen Rissmiller and Anthony Tran and Munish Taya and Kaushik Chowdhury and Peter Bex and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
journal = {Military medicine},
volume = {184},
number = {Supplement_1},
pages = {584--592},
publisher = {Oxford University Press},
abstract = {A new product prototype system for diagnosing vision and neurological disorders, called NeuroDotVR, is described herein: this system utilizes a novel wireless NeuroDot brain sensor [Versek C et al. J Neural Eng. 2018 Aug; 15(4):046027] that quantitatively measures visual evoked potentials and fields resulting from custom visual stimuli displayed on a smartphone housed in a virtual reality headset. The NeuroDot brain sensor is unique in that it can be operated both in regular electroencephalography mode, as well as a new electric field encephalography mode, which yields improvements in signal sensitivity and provides new diagnostic information. Steady state and transient visual evoked potentials and fields using reversing checkerboard stimuli are presented with case studies in amblyopia, glaucoma, and dark adaptation. These preliminary data sets highlight potential clinical applications that may be …},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Baldwin, Paige; Ohman, Anders W; Medina, Jamie Edward; McCarthy, Eric Timothy; Dinulescu, Daniela M; Sridhar, Srinivas
Nanoformulation of talazoparib delays tumor progression and ascites formation in a late stage cancer model Journal Article
In: Frontiers in oncology, vol. 9, pp. 353, 2019.
Abstract | BibTeX | Tags: Nanomedicine
@article{baldwin2019nanoformulation,
title = {Nanoformulation of talazoparib delays tumor progression and ascites formation in a late stage cancer model},
author = {Paige Baldwin and Anders W Ohman and Jamie Edward Medina and Eric Timothy McCarthy and Daniela M Dinulescu and Srinivas Sridhar},
year = {2019},
date = {2019-01-01},
journal = {Frontiers in oncology},
volume = {9},
pages = {353},
publisher = {Frontiers},
abstract = {Talazoparib, a potent PARP inhibitor, induces synthetic lethality in BRCA-deficient cancers making it an attractive candidate for ovarian cancer treatment. However, its potency lends itself to side effects associated more closely with traditional chemotherapeutics than other clinically approved PARP inhbitors. We sought to formulate Talazoparib in a nanoparticle delivery system, which allows the drug to be administered intraperitoneally. This was done to specifically target peritoneal dissemination of late stage metastatic ovarian cancer and increase talazoparib’s therapeutic efficacy while minimizing toxic side effects. NanoTalazoparib was developed and characterized with regard to its size, loading, and surface charge. Talazoparib and NanoTalazoparib were tested on a panel of murine and human BRCA cell lines and the dose response was compared to Olaparib’s, the currently used PARP inhibitor. Therapeutic efficacy was tested in vivo in a Brca peritoneal cancer model that mimics late stage disseminated disease. NanoTalazoparib has a diameter of about 70 nm with a neutral surface charge and ~75% encapsulation efficiency, which slowly releases the drug over several hours. Dose response analysis indicated that the murine cell lines with conditional BRCA1/2, PTEN, and TP53 deletions had the lowest IC50s. NanoTalazoparib administered on a schedule of three doses weekly slowed disease progression and resulted in significantly less mice with ascites at the end point compared to controls. These results indicate that the slow release nanoformulation, NanoTalazoparib, effectively delivers PARP inhibitor therapy to the peritoneal cavity for …},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Cheng, Ming J; Bal, Nandita N; Prabakaran, Priya; Kumar, Rajiv; Webster, Thomas J; Sridhar, Srinivas; Ebong, Eno E
Ultrasmall gold nanorods: synthesis and glycocalyx-related permeability in human endothelial cells Journal Article
In: International journal of nanomedicine, vol. 14, pp. 319, 2019.
Abstract | BibTeX | Tags: Nanomedicine
@article{cheng2019ultrasmall,
title = {Ultrasmall gold nanorods: synthesis and glycocalyx-related permeability in human endothelial cells},
author = {Ming J Cheng and Nandita N Bal and Priya Prabakaran and Rajiv Kumar and Thomas J Webster and Srinivas Sridhar and Eno E Ebong},
year = {2019},
date = {2019-01-01},
journal = {International journal of nanomedicine},
volume = {14},
pages = {319},
publisher = {Dove Press},
abstract = {Clinical data show shed endothelial glycocalyx (GCX) components in blood samples of atherosclerotic patients, linking atherosclerotic development to endothelial GCX integrity. Healthy GCX has pores no> 7 nm, and shed GCX has even larger pores. Therefore, we suggest targeting and treating atherosclerosis-prone blood vessels by using nanoscale vehicles to deliver drugs via the nanoscale GCX as it becomes dysfunctional.
