870 results on '"Farokhzad, Omid C."'
Search Results
202. Targeted nanoparticles for colorectal cancer
- Author
-
Cisterna, Bruno A, primary, Kamaly, Nazila, additional, Choi, Won Il, additional, Tavakkoli, Ali, additional, Farokhzad, Omid C, additional, and Vilos, Cristian, additional
- Published
- 2016
- Full Text
- View/download PDF
203. Ultra-pH-Responsive and Tumor-Penetrating Nanoplatform for Targeted siRNA Delivery with Robust Anti-Cancer Efficacy
- Author
-
Xu, Xiaoding, primary, Wu, Jun, additional, Liu, Yanlan, additional, Yu, Mikyung, additional, Zhao, Lili, additional, Zhu, Xi, additional, Bhasin, Sushant, additional, Li, Qing, additional, Ha, Emily, additional, Shi, Jinjun, additional, and Farokhzad, Omid C., additional
- Published
- 2016
- Full Text
- View/download PDF
204. Targeted Interleukin-10 Nanotherapeutics Developed with a Microfluidic Chip Enhance Resolution of Inflammation in Advanced Atherosclerosis
- Author
-
Kamaly, Nazila, primary, Fredman, Gabrielle, additional, Fojas, Jhalique Jane R., additional, Subramanian, Manikandan, additional, Choi, Won II, additional, Zepeda, Katherine, additional, Vilos, Cristian, additional, Yu, Mikyung, additional, Gadde, Suresh, additional, Wu, Jun, additional, Milton, Jaclyn, additional, Carvalho Leitao, Renata, additional, Rosa Fernandes, Livia, additional, Hasan, Moaraj, additional, Gao, Huayi, additional, Nguyen, Vance, additional, Harris, Jordan, additional, Tabas, Ira, additional, and Farokhzad, Omid C., additional
- Published
- 2016
- Full Text
- View/download PDF
205. Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release
- Author
-
Kamaly, Nazila, primary, Yameen, Basit, additional, Wu, Jun, additional, and Farokhzad, Omid C., additional
- Published
- 2016
- Full Text
- View/download PDF
206. Polymeric Nanoparticles Amenable to Simultaneous Installation of Exterior Targeting and Interior Therapeutic Proteins
- Author
-
Zhu, Xi, primary, Wu, Jun, additional, Shan, Wei, additional, Tao, Wei, additional, Zhao, Lili, additional, Lim, Jong-Min, additional, D'Ortenzio, Mathew, additional, Karnik, Rohit, additional, Huang, Yuan, additional, Shi, Jinjun, additional, and Farokhzad, Omid C., additional
- Published
- 2016
- Full Text
- View/download PDF
207. Microfluidic Platform for Combinatorial Synthesis and Optimization of Targeted Nanoparticles for Cancer Therapy
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Valencia, Pedro M., Pridgen, Eric M., Rhee, Minsoung, Langer, Robert, Farokhzad, Omid C., Karnik, Rohit, MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Valencia, Pedro M., Pridgen, Eric M., Rhee, Minsoung, Langer, Robert, Farokhzad, Omid C., and Karnik, Rohit
- Abstract
Taking a nanoparticle (NP) from discovery to clinical translation has been slow compared to small molecules, in part by the lack of systems that enable their precise engineering and rapid optimization. In this work we have developed a microfluidic platform for the rapid, combinatorial synthesis and optimization of NPs. The system takes in a number of NP precursors from which a library of NPs with varying size, surface charge, target ligand density, and drug load is produced in a reproducible manner. We rapidly synthesized 45 different formulations of poly(lactic-co-glycolic acid)-b-poly(ethylene glycol) NPs of different size and surface composition and screened and ranked the NPs for their ability to evade macrophage uptake in vitro. Comparison of the results to pharmacokinetic studies in vivo in mice revealed a correlation between in vitro screen and in vivo behavior. Next, we selected NP synthesis parameters that resulted in longer blood half-life and used the microfluidic platform to synthesize targeted NPs with varying targeting ligand density (using a model targeting ligand against cancer cells). We screened NPs in vitro against prostate cancer cells as well as macrophages, identifying one formulation that exhibited high uptake by cancer cells yet similar macrophage uptake compared to nontargeted NPs. In vivo, the selected targeted NPs showed a 3.5-fold increase in tumor accumulation in mice compared to nontargeted NPs. The developed microfluidic platform in this work represents a tool that could potentially accelerate the discovery and clinical translation of NPs., Prostate Cancer Foundation (Award in Nanotherapeutics), National Cancer Institute (U.S.) (Center of Cancer Nanotechnology Excellence at MIT-Harvard U54-CA151884, National Heart, Lung, and Blood Institute (Programs of Excellence in Nanotechnology HHSN268201000045C), National Science Foundation (U.S.). Graduate Research Fellowship, American Society for Engineering Education. National Defense Science and Engineering Graduate Fellowship, National Cancer Institute (U.S.) (Center of Cancer Nanotechnology Excellence. Graduate Research Fellowship)
- Published
- 2015
208. Transepithelial Transport of Fc-Targeted Nanoparticles by the Neonatal Fc Receptor for Oral Delivery
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Pridgen, Eric M., Alexis, Frank, Levy-Nissenbaum, Etgar, Karnik, Rohit, Langer, Robert, Farokhzad, Omid C., Kuo, Timothy T., Blumberg, Richard S., MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Pridgen, Eric M., Alexis, Frank, Levy-Nissenbaum, Etgar, Karnik, Rohit, Langer, Robert, Farokhzad, Omid C., Kuo, Timothy T., and Blumberg, Richard S.
- Abstract
Nanoparticles are poised to have a tremendous impact on the treatment of many diseases, but their broad application is limited because currently they can only be administered by parenteral methods. Oral administration of nanoparticles is preferred but remains a challenge because transport across the intestinal epithelium is limited. We show that nanoparticles targeted to the neonatal Fc receptor (FcRn), which mediates the transport of immunoglobulin G antibodies across epithelial barriers, are efficiently transported across the intestinal epithelium using both in vitro and in vivo models. In mice, orally administered FcRn-targeted nanoparticles crossed the intestinal epithelium and reached systemic circulation with a mean absorption efficiency of 13.7%*hour compared with only 1.2%*hour for nontargeted nanoparticles. In addition, targeted nanoparticles containing insulin as a model nanoparticle-based therapy for diabetes were orally administered at a clinically relevant insulin dose of 1.1 U/kg and elicited a prolonged hypoglycemic response in wild-type mice. This effect was abolished in FcRn knockout mice, indicating that the enhanced nanoparticle transport was specifically due to FcRn. FcRn-targeted nanoparticles may have a major impact on the treatment of many diseases by enabling drugs currently limited by low bioavailability to be efficiently delivered though oral administration., Prostate Cancer Foundation (Award in Nanotherapeutics), National Cancer Institute (U.S.) (Center for Cancer Nanotechnology Excellence U54-CA151884), National Heart, Lung, and Blood Institute (Program of Excellence in Nanotechnology Award Contract HHSN268201000045C), National Institutes of Health (U.S.) (Grant EB000244), National Institutes of Health (U.S.) (R01 Grant EB015419-01), American Society for Engineering Education. National Defense Science and Engineering Graduate Fellowship, National Cancer Institute (U.S.) (Center for Cancer Nanotechnology Excellence Graduate Research Fellowship 5 U54 CA151884-02)
- Published
- 2015
209. Nanoparticle Encapsulation of Mitaplatin and the Effect Thereof on In Vivo properties
- Author
-
Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Koch Institute for Integrative Cancer Research at MIT, Farokhzad, Omid C., Johnstone, Timothy, Lippard, Stephen J., Langer, Robert, Kulak, Nora, Pridgen, Eric M., Langer, Robert S, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Koch Institute for Integrative Cancer Research at MIT, Farokhzad, Omid C., Johnstone, Timothy, Lippard, Stephen J., Langer, Robert, Kulak, Nora, Pridgen, Eric M., and Langer, Robert S
- Abstract