Materials and methods
To test our idea, we investigated permeability of nanoparticles in endothelium, as related to a GCX expression. The present work involves nanorods, which are expected to interact with larger portions of endothelial cell (EC) membranes, due to surface area of the nanorod long axis. Conventional nanorod diameters are orders of magnitude larger than the GCX pore size, so we adapted conventional synthesis methods to fabricate ultrasmall gold nanorods …},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Materials and methods
To test our idea, we investigated permeability of nanoparticles in endothelium, as related to a GCX expression. The present work involves nanorods, which are expected to interact with larger portions of endothelial cell (EC) membranes, due to surface area of the nanorod long axis. Conventional nanorod diameters are orders of magnitude larger than the GCX pore size, so we adapted conventional synthesis methods to fabricate ultrasmall gold nanorods …
2018
Baldwin, Paige; Ohman, Anders W; Tangutoori, Shifalika; Dinulescu, Daniela M; Sridhar, Srinivas
Intraperitoneal delivery of NanoOlaparib for disseminated late-stage cancer treatment Journal Article
In: International journal of nanomedicine, vol. 13, pp. 8063, 2018.
Abstract | BibTeX | Tags: Nanomedicine
@article{baldwin2018intraperitoneal,
title = {Intraperitoneal delivery of NanoOlaparib for disseminated late-stage cancer treatment},
author = {Paige Baldwin and Anders W Ohman and Shifalika Tangutoori and Daniela M Dinulescu and Srinivas Sridhar},
year = {2018},
date = {2018-01-01},
journal = {International journal of nanomedicine},
volume = {13},
pages = {8063},
publisher = {Dove Press},
abstract = {Background
PARP inhibitors, such as Olaparib, have advanced the treatment of ovarian cancer by providing patients with an effective and molecularly-targeted maintenance therapy. However, all orally-administered drugs, including Olaparib, must undergo first-pass metabolism. In contrast, a nanoparticle delivery system has the advantage of administering Olaparib directly into the peritoneal cavity for local treatment. Consequently, we sought to optimize the sustained-release formulation NanoOlaparib, previously deemed effective as an intravenous solid tumor treatment, for the local treatment of disseminated disease via intraperitoneal (i.p.) therapy.
Methods
The tumor cell line 404, which was derived from a Brca2−/−, Tp53−/−, Pten−/− genetically engineered mouse model, exhibited high sensitivity to Olaparib in vitro. It was chosen for use in developing an i.p. spread xenograft for testing nanotherapy efficacy in vivo. NanoOlaparib as a monotherapy or in combination with cisplatin was compared to oral Olaparib alone or in combination using two different dose schedules. A pilot biodistribution study was performed to determine drug accumulation in various organs following i.p. administration.
Results
Daily administration of NanoOlaparib reduced tumor growth and decreased the variability of the treatment response observed with daily oral Olaparib administration. However, systemic toxicity was observed in both the NanoOlaparib and vehicle (empty nanoparticle) treated groups. Scaling back the administration to twice weekly was well tolerated up to 100 mg/kg but reduced the effect on tumor growth. Biodistribution profiles indicated that NanoOlaparib began accumulating in tissues within an hour of administration and persisted for at least 72 hours after a single dose, exiting the peritoneal cavity faster than expected.
Conclusion
NanoOlaparib must be modified for use against disseminated disease. Future avenues to develop NanoOlaparib as an i.p. therapy include a modified surface-coating to retain it in the peritoneal cavity and prevent entry into systemic circulation, in addition to targeting moieties for localization in tumor cells.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
PARP inhibitors, such as Olaparib, have advanced the treatment of ovarian cancer by providing patients with an effective and molecularly-targeted maintenance therapy. However, all orally-administered drugs, including Olaparib, must undergo first-pass metabolism. In contrast, a nanoparticle delivery system has the advantage of administering Olaparib directly into the peritoneal cavity for local treatment. Consequently, we sought to optimize the sustained-release formulation NanoOlaparib, previously deemed effective as an intravenous solid tumor treatment, for the local treatment of disseminated disease via intraperitoneal (i.p.) therapy.
Methods
The tumor cell line 404, which was derived from a Brca2−/−, Tp53−/−, Pten−/− genetically engineered mouse model, exhibited high sensitivity to Olaparib in vitro. It was chosen for use in developing an i.p. spread xenograft for testing nanotherapy efficacy in vivo. NanoOlaparib as a monotherapy or in combination with cisplatin was compared to oral Olaparib alone or in combination using two different dose schedules. A pilot biodistribution study was performed to determine drug accumulation in various organs following i.p. administration.
Results
Daily administration of NanoOlaparib reduced tumor growth and decreased the variability of the treatment response observed with daily oral Olaparib administration. However, systemic toxicity was observed in both the NanoOlaparib and vehicle (empty nanoparticle) treated groups. Scaling back the administration to twice weekly was well tolerated up to 100 mg/kg but reduced the effect on tumor growth. Biodistribution profiles indicated that NanoOlaparib began accumulating in tissues within an hour of administration and persisted for at least 72 hours after a single dose, exiting the peritoneal cavity faster than expected.
Conclusion
NanoOlaparib must be modified for use against disseminated disease. Future avenues to develop NanoOlaparib as an i.p. therapy include a modified surface-coating to retain it in the peritoneal cavity and prevent entry into systemic circulation, in addition to targeting moieties for localization in tumor cells.
Mitra, Ronodeep; Qiao, Ju; Madhavan, Sudharsan; O’Neil, Gerard L; Ritchie, Bailey; Kulkarni, Praveen; Sridhar, Srinivas; van de Ven, Anne L; Kemmerling, Erica Cherry M; Ferris, Craig; others,
The comparative effects of high fat diet or disturbed blood flow on glycocalyx integrity and vascular inflammation Journal Article
In: Translational medicine communications, vol. 3, no. 1, pp. 1–15, 2018.