Nanoparticle (NP) therapeutics have the potential to significantly alter the in vivo biological properties of the pharmaceutically active agents that they carry. Here we describe the development of a polymeric NP, termed M-NP, comprising poly(d,l-lactic-co-glycolic acid)-block-poly(ethylene glycol) (PLGA-PEG), stabilized with poly(vinyl alcohol) (PVA), and loaded with a water-soluble platinum(IV) [Pt(IV)] prodrug, mitaplatin. Mitaplatin, c,c,t-[PtCl[subscript 2](NH[subscript 3])[subscript 2](OOCCHCl[subscript 2])[subscript 2]], is a compound designed to release cisplatin, an anticancer drug in widespread clinical use, and the orphan drug dichloroacetate following chemical reduction. An optimized preparation of M-NP by double emulsion and its physical characterization are reported, and the influence of encapsulation on the properties of the platinum agent is evaluated in vivo. Encapsulation increases the circulation time of Pt in the bloodstream of rats. The biodistribution of Pt in mice is also affected by nanoparticle encapsulation, resulting in reduced accumulation in the kidneys. Finally, the efficacy of both free mitaplatin and M-NP, measured by tumor growth inhibition in a mouse xenograft model of triple-negative breast cancer, reveals that controlled release of mitaplatin over time from the nanoparticle treatment produces long-term efficacy comparable to that of free mitaplatin, which might limit toxic side effects., National Institutes of Health (U.S.) (Grant 5-U54-CA119349), National Institutes of Health (U.S.) (Grant 5-U54-CA151884), National Institutes of Health (U.S.) (Grant 5-R01-CA034992), National Institutes of Health (U.S.) (MIT-Harvard Center of Cancer Nanotechnology Excellence. Grant 5-U54-CA151884-02), National Institutes of Health (U.S.) (MIT-Harvard Center of Cancer Nanotechnology Excellence. Grant 5-U54-CA151884), German Academic Exchange Service (Research Fellowship), National Institutes of Health (U.S.) (1-S10-RR13886-01)
- Published
- 2015
210. Engineered nanomedicine for myeloma and bone microenvironment targeting
- Author
-
Koch Institute for Integrative Cancer Research at MIT, Basto, Pamela Antonia, Zhang, Sufeng, Bertrand, Nicolas, Swami, Archana, Reagan, Michaela R., Mishima, Yuji, Kamaly, Nazila, Glavey, Siobhan, Moschetta, Michele, Seevaratnam, Dushanth, Zhang, Yong, Liu, Jinhe, Memarzadeh, Masoumeh, Manier, Salomon, Shi, Jinjun, Lu, Zhi Ning, Nagano, Kenichi, Baron, Roland, Sacco, Antonio, Roccaro, Aldo M., Farokhzad, Omid C., Ghobrial, Irene M., Wu, Jun, 1968, Koch Institute for Integrative Cancer Research at MIT, Basto, Pamela Antonia, Zhang, Sufeng, Bertrand, Nicolas, Swami, Archana, Reagan, Michaela R., Mishima, Yuji, Kamaly, Nazila, Glavey, Siobhan, Moschetta, Michele, Seevaratnam, Dushanth, Zhang, Yong, Liu, Jinhe, Memarzadeh, Masoumeh, Manier, Salomon, Shi, Jinjun, Lu, Zhi Ning, Nagano, Kenichi, Baron, Roland, Sacco, Antonio, Roccaro, Aldo M., Farokhzad, Omid C., Ghobrial, Irene M., and Wu, Jun, 1968
- Abstract
Bone is a favorable microenvironment for tumor growth and a frequent destination for metastatic cancer cells. Targeting cancers within the bone marrow remains a crucial oncologic challenge due to issues of drug availability and microenvironment-induced resistance. Herein, we engineered bone-homing polymeric nanoparticles (NPs) for spatiotemporally controlled delivery of therapeutics to bone, which diminish off-target effects and increase local drug concentrations. The NPs consist of poly(d,l-lactic-co-glycolic acid) (PLGA), polyethylene glycol (PEG), and bisphosphonate (or alendronate, a targeting ligand). The engineered NPs were formulated by blending varying ratios of the synthesized polymers: PLGA-b-PEG and alendronate-conjugated polymer PLGA-b-PEG-Ald, which ensured long circulation and targeting capabilities, respectively. The bone-binding ability of Ald-PEG-PLGA NPs was investigated by hydroxyapatite binding assays and ex vivo imaging of adherence to bone fragments. In vivo biodistribution of fluorescently labeled NPs showed higher retention, accumulation, and bone homing of targeted Ald-PEG-PLGA NPs, compared with nontargeted PEG-PLGA NPs. A library of bortezomib-loaded NPs (bone-targeted Ald-Bort-NPs and nontargeted Bort-NPs) were developed and screened for optimal physiochemical properties, drug loading, and release profiles. Ald-Bort-NPs were tested for efficacy in mouse models of multiple myeloma (MM). Results demonstrated significantly enhanced survival and decreased tumor burden in mice pretreated with Ald-Bort-NPs versus Ald-Empty-NPs (no drug) or the free drug. We also observed that bortezomib, as a pretreatment regimen, modified the bone microenvironment and enhanced bone strength and volume. Our findings suggest that NP-based anticancer therapies with bone-targeting specificity comprise a clinically relevant method of drug delivery that can inhibit tumor progression in MM., United States. Dept. of Defense (Grant W81XWH-05-1-0390), Movember Foundation (Movember Prostate Cancer Foundation Challenge Award), National Research Foundation of Korea (K1A1A2048701), David H. Koch Institute for Integrative Cancer Research at MIT (David Koch-Prostate Cancer Foundation Award in Nanotherapeutics), Canadian Institutes of Health Research, National Institutes of Health (U.S.) (grant R00 CA160350), National Institutes of Health (U.S.) (grant R01 FD003743), National Institutes of Health (U.S.) (grant R01 CA154648), National Institutes of Health (U.S.) (grant CA151884)
- Published
- 2015
211. Tumour-associated macrophages act as a slow-release reservoir of nano-therapeutic Pt(IV) pro-drug
- Author
-
Massachusetts Institute of Technology. Department of Chemistry, Zheng, Yao-Rong, Lippard, Stephen J., Miller, Miles Aaron, Gadde, Suresh, Pfirschke, Christina, Zope, Harshal, Engblom, Camilla, Kohler, Rainer H., Iwamoto, Yoshiko, Yang, Katherine S., Askevold, Bjorn, Kolishetti, Nagesh, Pittet, Mikael, Farokhzad, Omid C., Weissleder, Ralph, Massachusetts Institute of Technology. Department of Chemistry, Zheng, Yao-Rong, Lippard, Stephen J., Miller, Miles Aaron, Gadde, Suresh, Pfirschke, Christina, Zope, Harshal, Engblom, Camilla, Kohler, Rainer H., Iwamoto, Yoshiko, Yang, Katherine S., Askevold, Bjorn, Kolishetti, Nagesh, Pittet, Mikael, Farokhzad, Omid C., and Weissleder, Ralph
- Abstract
Therapeutic nanoparticles (TNPs) aim to deliver drugs more safely and effectively to cancers, yet clinical results have been unpredictable owing to limited in vivo understanding. Here we use single-cell imaging of intratumoral TNP pharmacokinetics and pharmacodynamics to better comprehend their heterogeneous behaviour. Model TNPs comprising a fluorescent platinum(IV) pro-drug and a clinically tested polymer platform (PLGA-b-PEG) promote long drug circulation and alter accumulation by directing cellular uptake toward tumour-associated macrophages (TAMs). Simultaneous imaging of TNP vehicle, its drug payload and single-cell DNA damage response reveals that TAMs serve as a local drug depot that accumulates significant vehicle from which DNA-damaging Pt payload gradually releases to neighbouring tumour cells. Correspondingly, TAM depletion reduces intratumoral TNP accumulation and efficacy. Thus, nanotherapeutics co-opt TAMs for drug delivery, which has implications for TNP design and for selecting patients into trials., National Cancer Institute (U.S.) (Grant RO1-CA034992)
- Published
- 2015
212. Effects of ligands with different water solubilities on self-assembly and properties of targeted nanoparticles
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Valencia, Pedro M., Gao, Weiwei, Karim, Fawziya, Langer, Robert, Karnik, Rohit, Farokhzad, Omid C., Hanewich-Hollatz, Mikhail H., Valencia, Pedro Miguel, Langer, Robert S, MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Valencia, Pedro M., Gao, Weiwei, Karim, Fawziya, Langer, Robert, Karnik, Rohit, Farokhzad, Omid C., Hanewich-Hollatz, Mikhail H., Valencia, Pedro Miguel, and Langer, Robert S
- Abstract
The engineering of drug-encapsulated targeted nanoparticles (NPs) has the potential to revolutionize drug therapy. A major challenge for the smooth translation of targeted NPs to the clinic has been developing methods for the prediction and optimization of the NP surface composition, especially when targeting ligands (TL) of different chemical properties are involved in the NP self-assembly process. Here we investigated the self-assembly and properties of two different targeted NPs decorated with two widely used TLs that have different water solubilities, and developed methods to characterize and optimize NP surface composition. We synthesized two different biofunctional polymers composed of poly(lactide-co-glycolide)-b-polyethyleneglycol-RGD (PLGA-PEG-RGD, high water solubility TL) and PLGA-PEG-Folate (low water solubility TL). Targeted NPs with different ligand densities were prepared by mixing TL-conjugated polymers with non-conjugated PLGA-PEG at different ratios through nanoprecipitation. The NP surface composition was quantified and the results revealed two distinct nanoparticle assembly behaviors: for the case of PLGA-PEG-RGD, nearly all RGD molecules conjugated to the polymer were found to be on the surface of the NPs. In contrast, only ~20% of the folate from PLGA-PEG-Folate was present on the NP surface while the rest remained presumably buried in the PLGA NP core due to hydrophobic interactions of PLGA and folate. Finally, in vitro phagocytosis and cell targeting of NPs were investigated, from which a window of NP formulations exhibiting minimum uptake by macrophages and maximum uptake by targeted cells was determined. These results underscore the impact that the ligand chemical properties have on the targeting capabilities of self-assembled targeted nanoparticles and provide an engineering strategy for improving their targeting specificity., Prostate Cancer Foundation (Award in Nanotherapeutics), National Cancer Institute (U.S.) (Center of Cancer Nanotechnology Excellence at MIT-Harvard U54-CA151884), National Heart, Lung, and Blood Institute (Program of Excellence in Nanotechnology Award Contract HHSN268201000045C), National Science Foundation (U.S.). Graduate Research Fellowship
- Published
- 2015
213. Ultra-High Throughput Synthesis of Nanoparticles with Homogeneous Size Distribution Using a Coaxial Turbulent Jet Mixer
- Author
-
Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Lim, Jong-Min, Gilson, Laura M., Chopra, Sunandini, Choi, Sungyoung, Langer, Robert, Karnik, Rohit, Swami, Archana, Farokhzad, Omid C., Wu, Jun, 1968, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Lim, Jong-Min, Gilson, Laura M., Chopra, Sunandini, Choi, Sungyoung, Langer, Robert, Karnik, Rohit, Swami, Archana, Farokhzad, Omid C., and Wu, Jun, 1968
- Abstract
High-throughput production of nanoparticles (NPs) with controlled quality is critical for their clinical translation into effective nanomedicines for diagnostics and therapeutics. Here we report a simple and versatile coaxial turbulent jet mixer that can synthesize a variety of NPs at high throughput up to 3 kg/d, while maintaining the advantages of homogeneity, reproducibility, and tunability that are normally accessible only in specialized microscale mixing devices. The device fabrication does not require specialized machining and is easy to operate. As one example, we show reproducible, high-throughput formulation of siRNA-polyelectrolyte polyplex NPs that exhibit effective gene knockdown but exhibit significant dependence on batch size when formulated using conventional methods. The coaxial turbulent jet mixer can accelerate the development of nanomedicines by providing a robust and versatile platform for preparation of NPs at throughputs suitable for in vivo studies, clinical trials, and industrial-scale production., Prostate Cancer Foundation (Award in Nanotherapeutics), National Institutes of Health (U.S.) (Grant EB015419), National Institutes of Health (U.S.) (Grant CA119349)
- Published
- 2015
214. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance
- Author
-
Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Langer, Robert, Maldonado, Roberto A., LaMothe, Robert A., Ferrari, Joseph D., Zhang, Ai-Hong, Rossi, Robert J., Kolte, Pallavi N., Griset, Aaron P., O'Neil, Conlin P., Altreuter, David H., Browning, Erica A., Johnston, Lloyd P. M., Farokhzad, Omid C., Scott, David W., von Andrian, Ulrich H., Kishimoto, Takashi Kei, Langer, Robert S, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Langer, Robert, Maldonado, Roberto A., LaMothe, Robert A., Ferrari, Joseph D., Zhang, Ai-Hong, Rossi, Robert J., Kolte, Pallavi N., Griset, Aaron P., O'Neil, Conlin P., Altreuter, David H., Browning, Erica A., Johnston, Lloyd P. M., Farokhzad, Omid C., Scott, David W., von Andrian, Ulrich H., Kishimoto, Takashi Kei, and Langer, Robert S
- Abstract
Current treatments to control pathological or unwanted immune responses often use broadly immunosuppressive drugs. New approaches to induce antigen-specific immunological tolerance that control both cellular and humoral immune responses are desirable. Here we describe the use of synthetic, biodegradable nanoparticles carrying either protein or peptide antigens and a tolerogenic immunomodulator, rapamycin, to induce durable and antigen-specific immune tolerance, even in the presence of potent Toll-like receptor agonists. Treatment with tolerogenic nanoparticles results in the inhibition of CD4+ and CD8+ T-cell activation, an increase in regulatory cells, durable B-cell tolerance resistant to multiple immunogenic challenges, and the inhibition of antigen-specific hypersensitivity reactions, relapsing experimental autoimmune encephalomyelitis, and antibody responses against coagulation factor VIII in hemophilia A mice, even in animals previously sensitized to antigen. Only encapsulated rapamycin, not the free form, could induce immunological tolerance. Tolerogenic nanoparticle therapy represents a potential novel approach for the treatment of allergies, autoimmune diseases, and prevention of antidrug antibodies against biologic therapies., Juvenile Diabetes Research Foundation International
- Published
- 2015
215. Synergistic cytotoxicity of irinotecan and cisplatin in dual-drug targeted polymeric nanoparticles
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Valencia, Pedro M., Pridgen, Eric M., Langer, Robert, Lippard, Stephen J., Farokhzad, Omid C., Karnik, Rohit, Valencia, Pedro M, Pridgen, Eric M, Perea, Brian, Gadde, Suresh, Sweeney, Christopher, Kantoff, Philip W., Bander, Neil H., MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Valencia, Pedro M., Pridgen, Eric M., Langer, Robert, Lippard, Stephen J., Farokhzad, Omid C., Karnik, Rohit, Valencia, Pedro M, Pridgen, Eric M, Perea, Brian, Gadde, Suresh, Sweeney, Christopher, Kantoff, Philip W., and Bander, Neil H.
- Abstract
Aim: Two unexplored aspects for irinotecan and cisplatin (I&C) combination chemotherapy are: actively targeting both drugs to a specific diseased cell type, and delivering both drugs on the same vehicle to ensure their synchronized entry into the cell at a well-defined ratio. In this work, the authors report the use of targeted polymeric nanoparticles (NPs) to coencapsulate and deliver I&C to cancer cells expressing the prostate-specific membrane antigen. Materials & methods: Targeted NPs were prepared in a single step by mixing four different precursors inside microfluidic devices. Results: I&C were encapsulated in 55-nm NPs and showed an eightfold increase in internalization by prostate-specific membrane antigen-expressing LNCaP cells compared with nontargeted NPs. NPs coencapsulating both drugs exhibited strong synergism in LNCaP cells with a combination index of 0.2. Conclusion: The strategy of coencapsulating both I&C in a single NP targeted to a specific cell type could potentially be used to treat different types of cancer., Prostate Cancer Foundation (Nanotherapeutics Award), MIT-Harvard Center of Cancer Nanotechnology Excellence (U54-CA151884), National Science Foundation (U.S.). Graduate Research Fellowship Program, American Society for Engineering Education. National Defense Science and Engineering Graduate Fellowship
- Published
- 2015
216. Glutathione-Responsive Prodrug Nanoparticles for Effective Drug Delivery and Cancer Therapy
- Author
-
Ling, Xiang, Tu, Jiasheng, Wang, Junqing, Shajii, Aram, Kong, Na, Feng, Chan, Zhang, Ye, Yu, Mikyung, Xie, Tian, Bharwani, Zameer, Aljaeid, Bader M., Shi, Bingyang, Tao, Wei, and Farokhzad, Omid C.