Abstract | BibTeX | Tags: Nanomedicine
@article{mitra2018comparative,
title = {The comparative effects of high fat diet or disturbed blood flow on glycocalyx integrity and vascular inflammation},
author = {Ronodeep Mitra and Ju Qiao and Sudharsan Madhavan and Gerard L O’Neil and Bailey Ritchie and Praveen Kulkarni and Srinivas Sridhar and Anne L van de Ven and Erica Cherry M Kemmerling and Craig Ferris and others},
year = {2018},
date = {2018-01-01},
journal = {Translational medicine communications},
volume = {3},
number = {1},
pages = {1--15},
publisher = {BioMed Central},
abstract = {Background and aims
Endothelial surface glycocalyx shedding plays a role in endothelial dysfunction and increases vessel wall permeability, which can lead to inflammation and atherogenesis. We sought to elucidate whether a high fat diet (HFD) or disturbed blood flow conditions, both of which are atherogenic risk factors, would contribute more detrimentally to pre-atherosclerotic loss of endothelial glycocalyx integrity and vascular inflammation.
Methods
Six to seven week-old C57BL/6-background apolipoprotein-E-knockout (ApoE-KO) male mice were either fed a chow diet, fed a modified Western HFD, and/or subjected to a partial left carotid artery (LCA) ligation procedure to induce disturbed blood flow patterns in the LCA. Mice were sacrificed after 1 week of experimental conditions. Both LCA and right carotid artery (RCA) vessels were dissected and preserved to compare glycocalyx coverage and thickness as well as macrophage accumulation in carotid arterial walls amongst and between cohorts.
Results
Glycocalyx coverage of the endothelium was significantly reduced in the LCAs of HFD fed mice when compared to the control. More significant reduction in glycocalyx coverage occurred in the LCAs of mice exposed to disturbed flow by partial LCA ligation when compared to the control. No differences were found in glycocalyx coverage of RCAs from all cohorts. Regarding inflammation, no difference in macrophage accumulation in carotid arterial walls was observed when comparing the LCAs and RCAs of control and HFD fed mice. However, macrophage infiltration in vessel walls showed a 20-fold increase in the LCAs exposed to disturbed flow following ligation, when compared to control LCAs, while no such statistical difference was observed between the RCAs of the group.
Conclusions
In our mouse model, endothelial glycocalyx integrity was compromised more by disturbed blood flow patterns than by exposure of the carotid vessel to HFD conditions. The pathophysiological implications include endothelial dysfunction, which correlates to macrophage infiltration in vessel walls and promotes atherogenesis.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Endothelial surface glycocalyx shedding plays a role in endothelial dysfunction and increases vessel wall permeability, which can lead to inflammation and atherogenesis. We sought to elucidate whether a high fat diet (HFD) or disturbed blood flow conditions, both of which are atherogenic risk factors, would contribute more detrimentally to pre-atherosclerotic loss of endothelial glycocalyx integrity and vascular inflammation.
Methods
Six to seven week-old C57BL/6-background apolipoprotein-E-knockout (ApoE-KO) male mice were either fed a chow diet, fed a modified Western HFD, and/or subjected to a partial left carotid artery (LCA) ligation procedure to induce disturbed blood flow patterns in the LCA. Mice were sacrificed after 1 week of experimental conditions. Both LCA and right carotid artery (RCA) vessels were dissected and preserved to compare glycocalyx coverage and thickness as well as macrophage accumulation in carotid arterial walls amongst and between cohorts.
Results
Glycocalyx coverage of the endothelium was significantly reduced in the LCAs of HFD fed mice when compared to the control. More significant reduction in glycocalyx coverage occurred in the LCAs of mice exposed to disturbed flow by partial LCA ligation when compared to the control. No differences were found in glycocalyx coverage of RCAs from all cohorts. Regarding inflammation, no difference in macrophage accumulation in carotid arterial walls was observed when comparing the LCAs and RCAs of control and HFD fed mice. However, macrophage infiltration in vessel walls showed a 20-fold increase in the LCAs exposed to disturbed flow following ligation, when compared to control LCAs, while no such statistical difference was observed between the RCAs of the group.
Conclusions
In our mouse model, endothelial glycocalyx integrity was compromised more by disturbed blood flow patterns than by exposure of the carotid vessel to HFD conditions. The pathophysiological implications include endothelial dysfunction, which correlates to macrophage infiltration in vessel walls and promotes atherogenesis.
Zhang, Di; Baldwin, Paige; Sridhar, Srinivas; Liby, Karen
Nanoformulated Talazoparib enhances the efficacy and reduces the toxicity of this PARP inhibitor in a preclinical model of BRCA-deficient breast cancer Journal Article
In: The FASEB Journal, vol. 32, no. 1_supplement, pp. 565–10, 2018.