- Abstract
Spurred by recent progress in medicinal chemistry, numerous lead compounds have sprung up in the past few years, although the majority are hindered by hydrophobicity, which greatly challenges druggability. In an effort to assess the potential of platinum (Pt) candidates, the nanosizing approach to alter the pharmacology of hydrophobic Pt(IV) prodrugs in discovery and development settings is described. The construction of a self-assembled nanoparticle (NP) platform, composed of amphiphilic lipid-polyethylene glycol (PEG) for effective delivery of Pt(IV) prodrugs capable of resisting thiol-mediated detoxification through a glutathione (GSH)-exhausting effect, offers a promising route to synergistically improving safety and efficacy. After a systematic screening, the optimized NPs (referred to as P6NPs) exhibited small particle size (99.3 nm), high Pt loading (11.24%), reliable dynamic stability (∼7 days), and rapid redox-triggered release (∼80% in 3 days). Subsequent experiments on cells support the emergence of P6NPs as a highly effective means of transporting a lethal dose of cargo across cytomembranes through macropinocytosis. Upon reduction by cytoplasmic reductants, particularly GSH, P6NPs under disintegration released sufficient active Pt(II) metabolites, which covalently bound to target DNA and induced significant apoptosis. The PEGylation endowed P6NPs with in vivolongevity and tumor specificity, which were essential to successfully inhibiting the growth of cisplatin-sensitive and -resistant xenograft tumors, while effectively alleviating toxic side-effects associated with cisplatin. P6NPs are, therefore, promising for overcoming the bottleneck in the development of Pt drugs for oncotherapy.
- Published
- 2019
- Full Text
- View/download PDF
217. Predicting therapeutic nanomedicine efficacy using a companion magnetic resonance imaging nanoparticle
- Author
-
Miller, Miles A., primary, Gadde, Suresh, additional, Pfirschke, Christina, additional, Engblom, Camilla, additional, Sprachman, Melissa M., additional, Kohler, Rainer H., additional, Yang, Katherine S., additional, Laughney, Ashley M., additional, Wojtkiewicz, Gregory, additional, Kamaly, Nazila, additional, Bhonagiri, Sushma, additional, Pittet, Mikael J., additional, Farokhzad, Omid C., additional, and Weissleder, Ralph, additional
- Published
- 2015
- Full Text
- View/download PDF
218. Tumour-associated macrophages act as a slow-release reservoir of nano-therapeutic Pt(IV) pro-drug
- Author
-
Miller, Miles A., primary, Zheng, Yao-Rong, additional, Gadde, Suresh, additional, Pfirschke, Christina, additional, Zope, Harshal, additional, Engblom, Camilla, additional, Kohler, Rainer H., additional, Iwamoto, Yoshiko, additional, Yang, Katherine S., additional, Askevold, Bjorn, additional, Kolishetti, Nagesh, additional, Pittet, Mikael, additional, Lippard, Stephen J., additional, Farokhzad, Omid C., additional, and Weissleder, Ralph, additional
- Published
- 2015
- Full Text
- View/download PDF
219. Drug Delivery Nanocarriers from a Fully Degradable PEG‐Conjugated Polyester with a Reduction‐Responsive Backbone
- Author
-
Yameen, Basit, primary, Vilos, Cristian, additional, Choi, Won Il, additional, Whyte, Andrew, additional, Huang, Jining, additional, Pollit, Lori, additional, and Farokhzad, Omid C., additional
- Published
- 2015
- Full Text
- View/download PDF
220. Hydrophobic Cysteine Poly(disulfide)‐based Redox‐Hypersensitive Nanoparticle Platform for Cancer Theranostics
- Author
-
Wu, Jun, primary, Zhao, Lili, additional, Xu, Xiaoding, additional, Bertrand, Nicolas, additional, Choi, Won II, additional, Yameen, Basit, additional, Shi, Jinjun, additional, Shah, Vishva, additional, Mulvale, Matthew, additional, MacLean, James L., additional, and Farokhzad, Omid C., additional
- Published
- 2015
- Full Text
- View/download PDF
221. Polymeric nanoparticle drug delivery technologies for oral delivery applications
- Author
-
Pridgen, Eric M, primary, Alexis, Frank, additional, and Farokhzad, Omid C, additional
- Published
- 2015
- Full Text
- View/download PDF
222. Effect of PEG Pairing on the Efficiency of Cancer-Targeting Liposomes
- Author
-
Saw, Phei Er, primary, Park, Jinho, additional, Lee, Eunbeol, additional, Ahn, Sukyung, additional, Lee, Jinju, additional, Kim, Hyungjun, additional, Kim, Jinjoo, additional, Choi, Minsuk, additional, Farokhzad, Omid C., additional, and Jon, Sangyong, additional
- Published
- 2015
- Full Text
- View/download PDF
223. Adjuvant-carrying synthetic vaccine particles augment the immune response to encapsulated antigen and exhibit strong local immune activation without inducing systemic cytokine release
- Author
-
Harvard University--MIT Division of Health Sciences and Technology, Koch Institute for Integrative Cancer Research at MIT, Basto, Pamela Antonia, Radovic-Moreno, Aleksandar F., Langer, Robert, Ilyinskii, Petr O., Roy, Christopher J., O'Neil, Conlin P., Browning, Erica A., Pittet, Lynnelle A., Altreuter, David H., Alexis, Frank, Tonti, Elena, Shi, Jinjun, Iannacone, Matteo, Farokhzad, Omid C., von Andrian, Ulrich H., Johnston, Lloyd P. M., Kishimoto, Takashi Kei, Harvard University--MIT Division of Health Sciences and Technology, Koch Institute for Integrative Cancer Research at MIT, Basto, Pamela Antonia, Radovic-Moreno, Aleksandar F., Langer, Robert, Ilyinskii, Petr O., Roy, Christopher J., O'Neil, Conlin P., Browning, Erica A., Pittet, Lynnelle A., Altreuter, David H., Alexis, Frank, Tonti, Elena, Shi, Jinjun, Iannacone, Matteo, Farokhzad, Omid C., von Andrian, Ulrich H., Johnston, Lloyd P. M., and Kishimoto, Takashi Kei
- Abstract
Augmentation of immunogenicity can be achieved by particulate delivery of an antigen and by its co-administration with an adjuvant. However, many adjuvants initiate strong systemic inflammatory reactions in vivo, leading to potential adverse events and safety concerns. We have developed a synthetic vaccine particle (SVP) technology that enables co-encapsulation of antigen with potent adjuvants. We demonstrate that co-delivery of an antigen with a TLR7/8 or TLR9 agonist in synthetic polymer nanoparticles results in a strong augmentation of humoral and cellular immune responses with minimal systemic production of inflammatory cytokines. In contrast, antigen encapsulated into nanoparticles and admixed with free TLR7/8 agonist leads to lower immunogenicity and rapid induction of high levels of inflammatory cytokines in the serum (e.g., TNF-a and IL-6 levels are 50- to 200-fold higher upon injection of free resiquimod (R848) than of nanoparticle-encapsulated R848). Conversely, local immune stimulation as evidenced by cellular infiltration of draining lymph nodes and by intranodal cytokine production was more pronounced and persisted longer when SVP-encapsulated TLR agonists were used. The strong local immune activation achieved using a modular self-assembling nanoparticle platform markedly enhanced immunogenicity and was equally effective whether antigen and adjuvant were co-encapsulated in a single nanoparticle formulation or co-delivered in two separate nanoparticles. Moreover, particle encapsulation enabled the utilization of CpG oligonucleotides with the natural phosphodiester backbone, which are otherwise rapidly hydrolyzed by nucleases in vivo. The use of SVP may enable clinical use of potent TLR agonists as vaccine adjuvants for indications where cellular immunity or robust humoral responses are required.
- Published
- 2014
224. Surface Charge-Switching Polymeric Nanoparticles for Bacterial Cell Wall-Targeted Delivery of Antibiotics
- Author
-
Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Synthetic Biology Center, Radovic-Moreno, Aleksandar F., Lu, Timothy K., Puscasu, Vlad A., Yoon, Christopher J., Langer, Robert, Farokhzad, Omid C., Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Synthetic Biology Center, Radovic-Moreno, Aleksandar F., Lu, Timothy K., Puscasu, Vlad A., Yoon, Christopher J., Langer, Robert, and Farokhzad, Omid C.
- Abstract
Bacteria have shown a remarkable ability to overcome drug therapy if there is a failure to achieve sustained bactericidal concentration or if there is a reduction in activity in situ. The latter can be caused by localized acidity, a phenomenon that can occur as a result of the combined actions of bacterial metabolism and the host immune response. Nanoparticles (NP) have shown promise in treating bacterial infections, but a significant challenge has been to develop antibacterial NPs that may be suitable for systemic administration. Herein we develop drug-encapsulated, pH-responsive, surface charge-switching poly(d,l-lactic-co-glycolic acid)-b-poly(l-histidine)-b-poly(ethylene glycol) (PLGA-PLH-PEG) nanoparticles for treating bacterial infections. These NP drug carriers are designed to shield nontarget interactions at pH 7.4 but bind avidly to bacteria in acidity, delivering drugs and mitigating in part the loss of drug activity with declining pH. The mechanism involves pH-sensitive NP surface charge switching, which is achieved by selective protonation of the imidazole groups of PLH at low pH. NP binding studies demonstrate pH-sensitive NP binding to bacteria with a 3.5 ± 0.2- to 5.8 ± 0.1-fold increase in binding to bacteria at pH 6.0 compared to 7.4. Further, PLGA-PLH-PEG-encapsulated vancomycin demonstrates reduced loss of efficacy at low pH, with an increase in minimum inhibitory concentration of 1.3-fold as compared to 2.0-fold and 2.3-fold for free and PLGA-PEG-encapsulated vancomycin, respectively. The PLGA-PLH-PEG NPs described herein are a first step toward developing systemically administered drug carriers that can target and potentially treat Gram-positive, Gram-negative, or polymicrobial infections associated with acidity., National Institutes of Health (U.S.) (Grant CA151884), National Institutes of Health (U.S.) (Grant EB003647), Prostate Cancer Foundation (Award in Nanotherapeutics), United States. Dept. of Defense (Prostate Cancer Research Program PC 051156), MIT-Portugal Program, National Science Foundation (U.S.). Graduate Research Fellowship, National Institutes of Health (U.S.) (Office of the Director Grant DP2OD008435)
- Published
- 2014
225. Probing nanoparticle translocation across the permeable endothelium in experimental atherosclerosis
- Author
-
Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Lee Chung, Bomy, Becraft, Jacob Robert, Ma, Mingming, Langer, Robert, Kim, YongTae, Lobatto, Mark E., Kawahara, Tomohiro, Mieszawska, Aneta J., Sanchez-Gaytan, Brenda L., Fay, Francois, Senders, Max L., Calcagno, Claudia, Tun Saung, May, Gordon, Ronald E., Stroes, Erik S. G., Farokhzad, Omid C., Fayad, Zahi A., Mulder, Willem J. M., Langer, Robert S, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Lee Chung, Bomy, Becraft, Jacob Robert, Ma, Mingming, Langer, Robert, Kim, YongTae, Lobatto, Mark E., Kawahara, Tomohiro, Mieszawska, Aneta J., Sanchez-Gaytan, Brenda L., Fay, Francois, Senders, Max L., Calcagno, Claudia, Tun Saung, May, Gordon, Ronald E., Stroes, Erik S. G., Farokhzad, Omid C., Fayad, Zahi A., Mulder, Willem J. M., and Langer, Robert S
- Abstract
Therapeutic and diagnostic nanomaterials are being intensely studied for several diseases, including cancer and atherosclerosis. However, the exact mechanism by which nanomedicines accumulate at targeted sites remains a topic of investigation, especially in the context of atherosclerotic disease. Models to accurately predict transvascular permeation of nanomedicines are needed to aid in design optimization. Here we show that an endothelialized microchip with controllable permeability can be used to probe nanoparticle translocation across an endothelial cell layer. To validate our in vitro model, we studied nanoparticle translocation in an in vivo rabbit model of atherosclerosis using a variety of preclinical and clinical imaging methods. Our results reveal that the translocation of lipid–polymer hybrid nanoparticles across the atherosclerotic endothelium is dependent on microvascular permeability. These results were mimicked with our microfluidic chip, demonstrating the potential utility of the model system., National Heart, Lung, and Blood Institute (Contract HHSN268201000045C), National Cancer Institute (U.S.) (Grant CA151884), Prostate Cancer Foundation (Award in Nanotherapeutics)
- Published
- 2014
226. Enhancing tumor cell response to chemotherapy through nanoparticle-mediated codelivery of siRNA and cisplatin prodrug
- Author
-
Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Koch Institute for Integrative Cancer Research at MIT, Xu, Xiaoyang, Xie, Kun, Pridgen, Eric M., Park, Ga Young, Lippard, Stephen J., Langer, Robert, Walker, Graham C., Zhang, Xue-Qing, Cui, Danica S., Shi, Jinjun, Kantoff, Philip W., Farokhzad, Omid C., Langer, Robert S, Wu, Jun, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Koch Institute for Integrative Cancer Research at MIT, Xu, Xiaoyang, Xie, Kun, Pridgen, Eric M., Park, Ga Young, Lippard, Stephen J., Langer, Robert, Walker, Graham C., Zhang, Xue-Qing, Cui, Danica S., Shi, Jinjun, Kantoff, Philip W., Farokhzad, Omid C., Langer, Robert S, and Wu, Jun
- Abstract
Cisplatin and other DNA-damaging chemotherapeutics are widely used to treat a broad spectrum of malignancies. However, their application is limited by both intrinsic and acquired chemoresistance. Most mutations that result from DNA damage are the consequence of error-prone translesion DNA synthesis, which could be responsible for the acquired resistance against DNA-damaging agents. Recent studies have shown that the suppression of crucial gene products (e.g., REV1, REV3L) involved in the error-prone translesion DNA synthesis pathway can sensitize intrinsically resistant tumors to chemotherapy and reduce the frequency of acquired drug resistance of relapsed tumors. In this context, combining conventional DNA-damaging chemotherapy with siRNA-based therapeutics represents a promising strategy for treating patients with malignancies. To this end, we developed a versatile nanoparticle (NP) platform to deliver a cisplatin prodrug and REV1/REV3L-specific siRNAs simultaneously to the same tumor cells. NPs are formulated through self-assembly of a biodegradable poly(lactide-coglycolide)-b-poly(ethylene glycol) diblock copolymer and a self-synthesized cationic lipid. We demonstrated the potency of the siRNA-containing NPs to knock down target genes efficiently both in vitro and in vivo. The therapeutic efficacy of NPs containing both cisplatin prodrug and REV1/REV3L-specific siRNAs was further investigated in vitro and in vivo. Quantitative real-time PCR results showed that the NPs exhibited a significant and sustained suppression of both genes in tumors for up to 3 d after a single dose. Administering these NPs revealed a synergistic effect on tumor inhibition in a human Lymph Node Carcinoma of the Prostate xenograft mouse model that was strikingly more effective than platinum monotherapy., Prostate Cancer Foundation (Award in Nanotherapeutics), National Cancer Institute (U.S.). Centers of Cancer Nanotechnology Excellence, National Institutes of Health (U.S.) (Grant CA151884), National Institute of Environmental Health Sciences (Grant ES015818), National Institutes of Health (U.S.) (National Research Service Award Grant 1F32CA168163-01))
- Published
- 2014
227. Mass Production and Size Control of Lipid–Polymer Hybrid Nanoparticles through Controlled Microvortices
- Author
-
Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Kim, YongTae, Lee Chung, Bomy, Ma, Mingming, Langer, Robert, Mulder, Willem J. M., Fayad, Zahi A., Farokhzad, Omid C., Langer, Robert S, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Kim, YongTae, Lee Chung, Bomy, Ma, Mingming, Langer, Robert, Mulder, Willem J. M., Fayad, Zahi A., Farokhzad, Omid C., and Langer, Robert S
- Abstract
Lipid–polymer hybrid (LPH) nanoparticles can deliver a wide range of therapeutic compounds in a controlled manner. LPH nanoparticle syntheses using microfluidics improve the mixing process but are restricted by a low throughput. In this study, we present a pattern-tunable microvortex platform that allows mass production and size control of LPH nanoparticles with superior reproducibility and homogeneity. We demonstrate that by varying flow rates (i.e., Reynolds number (30–150)) we can control the nanoparticle size (30–170 nm) with high productivity (~3 g/hour) and low polydispersity (~0.1). Our approach may contribute to efficient development and optimization of a wide range of multicomponent nanoparticles for medical imaging and drug delivery., National Heart, Lung, and Blood Institute (Program of Excellence in Nanotechnology (PEN) Award Contract HHSN268201000045C), National Cancer Institute (U.S.) (Grant P01 CA151884), Prostate Cancer Foundation (Award in Nanotherapeutics)
- Published
- 2014
228. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance
- Author
-
Maldonado, Roberto A., primary, LaMothe, Robert A., additional, Ferrari, Joseph D., additional, Zhang, Ai-Hong, additional, Rossi, Robert J., additional, Kolte, Pallavi N., additional, Griset, Aaron P., additional, O’Neil, Conlin, additional, Altreuter, David H., additional, Browning, Erica, additional, Johnston, Lloyd, additional, Farokhzad, Omid C., additional, Langer, Robert, additional, Scott, David W., additional, von Andrian, Ulrich H., additional, and Kishimoto, Takashi Kei, additional
- Published
- 2014
- Full Text
- View/download PDF
229. Targeted Nanotherapeutics Encapsulating Liver X Receptor Agonist GW3965 Enhance Antiatherogenic Effects without Adverse Effects on Hepatic Lipid Metabolism in Ldlr−/− Mice.