Abstract | BibTeX | Tags: Nanomedicine
@article{zhang2018nanoformulated,
title = {Nanoformulated Talazoparib enhances the efficacy and reduces the toxicity of this PARP inhibitor in a preclinical model of BRCA-deficient breast cancer},
author = {Di Zhang and Paige Baldwin and Srinivas Sridhar and Karen Liby},
year = {2018},
date = {2018-01-01},
journal = {The FASEB Journal},
volume = {32},
number = {1_supplement},
pages = {565--10},
publisher = {The Federation of American Societies for Experimental Biology},
abstract = {BRCA mutations are the leading cause of hereditary breast cancer. PARP inhibitors have shown promising activities in clinical trials for breast cancer by inducing synthetic lethality, particularly in patients with BRCA deficiency. Moreover, the utility of PARP inhibitors could potentially extend beyond BRCA mutations by targeting defects in homologous recombination, and thus impact up to 75% of triple negative breast cancer patients and 33% of breast cancer patients overall. However, conventional oral delivery of PARP inhibitors is hindered by limited bioavailability and significant off-target toxicities, thus compromising the therapeutic benefits and quality of life in patients. Therefore, we developed a new nanoparticle delivery system for PARP inhibitors and hypothesize that nanoformulated Talazoparib can enhance efficacy by increasing drug concentrations in the tumor and reduce off-target toxicities. The nanoparticle formulation includes polymer brushes to prolong the circulation time, enabling tumor accumulation through the enhanced permeability and retention effect. The therapeutic efficacy of Nano-Talazoparib (Nano-TLZ) was assessed after i.v. injection in Brca1Co/Co;MMTV-Cre; p53+/− mice with established tumors and compared to vehicle control (saline, i.v.), empty nanoparticles (i.v.), free Talazoparib (i.v.), and free Talazoparib (gavage). Treatment was started when the tumor was 4 mm in diameter and ended when the tumor size reached defined IACUC endpoints. Nano-TLZ significantly (p<0.05) prolonged the life span of BRCA deficient mice from 11.6±2.6 days with saline injection and 18.3±3.6 days with empty nanoparticles injection to 82.2±10.5 days with i.v. Nano-TLZ. Nano-TLZ induced growth arrest in 100% of the tumors and regression (> 50% reduction in tumor volume) in 80% of the tumors. Established tumors regressed more rapidly, and therefore progression free survival significantly improved in the nanoformulated Talazoparib group (p<0.05). Moreover, Nano-TLZ is better tolerated than free Talazoparib with no significant weight lost or alopecia. In a biomarker study following 10 days of treatment, Nano-TLZ increased double strand DNA breaks (γ-H2AX) and decreased proliferation (PCNA) compared to the saline controls. Interestingly, Nano-TLZ significantly (p<0.05) decreased myeloid derived suppressor cells in both the tumor (41.6±4.7% to 11.2±2.9%) and spleen (10.0±3.2% to 3.7±0.6%) compared to the saline control. Nano-TLZ also significantly (p<0.05) decreased the percentage of tumor-associated macrophages in the mammary gland from 7.4±2.0% in the saline control group to 2.5±0.2% in the Nano-TLZ group. The changes in immune populations suggest potential immunomodulatory effects of Talazoparib. These results demonstrate that the delivery of Talazoparib as a nanoformulation induces superior treatment outcomes with reduced off-target toxicity in BRCA deficient mice, and provides a novel delivery strategy for PARP inhibitors in patients.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
Kunjachan, S; Kotb, S; Kumar, R; Pola, R; Pechar, M; Gremse, F; Taleei, R; Trichard, F; Motto-Ros, V; Sancey, L; others,
BEST IN PHYSICS (THERAPY): Enhanced Drug Delivery by Nanoparticle and Radiation-Mediated Tumor Vascular Modulation Proceedings Article
In: MEDICAL PHYSICS, pp. E535–E535, WILEY 111 RIVER ST, HOBOKEN 07030-5774, NJ USA 2018.
BibTeX | Tags: Nanomedicine
@inproceedings{kunjachan2018best,
title = {BEST IN PHYSICS (THERAPY): Enhanced Drug Delivery by Nanoparticle and Radiation-Mediated Tumor Vascular Modulation},
author = {S Kunjachan and S Kotb and R Kumar and R Pola and M Pechar and F Gremse and R Taleei and F Trichard and V Motto-Ros and L Sancey and others},
year = {2018},
date = {2018-01-01},
booktitle = {MEDICAL PHYSICS},
volume = {45},
number = {6},
pages = {E535--E535},
organization = {WILEY 111 RIVER ST, HOBOKEN 07030-5774, NJ USA},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {inproceedings}
}
Baldwin, Paige; Kumar, Rajiv; Sridhar, Srinivas
Targeted nanotherapy using the PARP inhibitor talazoparib for breast cancer treatment Miscellaneous
2018.