- Author
-
Yu, Mikyung, Amengual, Jaume, Menon, Arjun, Kamaly, Nazila, Zhou, Felix, Xu, Xiaoding, Saw, Phei Er, Lee, Seung‐Joo, Si, Kevin, Ortega, Carleena Angelica, Choi, Won Il, Lee, In‐Hyun, Bdour, Yazan, Shi, Jinjun, Mahmoudi, Morteza, Jon, Sangyong, Fisher, Edward A., and Farokhzad, Omid C.
- Published
- 2017
- Full Text
- View/download PDF
230. Aptamer bioconjugates for Cancer Therapy
- Author
-
Farokhzad, Omid C., primary and Langer, Robert, additional
- Full Text
- View/download PDF
231. A Solvent-Free Thermosponge Nanoparticle Platform for Efficient Delivery of Labile Proteins
- Author
-
Choi, Won Il, primary, Kamaly, Nazila, additional, Riol-Blanco, Lorena, additional, Lee, In-Hyun, additional, Wu, Jun, additional, Swami, Archana, additional, Vilos, Cristian, additional, Yameen, Basit, additional, Yu, Mikyung, additional, Shi, Jinjun, additional, Tabas, Ira, additional, von Andrian, Ulrich H., additional, Jon, Sangyong, additional, and Farokhzad, Omid C., additional
- Published
- 2014
- Full Text
- View/download PDF
232. Nanoparticles Containing a Liver X Receptor Agonist Inhibit Inflammation and Atherosclerosis
- Author
-
Zhang, Xue-Qing, primary, Even-Or, Orli, additional, Xu, Xiaoyang, additional, van Rosmalen, Mariska, additional, Lim, Lucas, additional, Gadde, Suresh, additional, Farokhzad, Omid C., additional, and Fisher, Edward A., additional
- Published
- 2014
- Full Text
- View/download PDF
233. Development of Multinuclear Polymeric Nanoparticles as Robust Protein Nanocarriers
- Author
-
Wu, Jun, primary, Kamaly, Nazila, additional, Shi, Jinjun, additional, Zhao, Lili, additional, Xiao, Zeyu, additional, Hollett, Geoffrey, additional, John, Rohit, additional, Ray, Shaunak, additional, Xu, Xiaoyang, additional, Zhang, Xueqing, additional, Kantoff, Philip W., additional, and Farokhzad, Omid C., additional
- Published
- 2014
- Full Text
- View/download PDF
234. Engineered nanomedicine for myeloma and bone microenvironment targeting
- Author
-
Swami, Archana, primary, Reagan, Michaela R., additional, Basto, Pamela, additional, Mishima, Yuji, additional, Kamaly, Nazila, additional, Glavey, Siobhan, additional, Zhang, Sufeng, additional, Moschetta, Michele, additional, Seevaratnam, Dushanth, additional, Zhang, Yong, additional, Liu, Jinhe, additional, Memarzadeh, Masoumeh, additional, Wu, Jun, additional, Manier, Salomon, additional, Shi, Jinjun, additional, Bertrand, Nicolas, additional, Lu, Zhi Ning, additional, Nagano, Kenichi, additional, Baron, Roland, additional, Sacco, Antonio, additional, Roccaro, Aldo M., additional, Farokhzad, Omid C., additional, and Ghobrial, Irene M., additional
- Published
- 2014
- Full Text
- View/download PDF
235. Ultra-High Throughput Synthesis of Nanoparticles with Homogeneous Size Distribution Using a Coaxial Turbulent Jet Mixer
- Author
-
Lim, Jong-Min, primary, Swami, Archana, additional, Gilson, Laura M., additional, Chopra, Sunandini, additional, Choi, Sungyoung, additional, Wu, Jun, additional, Langer, Robert, additional, Karnik, Rohit, additional, and Farokhzad, Omid C., additional
- Published
- 2014
- Full Text
- View/download PDF
236. Adjuvant-carrying synthetic vaccine particles augment the immune response to encapsulated antigen and exhibit strong local immune activation without inducing systemic cytokine release
- Author
-
Ilyinskii, Petr O., primary, Roy, Christopher J., additional, O’Neil, Conlin P., additional, Browning, Erica A., additional, Pittet, Lynnelle A., additional, Altreuter, David H., additional, Alexis, Frank, additional, Tonti, Elena, additional, Shi, Jinjun, additional, Basto, Pamela A., additional, Iannacone, Matteo, additional, Radovic-Moreno, Aleksandar F., additional, Langer, Robert S., additional, Farokhzad, Omid C., additional, von Andrian, Ulrich H., additional, Johnston, Lloyd P.M., additional, and Kishimoto, Takashi Kei, additional
- Published
- 2014
- Full Text
- View/download PDF
237. Development of Therapeutic Polymeric Nanoparticles for the Resolution of Inflammation
- Author
-
Gadde, Suresh, primary, Even-Or, Orli, additional, Kamaly, Nazila, additional, Hasija, Apoorva, additional, Gagnon, Philippe G., additional, Adusumilli, Krishna H., additional, Erakovic, Andrea, additional, Pal, Anoop K., additional, Zhang, Xue-Qing, additional, Kolishetti, Nagesh, additional, Shi, Jinjun, additional, Fisher, Edward A., additional, and Farokhzad, Omid C., additional
- Published
- 2014
- Full Text
- View/download PDF
238. Current Progress of Aptamer-Based Molecular Imaging
- Author
-
Wang, Andrew Z., primary and Farokhzad, Omid C., additional
- Published
- 2014
- Full Text
- View/download PDF
239. Parallel microfluidic synthesis of size-tunable polymeric nanoparticles using 3D flow focusing towards in vivo study
- Author
-
Lim, Jong-Min, primary, Bertrand, Nicolas, additional, Valencia, Pedro M., additional, Rhee, Minsoung, additional, Langer, Robert, additional, Jon, Sangyong, additional, Farokhzad, Omid C., additional, and Karnik, Rohit, additional
- Published
- 2014
- Full Text
- View/download PDF
240. Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biology
- Author
-
Bertrand, Nicolas, primary, Wu, Jun, additional, Xu, Xiaoyang, additional, Kamaly, Nazila, additional, and Farokhzad, Omid C., additional
- Published
- 2014
- Full Text
- View/download PDF
241. Probing nanoparticle translocation across the permeable endothelium in experimental atherosclerosis
- Author
-
Kim, YongTae, primary, Lobatto, Mark E., additional, Kawahara, Tomohiro, additional, Lee Chung, Bomy, additional, Mieszawska, Aneta J., additional, Sanchez-Gaytan, Brenda L., additional, Fay, Francois, additional, Senders, Max L., additional, Calcagno, Claudia, additional, Becraft, Jacob, additional, Tun Saung, May, additional, Gordon, Ronald E., additional, Stroes, Erik S. G., additional, Ma, Mingming, additional, Farokhzad, Omid C., additional, Fayad, Zahi A., additional, Mulder, Willem J. M., additional, and Langer, Robert, additional
- Published
- 2014
- Full Text
- View/download PDF
242. Microfluidic technologies for accelerating the clinical translation of nanoparticles
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Valencia, Pedro Miguel, Farokhzad, Omid C., Karnik, Rohit, Langer, Robert, MIT-Harvard Center for Cancer Nanotechnology Excellence, Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Valencia, Pedro Miguel, Farokhzad, Omid C., Karnik, Rohit, and Langer, Robert
- Abstract
Using nanoparticles for therapy and imaging holds tremendous promise for the treatment of major diseases such as cancer. However, their translation into the clinic has been slow because it remains difficult to produce nanoparticles that are consistent 'batch-to-batch', and in sufficient quantities for clinical research. Moreover, platforms for rapid screening of nanoparticles are still lacking. Recent microfluidic technologies can tackle some of these issues, and offer a way to accelerate the clinical translation of nanoparticles. In this Progress Article, we highlight the advances in microfluidic systems that can synthesize libraries of nanoparticles in a well-controlled, reproducible and high-throughput manner. We also discuss the use of microfluidics for rapidly evaluating nanoparticles in vitro under microenvironments that mimic the in vivo conditions. Furthermore, we highlight some systems that can manipulate small organisms, which could be used for evaluating the in vivo toxicity of nanoparticles or for drug screening. We conclude with a critical assessment of the near- and long-term impact of microfluidics in the field of nanomedicine., Prostate Cancer Foundation (Award in Nanotherapeutics), MIT-Harvard Center for Cancer Nanotechnology Excellence (U54-CA151884), National Heart, Lung, and Blood Institute (Programs of Excellence in Nanotechnology (HHSN268201000045C)), National Science Foundation (U.S.) (Graduate Research Fellowship)
- Published
- 2013
243. Engineering of Targeted Nanoparticles for Cancer Therapy Using Internalizing Aptamers Isolated by Cell-Uptake Selection
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Biology, Koch Institute for Integrative Cancer Research at MIT, Xiao, Zeyu, Levy-Nissenbaum, Etgar, Alexis, Frank, Teply, Benjamin A., Chan, Juliana Maria, Shi, JinJun, Cheng, Judy, Langer, Robert, Farokhzad, Omid C., Luptak, Andrej, Shi, Jinjun, Digga, Elise, MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Biology, Koch Institute for Integrative Cancer Research at MIT, Xiao, Zeyu, Levy-Nissenbaum, Etgar, Alexis, Frank, Teply, Benjamin A., Chan, Juliana Maria, Shi, JinJun, Cheng, Judy, Langer, Robert, Farokhzad, Omid C., Luptak, Andrej, Shi, Jinjun, and Digga, Elise
- Abstract
One of the major challenges in the development of targeted nanoparticles (NPs) for cancer therapy is to discover targeting ligands that allow for differential binding and uptake by the target cancer cells. Using prostate cancer (PCa) as a model disease, we developed a cell-uptake selection strategy to isolate PCa-specific internalizing 2′-O-methyl RNA aptamers (Apts) for NP incorporation. Twelve cycles of selection and counter-selection were done to obtain a panel of internalizing Apts, which can distinguish PCa cells from nonprostate and normal prostate cells. After Apt characterization, size minimization, and conjugation of the Apts with fluorescently labeled polymeric NPs, the NP–Apt conjugates exhibit PCa specificity and enhancement in cellular uptake when compared to nontargeted NPs lacking the internalizing Apts. Furthermore, when docetaxel, a chemotherapeutic agent used for the treatment of PCa, was encapsulated within the NP–Apt, a significant improvement in cytotoxicity was achieved in targeted PCa cells. Rather than isolating high-affinity Apts as reported in previous selection processes, our selection strategy was designed to enrich cancer cell-specific internalizing Apts. A similar cell-uptake selection strategy may be used to develop specific internalizing ligands for a myriad of other diseases and can potentially facilitate delivering various molecules, including drugs and siRNAs, into target cells., National Institutes of Health (U.S.) (grant CA151884), National Institutes of Health (U.S.) (grant EB003647), David H. Koch Institute for Integrative Cancer Research at MIT (Prostate Cancer Foundation Award in Nanotherapeutics), United States. Dept. of Defense (Prostate Cancer Research Program PC 051156)
- Published
- 2013
244. Engineering of lipid-coated PLGA nanoparticles with a tunable payload of diagnostically active nanocrystals for medical imaging
- Author
-
delete, Massachusetts Institute of Technology. Department of Chemical Engineering, Langer, Robert, Mieszawska, Aneta J., Gianella, Anita, Cormode, David P., Zhao, Yiming, Meijerink, Andries, Farokhzad, Omid C., Fayad, Zahi A., Mulder, Willem J. M., delete, Massachusetts Institute of Technology. Department of Chemical Engineering, Langer, Robert, Mieszawska, Aneta J., Gianella, Anita, Cormode, David P., Zhao, Yiming, Meijerink, Andries, Farokhzad, Omid C., Fayad, Zahi A., and Mulder, Willem J. M.
- Abstract
Polylactic-co-glycolic acid (PLGA) based nanoparticles are biocompatible and biodegradable and therefore have been extensively investigated as therapeutic carriers. Here, we engineered diagnostically active PLGA nanoparticles that incorporate high payloads of nanocrystals into their core for tunable bioimaging features. We accomplished this through esterification reactions of PLGA to generate polymers modified with nanocrystals. The PLGA nanoparticles formed from modified PLGA polymers that were functionalized with either gold nanocrystals or quantum dots exhibited favorable features for computed tomography and optical imaging, respectively., National Heart, Lung, and Blood Institute (Programs of Excellence in Nanotechnology (PEN) Award, Contract #HHSN268201000045C)), National Institutes of Health (U.S.) (R01 EB009638), National Institutes of Health (U.S.) (R01 CA155432), National Institutes of Health (U.S.) (K99 EB012165), Netherlands Organization for Scientific Research ((NWO) ECHO.06.B.047)
- Published
- 2013
245. Development and in vivo efficacy of targeted polymeric inflammation-resolving nanoparticles
- Author
-
Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Langer, Robert, Farokhzad, Omid C., Gadde, Suresh, Kamaly, Nazila, Fredman, Gabrielle, Subramanian, Manikandan, Pesic, Aleksandar, Cheung, Louis, Fayad, Zahi A., Tabas, Ira, Langer, Robert S, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Langer, Robert, Farokhzad, Omid C., Gadde, Suresh, Kamaly, Nazila, Fredman, Gabrielle, Subramanian, Manikandan, Pesic, Aleksandar, Cheung, Louis, Fayad, Zahi A., Tabas, Ira, and Langer, Robert S
- Abstract
Excessive inflammation and failed resolution of the inflammatory response are underlying components of numerous conditions such as arthritis, cardiovascular disease, and cancer. Hence, therapeutics that dampen inflammation and enhance resolution are of considerable interest. In this study, we demonstrate the proresolving activity of sub–100-nm nanoparticles (NPs) containing the anti-inflammatory peptide Ac2-26, an annexin A1/lipocortin 1-mimetic peptide. These NPs were engineered using biodegradable diblock poly(lactic-co-glycolic acid)-b-polyethyleneglycol and poly(lactic-co-glycolic acid)-b-polyethyleneglycol collagen IV–targeted polymers. Using a self-limited zymosan-induced peritonitis model, we show that the Ac2-26 NPs (100 ng per mouse) were significantly more potent than Ac2-26 native peptide at limiting recruitment of polymononuclear neutrophils (56% vs. 30%) and at decreasing the resolution interval up to 4 h. Moreover, systemic administration of collagen IV targeted Ac2-26 NPs (in as low as 1 µg peptide per mouse) was shown to significantly block tissue damage in hind-limb ischemia-reperfusion injury by up to 30% in comparison with controls. Together, these findings demonstrate that Ac2-26 NPs are proresolving in vivo and raise the prospect of their use in chronic inflammatory diseases such as atherosclerosis., National Heart, Lung, and Blood Institute (Program of Excellence in Nanotechnology Award Contract HHSN268201000045C), National Institutes of Health (U.S.) (Grant CA151884), Prostate Cancer Research Foundation (Award in Nanotherapeutics)
- Published
- 2013
246. DNA Self-Assembly of Targeted Near-Infrared-Responsive Gold Nanoparticles for Cancer Thermo-Chemotherapy
- Author
-
Massachusetts Institute of Technology. Institute for Medical Engineering & Science, MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Shi, JinJun, Pridgen, Eric Michael, Wu, Jun, Farokhzad, Omid C., Xiao, Zeyu, Ji, Changwei, Shi, Jinjun, Frieder, Jillian, Pridgen, Eric M., Massachusetts Institute of Technology. Institute for Medical Engineering & Science, MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Department of Chemical Engineering, Koch Institute for Integrative Cancer Research at MIT, Shi, JinJun, Pridgen, Eric Michael, Wu, Jun, Farokhzad, Omid C., Xiao, Zeyu, Ji, Changwei, Shi, Jinjun, Frieder, Jillian, and Pridgen, Eric M.