Abstract | BibTeX | Tags: Nanomedicine
@misc{baldwin2018targeted,
title = {Targeted nanotherapy using the PARP inhibitor talazoparib for breast cancer treatment},
author = {Paige Baldwin and Rajiv Kumar and Srinivas Sridhar},
year = {2018},
date = {2018-01-01},
publisher = {American Association for Cancer Research},
abstract = {Introduction: PARP inhibitors exploit defects in DNA repair pathways to selectively target cancerous cells. These drugs are currently delivered orally, but Talazoparib, the most potent PARP inhibitor, exhibits greater toxicity than the others. Systemic administration of nanoparticles bypasses the first-pass metabolism of oral drugs and nanoparticles preferentially accumulate in tumors due to the leaky tumor vasculature. Additionally, nanoparticles can be actively targeted to tumors by conjugating different moieties such as antibodies that recognize overexpressed markers on the tumor cells. NanoTalazoparib (NanoTLZ) has been previously formulated and extensively characterized in breast, ovarian, and lung cancer models. Here we describe further characterization of NanoTLZ and the development of a next generation fluorescently labeled EPCAM targeted formulation of NanoTLZ for the treatment of triple negative breast cancer (TNBC).
Methods: Animals with orthotopic TNBC xenografts were injected with NanoTLZ and at various time points tumor biopsies were taken to assay tumor PAR levels. Blood was collected at various time points and plasma separated for drug extraction and quantification. Pharmacokinetic modeling is underway. Fluorescently labeled NanoTLZ was developed with the addition of Cy5, followed by further improvement via conjugation of anti-EPCAM antibodies. Targeting capability was assessed via laser scanning confocal microscopy after treatment of the TNBC cell line MDA-MB-231 with either non-targeted or targeted NanoTLZ. Therapeutic efficacy studies with targeted NanoTLZ alone and in combination with radiation are underway in an MDA-MB-231 xenograft model.
Results: Pharmacodynamics indicated PAR suppression in tumors within 30 minutes of NanoTLZ treatment. Tumor PAR levels began to increase 24 hours after a single dose but remained significantly lower than control levels up to 72 hours (P<0.005). Characterization of NanoTLZ after the addition of Cy5 and after anti-EPCAM targeting indicated that the formulation remained under 100 nm in diameter with a charge of ~10 mV and equivalent drug loading and release. Cy 5 labeling allowed for visualization of nanoparticle uptake and intracellular fluorescence was 70% greater in cells when treated with EPCAM targeted NanoTLZ compared to non-targeted.
Conclusions: The sustained release of Talazoparib from the nanoformulation decreases tumor PAR levels for up to 72 hours after a single dose, allowing for less frequent administration than the current daily regime used for oral Talazoparib. The targeted formulation of NanoTLZ shares the same physicochemical properties as the untargeted formulation, but is taken up much faster in vitro, suggesting it will allow for greater accumulation at the tumor site making it more effective than the previously tested untargeted NanoTLZ. Supported by ARMY/W81XWH-16-1-0731 and Rivkin Foundation.
Citation Format: Paige Baldwin, Rajiv Kumar, Srinivas Sridhar. Targeted nanotherapy using the PARP inhibitor talazoparib for breast cancer treatment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3715.
©2018 American Association for Cancer Research.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Methods: Animals with orthotopic TNBC xenografts were injected with NanoTLZ and at various time points tumor biopsies were taken to assay tumor PAR levels. Blood was collected at various time points and plasma separated for drug extraction and quantification. Pharmacokinetic modeling is underway. Fluorescently labeled NanoTLZ was developed with the addition of Cy5, followed by further improvement via conjugation of anti-EPCAM antibodies. Targeting capability was assessed via laser scanning confocal microscopy after treatment of the TNBC cell line MDA-MB-231 with either non-targeted or targeted NanoTLZ. Therapeutic efficacy studies with targeted NanoTLZ alone and in combination with radiation are underway in an MDA-MB-231 xenograft model.
Results: Pharmacodynamics indicated PAR suppression in tumors within 30 minutes of NanoTLZ treatment. Tumor PAR levels began to increase 24 hours after a single dose but remained significantly lower than control levels up to 72 hours (P<0.005). Characterization of NanoTLZ after the addition of Cy5 and after anti-EPCAM targeting indicated that the formulation remained under 100 nm in diameter with a charge of ~10 mV and equivalent drug loading and release. Cy 5 labeling allowed for visualization of nanoparticle uptake and intracellular fluorescence was 70% greater in cells when treated with EPCAM targeted NanoTLZ compared to non-targeted.
Conclusions: The sustained release of Talazoparib from the nanoformulation decreases tumor PAR levels for up to 72 hours after a single dose, allowing for less frequent administration than the current daily regime used for oral Talazoparib. The targeted formulation of NanoTLZ shares the same physicochemical properties as the untargeted formulation, but is taken up much faster in vitro, suggesting it will allow for greater accumulation at the tumor site making it more effective than the previously tested untargeted NanoTLZ. Supported by ARMY/W81XWH-16-1-0731 and Rivkin Foundation.
Citation Format: Paige Baldwin, Rajiv Kumar, Srinivas Sridhar. Targeted nanotherapy using the PARP inhibitor talazoparib for breast cancer treatment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3715.
©2018 American Association for Cancer Research.
Vazquez-Pagan, Ana G; Baldwin, Paige; Ashtaputre, Ravina M; Kunjachan, Sijumon; Sridhar, Srinivas; Kumar, Rajiv; Berbeco, Ross
Nanoparticle-mediated concomitant radiation dose amplification and PARP inhibition in lung cancer Miscellaneous
2018.