- Abstract
Targeted cancer therapy: Inspired by the ability of DNA hybridization, a targeted near-infrared (NIR) light-responsive delivery system has been developed through simple DNA self-assembly (see picture; PEG=polyethylene glycol). This DNA-based platform shows the ability of releasing therapeutics upon near-infrared irradiation, and remarkable targeted thermo- and chemotherapeutic efficacy in vitro and in vivo., National Institutes of Health (U.S.) (Grant CA151884), Prostate Cancer Foundation (Program in Cancer Nanotherapeutics)
- Published
- 2013
247. Engineering of self-assembled nanoparticle platform for precisely controlled combination drug therapy
- Author
-
MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Kolishetti, Nagesh, Dhar, Shanta, Valencia, Pedro Miguel, Lin, Lucy Q., Karnik, Rohit, Lippard, Stephen J., Langer, Robert, Farokhzad, Omid C., MIT-Harvard Center for Cancer Nanotechnology Excellence, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Kolishetti, Nagesh, Dhar, Shanta, Valencia, Pedro Miguel, Lin, Lucy Q., Karnik, Rohit, Lippard, Stephen J., Langer, Robert, and Farokhzad, Omid C.
- Abstract
The genomic revolution has identified therapeutic targets for a plethora of diseases, creating a need to develop robust technologies for combination drug therapy. In the present work, we describe a self-assembled polymeric nanoparticle (NP) platform to target and control precisely the codelivery of drugs with varying physicochemical properties to cancer cells. As proof of concept, we codelivered cisplatin and docetaxel (Dtxl) to prostate cancer cells with synergistic cytotoxicity. A polylactide (PLA) derivative with pendant hydroxyl groups was prepared and conjugated to a platinum(IV) [Pt(IV)] prodrug, c,t,c-[Pt(NH[subscript 3])[subscript 2](O[subscript 2]CCH[subscript 2]CH[subscript 2]COOH)(OH)Cl[subscript 2]] [PLA-Pt(IV)]. A blend of PLA-Pt(IV) functionalized polymer and carboxyl-terminated poly(d,l-lactic-co-glycolic acid)-block-poly(ethylene glycol) copolymer in the presence or absence of Dtxl, was converted, in microfluidic channels, to NPs with a diameter of ∼100 nm. This process resulted in excellent encapsulation efficiency (EE) and high loading of both hydrophilic platinum prodrug and hydrophobic Dtxl with reproducible EEs and loadings. The surface of the NPs was derivatized with the A10 aptamer, which binds to the prostate-specific membrane antigen (PSMA) on prostate cancer cells. These NPs undergo controlled release of both drugs over a period of 48–72 h. Targeted NPs were internalized by the PSMA-expressing LNCaP cells via endocytosis, and formation of cisplatin 1,2-d(GpG) intrastrand cross-links on nuclear DNA was verified. In vitro toxicities demonstrated superiority of the targeted dual-drug combination NPs over NPs with single drug or nontargeted NPs. This work reveals the potential of a single, programmable nanoparticle to blend and deliver a combination of drugs for cancer treatment., National Cancer Institute (U.S.) (Grant CA119349), National Cancer Institute (U.S.) (Grant CA034992), National Institute of Biomedical Imaging and Bioengineering (U.S.) (Grant EB003647), David H. Koch (Prostate Cancer Foundation Award in Nanotherapeutics Award), National Science Foundation (U.S.) (Graduate Research Fellowship)
- Published
- 2013
248. Synthesis of size-tunable polymeric nanoparticles enabled by 3D hydrodynamic flow focusing in single-layer microchannels
- Author
-
Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Rhee, Minsoung, Valencia, Pedro Miguel, Rodriguez, Maria I., Langer, Robert, Karnik, Rohit, Farokhzad, Omid C., Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Rhee, Minsoung, Valencia, Pedro Miguel, Rodriguez, Maria I., Langer, Robert, Karnik, Rohit, and Farokhzad, Omid C.
- Abstract
Author Manuscript date: 2011 June 27, A versatile microfluidic platform to synthesize NPs by nanoprecipitation using 3D hydrodynamic flow focusing isolates the precipitating precursors from channel walls, eliminating fouling of the channels. It is shown that this new method enables robust nanoprecipitation without polymer aggregation, regardless of the polymer molecular weight or precursor concentration implemented, where the size of the resulting polymeric NPs is tunable., David H. Koch (Prostate Cancer Foundation Award in Nanotherapeutics), National Institutes of Health (U.S.) (Grant CA119349), National Science Foundation (U.S.) (Graduate Research Fellowship)
- Published
- 2013
249. Bioinspired Multivalent DNA Network for Capture and Release of Cells
- Author
-
Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Mechanical Engineering, Zhao, Weian, Cui, Cheryl, Bose, Suman, Guo, Dagang, Shen, Chong, Farokhzad, Omid C., Teo, Grace Sock Leng, Phillips, Joseph A., Karnik, Rohit, Karp, Jeffrey Michael, Wong, Wesley P., Halvorsen, Ken, Dorfman, David M., Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Mechanical Engineering, Zhao, Weian, Cui, Cheryl, Bose, Suman, Guo, Dagang, Shen, Chong, Farokhzad, Omid C., Teo, Grace Sock Leng, Phillips, Joseph A., Karnik, Rohit, Karp, Jeffrey Michael, Wong, Wesley P., Halvorsen, Ken, and Dorfman, David M.
- Abstract
Capture and isolation of flowing cells and particulates from body fluids has enormous implications in diagnosis, monitoring, and drug testing, yet monovalent adhesion molecules used for this purpose result in inefficient cell capture and difficulty in retrieving the captured cells. Inspired by marine creatures that present long tentacles containing multiple adhesive domains to effectively capture flowing food particulates, we developed a platform approach to capture and isolate cells using a 3D DNA network comprising repeating adhesive aptamer domains that extend over tens of micrometers into the solution. The DNA network was synthesized from a microfluidic surface by rolling circle amplification where critical parameters, including DNA graft density, length, and sequence, could readily be tailored. Using an aptamer that binds to protein tyrosine kinase-7 (PTK7) that is overexpressed on many human cancer cells, we demonstrate that the 3D DNA network significantly enhances the capture efficiency of lymphoblast CCRF-CEM cells over monovalent aptamers and antibodies, yet maintains a high purity of the captured cells. When incorporated in a herringbone microfluidic device, the 3D DNA network not only possessed significantly higher capture efficiency than monovalent aptamers and antibodies, but also outperformed previously reported cell-capture microfluidic devices at high flow rates. This work suggests that 3D DNA networks may have broad implications for detection and isolation of cells and other bioparticles., National Institutes of Health (U.S.) (Grant HL097172), National Institutes of Health (U.S.) (Grant HL095722)
- Published
- 2013
250. Intracellular Mechanistic Understanding of 2D MoS2Nanosheets for Anti-Exocytosis-Enhanced Synergistic Cancer Therapy
- Author
-
Zhu, Xianbing, Ji, Xiaoyuan, Kong, Na, Chen, Yunhan, Mahmoudi, Morteza, Xu, Xiaoding, Ding, Li, Tao, Wei, Cai, Ting, Li, Yujing, Gan, Tian, Barrett, Austin, Bharwani, Zameer, Chen, Hongbo, and Farokhzad, Omid C.
- Abstract
Emerging two-dimensional (2D) nanomaterials, such as transition-metal dichalcogenide (TMD) nanosheets (NSs), have shown tremendous potential for use in a wide variety of fields including cancer nanomedicine. The interaction of nanomaterials with biosystems is of critical importance for their safe and efficient application. However, a cellular-level understanding of the nano-bio interactions of these emerging 2D nanomaterials (i.e., intracellular mechanisms) remains elusive. Here we chose molybdenum disulfide (MoS2) NSs as representative 2D nanomaterials to gain a better understanding of their intracellular mechanisms of action in cancer cells, which play a significant role in both their fate and efficacy. MoS2NSs were found to be internalized through three pathways: clathrin → early endosomes → lysosomes, caveolae → early endosomes → lysosomes, and macropinocytosis → late endosomes → lysosomes. We also observed autophagy-mediated accumulation in the lysosomes and exocytosis-induced efflux of MoS2NSs. Based on these findings, we developed a strategy to achieve effective and synergistic in vivocancer therapy with MoS2NSs loaded with low doses of drug through inhibiting exocytosis pathway-induced loss. To the best of our knowledge, this is the first systematic experimental report on the nano-bio interaction of 2D nanomaterials in cells and their application for anti-exocytosis-enhanced synergistic cancer therapy.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.