Abstract | BibTeX | Tags: Nanomedicine
@misc{vazquez2018nanoparticle,
title = {Nanoparticle-mediated concomitant radiation dose amplification and PARP inhibition in lung cancer},
author = {Ana G Vazquez-Pagan and Paige Baldwin and Ravina M Ashtaputre and Sijumon Kunjachan and Srinivas Sridhar and Rajiv Kumar and Ross Berbeco},
year = {2018},
date = {2018-01-01},
publisher = {American Association for Cancer Research},
abstract = {Introduction: More than 50% of cancer patients receive radiation therapy at some point during their care. Gold nanoparticles (GNPs) can amplify the radiation dose by facilitating the ejection of low-energy photoelectrons, resulting in increased DNA damage. One of the main challenges of radiation therapy in cancer is to sustain this damage for longer durations. DNA single-strand breaks (SSBs) are repaired by base excision repair, which utilizes Poly(ADP-ribose) polymerase (PARP). PARP inhibition during radiotherapy provides an attractive alternative in maximizing treatment outcomes. Here, we explore a strategy to combine the radiosensitizing effect of GNPs with the DNA-repair inhibiting ability of NanoTalazoparib (nTLZ), a liposomal formulation of the PARP inhibitor, talazoparib (TLZ).
Methods: GNPs were synthesized by reducing AuCl4 using Tetrakis(hydroxymethyl)phosphonium chloride and further PEGylating using heterobifunctional PEGs. A liposomal formulation of TLZ was synthesized using the Nanoassemblr, a microfluidics-based device. Physicochemical characterization of GNPs and nTLZ was carried out using TEM, DLS and release kinetics studies. In vitro studies were carried out to assess the toxicity of the GNPs using MTT assay in non-small cell lung cancer (NSCLC) cell line Calu-6. The therapeutic efficacy of combination treatment using GNPs, nTLZ and radiation was done using clonogenic survival assays. Clonogenic assay was carried out using Calu 6 cells, which were sequentially treated with GNPs (1mg/mL), TLZ (0.5 uM) and nTLZ (0.5 uM) with and without radiation. The radiation doses varied from 0-10Gy for each set of treatments.
Results: PEGylated GNP's showed a hydrodynamic diameter of ~10-12 nm with a spherical morphology whereas nTLZ size was 70 nm encapsulating TLZ at a concentration of ~200 ug/ml. MTT assay showed no toxicity for PEGylated GNPs treated upto a concentration of 3.0 mg/mL. Cells treated with either GNPs, TLZ or nTLZ did not show significant reduction in colony formation, but were reduced with increasing doses of radiation. The survival plots showed a highly additive antiproliferative effect for the GNP + nTLZ combination at all radiation doses, while the free TLZ + GNPs combination was not as effective at inhibiting colony formation. In vivo experiments assessing the combination therapy in a subcutaneous xenograft mice model using Calu 6 are currently under way.
Conclusions: The preliminary in vitro results indicate that the combination of radiosensitizing GNPs with a potent DNA repair enzyme inhibitor, TLZ, has an immense potential as a complimentary combination therapy in conjunction with radiation therapy in treatment of lung cancer.
This work is supported by the CaNCURE program (grant #1CA174650-02), American Lung Association, Dana-Farber Cancer Institute and Brigham and Women's Hospital.
Citation Format: Ana G. Vazquez-Pagan, Paige Baldwin, Ravina M. Ashtaputre, Sijumon Kunjachan, Srinivas Sridhar, Rajiv Kumar, Ross Berbeco. Nanoparticle-mediated concomitant radiation dose amplification and PARP inhibition in lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3710.
©2018 American Association for Cancer Research.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Methods: GNPs were synthesized by reducing AuCl4 using Tetrakis(hydroxymethyl)phosphonium chloride and further PEGylating using heterobifunctional PEGs. A liposomal formulation of TLZ was synthesized using the Nanoassemblr, a microfluidics-based device. Physicochemical characterization of GNPs and nTLZ was carried out using TEM, DLS and release kinetics studies. In vitro studies were carried out to assess the toxicity of the GNPs using MTT assay in non-small cell lung cancer (NSCLC) cell line Calu-6. The therapeutic efficacy of combination treatment using GNPs, nTLZ and radiation was done using clonogenic survival assays. Clonogenic assay was carried out using Calu 6 cells, which were sequentially treated with GNPs (1mg/mL), TLZ (0.5 uM) and nTLZ (0.5 uM) with and without radiation. The radiation doses varied from 0-10Gy for each set of treatments.
Results: PEGylated GNP's showed a hydrodynamic diameter of ~10-12 nm with a spherical morphology whereas nTLZ size was 70 nm encapsulating TLZ at a concentration of ~200 ug/ml. MTT assay showed no toxicity for PEGylated GNPs treated upto a concentration of 3.0 mg/mL. Cells treated with either GNPs, TLZ or nTLZ did not show significant reduction in colony formation, but were reduced with increasing doses of radiation. The survival plots showed a highly additive antiproliferative effect for the GNP + nTLZ combination at all radiation doses, while the free TLZ + GNPs combination was not as effective at inhibiting colony formation. In vivo experiments assessing the combination therapy in a subcutaneous xenograft mice model using Calu 6 are currently under way.
Conclusions: The preliminary in vitro results indicate that the combination of radiosensitizing GNPs with a potent DNA repair enzyme inhibitor, TLZ, has an immense potential as a complimentary combination therapy in conjunction with radiation therapy in treatment of lung cancer.
This work is supported by the CaNCURE program (grant #1CA174650-02), American Lung Association, Dana-Farber Cancer Institute and Brigham and Women's Hospital.
Citation Format: Ana G. Vazquez-Pagan, Paige Baldwin, Ravina M. Ashtaputre, Sijumon Kunjachan, Srinivas Sridhar, Rajiv Kumar, Ross Berbeco. Nanoparticle-mediated concomitant radiation dose amplification and PARP inhibition in lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3710.
©2018 American Association for Cancer Research.
Zhang, Di; Baldwin, Paige; Sridhar, Srinivas; Liby, Karen
Developing a nanoformulation of the PARP inhibitor talazoparib as a novel delivery for treatment of BRCA-deficient breast cancer Miscellaneous
2018.
Abstract | BibTeX | Tags: Nanomedicine
@misc{zhang2018developing,
title = {Developing a nanoformulation of the PARP inhibitor talazoparib as a novel delivery for treatment of BRCA-deficient breast cancer},
author = {Di Zhang and Paige Baldwin and Srinivas Sridhar and Karen Liby},
year = {2018},
date = {2018-01-01},
urldate = {2018-01-01},
publisher = {American Association for Cancer Research},
abstract = {BRCA mutations are the leading cause of hereditary breast cancer. PARP inhibitors have shown promising activities in clinical trials for breast cancer by inducing synthetic lethality, particularly in patients with BRCA deficiency. Moreover, the utility of PARP inhibitors could potentially extend beyond BRCA mutations by targeting defects in homologous recombination, and thus impact up to 33% of breast cancer patients overall. However, conventional oral delivery of PARP inhibitors is hindered by limited bioavailability and significant off-target toxicities, thus compromising the therapeutic benefits and quality of life in patients. Therefore, we developed a new nanoparticle delivery system for PARP inhibitors and hypothesize that nanoformulated Talazoparib can enhance efficacy by increasing drug concentrations in the tumor and reduce off-target toxicities. The nanoparticle formulation includes polymer brushes to prolong the circulation time, enabling tumor accumulation through the enhanced permeability and retention effect. The therapeutic efficacy of Nano-Talazoparib (Nano-TLZ) was assessed after i.v. injection in Brca1Co/Co;MMTV-Cre;p53+/- mice with established tumors and compared to vehicle control (saline, i.v.), empty nanoparticles (i.v.), free Talazoparib (i.v.), and free Talazoparib (gavage). Treatment was started when the tumor was 4 mm in diameter and ended when the tumor size reached defined IACUC endpoints. Nano-TLZ significantly (p<0.05) prolonged the life span of BRCA deficient mice from 11.6±2.6 days with saline injection to 82.2±10.5 days with i.v. Nano-TLZ. Nano-TLZ induced growth arrest in 100% of the tumors and regression (> 50% reduction in tumor volume) in 80% of the tumors. Established tumors regressed more rapidly, and therefore progression free survival significantly improved in the nanoformulated Talazoparib group (p<0.05). Moreover, Nano-TLZ is better tolerated than free Talazoparib with no significant weight lost. In a biomarker study following 10 days of treatment, Nano-TLZ increased double strand DNA breaks (γ-H2AX) and decreased proliferation (PCNA) compared to controls. Interestingly, Nano-TLZ significantly (p<0.05) decreased myeloid derived suppressor cells in both the tumor (41.6±4.7% to 11.2±2.9%) and spleen (10.0±3.2% to 3.7±0.6%) compared to the saline control. Nano-TLZ also significantly (p<0.05) decreased the percentage of tumor-associated macrophages in the mammary gland from 7.4±2.0% in the saline control group to 2.5±0.2% in the Nano-TLZ group. The changes in immune populations suggest potential immunomodulatory effects of Talazoparib. These results demonstrate that the delivery of Talazoparib as a nanoformulation induces superior treatment outcomes with reduced off-target toxicity in BRCA deficient mice, and provides a novel delivery strategy for PARP inhibitors in patients.
Citation Format: Di Zhang, Paige Baldwin, Srinivas Sridhar, Karen Liby. Developing a nanoformulation of the PARP inhibitor talazoparib as a novel delivery for treatment of BRCA-deficient breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3874.
©2018 American Association for Cancer Research.},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {misc}
}
Citation Format: Di Zhang, Paige Baldwin, Srinivas Sridhar, Karen Liby. Developing a nanoformulation of the PARP inhibitor talazoparib as a novel delivery for treatment of BRCA-deficient breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3874.
©2018 American Association for Cancer Research.
Guthier, CV; D'Amico, AV; King, MT; Nguyen, PL; Orio, PF; Sridhar, S; Makrigiorgos, GM; Cormack, RA
Determining optimal eluter design by modeling physical dose enhancement in brachytherapy Journal Article
In: Medical physics, vol. 45, no. 8, pp. 3916–3925, 2018.
Abstract | BibTeX | Tags: Nanomedicine
@article{guthier2018determining,
title = {Determining optimal eluter design by modeling physical dose enhancement in brachytherapy},
author = {CV Guthier and AV D'Amico and MT King and PL Nguyen and PF Orio and S Sridhar and GM Makrigiorgos and RA Cormack},
year = {2018},
date = {2018-01-01},
journal = {Medical physics},
volume = {45},
number = {8},
pages = {3916--3925},
abstract = {Purpose
In situ drug release concurrent with radiation therapy has been proposed to enhance the therapeutic ratio of permanent prostate brachytherapy. Both brachytherapy sources and brachytherapy spacers have been proposed as potential eluters to release compounds, such as nanoparticles or chemotherapeutic agents. The relative effectiveness of the approaches has not been compared yet. This work models the physical dose enhancement of implantable eluters in conjunction with brachytherapy to determine which delivery mechanism provides greatest opportunity to enhance the therapeutic ratio.
Materials and methods
The combined effect of implanted eluters and radioactive sources were modeled in a manner that allowed the comparison of the relative effectiveness of different types of implantable eluters over a range of parameters. Prostate geometry, source, and spacer positions were extracted from treatment plans used for 125I permanent prostate implants. Compound concentrations were calculated using steady‐state solution to the diffusion equation including an elimination term characterized by the diffusion‐elimination modulus (ϕb). Does enhancement was assumed to be dependent on compound concentration up to a saturation concentration (csat). Equivalent uniform dose (EUD) was used as an objective to determine the optimal configuration of eluters for a range of diffusion‐elimination moduli, concentrations, and number of eluters. The compound delivery vehicle that produced the greatest enhanced dose was tallied for points in parameter space mentioned to determine the conditions under whether there are situations where one approach is preferable to the other.
Results
The enhanced effect of implanted eluters was calculated for prostate volumes from 14 to 45 cm3, ϕb from 0.01 to 4 mm−1, csat from 0.05 to 7.5 times the steady‐state compound concentration released from the surface of the eluter. The number of used eluters (ne) was simulated from 10 to 60 eluters. For the region of (c sat , Φ)‐space that results in a large fraction of the gland being maximally sensitized, compound eluting spacers or sources produce equal increase in EUD. In the majority of the remaining (c sat , Φ)‐space, eluting spacers result in a greater EUD than sources even where sources often produce greater maximal physical dose enhancement. Placing eluting implants in planned locations throughout the prostate results in even greater enhancement than using only source or spacer locations.
Conclusions
Eluting brachytherapy spacers offer an opportunity to increase EUD during the routine brachytherapy process. Incorporating additional needle placements permits compound eluting spacer placement independent of source placement and thereby allowing a further increase in the therapeutic ratio. Additional work is needed to understand the in vivo spatial distribution of compound around eluters, and to incorporate time dependence of both compound release and radiation dose.
},
keywords = {Nanomedicine},
pubstate = {published},
tppubtype = {article}
}
In situ drug release concurrent with radiation therapy has been proposed to enhance the therapeutic ratio of permanent prostate brachytherapy. Both brachytherapy sources and brachytherapy spacers have been proposed as potential eluters to release compounds, such as nanoparticles or chemotherapeutic agents. The relative effectiveness of the approaches has not been compared yet. This work models the physical dose enhancement of implantable eluters in conjunction with brachytherapy to determine which delivery mechanism provides greatest opportunity to enhance the therapeutic ratio.
Materials and methods
The combined effect of implanted eluters and radioactive sources were modeled in a manner that allowed the comparison of the relative effectiveness of different types of implantable eluters over a range of parameters. Prostate geometry, source, and spacer positions were extracted from treatment plans used for 125I permanent prostate implants. Compound concentrations were calculated using steady‐state solution to the diffusion equation including an elimination term characterized by the diffusion‐elimination modulus (ϕb). Does enhancement was assumed to be dependent on compound concentration up to a saturation concentration (csat). Equivalent uniform dose (EUD) was used as an objective to determine the optimal configuration of eluters for a range of diffusion‐elimination moduli, concentrations, and number of eluters. The compound delivery vehicle that produced the greatest enhanced dose was tallied for points in parameter space mentioned to determine the conditions under whether there are situations where one approach is preferable to the other.
Results
The enhanced effect of implanted eluters was calculated for prostate volumes from 14 to 45 cm3, ϕb from 0.01 to 4 mm−1, csat from 0.05 to 7.5 times the steady‐state compound concentration released from the surface of the eluter. The number of used eluters (ne) was simulated from 10 to 60 eluters. For the region of (c sat , Φ)‐space that results in a large fraction of the gland being maximally sensitized, compound eluting spacers or sources produce equal increase in EUD. In the majority of the remaining (c sat , Φ)‐space, eluting spacers result in a greater EUD than sources even where sources often produce greater maximal physical dose enhancement. Placing eluting implants in planned locations throughout the prostate results in even greater enhancement than using only source or spacer locations.
Conclusions
Eluting brachytherapy spacers offer an opportunity to increase EUD during the routine brachytherapy process. Incorporating additional needle placements permits compound eluting spacer placement independent of source placement and thereby allowing a further increase in the therapeutic ratio. Additional work is needed to understand the in vivo spatial distribution of compound around eluters, and to incorporate time dependence of both compound release and radiation dose.